Cytogenetics and Molecular Cytogenetics - Medical Genomics and Proteomics - Thomas Liehr - CRC Press (2022)
Cytogenetics and Molecular Cytogenetics - Medical Genomics and Proteomics - Thomas Liehr - CRC Press (2022)
Molecular Cytogenetics
Genomic technologies provide the means of diagnosis and management of many
human diseases. Without insights from cytogenetics, correct interpretation of
modern high-throughput results is diffcult, if not impossible. This book summarizes
applications of cytogenetics and molecular cytogenetics for students, clinicians and
researchers in genetics, genomics and diagnostics. The book combines the state-of-
the-art knowledge and practical expertise from leading researchers and clinicians
and provides a comprehensive overview of current medical and research applications
of many of these technologies.
KEY FEATURES
• Provides clear summaries of fuorescence in situ hybridization technologies
and others
• Comprehensively covers established and emerging methods
• Chapters from an international team of leading researchers
• Useful for students, researchers and clinicians
Medical Genomics and Proteomics
Series Editors
Scott O. Rogers
Science publisher
Charles R. Crumly—CRC Press/Taylor & Francis Group
Published Titles
Molecular Analyses
Edited by Scott O. Rogers
Edited by
Thomas Liehr
Legend for fgure in titlepage:
Central white Figure part consists (from left to right) of: Two schemes showing multicolor-banding
and chromosomal translocations were provided by Leon Liehr (Jena, Germany; https://leonliehr.
jimdofree.com/), the partial M-FISH karyogramm, the interphase nucleus and the chromosomal
microarray fgures are derived from chapters 14 (part of 1), 6 (part of 1D) and 21 (part of 2A),
respectively.
First Edition published 2023
by CRC Press
6000 Broken Sound Parkway NW, Suite 300, Boca Raton, FL 33487–2742
and by CRC Press
4 Park Square, Milton Park, Abingdon, Oxon, OX14 4RN
CRC Press is an imprint of Taylor & Francis Group, LLC
© 2023 selection and editorial matter, Thomas Liehr; individual chapters, the contributors
Reasonable efforts have been made to publish reliable data and information, but the author and
publisher cannot assume responsibility for the validity of all materials or the consequences of
their use. The authors and publishers have attempted to trace the copyright holders of all material
reproduced in this publication and apologize to copyright holders if permission to publish in this
form has not been obtained. If any copyright material has not been acknowledged please write and
let us know so we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced,
transmitted, or utilized in any form by any electronic, mechanical, or other means, now known or
hereafter invented, including photocopying, microflming, and recording, or in any information
storage or retrieval system, without written permission from the publishers.
For permission to photocopy or use material electronically from this work, access www.
copyright.com or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive,
Danvers, MA 01923, 978–750–8400. For works that are not available on CCC please contact
[email protected]
Trademark notice: Product or corporate names may be trademarks or registered trademarks and are
used only for identifcation and explanation without intent to infringe.
Library of Congress Cataloging‑in‑Publication Data
Names: Liehr, Thomas, 1965-editor.
Title: Cytogenetics and molecular cytogenetics / edited by Thomas Liehr.
Description: First edition. | Boca Raton : CRC Press, 2023. | Series: Medical genomics and
proteomics | Includes bibliographical references and index.
Identifers: LCCN 2022026812 (print) | LCCN 2022026813 (ebook) | ISBN 9781032121628
(hardback) | ISBN 9781032122212 (paperback) | ISBN 9781003223658 (ebook)
Subjects: LCSH: Cytogenetics. | Molecular genetics.
Classifcation: LCC QH441.5 .C98 2023 (print) | LCC QH441.5 (ebook) | DDC 572.8—dc23/
eng/20220713
LC record available at https://lccn.loc.gov/2022026812
LC ebook record available at https://lccn.loc.gov/2022026813
ISBN: 978-1-032-12162-8 (hbk)
ISBN: 978-1-032-12221-2 (pbk)
ISBN: 978-1-003-22365-8 (ebk)
DOI: 10.1201/9781003223658
Typeset in Times
by Apex CoVantage, LLC
To my two teachers in Human Genetics and Chromosomics:
Prof. Erich Gebhart (Erlangen, Germany), who showed
to me the importance of publishing results, and
Prof. Uwe Claussen (Jena, Germany), who was
a role model for unconventional thinking.
Contents
Series Preface............................................................................................................xi
Scott O. Rogers
Preface.................................................................................................................... xiii
Acknowledgements .................................................................................................. xv
Editor .....................................................................................................................xvii
List of Contributors.................................................................................................xix
vii
viii Contents
Index...................................................................................................................... 357
Series Preface
In 2005, the Encyclopedia of Medical Genomics and Proteomics was published.
It consisted of two large volumes of analytical methods and equipment that were
applicable to medical and other studies of genomics and proteomics. While it was
a comprehensive collection at the time, many variations, improvements, and new
technological developments have occurred during the past two decades. Also, the
alphabetical arrangements of the chapters (by title) made searching by topic some-
what cumbersome. Finally, the Encyclopedia was originally available in print ver-
sion only. All of these factors necessitated an extensive revision in content, format,
and organization. The new collection is entitled Series in Medical Genomics and
Proteomics. The revised version will be offered as volumes, organized by topic,
and will be published in print and electronic formats. Currently, at least one dozen
volumes are planned.
This is the second volume in the Series, entitled Cytogenetics and Molecular
Cytogenetics. Because of their importance in several felds, the methods described
in this volume have been developed and used increasingly over the past several
decades to study chromosomal loci, genetic alterations associated with diseases,
chromosomal translocations, congenital abnormalities, tissue-specifc changes,
development, gene expression, molecular taxonomy, evolution, epigenetic changes
associated with diseases (including cancer), and others. The chapters included in this
volume provide a detailed comprehensive collection of descriptions of cytogenetic
and molecular cytogenetic methods, with special emphasis on FISH (fuorescence in
situ hybridization) procedures and applications. This includes an overview of FISH
methods, as well as descriptions of techniques used to identify specifc components
of cells (e.g., centromeres, portions of synaptonemal complexes), disease diagnoses
(e.g., leukemia, chromosome rearrangements), and protocols to characterize cellular
components in various organisms (e.g., birds, fshes, plants). Additionally, chapters
are included on basic cytogenetics, chromosome banding techniques, molecular
karyotyping, and microdissection. This volume is poised to be a valuable resource
for those in cell, molecular, and medical sciences.
Scott O. Rogers
Medical Genomics and Proteomics Book Series Editor
xi
Preface
With this book, a contribution to the highly actual feld of cytogenetics, cytogenom-
ics and chromosomics is provided. The book presents applications and protocols
in clinical and research applications. It is not restricted to human genetics, but also
includes animal and plant cytogenetics and provides some insights in actual develop-
ments. This book is divided in following sections:
xiii
Acknowledgements
The Editor thanks to all authors investing time and work to provide chapters to this
book on cytogenetics and molecular cytogenetics. Only together could we provide
this important contribution to the feld of cytogenomics and chromosomics.
xv
Editor
Thomas Liehr has been working in human genetics since 1991. He is a biologist
by education and head of the molecular cytogenetic group in Jena, Germany.
Research felds include clinical genetics, leukemia cytogenetics, interphase structure
of human chromosomes, and research on chromosomal evolution. The results of
his research are published in >10 books, ~100 book chapters, >800 referred papers,
and ~1000 abstracts. His particular expertise includes small supernumerary marker
chromosomes (sSMC), chromosomal heteromorphisms and uniparental disomy—
see ChromosOmics databases (http://cs-tl.de/DB.html). He is active member of the
European Board of Medical Genetics and received multiple prizes, two invited pro-
fessorships and a Dr.h.c. as summarized at http://cs-tl.de/.
xvii
Contributors
Rouben Aroutiounian Gerson Ferreira
Yerevan State University Instituto Nacional de Câncer José
Yerevan, Armenia de Alencar Gomes da Silva
(INCA-RJ)
Sarah Breitenbach Rio de Janeiro, Brazil
Technische Universität Dresden
Dresden, Germany Susana Isabel Ferreira
Faculty of Medicine of the University
Isabel Marques Carreira of Coimbra
Faculty of Medicine of the University Coimbra, Portugal
of Coimbra
Coimbra, Portugal Amanda Figueiredo
Federal University of Rio de Janeiro
Hongyan Chai Rio de Janeiro, Brazil
Yale School of Medicine
New Haven, CT, USA Jelena Filipović Tričković
University of Belgrade
Chenghua Cui Belgrade, Serbia
Chinese Academy of Medical Sciences
& Peking Union Medical College Vladimir E. Gokhman
Tianjin, China Moscow State University
Moscow, Russia
Marcelo de Bello Cioff
Universidade Federal de São Carlos Tigran Harutyunyan
São Carlos, SP, Brazil Yerevan State University
Yerevan, Armenia
Edivaldo Herculano Correa de Oliveira
Universidade Federal do Pará Tony Heitkam
Belém, Brazil Technische Universität Dresden
Dresden, Germany
Ivanete de Oliveira Furo
Universidade Federal Rural da Galina Hovhannisyan
Amazônia (UFRA) Yerevan State University
Parauapebas, Brazil Yerevan, Armenia
xix
xx Contributors
Mariana Val
Faculty of Medicine of the University of
Coimbra
Coimbra, Portugal
1 Cytogenetics and
Chromosomics
Thomas Liehr
CONTENTS
Introduction................................................................................................................ 1
Chromosomics and Cytogenomics ............................................................................ 1
Application Fields of (Molecular) Cytogenetics........................................................ 2
Classical/Solid Staining .................................................................................... 3
C-Banding and NOR Silver Staining................................................................ 3
Banding Cytogenetics....................................................................................... 3
Fluorescence In Situ Hybridization (FISH)...................................................... 3
Primed In Situ Hybridization (PRINS)............................................................. 4
Comparative Genomic Hybridization (CGH)................................................... 4
Molecular Combing .......................................................................................... 4
Conclusions................................................................................................................ 4
References.................................................................................................................. 4
INTRODUCTION
This chapter is a general introduction to the book Cytogenetics and Molecular
Cytogenetics published in this new “Medical Genomics and Proteomics” book series.
The specifc topic of this book on (molecular) cytogenetics is embedded in the feld
of chromosomics, which can be only realized based on cytogenomic approaches.[1, 2]
DOI: 10.1201/9781003223658-1 1
2 Cytogenetics and Molecular Cytogenetics
TABLE 1.1
Cytogenomic Approaches Adapted acc. to[4] Are Listed Here
Cytogenomic Field Cytogenomic Technique
Cytogenetics – Classical/solid staining
– C-banding
– NOR silver staining
– Banding cytogenetics
Molecular Cytogenetics – Fluorescence in situ hybridization
– Primed in situ hybridization
– Comparative genomic hybridization (CGH)
– Molecular combing
Molecular Genetics – Restriction fragment polymorphism analyses
(classical approaches) – DNA-cloning in vectors
– Blotting approaches like Southern blotting
– DNA-fngerprinting
– PCR approaches incl. MLPA
– Sanger sequencing
Molecular Genetics – Array-CGH/chromosomal microarray analyses (CMA)
(modern approaches) – Second generation sequencing
– Third generation sequencing
– Gene editing (CRISP/CAS9)
Others – Electron microscopy–based approaches
– Laser scanning–based approaches
– Optical mapping approaches
– Uniparental disomy/Epigenetic changes oriented approaches—incl.
studies on long non-coding RNAs, etc.
– Bioinformatics
CLASSICAL/SOLID STAINING
Classical solid Giemsa or Orcein staining—also called classical cytogenetics[8]—is
the basic approach used to determine constitutional chromosome numbers in spe-
cies previously not studied by cytogenetics.[9] Also, there are species in which chro-
mosome banding is not applicable[10, 11]; here classical cytogenetics is necessary in
research.
Solid chromosome stains are also applied in research settings of radiation or
mutagenesis—and here also diagnostic applications are reported to determine num-
ber of chromosomal breaks per metaphases after irradiation and/or exposure to a
mutagen.[12]
BANDING CYTOGENETICS
Banding cytogenetics was introduced by Lore Zech in the 1970s.[15] Nowadays most
countries apply GTG-banding = G-bands by trypsin using Giemsa for routine band-
ing cytogenetics in prenatal, postnatal and tumor cytogenetic diagnostics. Besides,
banding cytogenetics is used in animal cytogenetics, in case the corresponding chro-
mosomes allow for introduction of a protein-based banding pattern.[16]
MOLECULAR COMBING
So-called fber-FISH is a several decades–old variant of FISH, where DNA-probes
are hybridized instead of on interphases or metaphases on to extended DNA-fbers,
providing thus a higher in situ resolution. Recently, fber-FISH on extremely stretched
DNA-fbers became available as a standardized protocol, and is commercially avail-
able as “molecular combing”. This enables applications in research and in diagnos-
tics for studying repetitive DNA-stretches yet not reliably accessible by any other
cytogenomic approach.[22]
CONCLUSIONS
Chromosomic research and diagnostics, in general are enabled and in parts moti-
vated by new technical developments.[4] New cytogenomic approaches are always
welcome; however, due to specifc advantages of each technique, older ones should
not be considered too hastily as outdated.[7] As shown exemplarily here and with
no claim to completeness, cytogenetics and molecular cytogenetics are still, and
also will be in future, the most relevant participants in the concert of actual cytoge-
nomic approaches being necessary to get as comprehensive as possible chromosomic
insights.[1, 2]
REFERENCES
1. Liehr, T. Molecular cytogenetics in the era of chromosomics and cytogenomic
approaches. Front. Genet. 2021, 12, 720507.
2. Liehr, T. From human cytogenetics to human chromosomics. Int J Mol Sci. 2019, 20,
E826.
3. Claussen, U. Chromosomics. Cytogenet. Genome Res. 2005, 111, 101–106.
4. Liehr, T. A Defnition for cytogenomics: Also may be called chromosomics. In:
Cytogenomics; Liehr, T., Ed. Academic Press, London, 2021, pp. 1–7.
Cytogenetics and Chromosomics 5
5. Reichwald, K.; Petzold, A.; Koch, P.; Downie, B.R.; Hartmann, N.; Pietsch, S.; Baumgart,
M.; Chalopin, D.; Felder, M.; Bens, M.; Sahm, A.; Szafranski, K.; Taudien, S.; Groth, M.;
Arisi, I.; Weise, A.; Bhatt, S.S.; Sharma, V.; Kraus, J.M.; Schmid, F.; Priebe, S.; Liehr,
T.; Görlach, M.; Than, M.E.; Hiller, M.; Kestler, H.A.; Volff, J.N.; Schartl, M.; Cellerino,
A.; Englert, C.; Platzer, M. Insights into sex chromosome evolution and aging from the
genome of a short-lived fsh. Cell. 2015, 163, 1527–1538.
6. Yano, C.F.; Sember, A.; Kretschmer, R.; Bertollo, L.A.C.; Ezaz, T.; Hatanaka, T.; Liehr,
T.; Ráb, P.; Al-Rikabi, A.; Ferreira Viana, P.; Feldberg, E.; de Oliveira, E.A.; Toma,
G.A.; Cioff, M.d.B. Against the mainstream: Exceptional evolutionary stability of l ZW
sex chromosomes across fsh families Triportheidae and Gasteropelecidae (Teleostei:
Characiformes). Chromosome Res. 2021, 29, 391–416.
7. Liehr, T.; Mrasek, K.; Klein, E.; Weise, A. Modern high throughput approaches are not
meant to replace ‘old fashioned’ but robust techniques. J. Genet. Genomes. 2017, 1, e101.
8. Liehr, T. “Classical cytogenetics” is not equal to “banding cytogenetics”. Mol Cytogenet.
2017, 10, 3.
9. Chaiyasan, P.; Mingkwan, B.; Jantarat, S.; Suwannapoom, C.; Cioff, M.d.B.; Liehr, T.;
Talumphai, S.; Tanomtong, A.; Supiwong, W. Classical and molecular cytogenetics of
Belontia hasselti (Perciformes: Osphronemidae): Insights into the ZZ/ZW sex chromo-
some system. Biodiversitas. 2021, 22, 546–554.
10. D’Amato, G.; Bianchi, G.; Capineri, R.; Marchi, P. N-band staining in plant chromo-
somes with a HCl-Giemsa technique. Caryologia. 1979, 32, 455–459.
11. Martínez-Lage, A.; González-Tizón, A.; Méndez, J. Characterization of different chro-
matin types in Mytilus galloprovincialis L. after C-banding, fuorochrome and restric-
tion endonuclease treatments. Heredity. 1994, 72, 242–249.
12. Natarajan, A.T. Chromosome aberrations: Past, present and future. Mutat. Res. 2002,
504, 3–16.
13. Weise, A.; Liehr, T. Cytogenetics. In: Cytogenomics; Liehr, T., Ed. Academic Press,
London, 2021, pp. 25–34.
14. Liehr, T. Cases with heteromorphisms. http://cs-tl.de/DB/CA/HCM/0-Start.html
[accessed on 01/12/2022].
15. Schlegelberger, B. In memoriam: Prof. Dr. rer. nat. Dr. med. h.c. Lore Zech; 24.9.1923–
13.3.2013: Honorary member of the European Society of Human Genetics, Honorary
member of the German Society of Human Genetics, Doctor laureate, the University of
Kiel, Germany. Mol. Cytogenet. 2013, 6, 20.
16. Claussen, U.; Michel, S.; Mühlig, P.; Westermann, M.; Grummt, U.W.; Kromeyer-
Hauschild, K.; Liehr, T. Demystifying chromosome preparation and the implications for
the concept of chromosome condensation during mitosis. Cytogenet. Genome Res. 2002,
98, 136–146.
17. Nurk, S.; Koren, S.; Rhie, A.; Rautiainen, M.; Bzikadze, A.V.; Mikheenko, A.; Vollger,
M.R.; Altemose, N.; Uralsky, L.; Gershman, A.; Aganezov, S.; Hoyt, S.J.; Diekhans, M.;
Logsdon, G.A.; Alonge, M.; Antonarakis, S.E.; Borchers, M.; Bouffard, G.G.; Brooks,
S.Y.; Caldas, G.V.; Cheng, H.; Chin, C.-S.; Chow, W.; de Lima, L.G.; Dishuck, P.C.;
Durbin, R.; Dvorkina, T.; Fiddes, I.T.; Formenti, G.; Fulton, R.S.; Fungtammasan, A.;
Garrison, E.; Grady, P.G.S.; Graves-Lindsay, T.-A.; Hall, I.M.; Hansen, N.F.; Hartley,
G.A.; Haukness, M.; Howe, K.; Hunkapiller, M.W.; Jain, C.; Jain, M.; Jarvis, E.D.;
Kerpedjiev, P.; Kirsche, M.; Kolmogorov, M.; Korlach, J.; Kremitzki, M.; Li, H.; Maduro,
V.V.; Marschall, T.; McCartney, A.M.; McDaniel, J.; Miller, D.E.; Mullikin, J.C.; Myers,
E.W.; Olson, N.D.; Paten, B.; Peluso, P.; Pevzner, P.A.; Porubsky, D.; Potapova, T.; Rogaev,
E.I.; Rosenfeld, J.A.; Salzberg, S.L.; Schneider, V.A.; Sedlazeck, F.J.; Shafn, K.; Shew,
C.J.; Shumate, A.; Sims, Y.; Smit, A.F.A.; Soto, D.C.; Sović, I.; Storer, J.M.; Streets, A.;
6 Cytogenetics and Molecular Cytogenetics
Sullivan, B.A.; Thibaud-Nissen, F.; Torrance, J.; Wagner, J.; Walenz, P.P.; Wenger, A.;
Wood, J.M.D.; Xiao, C.; Yan, S.M.; Young, A.C.; Zarate, S.; Surti, U.; McCoy, R.C.;
Dennis, M.Y.; Alexandrov, I.A.; Gerton, J.L.; O’Neill, R.J.; Timp, W.; Zook, J.M.;
Schatz, M.C.; Eichler, E.E.; Miga, K.H.; Phillippy, A.M.The complete sequence of a
human genome. bioRxiv. 2021. preprint doi: https://doi.org/10.1101/2021.05.26.445798.
18. Liehr, T. Basics and literature on multicolor fuorescence in situ hybridization applica-
tion. http://cs-tl.de/DB/TC/mFISH/0-Start.html [accessed on 01/12/2022].
19. Pellestor, F. Development and adaptation of the PRINS technology: An overview.
Methods Mol. Biol. 2006, 334, 211–220.
20. Kallioniemi, A.; Kallioniemi, O.P.; Sudar, D.; Rutovitz, D.; Gray, J.W.; Waldman, F.;
Pinkel, D. Comparative genomic hybridization for molecular cytogenetic analysis of
solid tumors. Science. 1992, 258, 818–821.
21. Weise, A.; Liehr, T. Molecular karyotyping. In: Cytogenomics; Liehr, T., Ed. Academic
Press, London, 2021, pp. 72–85.
22. Bisht, P.; Avarello, M.D.M. Molecular combing solutions to characterize replication
genetics and genome rearrangements. In: Cytogenomics; Liehr, T., Ed. Academic Press,
London, 2021, pp. 47–72.
2 Banding Cytogenetics
Hongyan Chai and Peining Li
CONTENTS
Introduction................................................................................................................ 7
Chromosome Structure and Various Banding Techniques................................ 8
Human Chromosome Structure in Interphases and Metaphases .......... 8
Various Banding Techniques ................................................................ 9
Molecular Basis of Banding in Human Chromosomes ...................... 10
G-Banding in Normal Population................................................................... 12
International System for Human Cytogenetic/Cytogenomic
Nomenclature (ISCN).......................................................... 12
Chromosome Heteromorphisms in Humans....................................... 13
Inducible Fragile Sites in Human Chromosomes ............................... 15
Clinical Application of Banding Cytogenetics ............................................... 16
Detection of Constitutional Chromosome Abnormalities................... 16
Detection of Somatic Chromosome Abnormalities ............................ 18
Report and Interpretation of Diagnostic Karyotype ........................... 20
Conclusions.............................................................................................................. 20
Web Resources......................................................................................................... 21
Acknowledgement ................................................................................................... 21
References................................................................................................................ 21
INTRODUCTION
Cytogenetics is an important feld of genetics focusing on the study of chromosome
structure and related functions in heredity and evolution. In 1902, W. Sutton and
T. Boveri independently developed the chromosome theory of inheritance that identi-
fes chromosomes as the carrier of genetic material.[1, 2] Earlier studies of human chro-
mosomes struggled with the correct number of chromosomes.[3] In 1923, T.S. Painter
analyzed chromosomes in human spermatogonial cells at mitotic prophase and made
three important fndings: i) the size and shape of the chromosomes were described,
and the diploid chromosome number was determined to be in the range of 46–48 with
an emphasis on 48; ii) human sex was found to be determined by an X/Y heteromor-
phic chromosomal pair mechanism; and iii) Caucasian and African-American males
had indistinguishable chromosome sets.[4, 5] In 1952, T.C. Hsu introduced a hypotonic
shock method to improve the quality of chromosome preparation from in vitro cul-
tured cells.[6] In 1956, J.H. Tjio and A. Levan used this improved technique to rec-
ognize the correct number of 46 chromosomes in a human genome.[7] This certainty
of chromosome number in normal individuals facilitated cytogenetic investigations
DOI: 10.1201/9781003223658-2 7
8 Cytogenetics and Molecular Cytogenetics
pair of sex chromosomes was named XY for male and XX for female. Each chromo-
some has a primary constriction designated as centromere. Based on the position
of the centromeres, these numbered autosomes were further divided into: group A
of metacentric chromosomes 1, 2, and 3, group B of submetacentric chromosomes
4 and 5, group C of submetacentric chromosomes 6 to 12, group D of acrocentric
chromosomes 13, 14, and 15, group E of submetacentric chromosomes 16, 17, and 18,
group F of metacentric chromosomes 19 and 20, and group G of acrocentric chromo-
somes 21 and 22. Chromosomes were ordered according to their size, and fnally it
was determined that indeed chromosome 21 is the smallest human chromosome and
not 22; still the designations were not changed. The constricted or narrow regions
outside the centromere in some chromosomes are termed secondary constrictions.
This original solid stained karyotype has been the reference for detecting numeri-
cal chromosomal abnormalities in patients, but the diffculty in distinguishing indi-
vidual chromosomes within a group severely affects its accuracy.
laboratories and has been the most widely used banding method in chromosome
analysis. When incubating chromosome preparations in phosphate buffer at a high
temperature followed by Giemsa staining, an R-band pattern with dark and light
staining reverse of the G-band pattern was noted. Extending this treatment to a lon-
ger time or a higher temperature could erase most of the R bands but retain terminal
bands, so called T-banding at the end of each chromosome. Further improvement
in the banding technique involved the synchronization of cell cycle to capture late
prophase and prometaphase and the integration of DNA intercalating agents such
as ethidium bromide to elongate chromosomes for a high-resolution G-banding
pattern.[25, 26] Standardized protocols for these traditional banding of chromosomes
for cytogenetic analysis have been published.[27–29] A three-letter code with the frst
letter denoting the type of banding, the second letter for method of chromosome
treatment, and the third letter for the staining dye is used to describe different
banding techniques.[13] For example, GTG refers to G-Banding by Trypsin treat-
ment and Giemsa staining, and CBG refers to C-banding by Barium hydroxide
treatment and Giemsa staining. All these banding patterns refect the organization
of heterochromatin and euchromatin in human chromosomes and thus have differ-
ent applications.
TABLE 2.1
Various Banding Techniques and Related Staining Patterns and Genomic
Sequences
Banding Staining Pattern Genomic Sequences Replication
Technique
Q-banding Brighter Q band AT-rich
G-banding G-band positive (dark stain) AT-rich, Alu poor, LINE rich, late
gene poor
G-band negative (light stain) GC-rich, Alu rich, LINE poor, early
gene rich
R-banding R-band positive (dark stain) (similar to G-band negative)
R-band negative (light stain) (similar to G-band positive)
C-banding C-band positive (dark stain) centromeric satellite sequences and
Yq12
Silver staining Positive on short arm of proteins on 5S/18S/28S ribosomal
D/G group chromosomes DNA at active NORs
T-banding Positive on both ends of subtelomeric and telomeric repeats
each chromosome
the various banding techniques and their related staining patterns and genomic
sequence organizations.
Studies from genome sequencing further defned the organization of heterochro-
matin and euchromatin in human chromosomes.[34, 35] The primary constriction or
centromere in each chromosome contains heterochromatin consisting largely of
high-order arrays of α-satellite DNA and other satellite repeats (beta, gamma, I,
II, and III). The α-satellite DNA monomers, consisting of an approximately 171 bp
repeat unit with 40% sequence divergence in different chromosomes, are tandemly
organized into distinct high-order repeat units at the centromere. A 9 bp degenerate
motif in the α-satellite serves as the binding site for centromere protein B. There are
centromeric transition regions with euchromatic gene-containing segments dupli-
catively transposed toward the pericentric regions.[35] The secondary constrictions
in the fve acrocentric chromosome arms 13p, 14p, 15p, 21p, and 22p encode the
5S, 18S, and 28S ribosomal DNA genes, which lie on a 43-kb sequence present in
approximately 50 tandem copies on each arm and are fanked by additional repeats
arranged in a complex structure.[34] Silver staining reveals the protein components
associated with these ribosomal gene transcriptions. Finally, there is a single large
region on distal Yq composed primarily of thousands of copies of several repeat
families. The heterochromatic regions all tend to be highly polymorphic in the
human population. The euchromatin of the human genome are bounded proximally
by heterochromatin and distally by a telomere consisting of several kilobases of the
hexamer repeat TTAGGG.[34]
12 Cytogenetics and Molecular Cytogenetics
in chromosomes 1, 9, 16, 18, and Y and estimated the relative frequency in 3,476
Caucasian and 807 African-American infants.[39] Further studies of chromosome
variations in Q-band and C-band recognized increase (+) or decrease (-) in length of
heterochromatin in the long arm (q) of chromosomes 1, 9, 16, and Y, and in the short
arm (p) or centromere (cen) of chromosomes of 13, 14, 15, 21, and 22 and X.[40] The
molecular basis of these variations related to the large arrays of tandemly repeated
DNA sequences.
Another type of human chromosome heteromorphism is pericentric inversions
likely resulting from the shuffing of satellite DNA in the heterochromatic regions.
An early study on heterochromatin size and pericentric inversion by C-banding on
80 normal Caucasians noted that the frequency of size heteromorphisms for chro-
mosomes 1, 9, and 16 were 11.3%, 47.5%, and 7.5%, respectively, and pericentric
inversions of chromosomes 1 and 9 were 20% and 23%, respectively. These was
no signifcant difference between males and females for size and position of these
heteromorphisms.[41] A more recent large case series of 1,276 individuals from India
detected pericentric inversion of chromosome 9, inv(9)(p11q13), and Robertsonian
translocation of chromosomes 13 and 14, rob(13;14)(q10;q10), in 3.68% and 1.25%
of individuals, respectively; while other chromosomal heteromorphisms of chromo-
somes 1, 9, 13, 14, 15, 21, 22, and Y were seen in 0.15%–1.95% of individuals.[42] A
molecular cytogenetic study of 334 carriers of heterochromatic variants of chromo-
some 9 recognized the presence of different type of variants and absence of evidence
linking these variants to infertility.[43] The pericentromeric region of chromosome 9
is the most obvious heteromorphism in human chromosomes; the proximal 5-Mb on
9p11 to distal 4-Mb on 9q12 comprise a mere 0.3% of the genome. These two regions
have a high density of segmental duplications and a high degree of intrachromosomal
sequence identity (98.7%). This high sequence similarity between the two regions is
likely to be the reason for a polymorphic inv(9) present in approximately 1% of
the human population.[34] A study of 2,970 prenatal cases in China showed an inci-
dence of approximately 8.8% for chromosomal heteromorphisms. The most frequent
was found to be in chromosome Yqh (5.28% males), followed by chromosome 1qh
(1.65%), inv(9) (0.94%), 22p/cen (0.77%), 15p/cen (0.64 %), 9qh (0.58%), and 16qh
(0.34%). The frequency of common chromosomal heteromorphisms and pericentric
inversions from three large case series of different populations are reviewed and
summarized in Table 2.2. The earlier New Haven study showed a higher frequency
of heteromorphisms likely due to the solid staining and related size estimation for
variant patterns.[39] Recent studies showed comparable frequencies of heteromor-
phisms between Indian and Chinese populations; furthermore, there was no evi-
dence for an association between chromosome heteromorphisms and infertility or
recurrent spontaneous abortions.[42, 44]
Chromosome heteromorphisms are inheritable traits within a family, so it is a
useful biomarker to track parental origin of chromosomal abnormalities such as the
maternal or paternal origin of trisomy 21, derivative chromosomes, and uniparental
disomy of chromosome 15.[45–47] The reporting practice on chromosome heteromor-
phisms varies among clinical cytogenetics laboratories. Survey results from more
than 200 cytogeneticists indicated that 61% of them would include selected hetero-
morphism data in a clinical report. More than 90% considered prominent short arms,
Banding Cytogenetics 15
TABLE 2.2
The Estimated Frequency of Common Chromosomal Heteromorphisms from
Different Populations
Caucasian African-American[39] Indians[42] Chinese[44]
Infants[39] (n=807, 411 males) (n=1276, (n=2970,
(n=3476, 638 males) 1533 males)
1741 males)
qh: long arm heterochromatin, cenh: centromeric heterochromatin, ps: short arm satellite, pstk: short arm
stalk, inv: inversion
* frequency given by D or G groups
chromosomes such as a giant ring or marker chromosome have been seen in lipo-
sarcoma.[75] A review of profciency test results from over 200 cytogenetics labo-
ratories for a 20-year period showed that more than 96% of laboratories provided
correct interpretation of chromosomal abnormalities of hematological neoplasms.[76]
Online databases such as the ‘Atlas of Genetics and Cytogenetics in Oncology and
20 Cytogenetics and Molecular Cytogenetics
CONCLUSIONS
Banding cytogenetics has provided structural basis for the understanding of
sequence organization in human chromosomes and cytological landmarks for draft-
ing the human genome. Technical standards and guidelines of G-band analysis have
Banding Cytogenetics 21
been developed and implemented for clinical cytogenetics. Current ISCN 2020 pro-
vides general principles applicable to multiple molecular and genomic techniques
and standardized documentation of chromosomal abnormalities, pathogenic copy
number variants, and gene rearrangements. Banding cytogenetics has been and con-
tinues to be an effective and highly profcient cell-based approach to detect constitu-
tional numerical and structural chromosomal abnormalities in prenatal and pediatric
patients and somatic clonal chromosomal abnormalities in various types of cancers.
WEB RESOURCES
Atlas of Genetics and Cytogenetics in Oncology and Haematology (http://
atlasgeneticsoncology.org/)
Cases with heteromorphisms (http://cs-tl.de/DB/CA/HCM/0-Start.html)
ClinVar (www.ncbi.nlm.nih.gov/clinvar/)
ClinGen (https://clinicalgenome.org/)
DECIPHER (www.deciphergenomics.org/)
Human Genome Browser (https://genome.ucsc.edu/)
ACKNOWLEDGEMENT
Special thanks to Autumn DiAdamo and Jiadi Wen of Yale Clinical Cytogenetics
Laboratory for helpful discussion and careful editing on the content of this chapter.
REFERENCES
1. Boveri, O.G.M. Über Mitosen bei einseitiger Chromosomenbindung. Jenaische
Zeitschrift für Naturwissenschaft. 1903, 37, 401–443.
2. Sutton, S.W. The chromosomes in heredity. Biol. Bull. 1903, 4, 231–251.
3. Gartler, S.M. The chromosome number in humans: A brief history. Nat. Rev. Genet.
2006, 7, 655–660.
4. Painter, S.T. Studies in mammalian spermatogenesis. II: The spermatogenesis of man.
J. Exp. Zool. 1923, 37, 291–338.
5. Ruddle, F.H. Theophilus painter: First steps toward an understanding of the human
genome. J. Exp. Zool. A. Comp. Exp. Biol. 2004, 301, 375–377.
6. Hsu, T.C. Mammalian chromosomes in vitro, I. Karyotype of man. J. Heredity. 1952, 43,
167–172.
7. Tjio, J.H.; Levan, A. The chromosome number of man. Hereditas. 1956, 42, U1–6.
8. Ford, C.E.; Jones, K.W.; Polani, P.E.; De Almeida, J.C.; Briggs, J.H. A sex-chromosome
anomaly in a case of gonadal dysgenesis (Turner’s syndrome). Lancet. 1959, 1, 711–713.
9. Jacobs, P.A.; Strong, J.A. Case of human intersexuality having a possible XXY sex-
determining mechanism. Nature. 1959, 183, 302–303.
10. Lejeune, J.; Turpin, R.; Gautier, M. Chromosomic diagnosis of mongolism. Arch. Fr.
Pediatr. 1959, 16, 962–963.
11. Nowell, P.C.; Hungerford, D.A. A minute chromosome in human chronic granulocytic
leukemia. Science. 1960, 132, 1497.
12. Priest, J.H. Medical Cytogenetics and Cell Culture. Henry Kimpton Publisher, London;
1977.
22 Cytogenetics and Molecular Cytogenetics
13. McGowan-Jordan, J.; Hastings, R.J.; Moore, S. ISCN 2020: An International System for
Human Cytogenomic Nomenclature. Karger, Basel; 2020.
14. Maeshima, K.; Iida, S.; Tamura, S. Physical nature of chromatin in the nucleus. Cold
Spring Harb. Perspect. Biol. 2021, 13, a040675.
15. Brown, S.W. Heterochromatin. Science. 1966, 151, 417–425.
16. Barr, M.L. Dysgenesis of the seminiferous tubules. Br. J. Urol. 1957, 29, 251–257.
17. Arrighi, F.E.; Hsu, T.C. Localization of heterochromatin in human chromosomes.
Cytogenetics. 1971, 10, 81–86.
18. Barr, M.L. Sex chromatin and phenotype in man. Science. 1959, 130, 679–685.
19. Pardue, M.L.; Gall, J.G. Chromosomal localization of mouse satellite DNA. Science.
1970, 168, 1356–1358.
20. Corneo, G.; Ginelli, E.; Polli, E. Repeated sequences in human DNA. J. Mol. Biol. 1970,
48, 319–327.
21. Ginelli, E.; Corneo, G. The organization of repeated DNA sequences in the human
genome. Chromosoma. 1976, 56, 55–68.
22. Tantravahi, R.; Miller, D.A.; Dev, V.G.; Miller, O.J. Detection of nucleolus orga-
nizer regions in chromosomes of human, chimpanzee, gorilla, orangutan and gibbon.
Chromosoma. 1976, 56, 15–27.
23. Caspersson, T.; Farber, S.; Foley, G.E.; Kudynowski, J.; Modest, E.J.; Simonsson, E.;
Wagh, U.; Zech, L. Chemical differentiation along metaphase chromosomes. Exp. Cell.
Res. 1968, 49, 219–222.
24. Seabright, M. A rapid banding technique for human chromosomes. Lancet. 1971, 2,
971–972.
25. Hoo, J.J.; Jamro, H.; Schmutz, S.; Lin, C.C. Preparation of high resolution chromosomes
from amniotic fuid cells. Prenat. Diagn. 1983, 3, 265–267.
26. Yunis, J.J.; Sawyer, J.R.; Ball, D.W. The characterization of high-resolution G-banded
chromosomes of man. Chromosoma. 1978, 67, 293–307.
27. Arsham, M.S.; Barch, M.J.; Lawce, H.J. The AGT Laboratory Manual. 4th Edition.
Wiley-Blackwell, Hoboken, NJ; 2017.
28. Rooney, D.E.; Czepulkowski, B.H. Human Cytogenetics: A Practical Approach Volume
I: Constitutional Analysis. 2nd Edition. Oxford University Press, Oxford; 1992.
29. Rooney, D.E.; Czepulkowski, B.H. Human Cytogenetics: A Practical Approach Volume
II. Malignancy and Acquired Abnormalities. 2nd Edition. Oxford University Press,
Oxford; 1992.
30. Bickmore, W.A. Patterns in the genome. Heredity (Edinb). 2019, 123, 50–57.
31. Gardiner, K. Human genome organization. Curr. Opin. Genet. Dev. 1995, 5, 315–322.
32. Ushiki, T.; Hoshi, O.; Iwai, K.; Kimura, E.; Shigeno, M. The structure of human meta-
phase chromosomes: Its histological perspective and new horizons by atomic force
microscopy. Arch. Histol. Cytol. 2002, 65, 377–390.
33. Chicano, A.; Crosas, E.; Oton, J.; Melero, R.; Engel, B.D.; Daban, J.R. Frozen-hydrated
chromatin from metaphase chromosomes has an interdigitated multilayer structure.
EMBO J. 2019, 38, e99769.
34. Collins, F.S.; Lander, E.S.; Rogers, J.; Waterston, R.H.; Conso, I.H.G.S. Finishing the
euchromatic sequence of the human genome. Nature. 2004, 431, 931–945.
35. She, X.W.; Horvath, J.E.; Jiang, Z.S.; Liu, G.; Furey, T.S.; Christ, L.; Clark, R.; Graves,
T.; Gulden, C.L.; Alkan, C.; Bailey, J.A.; Sahinalp, C.; Rocchi, M.; Haussler, D.; Wilson,
R.K.; Miller, W.; Schwartz, S.; Eichler, E.E. The structure and evolution of centromeric
transition regions within the human genome. Nature. 2004, 430, 857–864.
Banding Cytogenetics 23
36. Geiersbach, K.B.; Gardiner, A.E.; Wilson, A.; Shetty, S.; Bruyère, H.; Zabawski, J.; Saxe,
D.F.; Gaulin, R.; Williamson, C.; Van Dyke, D.L. Subjectivity in chromosome band-level
estimation: A multicenter study. Genet. Med. 2014, 16, 170–175.
37. Bishop, A.; Blank, C.E.; Hunter, H. Heritable variation in the length of the Y chromo-
some. The Lancet. 1962, 280, 18–20.
38. Starkman, M.N.; Shaw, M.W. Atypical acrocentric chromosomes in Negro and Caucasian
Mongols. Am. J. Hum. Genet. 1967, 19, 162–173.
39. Lubs, H.A.; Ruddle, F.H. Chromosome polymorphism in American Negro and White
populations. Nature. 1971, 233, 134–136.
40. McKenzie, W.H.; Lubs, H.A. Human Q and C chromosomal variations: Distribution and
incidence. Cytogenet. Cell Genet. 1975, 14, 97–115.
41. Verma, R.S.; Dosik, H.; Lubs, H.A. Size and pericentric inversion heteromorphisms of
secondary constriction regions (h) of chromosomes 1, 9, and 16 as detected by CBG
technique in Caucasians: Classifcation, frequencies, and incidence. Am. J. Med. Genet.
1978, 2, 331–339.
42. Rawal, L.; Kumar, S.; Mishra, S.R.; Lal, V.; Bhattacharya, S.K. Clinical manifestations
of chromosomal anomalies and polymorphic variations in patients suffering from repro-
ductive failure. J. Hum. Reprod. Sci. 2020, 13, 209–215.
43. Kosyakova, N.; Grigorian, A.; Liehr, T.; Manvelyan, M.; Simonyan, I.; Mkrtchyan,
H.; Aroutiounian, R.; Polityko, A.D.; Kulpanovich, A.I.; Egorova, T.; Jaroshevich, E.;
Frolova, A.; Shorokh, N.; Naumchik, I.V.; Volleth, M.; Schreyer, I.; Nelle, H.; Stumm,
M.; Wegner, R.D.; Reising-Ackermann, G.; Merkas, M.; Brecevic, L.; Martin, T.;
Rodríguez, L.; Bhatt, S.; Ziegler, M.; Kreskowski, K.; Weise, A.; Sazci, A.; Vorsanova,
S.; Cioff, M.deB.; Ergul, E. Heteromorphic variants of chromosome 9. Mol. Cytogenet.
2013, 6, 14.
44. Zhu, J.J.; Qi, H.; Cai, L.; Wen, X.H.; Zeng, W.; Tang, G.D.; Luo, Y.; Meng, R.; Mao,
X.Q.; Zhang, S.Q. C-banding and AgNOR-staining were still effective complementary
methods to indentify chromosomal heteromorphisms and some structural abnormalities
in prenatal diagnosis. Mol. Cytogenet. 2019, 12, 41.
45. Ceylaner, G.; Ceylaner, S.; Danişman, N.; Ergün, A.; Ekici, E.; Schinzel, A.; Baumer,
A. Chromosomal heteromorphisms may help for the diagnosis of uniparental disomy
(UPD): A case report. Prenat. Diag. 2007, 27, 1072–1074.
46. Magenis, R.E.; Overton, K.M.; Chamberlin, J.; Brady, T.; Lovrien, E. Parental origin of
the extra chromosome in Down’s syndrome. Hum. Genet. 1977, 37, 7–16.
47. Magenis, R.E.; Sheehy, R.R.; Brown, M.G.; McDermid, H.E.; White, B.N.; Zonana, J.;
Weleber, R. Parental origin of the extra chromosome in the cat eye syndrome: Evidence
from heteromorphism and in situ hybridization analysis. Am. J. Med. Genet. 1988, 29,
9–19.
48. Brothman, A.R.; Schneider, N.R.; Saikevych, I.; Cooley, L.D.; Butler, M.G.; Patil, S.;
Mascarello, J.T.; Rao, K.W.; Dewald, G.W.; Park, J.P.; Persons, D.L.; Wolff, D.J.; Vance,
G.H.; Cytogenetics Resource Committee.; College of American Pathologists/American
College of Medical Genetics. Cytogenetic heteromorphisms: Survey results and report-
ing practices of Giemsa-band regions that we have pondered for years. Arch. Pathol.
Lab. Med. 2006, 130, 947–949.
49. Lukusa, T.; Fryns, J.P. Human chromosome fragility. Biochim. Biophys. Acta. 2008,
1779, 3–16.
50. Sutherland, G.R.; Richards, R.I. The molecular basis of fragile sites in human chromo-
somes. Curr. Opin. Genet. Dev. 1995, 5, 323–327.
24 Cytogenetics and Molecular Cytogenetics
51. Xie, X.; Chai, H.; DiAdamo, A.; Grommisch, B.; Wen, J.; Zhang, H.; Li, P. Genotype-
phenotype correlations for putative haploinsuffcient genes in deletions of 6q26-q27:
Report of eight patients and review of literature. Glob. Med. Genet. 2022, 9, 166–174.
52. Warren, S.T.; Nelson, D.L. Advances in molecular analysis of fragile X syndrome.
JAMA. 1994, 271, 536–542.
53. Mikhail, F.M.; Heerema, N.A.; Rao, K.W.; Burnside, R.D.; Cherry, A.M.; Cooley, L.D.
Section E6.1–6.4 of the ACMG technical standards and guidelines: Chromosome stud-
ies of neoplastic blood and bone marrow-acquired chromosomal abnormalities. Genet.
Med. 2016, 18. 635–642.
54. Silva, M.; de Leeuw, N.; Mann, K.; Schuring-Blom, H.; Morgan, S.; Giardino, D.; Rack,
K.; Hastings, R. European guidelines for constitutional cytogenomic analysis. Eur. J.
Hum. Genet. 2019, 27, 1–16.
55. Hook, E.B. Exclusion of chromosomal mosaicism: Tables of 90%.; 95% and 99% conf-
dence limits and comments on use. Am. J. Hum. Genet. 1977, 29, 94–97.
56. Zhou, Q.H.; Wu, S.Y.; Amato, K.; DiAdamo, A.; Li, P.N. Spectrum of cytogenomic
abnormalities revealed by array comparative genomic hybridization on products of con-
ception culture failure and normal karyotype samples. J. Genet Genomics. 2016, 43,
121–131.
57. Chai, H.; DiAdamo, A.; Grommisch, B.; Boyle, J.; Amato, K.; Wang, D.; Wen, J.; Li,
P. Integrated FISH, karyotyping and aCGH analyses for effective prenatal diagnosis of
common aneuploidies and other cytogenomic abnormalities. Med. Sci. (Basel). 2019,
7, 16.
58. Meng, J.; Matarese, C.; Crivello, J.; Wilcox, K.; Wang, D.; DiAdamo, A.; Xu, F.; Li,
P. Changes in and effcacies of indications for invasive prenatal diagnosis of cytoge-
nomic abnormalities: 13 years of experience in a single center. Med. Sci. Monit. 2015,
21, 1942–1948.
59. Chai, H.; DiAdamo, A.; Grommisch, B.; Xu, F.; Zhou, Q.; Wen, J.; Mahoney, M.; Bale,
A.; McGrath, J.; Spencer-Manzon, M.; Li, P.; Zhang, H. A retrospective analysis of
10-year data assessed the diagnostic accuracy and effcacy of cytogenomic abnormali-
ties in current prenatal and pediatric settings. Front. Genet. 2019, 10, 1162.
60. Xie, X.; Tan, W.; Li, F.; Ramirez, P.; DiAdamo, A.; Grommisch, B.; Amato, K.; Chai,
H.; Wen, J.; Li, P. Diagnostic cytogenetic testing following positive noninvasive prenatal
screening results of sex chromosome abnormalities: Report of fve cases and systematic
review of evidence. Mol. Genet. Genomic Med. 2020, 8, e1297.
61. Nagaoka, S.I.; Hassold, T.J.; Hunt, P.A. Human aneuploidy: Mechanisms and new
insights into an age-old problem. Nat. Rev. Genet. 2012, 13, 493–504.
62. Dumont, M.; Gamba, R.; Gestraud, P.; Klaasen, S.; Worrall, J.T.; De Vries, S.G.;
Boudreau, V.; Salinas-Luypaert, C.; Maddox, P.S.; Lens, S.M.; Kops, G.J.; McClelland,
S.E.; Miga, K.H.; Fachinetti, D. Human chromosome-specifc aneuploidy is infuenced
by DNA-dependent centromeric features. EMBO J. 2020, 39, e102924.
63. Rossi, M.R.; DiMaio, M.S.; Xiang, B.; Lu, K.; Kaymakcalan, H.; Seashore, M.; Mahoney,
M.J.; Li, P. Clinical and genomic characterization of distal duplications and deletions of
chromosome 4q: Study of two cases and review of the literature. Am. J. Med. Genet. A.
2009, 149A, 2788–2794.
64. Wei, Y.; Gao, X.; Yan, L.; Xu, F.; Li, P.; Zhao, Y. Prenatal diagnosis and postnatal fol-
lowup of partial trisomy 13q and partial monosomy 10p: A case report and review of the
literature. Case Rep. Genet. 2012, 2012, 821347.
65. Zhang, H.Z.; Xu, F.; Seashore, M.; Li, P. Unique genomic structure and distinct mitotic
behavior of ring chromosome 21 in two unrelated cases. Cytogenet. Genome Res. 2012,
136, 180–187.
Banding Cytogenetics 25
66. Cook, S.; Wilcox, K.; Grommisch, B.; Li, P.; Xu, F. Prenatal diagnosis of Xq26.1-q26.3
duplication in two fetuses of a woman with gonadal mosaicism. North Am. J. Med. Sci.
2014, 7, 176–179.
67. Xu, F.; Li, L.; Schulz, V.P.; Xiang, B.; Zhao, H.; Li, P. Cytogenomic mapping and bioin-
formatic mining reveal interacting brain expressed genes for intellectual disability. Mol.
Cytogenet. 2014, 7, 4.
68. Qumsiyeh, M.B.; Li, P. Chapter 5: Molecular Biology of Cancer: Cytogenetics.
Philadelphia: Lippincott Williams & Wilkins Publishers; 2001.
69. Nowell, P.C.; Hungerford, D.A. Chromosome studies on normal and leukemic human
leukocytes. J. Natl. Cancer. Inst. 1960, 25, 85–109.
70. Rowley, J.D. Genetics: A story of swapped ends. Science. 2013, 340, 1412–1413.
71. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.;
Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; Jaffe, E.S. The 2016 revision of the World
Health Organization classifcation of lymphoid neoplasms. Blood. 2016, 127, 2375–2390.
72. Moorman, A.V. New and emerging prognostic and predictive genetic biomarkers in
B-cell precursor acute lymphoblastic leukemia. Haematologica. 2016, 101, 407–416.
73. Couture, T.; Amato, K.; DiAdamo, A.; Li, P. Jumping translocations of 1q in myelo-
dysplastic syndrome and acute myeloid leukemia: Report of three cases and review of
literature. Case Rep. Genet. 2018, 2018, 8296478.
74. Marino-Enriquez, A.; Li, P.; Samuelson, J.; Rossi, M.R.; Reyes-Mugica, M. Congenital
fbrosarcoma with a novel complex 3-way translocation t(12;15;19) and unusual histo-
logic features. Hum. Pathol. 2008, 39, 1844–1848.
75. Chai, H.; Xu, F.; DiAdamo, A.; Grommisch, B.; Mao, H.; Li, P. Cytogenomic character-
ization of giant ring or rod marker chromosomes on four cases of well-differentiated and
dedifferentiated liposarcomas. Case Rep. Genet. 2022, 2022, 6341207 .
76. Larson, D.P.; Akkari, Y.M.; Van Dyke, D.L.; Raca, G.; Gardner, J.A.; Rehder, C.W.;
Kaiser-Rogers, K.A.; Eagle, P.; Yuhas, J.A.; Gu, J.; Toydemir, R.M.; Kearney, H.; Conlin,
L.K.; Tang, G.; Dolan, M.M.; Ketterling, R.P.; Peterson, J.F. Conventional cytogenetic
analysis of hematologic neoplasms: A 20-year review of profciency test results from the
college of American pathologists/American college of medical genetics and genomics
cytogenetics committee. Arch. Pathol. Lab. Med. 2021, 145, 176–190.
77. Giersch, A.B.S.; Bieber, F.R.; Dubuc, A.M.; Fletcher, J.A.; Ligon, A.H.; Mason-Suares,
H.; Morton, C.C.; Weremowicz, S.; Xiao, S.; Cin, P.D. Reporting of diagnostic cytoge-
netic results. Curr. Protoc. Hum. Genet. 2016, 89, A 1D 1-A 1D 23.
78. Chun, K.; Hagemeijer, A.; Iqbal, A.; Slovak, M.L. Implementation of standardized inter-
national karyotype scoring practices is needed to provide uniform and systematic evalu-
ation for patients with myelodysplastic syndrome using IPSS criteria: An International
Working Group on MDS cytogenetics study. Leuk. Res. 2010, 34, 160–165.
79. Grimwade, D.; Hills, R.K.; Moorman, A.V.; Walker, H.; Chatters, S.; Goldstone, A.H.;
Wheatley, K.; Harrison, C.J.; Burnett, A.K; National Cancer Research Institute Adult
Leukaemia Working Group. Refnement of cytogenetic classifcation in acute myeloid
leukemia: Determination of prognostic signifcance of rare recurring chromosomal
abnormalities among 5876 younger adult patients treated in the United Kingdom Medical
Research Council trials. Blood. 2010, 116, 354–365.
80. Baliakas, P.; Jeromin, S.; Iskas, M.; Puiggros, A.; Plevova, K.; Nguyen-Khac, F.; Davis,
Z.; Rigolin, G.M.; Visentin, A.; Xochelli, A.; Delgado, J.; Baran-Marszak, F.; Stalika,
E.; Abrisqueta, P.; Durechova, K.; Papaioannou, G.; Eclache, V.; Dimou, M.; Iliakis,
T.; Collado, R.; Doubek, M.; Calasanz, M.J.; Ruiz-Xiville, N.; Moreno, C.; Jarosova,
M.; Leeksma, A.C.; Panayiotidis, P.; Podgornik, H.; Cymbalista, F.; Anagnostopoulos,
A.; Trentin, L.; Stavroyianni, N.; Davi, F.; Ghia, P.; Kater, A.P.; Cuneo, A.; Pospisilova,
26 Cytogenetics and Molecular Cytogenetics
S.; Espinet, B.; Athanasiadou, A.; Oscier, D.; Haferlach, C.; Stamatopoulos, K.; ERIC.;
the European Research Initiative on CLL. Cytogenetic complexity in chronic lympho-
cytic leukemia: Defnitions, associations, and clinical impact. Blood. 2019, 133(11),
1205–1216.
81. Haase, D.; Stevenson, K.E.; Neuberg, D.; Maciejewski, J.P.; Nazha, A.; Sekeres,
M.A.; Ebert, B.L.; Garcia-Manero, G.; Haferlach, C.; Haferlach, T.; Kern, W.; Ogawa,
S.; Nagata, Y.; Yoshida, K.; Graubert, T.A.; Walter, M.J.; List, A.F.; Komrokji, R.S.;
Padron, E.; Sallman, D.; Papaemmanuil, E.; Campbell, P.J.; Savona, M.R.; Seegmiller,
A.; Adès, L.; Fenaux, P.; Shih, L.Y.; Bowen, D.; Groves, M.J.; Tauro, S.; Fontenay, M.;
Kosmider, O.; Bar-Natan, M.; Steensma, D.; Stone, R.; Heuser, M.; Thol, F.; Cazzola,
M.; Malcovati, L.; Karsan, A.; Ganster, C.; Hellström-Lindberg, E.; Boultwood, J.;
Pellagatti, A.; Santini, V.; Quek, L.; Vyas, P.; Tüchler, H.; Greenberg, P.L.; Bejar, R.;
International Working Group for MDS Molecular Prognostic Committee. TP53 muta-
tion status divides myelodysplastic syndromes with complex karyotypes into distinct
prognostic subgroups. Leukemia. 2019, 33, 1747–1758.
3 Generation of
Microdissection-
Derived Painting
Probes from Single
Copy Chromosomes
Svetlana Romanenko and Vladimir Trifonov
CONTENTS
Introduction.............................................................................................................. 27
Materials .................................................................................................................. 28
Instruments ..................................................................................................... 28
Reagents.......................................................................................................... 28
Solutions ......................................................................................................... 29
Description of Methods............................................................................................ 29
Micro-Needle and Micropipette Preparation .................................................. 29
Slide Preparation............................................................................................. 29
Chromosome Staining .................................................................................... 29
Microdissection............................................................................................... 30
Generation of Paint Probe by Whole Genome Amplifcation (WGA) ........... 31
Expected Results...................................................................................................... 31
Acknowledgements.................................................................................................. 32
References................................................................................................................ 32
INTRODUCTION
The method of chromosome microdissection with a subsequent DNA isolation was
developed over 40 years ago. It allows physically isolating either a whole or a large
fragment of a chromosome. After early works on Drosophila polytene,[1] murine,[2]
and human[3] metaphase chromosomes, the method was considerably improved by
Senger and colleagues,[4] who frst employed a combination of an inverted micro-
scope equipped with a rotating plate, including a deproteinization step, followed
by DNA amplifcation. From 1992 onwards, most paints for fuorescence in situ
hybridization (FISH) have been made by direct polymerase chain reaction (PCR)
amplifcation, using a degenerate universal primer (DOP-PCR).[5] Subsequent major
DOI: 10.1201/9781003223658-3 27
28 Cytogenetics and Molecular Cytogenetics
MATERIALS
INSTRUMENTS
• Inverted microscope the IX Series (Olympus Shinjuku, Tokyo, Japan) or
Axiovert 10 or 135 (Zeiss, Jena, Germany)
• Right-handed and left-handed micromanipulators (Narishige, Tokyo, Japan
or Eppendorf, Germany)
• Pipette puller (Narishige, Japan)
• Glass rods, 2mm diameter (Schott Glas, Mainz, Germany)
• Pasteur pipettes, 230mm (Deltalab, Barcelona, Spain, or Assistent,
Sondheim, Germany)
• Aspirator with trap fask (“Grant”, UK).
REAGENTS
• Glycerol (Sigma-Aldrich)
• Proteinase K from Tritirachium album (Sigma-Aldrich, USA)
• Trypsin (Sigma-Aldrich, USA)
• 2×SSC
• 1×PBS
• Giemsa (Merck)
• Carbon tetrachloride (Sigma-Aldrich, USA)
• Dimethyldichlorosilane (Sigma-Aldrich, USA)
• GenomePlex® Whole Genome Amplifcation (WGA) Kit, WGA1 (Sigma-
Aldrich, USA)
Generation of Microdissection-Derived Painting Probes 29
SOLUTIONS
• Collection drop solution: 30% Glycerol, 10mM Tris-HCl pH 7.5, 10mM
NaCl, 0.1% SDS, 1mM EDTA, 0.1% Trition X100, 1.44mg/ml Proteinase K.
DESCRIPTION OF METHODS
MICRO-NEEDLE AND MICROPIPETTE PREPARATION
1. Prepare micro-needles from 2mm glass rods on a vertical two-step puller,
to get very sharp but not very long edges.
2. Expose the needles to UV light for at least 30min.
3. Prepare micropipettes on the same puller from Pasteur pipettes.
4. Break the micropipette tips carefully to obtain a small round opening
(~30–70µm).
5. Siliconize micropipettes by immersing into 1% dimethyldichlorosilane in
carbon tetrachloride using an aspirator with a trap fask (“Grant”).
6. Air dry and wash twice in 1mM EDTA (pH 7.5).
7. Incubate micropipettes for 30min at 100°C.
8. Keep micropipettes and micro-needles in closed boxes prior to use.
Caution: Dimethyldichlorosilane and carbon tetrachloride are highly
toxic—all manipulations should be done under the draft hood. Two ml of
siliconization solution is enough to siliconize more than 30 micropipettes.
SLIDE PREPARATION
Traditionally, preparations for microdissection were dropped on coverslips.[15]
However, in our experience, the use of microscope slides is more convenient. To
achieve a good spreading of chromosomes, the conditions for slides preparation
should be optimized. In some cases, for better spreading, it is recommended to keep
the slide over a water bath, or add more fxative over the freshly dropped preparation.
CHROMOSOME STAINING
Traditionally, chromosome staining was made using trypsin and phosphate buf-
fer incubation.[15] However, conventional GTG-banding[16] allows for better stain-
ing of chromosomes and does not affect the quality of probes. In cases where the
30 Cytogenetics and Molecular Cytogenetics
MICRODISSECTION
1. Prepare 10–20µl of collection drop solution.
2. Use a 10× or 20× objective.
3. Move the objective down to adjust the needle.
4. Load a needle into the holder, and then load onto the micromanipulator, so
that the tip of the needle is close to the center of visual feld of the objective.
5. Find the needle in the visual feld and center it; then switch to 40× objec-
tive and ensure that the tip of the needle can be seen with the 40× objective.
6. Put the slide on the specimen stage and fnd a suitable metaphase spread.
By rotating the table, place the chromosome to be cut at a right angle to
the needle.
7. Bring the needle down carefully until it is just above the chromosome.
Forward movement of the tip will lead to the excision of the chromosome
material.
8. Touch the excised fragment with the tip of the needle several times until it
is attached to the needle. Carefully elevate the needle.
9. Switch to a 10× or 20× objective.
10. Remove the microscope slides.
11. Take a siliconized pipette; gently touch the surface of the collection drop
solution using the siliconized pipette, so that a tiny amount of collection
drop solution will be sucked up by capillary force.
12. Suspend the pipette above the objective.
13. Place the needle to the same plane as the pipette tip using micromanipu-
lator. Transfer the chromosome fragment by touching the solution inside
of the pipette with the needle, and then withdraw the needle. Large frag-
ments will remain visible in the collection drop for several seconds.
14. Put the pipette in a humidifed tray at 60°C for 1–2h.
Generation of Microdissection-Derived Painting Probes 31
EXPECTED RESULTS
The resulted libraries may demonstrate different qualities depending on the repetitive
DNA content in studied genomes and chromosomes. Therefore, we recommend using
COT DNA for repetitive DNA suppression during FISH.[17] Figure 3.1 demonstrates
expected results of FISH with painting probes obtained from single-copies of chro-
mosomes 15 and 16 of the Nile crocodile (Crocodylus niloticus). Although both
chromosomes are specifcally painted by the respective libraries, heterochromatic
32 Cytogenetics and Molecular Cytogenetics
FIGURE 3.1 FISH-result on a metaphase spread from Nile crocodile CNO (Crocodylus
niloticus) hybridized with painting probes obtained from single-copies of chromosomes 15
(red) and 16 (green) of this species. Painting probe CNO15 additionally paints heterochro-
matic regions of most CNO chromosomes, despite suppression with sonicated total genomic
CNO-DNA.
regions on other chromosomes are also highlighted with the CNI15 probe, despite
suppression with sonicated total genomic DNA of Crocodylus niloticus.
ACKNOWLEDGEMENTS
While this protocol was written, SR and VT were supported by RFBR grant
# 19-54-26017.
REFERENCES
1. Scalenghe, F.; Turco, E.; Edstrom, J.E.; Pirrotta, V.; Melli, M. Microdissection and clon-
ing of DNA from a specifc region of Drosophila melanogaster polytene chromosomes.
Chromosoma. 1981, 82, 205–216.
2. Röhme, D.; Fox, H.; Herrmann, B.; Frischauf, A.M.; Edström, J.E.; Mains, P.; Silver,
L.M.; Lehrach, H. Molecular clones of the mouse t complex derived from microdissected
metaphase chromosomes. Cell. 1984, 36, 783–788.
3. Bates, G.P.; Wainwright, B.J.; Williamson, R.; Brown, S.D. Microdissection and
microcloning from the short arm of human chromosome 2. Mol. Cell. Biol. 1986, 6,
3826–3830.
Generation of Microdissection-Derived Painting Probes 33
CONTENTS
Introduction.............................................................................................................. 35
FISH Probes ............................................................................................................. 36
Repetitive Probes ............................................................................................ 36
Locus-Specifc Probes .................................................................................... 36
Partial Chromosome Paints............................................................................. 37
Whole Chromosome Paints ............................................................................ 37
Whole Genome ............................................................................................... 37
FISH Probe Sets.............................................................................................. 37
Basic FISH-Protocol ................................................................................................ 38
Yield = FISH Applications....................................................................................... 38
Conclusions.............................................................................................................. 39
References................................................................................................................ 39
INTRODUCTION
FISH = fuorescence in situ hybridization is one of two basic approaches originally
being considered to constitute the feld of molecular cytogenetics.[1] FISH is the
approach that at present is widely applied in chromosomic research, while its sister
approach PRINS = primed in situ hybridization (also sometimes called primed in
situ amplifcation, or primed in situ labeling) is practically no more in use.[1, 2] FISH
can be used to visualize DNA or RNA in tissues, interphase cells and metaphases.[1, 3]
Here only DNA-oriented FISH is considered.
FISH is in general single cell–oriented; single to multiple loci in a genome can
be accessed by single to multicolor-FISH approaches. The possibilities of FISH are
only limited by ideas whose questions can be solved by this approach.[1] Thus, since
the frst radioactive ISH experiments in the 1970s[4] and FISH-experiments in the
1980s,[5] practically each year new applications in human genetic, animal- and plant-
genetics and even in single-cell eukaryotic and prokaryotic research are reported.[1, 6]
The FISH-principle is depicted in Figure 4.1. Necessary for FISH are only a target-
DNA and a probe-DNA to hybridize.[3]
In the following, basics on DNA-probes being used in FISH and some examples
for application of FISH—mainly in human genetics—are provided.
DOI: 10.1201/9781003223658-4 35
36 Cytogenetics and Molecular Cytogenetics
FISH PROBES
FISH probes = probe-DNA in human genetics are nowadays mainly commercially
available ones, being ready to use and labeled already with fuorophores or other
haptens like biotin or digoxigenin.[1, 3] Besides, there also can be used homemade/
homebrewed probes being labeled either by PCR-methods[7] or Nick-translation.[8]
Here fve basic probe DNA types suitable and used for FISH are listed:
REPETITIVE PROBES
Repetitive and/or heterochromatic DNA stretches can be easily visualized by FISH.[9]
Basic units of the repeats are 1 to 200 base pairs; however, they become detectable
by FISH as soon as they span 10s to 1000s of kilo base pairs.[9] In FISH, randomly
selected 2n to 6n repeats, telomeric repeats (6n), centromeric repeats (~170n) and
repeats being present in nucleolus organizing regions (NOR) are applied as FISH-
probes in general.[10] As in human chromosomes most centromeres have a private
satellite-DNA sequence, these centromeric/alphoid besides telomeric probes are
popular, commercially available probes.[3]
LOCUS-SPECIFIC PROBES
Locus-specifc probes (LSPs) are normally euchromatic DNA-stretches cloned into
genetic vectors; nowadays in most cases bacterial artifcial chromosome (BAC)
clones are used as basis for LSPs.[3] To produce good and reproducible FISH-signals,
FISH—An Overview 37
a vector needs to carry inserts of at least 12 kb,[11] while for mapping purposes excep-
tionally 0.44 kb–sized cDNA probes can be useful.[12]
WHOLE GENOME
FISH is normally used to access single cells. However, when doing compara-
tive genomic hybridization (CGH),[22] the single cell–derived metaphase chromo-
somes of a normal individual is just used as a kind of matrix, or low-resolution
DNA-chip. Towards this matrix, DNA extracted from millions of cells derived
from two different body tissues or even two closely related species is hybrid-
ized. Certainly tested DNAs are labeled in two different colors. Thus, it is pos-
sible to detect copy number gains and losses between the two probes. This CGH
approach has been used in human genetic research for solid tumors and has
been further evolved as an important tool in diagnostics nowadays as chromo-
some microarray (CMA) technique.[23, 24] In addition, CGH is currently applied
in evolution-research.[10]
BASIC FISH-PROTOCOL
As each laboratory uses a specifc protocol to do FISH, here just basic steps to be
included in each metaphase FISH-protocol are listed—see also ref[3]:
CONCLUSIONS
As a conclusion, still best suited is a statement of Serakinci and Koelvraa from 2009:
FISH techniques were originally developed as extra tools in attempts to map genes and
a number of advances were achieved with this new technique. However, it soon became
apparent that the FISH concept offered promising possibilities also in a number of
other areas in biology and its use spread into new areas of research and also into the
area of clinical diagnosis. In very general terms the virtues of FISH are in two areas
of biology, namely genome characterization and cellular organization, function and
diversity. . . . To what extend FISH technology will be further developed and applied
in new areas of research in the future remains to be seen.[42]
REFERENCES
1. Liehr, T. Molecular cytogenetics in the era of chromosomics and cytogenomic
approaches. Front. Genet. 2021, 12, 720507.
2. Pellestor, F. Development and adaptation of the PRINS technology: An overview.
Methods Mol. Biol. 2006, 334, 211–220.
3. Liehr, T. (Ed.). Fluorescence in situ Hybridization (FISH): Application Guide. 2nd
Edition. Springer, Berlin; 2017.
4. Gall, J.G.; Pardue, M.L. Formation and detection of RNA-DNA hybrid molecules in
cytological preparations. Proc. Natl. Acad. Sci. U. S. A. 1969, 63, 378–383.
5. Langer, P.; Waldrop, A.; Ward, D. Enzymatic synthesis of biotin-labeled olynucleo-
tides: Novel nucleic acid affnity probes. Proc. Natl. Acad. Sci. U. S. A. 1981, 78,
6633–6637.
6. Liehr, T. Basics and literature on multicolor fuorescence in situ hybridization applica-
tion. http://cs-tl.de/DB/TC/mFISH/0-Start.html [accessed on 01/12/2022].
7. Telenius, H.; Carter, N.P.; Bebb, C.E.; Nordenskjöld, M.; Ponder, B.A.J.; Tunnacliffe,
A. Degenerate oligonucleotide-primed PCR: General amplifcation of target DNA by a
single degenerate primer. Genomics. 1992, 13, 718–725.
8. Rigby, P.W.J.; Dieckmann, M.; Rhodes, C.; Berg, P. Labeling deoxyribonucleic acid to
high specifc activity in vitro by nick translation with DNA polymerase I. J. Mol. Biol.
1977, 113, 237–251.
9. Liehr, T. Repetitive elements in humans. Int. J. Mol. Sci. 2021, 22, 2072.
10. Xu, D.; Sember, A.; Zhu, Q.; de Oliveira, E.A.; Liehr, T.; Al-Rikabi, A.B.H.; Xiao, Z.;
Song, H.; de Bello Cioff, M. Deciphering the origin and evolution of the X1X2Y system
in two closely-related oplegnathus species (Oplegnathidae and Centrarchiformes). Int. J.
Mol. Sci. 2019, 20, 3571.
11. Liehr, T.; Thoma, K.; Kammler, K.; Gehring, C.; Ekici, A.; Bathke, K.; Grehl, H.;
Rautenstrauss, B. Direct preparation of uncultured EDTA-treated or heparinized blood
for interphase FISH analysis. Appl. Cytogenet. 1995, 21, 185–188.
40 Cytogenetics and Molecular Cytogenetics
12. Pröls, F.; Liehr, T.; Rinke, R.; Rautenstrauss, B. Assignment of the microvascular endo-
thelial differentiation gene 1 (MDG1) to human chromosome band 14q24.2→q24.3 by
fuorescence in situ hybridization. Cytogenet. Genome Res. 1997, 79, 149–150.
13. Liehr, T. Human Genetics: Edition 2020: A Basic Training Package. Epubli, Berlin;
2020.
14. Al-Rikabi, A.B.H.; Cioff, M.d.B.; Liehr, T. Chromosome microdissection on semi-
archived material. Cytometry Part A. 2019, 95, 1285–1288.
15. Kytölä, S.; Rummukainen, J.; Nordgren, A.; Karhu, R.; Farnebo, F.; Isola, J.; Larsson, C.
Chromosomal alterations in 15 breast cancer cell lines by comparative genomic hybrid-
ization and spectral karyotyping. Genes Chromosomes Cancer. 2000, 28, 308–317.
16. Senger, G.; Lüdecke, H.J.; Horsthemke, B.; Claussen, U. Microdissection of banded
human chromosomes. Hum. Genet. 1990, 84, 507–511.
17. Zlotina, A.; Kulikova, T.; Kosyakova, N.; Liehr, T.; Krasikova, A. Microdissection of
lampbrush chromosomes as an approach for generation of locus-specifc FISH-probes
and samples for high-throughput sequencing. BMC Genomics. 2016, 17, 126.
18. Guyon, R.; Rakotomanga, M.; Azzouzi, N.; Coutanceau, J.; Bonillo, C.; D’Cotta, H.;
Pepey, E.; Soler, L.; Rodier-Goud, M.; D’Hont, A.; Conte, M.; van Bers, N.; Penman,
D.; Hitte, C.; Crooijmans, R.; Kocher, T.; Ozouf-Costaz, C.; Baroiller, J.; Galibert, F.
A high-resolution map of the Nile tilapia genome: A resource for studying cichlids and
other percomorphs. BMC Genomics. 2012, 13, 222.
19. Müller, S.; Wienberg, J. “Bar-coding” primate chromosomes: Molecular cytogenetic
screening for the ancestral hominoid karyotype. Hum. Genet. 2001, 109, 85–94.
20. Ferguson-Smith, M.; Yang, F.; Rens, W.; O’Brien, P. The impact of chromosome sorting
and painting on the comparative analysis of primate genomes. Cytogenet. Genome Res.
2005, 108, 112–121.
21. Sabile, A.; Poras, I.; Cherif, D.; Goodfellow, P.; Avner, P. Isolation of monochromosomal
hybrids for mouse chromosomes 3, 6, 10, 12, 14, and 18. Mamm. Genome. 1997, 8,
81–85.
22. Kallioniemi, A.; Kallioniemi, O.P.; Sudar, D.; Rutovitz, D.; Gray, J.W.; Waldman, F.;
Pinkel, D. Comparative genomic hybridization for molecular cytogenetic analysis of
solid tumors. Science. 1992, 258, 818–821.
23. Pinkel, D.; Segraves, R.; Sudar, D.; Clark, S.; Poole, I.; Kowbel, D.; Collins, C.; Kuo,
W.L.; Chen, C.; Zhai, Y.; Dairkee, S.H.; Ljung, B.M.; Gray, J.W.; Albertson, D.G. High
resolution analysis of DNA copy number variation using comparative genomic hybrid-
ization to microarrays. Nat. Genet. 1998, 20, 207–211.
24. Brady, P.D.; Vermeesch, J.R. Genomic microarrays: A technology overview. Prenat.
Diagn. 2012, 32, 336–343.
25. Schröck, E.; du Manoir, S.; Veldman, T.; Schoell, B.; Wienberg, J.; Ferguson-Smith, M.;
Ning, Y.; Ledbetter, D.; Bar-Am, I.; Soenksen, D.; Garini, Y.; Ried, T. Multicolor spec-
tral karyotyping of human chromosomes. Science. 1996, 273, 494–497.
26. Speicher, M.; Gwyn Ballard, S.; Ward, D. Karyotyping human chromosomes by combi-
natorial multi-fuor FISH. Nat. Genet. 1996, 12, 368–375.
27. Nagai, T.; Okamura, T.; Yanase, T.; Chaya, R.; Moritoki, Y.; Kobayashi, D.; Akita, H.;
Yasui, T. Examination of diagnostic accuracy of UroVysion fuorescence in situ hybrid-
ization for bladder cancer in a single community of Japanese hospital patients. Asian
Pac. J. Cancer. Prev. 2019, 20, 1271–1273.
28. Eiben, B.; Trawicki, W.; Hammans, W.; Goebel, R.; Pruggmayer, M.; Epplen, J. Rapid
prenatal diagnosis of aneuploidies in uncultured amniocytes by fuorescence in situ
hybridization: Evaluation of >3,000 cases. Fetal. Diagn. Ther. 1999, 14, 193–197.
FISH—An Overview 41
29. Nguyen, K.; Broucqsault, N.; Chaix, C.; Roche, S.; Robin, J.; Vovan, C.; Gerard, L.;
Mégarbané, A.; Urtizberea, J.; Bellance, R.; Barnérias, C.; David, A.; Eymard, B.; Fradin,
M.; Manel, V.; Sacconi, S.; Tiffreau, V.; Zagnoli, F.; Cuisset, J.M.; Salort-Campana, E.;
Attarian, S.; Bernard, R.; Lévy, N.; Magdinier, F. Deciphering the complexity of the 4q
and 10q subtelomeres by molecular combing in healthy individuals and patients with
facioscapulohumeral dystrophy. J. Med. Genet. 2019, 56, 590–601.
30. Vorsanova, S.G.; Zelenova, M.A.; Yurov, Y.B.; Iourov, I.Y. Behavioral variability and
somatic mosaicism: A cytogenomic hypothesis. Curr. Genomics. 2018, 19, 158–162.
31. Mkrtchyan, H.; Gross, M.; Hinreiner, S.; Polytiko, A.; Manvelyan, M.; Mrasek, K.;
Kosyakova, N.; Ewers, E.; Nelle, H.; Liehr, T.; Bhatt, S.; Thoma, K.; Gebhart, E.;
Wilhelm, S.; Fahsold, R.; Volleth, M.; Weise, A. The human genome puzzle: The role of
copy number variation in somatic mosaicism. Curr. Genomics. 2010, 11, 426–431.
32. Jancuskova, T.; Plachy, R.; Stika, J.; Zemankova, L.; Hardekopf, D.W.; Liehr, T.;
Kosyakova, N.; Cmejla, R.; Zejskova, L.; Kozak, T.; Zak, P.; Zavrelova, A.; Havlikova,
P.; Karas, M.; Junge, A.; Ramel, C.; Pekova, S. A method to identify new molecular
markers for assessing minimal residual disease in acute leukemia patients. Leuk. Res.
2013, 37, 1363–1373.
33. Hovhannisyan, G.G. Fluorescence in situ hybridization in combination with the comet
assay and micronucleus test in genetic toxicology. Mol. Cytogenet. 2010, 3, 17.
34. Shimizu, N. Extrachromosomal double minutes and chromosomal homogeneously
staining regions as probes for chromosome research. Cytogenet. Genome Res. 2009,
124, 312–326.
35. Koltsova, A.; Pendina, A.; Efmova, O.; Chiryaeva, O.; Kuznetzova, T.; Baranov, V.S. On
the complexity of mechanisms and consequences of chromothripsis: An update. Front.
Genet. 2019, 10, 393.
36. Nazaryan-Petersen, L.; Bjerregaard, V.; Nielsen, F.; Tommerup, N.; Tümer, Z.
Chromothripsis and DNA repair disorders. J. Clin. Med. 2020, 9, 613.
37. Spangenberg, V.; Arakelyan, M.; Cioff, M.; Liehr, T.; Al-Rikabi, A.; Martynova, E.;
Danielyan, F.; Stepanyan, I.; Galoyan, E.; Kolomiets, O. Cytogenetic mechanisms of
unisexuality in rock lizards. Sci. Rep. 2020, 10, 8697.
38. Lemke, J.; Claussen, J.; Michel, S.; Chudoba, I.; Mühlig, P.; Westermann, M.; Sperling,
K.; Rubtsov, N.; Grummt, U.; Ullmann, P.; Kromeyer-Hauschild, K.; Liehr, T.; Claussen,
U. The DNA-based structure of human chromosome 5 in interphase. Am. J. Hum. Genet.
2002, 71, 1051–1059.
39. Cremer, T.; Cremer, M.; Hübner, B.; Silahtaroglu, A.; Hendzel, M.; Lanctôt, C.;
Strickfaden, H.; Cremer, C. The interchromatin compartment participates in the struc-
tural and functional organization of the cell nucleus. Bioessays. 2020, 42, e1900132.
40. Daban, J. Supramolecular multilayer organization of chromosomes: Possible functional
roles of planar chromatin in gene expression and DNA. FEBS Lett. 2020, 594, 395–411.
41. Weise, A.; Othman, M.A.K.; Bhatt, S.; Löhmer, S.; Liehr, T. Application of BAC-probes
to visualize copy number variants (CNVs). Meth. Mol. Biol. (Clifton, N.J.). 2015, 1227,
299–307.
42. Serakinci, N.; Koelvraa, S. Molecular cytogenetic applications in diagnostics and
research: An overview. In In Fluorescence In Situ Hybridization (FISH): Application
Guide; Liehr, T., Ed. 1st Edition. Springer, Berlin, 2009.
5 FISH—Microscopy
and Evaluation
Ivan Y. Iourov
CONTENTS
Introduction .............................................................................................................. 43
Fluorescence Microscopy ........................................................................................ 44
Imaging and Image Analysis.................................................................................... 45
Human Eye versus Electric Eye ............................................................................... 46
Conclusions .............................................................................................................. 46
Acknowledgments.................................................................................................... 47
References ................................................................................................................ 47
INTRODUCTION
In the postgenomic era, microscopic (visual) analysis of human chromosomes is
often regarded as an optional procedure in genomic research. However, the avail-
ability of fuorescence-based microscopic (genomic) techniques (like fuorescence
in situ hybridization = FISH) gradually expands the area of genetic and genomic
research.[1–3] Moreover, practical issues in medical genetics cannot be properly
addressed without FISH, the essence of which lies in the fuorescence microscopy,
visualization approaches, and DNA-based chromosome research.[1, 3, 4] Accordingly,
to understand the role of FISH in current bio- and medical science,[5–7] fuorescence
microscopy is an area to start with. Currently, FISH-based assays are required for
analyzing structure and numbers of human chromosomes in interphase (the longest
part of the cell cycle or the most usual state of a cell of an eukaryotic organism),[7–9]
genome organization at the chromosomal level,[10, 11] developing single-cell genomic
approaches,[12] and cytogenetic genotoxicological studies.[13] In total, there is a huge
and increasing amount of FISH-based assays applicable for different biomedical
tasks.[5, 7, 8, 14] Fluorescence microscopy is the basis for evaluation of FISH results,
which determines the resolution and effciency of the molecular cytogenetic analysis.
Therefore, fuorescence microscopy and evaluation are of prime importance for suc-
cessful application of FISH-based molecular cytogenetic technologies. Here, a brief
overview of fuorescence microscopy and FISH results evaluation is provided.
DOI: 10.1201/9781003223658-5 43
44 Cytogenetics and Molecular Cytogenetics
FLUORESCENCE MICROSCOPY
The frst key concept in fuorescence microscopy is the resolution. The smallest vis-
ible molecular target in the feld of the vision determines the microscopic resolution
of FISH. Usually, FISH analysis is referred to as the evaluation of small fuorescing
objects of roundish, globular, or spherical shape (i.e. FISH signals or fuorescence
signals). The diffraction may cause problems in distinguishing the closest fuores-
cence signals. Accordingly, fuorescence microscopy resolution is related to the
determination of the closest distinguishable visible objects. The resolution depends
on the wavelength (λ), the numerical aperture (NA), the magnifcation, and detection
devices. The smallest diameter of a visible FISH signal is determined by the follow-
ing ratio: d=1.22 λ/NA. Visible FISH signals are still distinguished when the dis-
tance between fuorescing objects is more than half a diameter, determined by the
aforementioned ratio.[5]
The second key concept in fuorescence microscopy is the background autofuo-
rescence. The related phenomena affect actually all the visualization-based fores-
cence assays. The decrease of the effective contrast of the adjacent FISH signals
impeding distinguishing between the signals may help to autofuorescence-related
problems in visualization of FISH results. However, reducing the image contrast
would result in resolution reduction. This dependence determines interplay between
autofuorescence and resolution (i.e. the frst and the second key concept of fuores-
cence microscopy). Actually, the background may be removed by imaging software
(imaging techniques). A number of procedures for pre-treatment of microscopic
slides before or after FISH is able to increase the signal removing background auto-
fuorescence are available, as well.[5]
The third key concept in fuorescence microscopy is the detection. The use of
specifc detection systems or algorithms is the way to increase the microscopic res-
olution of FISH-based techniques. Generally, a digital camera is used as a device
for FISH detection. The resolution may be modifed by an increase in the quantum
effciency of the detection system. Automatically or manually, the increase of the
quantum effciency reduces the noise and optical NA of the microscope and cam-
era. The dependence between NA and d is as follows: increasing NA decreases
d. In the detection context, FISH resolution may also be affected by the maxi-
mum number of distinguishable intensity levels. This number is defned by the
parameters of the detection system; exposure time may be adjusted manually or
automatically. Alternatively, detection using digitalization for quantitative analy-
sis allows differentiation between FISH signals and background autofuorescence.
The digitalization and quantitative analysis underlie quantitative FISH (QFISH),
which is discussed hereafter. For details about fuorescence microscopy concepts
considered in the FISH context, readers are recommended to address following
sources.[5–7]
The essential device for FISH-based microscopic analysis is the fuorescence
microscope. The device is required to include a fuorescence light source and a set
of fuorescence flters, which are composed of dichroic mirror and excitation and
emission flters. For FISH-based molecular cytogenetic analyses, a high-pressure
Hg (mercury) lamp is generally the most preferable to be used as a light source. The
FISH—Microscopy and Evaluation 45
latter provides suitable high-energy excitations at wavelengths specifc for the major-
ity of FISH-based techniques. The lamp produces heat. Accordingly, lamp cooling is
systematically requited. The hot lamp should not be turned on. Usually, the overall
working time of these lamps is less than 200 burning hours. Since recently, light-
emitting diodes (LED) are an Hg-free alternative as light source, which also have
longer ‘burning times’ in the range of 2000 hours or more.[15] FISH specimens are
not to be exposed to the light. To avoid fading, light shutter is used to block the expo-
sure. Multicolor nature of FISH is realized through the application of fuorescence
flters. The combination of flters has to provide proper contrast between fuoro-
chromes used for probe labeling.[5]
CONCLUSIONS
Microscopy is the basis of FISH analysis. Actually, a number of biomedical areas
require FISH to have an important place in the methodology. Somatic human
genetics/genomics, cytogenomics, and cancer research are certainly among these
areas.[27–29] Therefore, knowledge of basic principles underlying FISH may be useful
FISH—Microscopy and Evaluation 47
for researchers undertaking related biomedical studies. Since FISH and microscopy
are intimately linked to each other, it is to acknowledge the importance of knowing
opportunities and limitations of fuorescence microscopy and evaluations in the con-
text of this molecular cytogenetic platform.
ACKNOWLEDGMENTS
The chapter is dedicated to professors Svetlana G. Vorsanova and Yuri B. Yurov
and to Dr. Ilia V. Soloviev. The author’s work is supported by the Government
Assignment of the Russian Ministry of Science and Higher Education, Assignment
no. AAAA-A19-119040490101-6 and by the Government Assignment of the Russian
Ministry of Health, Assignment no. 121031000238-1.
REFERENCES
1. Liehr, T. From human cytogenetics to human chromosomics. Int J Mol Sci. 2019, 20,
826.
2. Ye, C.J.; Stilgenbauer, L.; Moy, A.; Liu, G.; Heng, H.H. What is karyotype coding and
why is genomic topology important for cancer and evolution? Front Genet. 2019, 10,
1082.
3. Iourov, I.Y.; Yurov, Y.B.; Vorsanova, S.G. Chromosome-centric look at the genome. In:
Human Interphase Chromosomes: Biomedical Aspects; Iourov, I.Y.; Vorsanova, S.G.;
Yurov, Y.B., Eds. Springer, Berlin, 2020, pp. 157–170.
4. Gersen, S.L.; Keagle, M.B. The Principles of Clinical Cytogenetics. 2nd Edition.
Humana, Totowa, NJ; 2005.
5. Liehr, T. (Ed.). Fluorescence In Situ Hybridization (FISH): Application Guide. Springer,
Berlin, Heidelberg, Germany; 2017.
6. Garini, Y.; Young, I.T.; McNamara, G. Spectral imaging: Principles and applications.
Cytometry A. 2006, 69, 735–747.
7. Yurov, Y.B.; Vorsanova, S.G.; Iourov, I.Y. Human Interphase Chromosomes: Biomedical
Aspects. Springer, Berlin; 2013.
8. Vorsanova, S.G.; Yurov, Y.B.; Iourov, I.Y. Human interphase chromosomes: A review of
available molecular cytogenetic technologies. Mol Cytogenet. 2010, 3, 1.
9. Bakker, B.; van den Bos, H.; Lansdorp, P.M.; Foijer, F. How to count chromosomes in a
cell: An overview of current and novel technologies. Bioessays. 2015, 37, 570–577.
10. Manvelyan, M.; Hunstig, F.; Bhatt, S.; Mrasek, K.; Pellestor, F.; Weise, A.; Simonyan, I.;
Aroutiounian, R.; Liehr, T. Chromosome distribution in human sperm: A 3D multicolor
banding-study. Mol Cytogenet. 2008, 1, 25.
11. McClelland, S.E. Single-cell approaches to understand genome organisation throughout
the cell cycle. Essays Biochem. 2019, 63, 209–216.
12. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B. Single cell genomics of the brain: Focus on
neuronal diversity and neuropsychiatric diseases. Curr Genomics. 2012, 13, 477–488.
13. Hovhannisyan, G.G. Fluorescence in situ hybridization in combination with the comet
assay and micronucleus test in genetic toxicology. Mol Cytogenet. 2010, 3, 17.
14. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B. Recent patents on molecular cytogenetics.
Recent Pat DNA Gene Seq. 2008, 2, 6–15.
15. Lang, D.S.; Zeiser, T.; Schultz, H.; Stellmacher, F.; Vollmer, E.; Zabel, P.; Goldmann, T.
LED-FISH: Fluorescence microscopy based on light emitting diodes for the molecular
analysis of Her-2/neu oncogene amplifcation. Diagn Pathol. 2008, 3, 49.
48 Cytogenetics and Molecular Cytogenetics
16. Iourov, I.Y.; Soloviev, I.V.; Vorsanova, S.G.; Monakhov, V.V.; Yurov, Y.B. An approach
for quantitative assessment of fuorescence in situ hybridization (FISH) signals for
applied human molecular cytogenetics. J Histochem Cytochem. 2005, 53, 401–408.
17. Konsti, J.; Lundin, J.; Jumppanen, M.; Lundin, M.; Viitanen, A.; Isola, J. A public-domain
image processing tool for automated quantifcation of fuorescence in situ hybridisation
signals. J Clin Pathol. 2008, 61, 278–282.
18. Amakawa, G.; Ikemoto, K.; Ito, H.; Furuya, T.; Sasaki, K. Quantitative analysis of cen-
tromeric FISH spots during the cell cycle by image cytometry. J Histochem Cytochem.
2013, 61, 699–705.
19. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B. Fluorescence intensity profles of in situ
hybridization signals depict genome architecture within human interphase nuclei. Tsitol
Genet. 2008, 42, 3–8.
20. Vorsanova, S.G.; Iourov, I.Y.; Beresheva, A.K.; Demidova, I.A.; Monakhov, V.V.; Kravets,
V.S.; Bartseva, O.B.; Goyko, E.A.; Soloviev, I.V.; Yurov, Y.B. Non-disjunction of chro-
mosome 21, alphoid DNA variation, and sociogenetic features of Down syndrome. Tsitol
Genet. 2005, 39, 30–36.
21. Weise, A.; Gross, M.; Mrasek, K.; Mkrtchyan, H.; Horsthemke, B.; Jonsrud, C.; Von
Eggeling, F.; Hinreiner, S.; Witthuhn, V.; Claussen, U.; Liehr, T. Parental-origin-
determination fuorescence in situ hybridization distinguishes homologous human chro-
mosomes on a single-cell level. Int J Mol Med. 2008, 21, 189–200.
22. Iourov, I.Y. Quantitative fuorescence in situ hybridization (QFISH). Methods Mol Biol.
2017, 1541, 143–149.
23. Liehr, T.; Heller, A.; Starke, H.; Rubtsov, N.; Trifonov, V.; Mrasek, K.; Weise, A.;
Kuechler, A.; Claussen, U. Microdissection based high resolution multicolor banding for
all 24 human chromosomes. Int J Mol Med. 2002, 9, 335–339.
24. Alhourani, E.; Aroutiounian, R.; Harutyunyan, T.; Glaser, A.; Schlie, C.; Pohle, B.;
Liehr, T. Interphase molecular cytogenetic detection rates of chronic lymphocytic
leukemia-specifc aberrations are higher in cultivated cells than in blood or bone mar-
row smears. J Histochem Cytochem. 2016, 64, 495–501.
25. Iourov, I.Y.; Liehr, T.; Vorsanova, S.G.; Kolotii, A.D.; Yurov, Y.B. Visualization of inter-
phase chromosomes in postmitotic cells of the human brain by multicolour banding
(MCB). Chromosome Res. 2006, 14, 223–229.
26. Iourov, I.Y.; Liehr, T.; Vorsanova, S.G.; Yurov, Y.B. Interphase chromosome-specifc
multicolor banding (ICS-MCB): A new tool for analysis of interphase chromosomes in
their integrity. Biomol Eng. 2007, 24, 415–417.
27. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B.; Kutsev, S.I. Ontogenetic and pathogenetic
views on somatic chromosomal mosaicism. Genes. 2019, 10, 379.
28. Ye, C.J.; Sharpe, Z.; Heng, H.H. Origins and consequences of chromosomal instability:
From cellular adaptation to genome chaos-mediated system survival. Genes. 2020, 11,
1162.
29. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B. Systems cytogenomics: Are we ready yet?
Curr Genomics. 2021, 22, 75–78.
6 FISH—in Routine
Diagnostic Settings
Chenghua Cui and Peining Li
CONTENTS
Introduction.............................................................................................................. 49
Analytic and Clinical Validity of FISH Assays........................................................ 50
Design of Locus-Specifc, Gene-Targeted, Regional or Whole
Chromosome Painting Probes............................................................. 50
Validation and Verifcation of FISH Assays for Clinical Testing.................... 50
Detection of Constitutional Cytogenomic Abnormalities........................................ 52
Rapid Prenatal Screening of Common Aneuploidy........................................ 53
Detection of Microdeletion and Microduplication Syndromes ...................... 53
Detection of Numerical and Structural Chromosomal Abnormalities............ 55
Detection of Somatic Numerical and Structural Chromosomal
Abnormalities ..................................................................................... 57
Detection of Recurrent Rearrangements in Hematopoietic
and Lymphoid Tissues ........................................................................ 57
Detection of Recurrent Rearrangements in Solid Tumors.............................. 60
Conclusions.............................................................................................................. 64
Acknowledgements.................................................................................................. 65
References................................................................................................................ 65
INTRODUCTION
Fluorescence in situ hybridization (FISH) is a cell-based macromolecule rec-
ognition technique based on the complementary nature of DNA double strands.
Fluorophore-coupled nucleotides can be enzymatically incorporated into cloned or
amplifed DNA fragments. These fuorescence-labeled DNA fragments can be used
as probes to hybridize onto the complementary sequences in interphase nuclei and
metaphase chromosomes, and then visualized through a fuorescence microscope.
This technique was initially developed to replace radioactive probes for physical
mapping of genes to chromosomes.[1–4] Human genomic DNA fragments of 100 to
300 kilobases (kb) cloned in bacterial artifcial chromosomes (BAC) were com-
piled in a database and mapped to chromosomes by FISH.[5] These mapped BAC
clones provided frstly cytogenetic landmarks for the Human Genome Project and
later a resource for developing locus-specifc and gene-targeted FISH probes. FISH
assays have higher analytical resolution than karyotyping and high sensitivity and
DOI: 10.1201/9781003223658-6 49
50 Cytogenetics and Molecular Cytogenetics
BAC clones mapped across the human genome in the genome browser (https://
genome.ucsc.edu/) are a reliable resource for developing FISH probes target-
ing to specifc loci and genes.[10] A wide variety of fuorophores are available,
and several enzymatic reactions such as nick translation, random priming, and
degenerative oligonucleotide primed polymerase chain reaction have been used
to incorporate fuorophore-coupled nucleotides.[11] Locus-specifc, gene-targeted,
regional or whole chromosome painting probes have been developed for diagnos-
tic uses. Enumerative locus-specifc probes containing highly repetitive α-satellite
sequences at centromere and unique DNA sequences at pericentric or specifc
chromosome loci are used to detect numerical chromosome abnormalities.[6, 10]
Probes containing repetitive sequences at telomeres and subtelomeric sequences
are used to detect telomeric integrity and subtelomeric rearrangements.[12] Gene-
targeted design of dual-color double fusion probes for two genes are used to detect
conjugated signals for a reciprocal translocation involving the two genes, while
dual-color break apart probes juxtaposing the 5′ and 3′ regions of a gene are used
to detect separated 5′ and 3′ signals for rearrangements of the gene.[13, 14] Contig
probes for a chromosome band or collection of probes for a whole chromosome
can be labeled in a multicolor pattern to generate a painting effect for an individual
chromosome or a whole metaphase.[15–17] Due to the standardized probe design and
widespread diagnostic usage, all these probes are available commercially or can be
made custom. Figure 6.1 shows various designs of FISH probes and typical signal
patterns on interphase nuclei and metaphase cells.
FIGURE 6.1 FISH probes designs and typical signal patterns on interphase nuclei and
metaphase chromosomes. A. FISH using locus-specifc probes targeted to chromosomes 13
and 21 shows an abnormal pattern of three signals for the KCNJ6 probe indicating trisomy
21 in a patient and a normal two signals for both RB1 and KCNJ6 probes in a normal control
(NL). B. FISH using whole chromosome painting (WCP) probes for chromosomes 2 and 16
shows translocated segment of 2q onto a 16p. C. FISH using locus-specifc probes for 5p and
5q detects a deletion of 5q in an abnormal cell and a disomic pattern in a normal cell. D. FISH
using gene-specifc dual-color double fusion probes for the ABL1 and BCR genes shows one
independent signal and two conjugated signals for BCR‑ABL1 rearrangement (pointed by
arrow).
of FISH probes onto nuclei or chromosomes, the targeted region of unique sequence
FISH probes for clinical use is in the range of 100–200 kb. This has been generally
accepted as the analytic resolution for FISH assays using a fuorescence microscope.
The size of G-band in the 550-band high resolution level is in an average of 5–6
megabases (Mb) per band. The analytical resolution of FISH is approximately 50
times higher than karyotyping.
To comply with clinical laboratory standards and guidelines, FISH assays need to
be verifed and validated before their usage on clinical specimens.[18, 19] Verifcation
refers to analytical accuracy by ensuring the location of hybridized probes on the
expected chromosome loci and analytical precision by measuring quantitative agree-
ment between repeated assessments of the same sample.[18] Accuracy of FISH probes
can be evaluated by a hybridization to normal control metaphases for a correct signal
pattern onto the expected loci in chromosomes and to abnormal metaphases for the
52 Cytogenetics and Molecular Cytogenetics
intended abnormal signal pattern. The latter approach has the advantage of con-
frming both chromosomal location and numerical or structural rearrangements.
Accuracy can be evaluated by scoring a minimum of fve metaphase cells to verify
the presence of hybridized probes to the appropriate chromosome loci and absence
of cross-hybridization to other chromosomes. Precision of FISH assays measures the
reproducibility by performing consecutive three to ten repeat FISH assays over three
to ten days on a selected specimen with a known proportional of normal and abnor-
mal cells. The precision is calculated as the mean, standard deviation, and range of
the results; intra-assay and inter-assay concordance for signal pattern scoring should
be ≥ 95%.[20, 21]
Validation refers to the assessment of analytical sensitivity, specifcity, reportable
range, and other performance characteristics required to ensure assay performance.
For the examination of a FISH signal at the targeted location, probe sensitivity and
specifcity are equivalent to analytical sensitivity and specifcity.[18] Probe sensi-
tivity is the percentage of scorable metaphase chromosomes or interphase nuclei
with the expected probe signal pattern. Probe specifcity is the percentage of all
scored signals that occur at the expected location at metaphase chromosomes or
at the expected signal pattern at interphase nuclei. Probe sensitivity and specifcity
should be established by analyzing signal patterns of probe hybridization to at least
40 chromosomes in 20 metaphases or 100 interphase nuclei. Reportable range is
defned by a cut-off value for normal signal patterns.[20, 21] This cut-off value for the
upper limit at the 95% and 99% confdence intervals can be easily ascertained using
the β inversion function (BETAINV) in a Microsoft excel spreadsheet.[22] In addition
to the pre-clinical verifcation and validation, quality control, quality assurance, and
quality management procedures should be implemented in the pre-analytic, ana-
lytic, and post-analytic phases for each FISH assay. Interpretation and reporting of
FISH results should follow the technical standards and guidelines.[18, 19] The results
of FISH assays should be described following the current International System for
Human Cytogenomic Nomenclature (ISCN 2020).[23] Profciency testing for labo-
ratories performing FISH assays showed a consensus of 91.7% of participants with
the same diagnostic conclusion, indicating the reliability of current FISH assays in
detecting constitutional and somatic chromosomal abnormalities.[24]
TABLE 6.1
FISH Probes for Common Aneuploidies and Microdeletion/Duplication
Syndromes
Diseases Chromosomal FISH Probes
Abnormalities
Aneuploidy
Sex chromosome aneuploidies 45,X, 47,XXX, DXZ1, DYZ3
47,XXY, 47,XYY
Pätau syndrome trisomy 13 RB1, KCNJ6
Down syndrome trisomy 21
Edwards syndrome trisomy 18 D18Z1
Microdeletion/microduplication syndromes
Kallman syndrome del(X)(p22.31p22.31) KAL1
X-linked ichthyosis del(X)(p22.31p22.31) STS
Cri-du-Chat syndrome del(5)(p15.2p15.2) CTNND2
Sotos syndrome del(5)(q35q35) NSD1
Saethre-Chotzen syndrome del(7)(p21.1p21.1) TWIST1
Williams-Beuren syndrome del(7)(q11.23q11.23) ELN
Williams-Beuren region duplication syndrome dup(7)(q11.23q11.23) ELN
Langer-Giedion Syndrome del(8)(q23.3q23.3) TRPS1
Multiple hereditary exotoses del(8)(q24.1q24.1) EXT1
Prader-Willi/Angelman syndromes del(15)(q11q13) SNRPN, UBE3A
15q11-q13 duplication syndrome dup(15)(q11q13) SNRPN, UBE3A
Smith-Magenis syndrome del(17)(p11.2p11.2) RAI1
Miller-Dieker syndrome del(17)(p13.3p13.3) LIS1
DiGeorge syndrome del(22)(q11.21q11.21) TUPLE1
22q11.2 duplication syndrome dup(22)(q11.21q11.21) TUPLE1
Phelan-McDermid syndrome del(22)(q13q13) SHANK3
interphase cells. For example, FISH mapping identifed the presence of terminal
deletions in a ring chromosome 6 and the association of a deletion of the FOXC1
gene with eye anomalies in the patient.[45] Cases of ring chromosomes 13 and 21
with terminal deletions and interstitial duplication detected by CMA were further
confrmed by FISH.[46, 47] Figure 6.2A shows the FISH assays performed on a case
with a ring chromosome 13 and the tracking of a variant ring and ring chromosome
loss in interphase cells.
CMA has been the method of choice to defne genomic imbalances, but karyotyp-
ing and FISH are needed to construct these imbalances into the rearranged chromo-
somes.[48, 49] For example, in a case with a reciprocal translocation t(5;8)(p15.3;p23.1)
and derivative chromosomes 11 and 16 from likely an 11q/16q rearrangement by band-
ing analysis and deletions in an 11q and a 16q defned by CMA, FISH assays using
whole chromosome painting probes, subtelomeric probes, and locus-specifc probes
on metaphase chromosomes revealed multiple-breakpoint inter-/intra-chromosome
rearrangements during the formation of these derivative chromosomes and identifed
FZD4 haploinsuffciency for exudative vitreoretinopathy in this patient.[50] In a case of
spontaneous abortion with a 1.5 Mb deletion at 5p15.33 detected by CMA, chromo-
some analysis and FISH assay revealed inverted duplication, triplication, and quin-
tuplication resulted from sequential breakage–fusion–bridge events (Figure 5.2B).[51]
For balanced translocations with clinical symptoms, physical mapping of breakpoints
by FISH probes could lead to the identifcation of candidate genes. For example, in a
unique case with a balanced translocation t(9;13), the delineation of the breakpoint and
FIGURE 6.2 FISH analysis for chromosomal structural abnormalities. A. FISH using
probes for the RB1 gene at 13q14.2 and the D13S1825 locus at 13q34 on a metaphase shows a
ring chromosome 13 with an interstitial duplication and a terminal deletion. Further examina-
tion of signal pattern on interphase nuclei noted this ring 13, a dicentric ring 13, and loss of
this ring chromosome 13 for monosomy 13 in 90%, 1%, and 9% of cells. B. FISH using dual-
color probes for distal 5p detects three types of rearrangements. A diagram shows a sequen-
tial breakage–fusion–bridge process through mitosis in the formation of this mosaic pattern.
FISH—in Routine Diagnostic Settings 57
further expression analysis on the gene at the junction identifed that increased expres-
sion of the α‑Kloth gene results in hypophosphatemic rickets and hyperparathyroid-
ism.[52] These cases demonstrated that FISH is an integral part of cytogenomic analysis
to facilitate accurate reporting and interpretation.
partner genes; these abnormal gene fusions encode constitutively activated tyrosine
kinases. Routine karyotype can detect most of these rearrangements but escape
cryptic rearrangements such as a submicroscopic deletion del(4)(q12q12) for FIP1L1‑
PDGFRA gene fusion. FISH assays using dual-color break apart probes for these
genes can detect cryptic rearrangements and infer the partner genes in chromosom-
ally observed or unobserved rearrangements. Imatinib is the frst line therapy for
patients with rearrangements of the PDGFRA and PDGFRB genes, where patients
with FGFR1 gene rearrangements are resistant to this therapy and carry a poor
prognosis.[65]
A panel of 20 probes for 10 FISH assays was utilized to detect recurrent numeri-
cal and structural rearrangements for myelodysplastic syndromes (MDS) and acute
myeloid leukemia (AML); the concordance rate between karyotyping and FISH
was more than 98%.[66] Dual-color two loci enumeration probes for 5q, 7q, and
8q/20q detect −5/5q−, −7/7q−, +8, and −20/20q−, respectively. Dual-color double
fusion probes for the RUNX1‑RUNX1T1, BCR‑ABL1, PML‑RARA, and CBFB‑
MYH11 genes can detect translocations t(8;21)(q22;q22), t(9;22)(q34.1;q11.2), t(15;17)
(q24;q21), and inversion inv(16)(p13q22), respectively. Dual-color break apart probes
for the MECOM gene at 3q26.2, the KMT2A gene at 11q23, and the NUP98 gene at
11p15.4 can detect translocations t(3q;var), t(11q;var), and t(11p;var) for rearrange-
ments of these genes with multiple partner genes. Patients with translocations t(8;21),
t(15;17), and inversion inv(16) have a favorable prognosis, trisomy 8 for an intermedi-
ate prognosis, and 5q-, 7q-, translocation t(11;var), and translocation t(9;22) for unfa-
vorable prognosis. Rapid FISH can give a result within a few hours, which could be
vital for specifc leukemias, such as acute promyelocytic leukemia (APL). APL has
characteristic reciprocal translocation t(15;17) and variant rearrangements involving
the PML‑RARA gene fusion. APL is prone to intra-vascular coagulation and causes
a high risk of mortality due to cerebral hemorrhage. All-trans retinoic acid therapy
is essential for complete remission in these patients. Rapid FISH test can provide
important evidence for the correct diagnosis and treatment of APL, which leads to
more possibilities for patients to survive.
B-cell precursor acute lymphoblastic leukemia (pre B-ALL) is a heterogeneous
disease with associated recurrent chromosomal abnormalities for diagnostic sub-
typing, prognostic stratifcation, and treatment strategy.[67] Current FISH testing for
B-ALL uses two panels of FISH probes and additional FISH probes for specifc rear-
rangements. The primary panel has four sets of probes including the ETV6‑RUNX1
genes for translocation t(12;21)(p13;q22) and an intrachromosomal amplifcation of
21q (iAMP21), the CEP4/CEP10/CEP17 centromeric probes for hyperdiploidy and
low hypodiploidy, the KMT2A gene for an 11q23 translocation t(11q;var) with vari-
ous partners, and the BCR‑ABL1 genes for translocation t(9;22). The detection of
iAMP21 by FISH is defned by fve or more signals for the RUNX1 probe in inter-
phases or three or more signals in a single chromosome 21. Additionally, FISH using
probes for the TCF3‑PBX1 genes for translocation t(1;19)(q23;p13.3) and the TCF3‑
HLF genes for translocation t(17;19)(q22;p13.3) can be performed. The secondary
panel uses dual-color break apart probes for the ABL2 gene at 1q25, the PDGFRB
gene at 5q32, the JAK2 gene at 9p24.1, the ABL1 gene at 9q34, and the CRLF2 gene
at Xp22.3 or Yp11.2 to detect rearrangements for BCR‑ABL1-like B-ALL (Ph-like
FISH—in Routine Diagnostic Settings 59
lymphoma (DLBCL) with BCL6 gene rearrangement and Burkitt lymphoma (BL)
with MYC gene rearrangement can also be detected by this FISH panel for lymphoma.
For cutaneous T-cell lymphoma (CTCL), a panel of 11 FISH probes was developed
and validated to capture gene copy number alterations. This panel includes enumera-
tion probes for genes ARID1A at 1p35.3, DNMT3A at 2p23, CARD11 at 7p22, MYC at
8q24.2, CDKN2A at 9p21.3, ZEB1 at 10p11.2, FAS at 10q24.1, ATM at 11q22.3, RB1
at 13q14.2, TP53 at 17p13.1, and STAT3/5B at 17q21.3.[72]
In general, FISH is an effective way to detect recurrent rearrangements with sig-
nifcant clinical value on diagnosis, prognosis, and treatment. FISH is mandatory
when the karyotype is normal or analyzable metaphases are not present. Different
kinds of specimens such as bone marrow, blood, and lymph node are available for
FISH testing in hematology and lymphoma. Selecting a suitable specimen is criti-
cal for a precise result. It is better to use bone marrow for myeloid diseases, bone
marrow or blood for CLL, CD138+ sorted plasma cells for MGUS/PCM, and lymph
nodes for lymphoma. FISH assays offer a rapid detection of recurrent clonal rear-
rangements, many of which are cryptic and escape the detection by banding analysis,
such as translocation t(4;14)(p16.3;q32) with IGH‑FGFR3 gene fusions, translocation
t(12;21)(p13;q22) with ETV6‑RUNX1 gene fusions, gene deletions of 9p21(CDKN2A),
12p13(ETV6), 13q14.2(RB1), and 17p13.1(TP53), and rearrangements leading to
Ph-like or ETV6‑RUNX1-like ALL. Some chromosome rearrangements involve
genes with many partner genes, such as KMT2A, MYC, IGH, and ETV6. FISH is
recommended to detect rearrangements for these genes. For example, KMT2A rear-
rangement is one of the major drivers and generally regarded as a poor prognostic
factor in acute leukemias. There are more than 120 partner genes of KMT2A rear-
rangement identifed currently, many of which are cryptic and unidentifed by chro-
mosome analysis. FISH is a good approach to detect KMT2A rearrangement using
the break-apart probe. Additionally, FISH assay using probes for sex chromosomes
X and Y has been used to check engraftment of donor cells from sex-mismatched
bone marrow and blood stem cell transplantation. Online resources such as the ‘Atlas
of Genetics and Cytogenetics in Oncology and Haematology’ (http://atlasgeneticson-
cology.org/) are very helpful in interpreting chromosome and FISH results. Table 6.2
lists the routinely used FISH panels for tumors of hematopoietic and lymphoid tis-
sues in clinical cytogenetics.
(Continued)
61
62
TABLE 6.2
(Continued)
Disease Recurrent Rearrangements Gene Rearrangement FISH Probes Prognosis Targeted Therapy
B-cell precursor acute lymphoblastic leukemia (B-ALL)
t(9;22)(q34.1;q11.2) BCR‑ABL1 BCR, ABL1 poor tyrosine kinase inhibitors
t(11q23;var) KMT2A KMT2A poor
t(12;21)(p13.2;q22.1), iAMP21 ETV6‑RUNX1 ETV6, RUNX1 very favorable
Hyperdiploid 4/10/17 D4Z1, D10Z1, D17Z1 very favorable
t(1;19)(q23;p13.3) TCF3‑PBX1 TCF3, PBX1 intermediate risk intensive therapy
t(17;19)(q22;p13.3) TCF3‑HLB TCF3, HLB
Ph-like B-cell ALL
t(1q25;var) ABL2 ABL2 high risk ABL-class tyrosine
kinase inhibitors
63
64 Cytogenetics and Molecular Cytogenetics
CONCLUSIONS
In summary, FISH assays are a clinically validated and cell-based diagnostic
tool capable of detecting numerical and structural chromosomal abnormalities
and genomic copy number alterations. FISH assays have been routinely used in
rapid prenatal screening of common aneuploidies, detection of microdeletion and
microduplication syndromes, and further characterization of sSMC, subtelomeric
FISH—in Routine Diagnostic Settings 65
ACKNOWLEDGEMENTS
Special thanks to Autumn DiAdamo and Jiadi Wen of Yale Clinical Cytogenetics
Laboratory (USA) for helpful discussion and careful editing on the content of this
chapter.
REFERENCES
1. Gall, J.G.; Pardue, M.L. Formation and detection of RNA-DNA hybrid molecules in
cytological preparations. Proc. Natl. Acad. Sci. U. S. A. 1969, 63, 378–383.
2. Rudkin, G.T.; Stollar, B.D. High resolution detection of DNA-RNA hybrids in situ by
indirect immunofuorescence. Nature. 1977, 265, 472–473.
3. Lichter, P.; Cremer, T.; Borden, J.; Manuelidis, L.; Ward, D.C. Delineation of individual
human chromosomes in metaphase and interphase cells by in situ suppression hybridiza-
tion using recombinant DNA libraries. Hum. Genet. 1988, 80, 224–234.
4. Lichter, P.; Tang, C.J.; Call, K.; Hermanson, G.; Evans, G.A.; Housman, D.; Ward, D.C.
High-resolution mapping of human chromosome 11 by in situ hybridization with cosmid
clones. Science. 1990, 247, 64–69.
5. Cheung, V.G.; Nowak, N.; Jang, W.; Kirsch, I.R.; Zhao, S.; Chen, X.N.; Furey, T.S.; Kim,
U.J.; Kuo, W.L.; Olivier, M.; Conroy, J.; Kasprzyk, A.; Massa, H.; Yonescu, R.; Sait, S.;
Thoreen, C.; Snijders, A.; Lemyre, E.; Bailey, J.A.; Bruzel, A.; Burrill, W.D.; Clegg,
S.M.; Collins, S.; Dhami, P.; Friedman, C.; Han, C.S.; Herrick, S.; Lee, J.; Ligon, A.H.;
Lowry, S.; Morley, M.; Narasimhan, S.; Osoegawa, K.; Peng, Z.; Plajzer-Frick, I.; Quade,
B.J.; Scott, D.; Sirotkin, K.; Thorpe, A.A.; Gray, J.W.; Hudson, J.; Pinkel, D.; Ried, T.;
Rowen, L.; Shen-Ong, G.L.; Strausberg, R.L.; Birney, E.; Callen, D.F.; Cheng, J.F.; Cox,
D.R.; Doggett, N.A.; Carter, N.P.; Eichler, E.E.; Haussler, D.; Korenberg, J.R.; Morto,
C.C.; Albertson, D.; Schuler, G.; de Jong, P.J.; Trask, B.J.; BAC Resource Consortium.
Integration of cytogenetic landmarks into the draft sequence of the human genome.
Nature. 2001, 409, 953–958.
6. Ried, T.; Landes, G.; Dackowski, W.; Klinger, K.; Ward, D.C. Multicolor fuorescence in
situ hybridization for the simultaneous detection of probe sets for chromosomes 13, 18,
21, X and Y in uncultured amniotic fuid cells. Hum. Mol. Genet. 1992, 1, 307–313.
7. Weise, A.; Liehr, T. Rapid prenatal aneuploidy screening by fuorescence in situ hybrid-
ization (FISH). Methods Mol. Biol. 2019, 1885, 129–137.
8. Speicher, M.R.; Ward, D.C. The coloring of cytogenetics. Nat. Med. 1996, 2, 1046–1048.
9. Cui, C.; Shu, W.; Li, P. Fluorescence in situ hybridization: Cell-based genetic diagnostic
and research applications. Front. Cell. Dev. Biol. 2016, 4, 89.
10. Castronovo, C.; Valtorta, E.; Crippa, M.; Tedoldi, S.; Romitti, L.; Amione, M.C.;
Guerneri, S.; Rusconi, D.; Ballarati, L.; Milani, D.; Grosso, E.; Cavalli, P.; Giardino, D.;
Bonati, M.T.; Larizza, L.; Finelli, P. Design and validation of a pericentromeric BAC
clone set aimed at improving diagnosis and phenotype prediction of supernumerary
marker chromosomes. Mol. Cytogenet. 2013, 6, 45.
11. Morrison, L.E.; Ramakrishnan, R.; Ruffalo, T.M.; Wilber, K.A. Labeling fuorescence in
situ hybridization probes for genomic targets. Methods Mol. Biol. 2002, 204, 21–40.
66 Cytogenetics and Molecular Cytogenetics
12. Ning, Y.A.R.; Smith, A.C.M.; Macha, M.; Precht, K.; Riethman, H.; Ledbetter, D.H.; Flint,
J.; Horsley, S.; Regan, R.; Kearney, L.; Knight, S.; Kvaloy, K.; Brown, W.R.A. A complete
set of human telomeric probes and their clinical application: National Institutes of Health
and Institute of Molecular Medicine collaboration. Nat. Genet. 1996, 14, 86–89.
13. Bentz, M.; Cabot, G.; Moos, M.; Speicher, M.R.; Ganser, A.; Lichter, P.; Döhner, H.
Detection of chimeric BCR-ABL genes on bone marrow samples and blood smears in
chronic myeloid and acute lymphoblastic leukemia by in situ hybridization. Blood. 1994,
83, 1922–1928.
14. van der Burg, M.; Beverloo, H.B.; Langerak, A.W.; Wijsman, J.; van Drunen, E.; Slater,
R.; van Dongen, J.J. Rapid and sensitive detection of all types of MLL gene transloca-
tions with a single FISH probe set. Leukemia. 1999, 13, 2107–2113.
15. Pinkel, D.; Landegent, J.; Collins, C.; Langerak, A.W.; Wijsman, J.; van Drunen, E.;
Slater, R.; van Dongen, J.J. Fluorescence in situ hybridization with human chromosome-
specifc libraries: Detection of trisomy 21 and translocations of chromosome 4. Proc.
Natl. Acad. Sci. U. S A. 1988, 85, 9138–9142.
16. Meltzer, P.S.; Guan, X.Y.; Burgess, A.; Trent, J.M. Rapid generation of region specifc
probes by chromosome microdissection and their application. Nat. Genet. 1992, 1,
24–28.
17. Speicher, M.R.; Gwyn Ballard, S.; Ward, D.C. Karyotyping human chromosomes by
combinatorial multi-fuor FISH. Nat. Genet. 1996, 12, 368–375.
18. Mascarello, J.T.; Hirsch, B.; Kearney, H.M.; Ketterling, R.P.; Olson, S.B.; Quigley,
D.I.; Rao, K.W.; Tepperberg, J.H.; Tsuchiya, K.D.; Wiktor, A.E; Working Group of the
American College of Medical Genetics Laboratory Quality Assurance Committee.
Section E9 of the American College of Medical Genetics technical standards and guide-
lines: Fluorescence in situ hybridization. Genet. Med. 2011, 13, 667–675.
19. (CLSI) CaLSI. MM07-A2. Fluorescence In Situ Hybridization Methods for Clinical
Laboratories Approved. Guideline. 2nd Edition. Vol. 33. Clinical and Laboratory
Standards Institute, Wayne, PA; Aug. 2013.
20. Wiktor, A.E.; Van Dyke, D.L.; Stupca, P.J.; Ketterling, R.P.; Thorland, E.C.; Shearer,
B.M.; Fink, S.R.; Stockero, K.J.; Majorowicz, J.R.; Dewald, G.W. Preclinical validation
of fuorescence in situ hybridization assays for clinical practice. Genet. Med. 2006, 8,
16–23.
21. Zneimer, S.M. Validation of fuorescence in situ hybridization (FISH) for chromosome 5
monosomy and deletion. Curr. Prot. Hum. Genet. 2020, 105, e96.
22. Ciolino, A.L.; Tang, M.E.; Bryant, R. Statistical treatment of fuorescence in situ hybrid-
ization validation data to generate normal reference ranges using Excel functions. J. Mol.
Diagn. 2009, 11, 330–333.
23. McGowan-Jordan, J.; Hastings, A.; Moore, S. ISCN 2020 an International System for
Human Cytogenomic Nomenclature (2020). Karger, Basel/Freiburg; 2020.
24. Mascarello, J.T.; Brothman, A.R.; Davison, K.; Dewald, G.W.; Herrman, M.; McCandless,
D.; Park, J.P.; Persons, D.L.; Rao, K.W.; Schneider, N.R.; Vance, G.H.; Cooley, L.D;
Cytogenetics Resource Committee of the College of American Pathologists and
American College of Medical Genetics. Profciency testing for laboratories performing
fuorescence in situ hybridization with chromosome-specifc DNA probes. Arch. Pathol.
Lab. Med. 2002, 126, 1458–1462.
25. Chai, H.; DiAdamo, A.; Grommisch, B.; Xu, F.; Zhou, Q.; Wen, J.; Mahoney, M.; Bale,
A.; McGrath, J.; Spencer-Manzon, M.; Li, P.; Zhang, H. A retrospective analysis of
10-year data assessed the diagnostic accuracy and effcacy of cytogenomic abnormali-
ties in current prenatal and pediatric settings. Front. Genet. 2019, 10, 1162.
FISH—in Routine Diagnostic Settings 67
26. Liehr, T.; Ziegler, M. Rapid prenatal diagnostics in the interphase nucleus: Procedure
and cut-off rates. J. Histochem. Cytochem. 2005, 53, 289–291.
27. Tepperberg, J.; Pettenati, M.J.; Rao, P.N.; Lese, C.M.; Rita, D.; Wyandt, H.; Gersen, S.;
White, B.; Schoonmaker, M.M. Prenatal diagnosis using interphase fuorescence in situ
hybridization (FISH): 2-year multi-center retrospective study and review of the litera-
ture. Prenat. Diagn. 2001, 21, 293–301.
28. Lall, M.; Mahajan, S.; Saviour, P.; Paliwal, P.; Joshi, A.; Setai, N.; Verma, J.C. FISH is
not suitable as a standalone test for detecting fetal chromosomal abnormalities. J. Fet.
Med. 2015, 2, 53–59.
29. Chai, H.; DiAdamo, A.; Grommisch, B.; Boyle, J.; Amato, K.; Wang, D.; Wen, J.; Li,
P. Integrated FISH, karyotyping and aCGH analyses for effective prenatal diagnosis of
common aneuploidies and other cytogenomic abnormalities. Med. Sci. (Basel). 2019,
7, 16.
30. Xie, X.; Tan, W.; Li, F.; Carrano, E.; Ramirez, P.; DiAdamo, A.; Grommisch, B.; Amato,
K.; Chai, H.; Wen, J.; Li, P. Diagnostic cytogenetic testing following positive noninvasive
prenatal screening results of sex chromosome abnormalities: Report of fve cases and
systematic review of evidence. Mol. Genet. Genomic Med. 2020, 8, e1297.
31. Schinzel, A. Microdeletion syndromes, balanced translocations, and gene mapping. J.
Med. Genet. 1988, 25, 454–462.
32. Kuwano, A.; Ledbetter, S.A.; Dobyns, W.B.; Emanuel, B.S.; Ledbetter, D.H. Detection
of deletions and cryptic translocations in Miller-Dieker syndrome by in situ hybridiza-
tion. Am. J. Hum. Genet. 1991, 49, 707–714.
33. Desmaze, C.; Prieur, M.; Amblard, F.; Aikem, M.; LeDeist, F.; Demczuk, S.; Zucman,
J.; Plougastel, B.; Delattre, O.; Croquette, M.F.; Brevière, G.-M.; Huon, C.; Le Merrer,
M.; Mathieu, M.; Sidi, D.; Stephan, J.-L.; Aurias, A. Physical mapping by FISH of the
DiGeorge critical region (DGCR): Involvement of the region in familial cases. Am. J.
Hum. Genet. 1993, 53, 1239–1249.
34. Stankiewicz, P.; Lupski, J.R. Genome architecture, rearrangements and genomic disor-
ders. Trends Genet. 2002, 18, 74–82.
35. Ensenauer, R.E.; Adeyinka, A.; Flynn, H.C.; Michels, V.V.; Lindor, N.M.; Dawson, D.B.;
Thorland, E.C.; Lorentz, C.P.; Goldstein, J.L.; McDonald, M.T.; Smith, W.E.; Simon-
Fayard, E.; Alexander, A.A.; Kulharya, A.S.; Ketterling, R.P.; Clark, R.D.; Jalal, S.M.
Microduplication 22q11.2, an emerging syndrome: Clinical, cytogenetic, and molecular
analysis of thirteen patients. Am. J. Hum. Genet. 2003, 73, 1027–1040.
36. Liehr, T.; Claussen, U.; Starke, H. Small supernumerary marker chromosomes (sSMC)
in humans. Cytogenet. Genome Res. 2004, 107, 55–67.
37. Henegariu, O.; Bray-Ward, P.; Artan, S.; Vance, G.H.; Qumsyieh, M.; Ward, D.C. Small
marker chromosome identifcation in metaphase and interphase using centromeric mul-
tiplex fsh (CM-FISH). Lab. Invest. 2001, 81, 475–481.
38. Brecevic, L.; Michel, S.; Starke, H.; Müller, K.; Kosyakova, N.; Mrasek, K.; Weise, A.;
Liehr, T. Multicolor FISH used for the characterization of small supernumerary marker
chromosomes (sSMC) in commercially available immortalized cell lines. Cytogenet,
Genome Res. 2006, 114, 319–324.
39. Manolakos, E.; Kefalas, K.; Neroutsou, R.; Lagou, M.; Kosyakova, N.; Ewers, E.;
Ziegler, M.; Weise, A.; Tsoplou, P.; Rapti, S.M.; Papoulidis, I.; Anastasakis, E.; Garas,
A.; Sotiriou, S.; Eleftheriades, M.; Peitsidis, P.; Malathrakis, D.; Thomaidis, L.; Kitsos,
G.; Orru, S.; Liehr, T.; Petersen, M.B.; Kitsiou-Tzeli, S. Characterization of 23 small
supernumerary marker chromosomes detected at pre-natal diagnosis: The value of fuo-
rescence in situ hybridization. Mol. Med. Rep. 2010, 3, 1015–1022.
68 Cytogenetics and Molecular Cytogenetics
40. Liehr, T.; Ewers, E.; Hamid, A.B.; Kosyakova, N.; Voigt, M.; Weise, A.; Manvelyan, M.
Small supernumerary marker chromosomes and uniparental disomy have a story to tell.
J. Histochem. Cytochem. 2011, 59, 842–848.
41. Liehr, T.; Klein, E.; Mrasek, K.; Kosyakova, N.; Guilherme, R.S.; Aust, N.; Venner, C.;
Weise, A.; Hamid, A.B. Clinical impact of somatic mosaicism in cases with small super-
numerary marker chromosomes. Cytogenet. Genome Res. 2013, 139, 158–163.
42. Armanet, N.; Tosca, L.; Brisset, S.; Liehr, T.; Tachdjian, G. Small supernumerary marker
chromosomes in human infertility. Cytogenet Genome Res. 2015, 146, 100–108.
43. Ravnan, J.B.; Tepperberg, J.H.; Papenhausen, P.; Lamb, A.N.; Hedrick, J.; Eash, D.;
Ledbetter, D.H.; Martin, C.L. Subtelomere FISH analysis of 11 688 cases: An evaluation
of the frequency and pattern of subtelomere rearrangements in individuals with develop-
mental disabilities. J. Med. Genet. 2006, 43, 478–489.
44. Hu, Q.; Chai, H.; Shu, W.; Li, P. Human ring chromosome registry for cases in the
Chinese population: Re-emphasizing cytogenomic and clinical heterogeneity and
reviewing diagnostic and treatment strategies. Mol. Cytogenet. 2018, 11, 19.
45. Zhang, H.Z.; Li, P.; Wang, D.; Huff, S.; Nimmakayalu, M.; Qumsiyeh, M.; Pober, B.R.
FOXC1 gene deletion is associated with eye anomalies in ring chromosome 6. Am. J.
Med. Genet. 2004, 124A, 280–287.
46. Zhang, H.Z.; Xu, F.; Seashore, M.; Li, P. Unique genomic structure and distinct mitotic
behavior of ring chromosome 21 in two unrelated cases. Cytogenet. Genome Res. 2012,
136, 180–187.
47. Xu, F.; DiAdamo, A.J.; Grommisch, B.; Li, P. Interstitial duplication and distal deletion
in a ring chromosome 13 with pulmonary atresia and ventricular septal defect: A case
report and review of literature. N. Am. J. Med. Sci. 2013, 6, 208–212.
48. Rossi, M.R.; DiMaio, M.S.; Xiang, B.; Lu, K.; Kaymakcalan, H.; Seashore, M.; Mahoney,
M.J.; Li, P. Clinical and genomic characterization of distal duplications and deletions of
chromosome 4q: Study of two cases and review of the literature. Am. J. Med. Genet.
2009, 149A, 2788–2794.
49. Khattab, M.; Xu, F.; Li, P.; Bhandari, V. A de novo 3.54 Mb deletion of 17q22-q23.1 asso-
ciated with hydrocephalus: A case report and review of literature. Am. J. Med. Genet.
2011, 155A, 3082–3086.
50. Li, P.; Zhang, H.Z.; Huff, S.; Nimmakayalu, M.; Qumsiyeh, M.; Yu, J.; Szekely, A.; Xu,
T.; Pober, B.R. Karyotype-phenotype insights from 11q14.1-q23.2 interstitial deletions:
FZD4 haploinsuffciency and exudative vitreoretinopathy in a patient with a complex
chromosome rearrangement. Am. J. Med. Genet 2006, 140A, 2721–2729.
51. Chai, H.; Grommisch, B.; DiAdamo, A.; Wen, J.; Hui, P.; Li, P. Inverted duplication:
Triplication and quintuplication through sequential breakage-fusion-bridge events
induced by a terminal deletion at 5p in a case of spontaneous abortion. Mol. Genet.
Genomic. Med. 2019, 7, e00965.
52. Brownstein, C.A.; Adler, F.; Nelson-Williams, C.; Iijima, J.; Li, P.; Imura, A.; Nabeshima,
Y.; Reyes-Mugica, M.; Carpenter, T.O.; Lifton, R.P. A translocation causing increased
alpha-klotho level results in hypophosphatemic rickets and hyperparathyroidism. Proc.
Natl. Acad. Sci. U. S. A. 2008, 105, 3455–3460.
53. Nowell, P.C.; Hungerford, D.A. Chromosome studies on normal and leukemic human
leukocytes. J. Natl. Cancer Inst. 1960, 25, 85–109.
54. Rowley, J.D. Genetics: A story of swapped ends. Science. 2013, 340, 1412–1413.
55. Tkachuk, D.C.; Westbrook, C.A.; Andreeff, M.; Donlon, T.A.; Cleary, M.L.;
Suryanarayan, K.; Homge, M.; Redner, A.; Gray, J.; Pinkel, D. Detection of bcr-abl
fusion in chronic myelogeneous leukemia by in situ hybridization. Science. 1990, 250,
559–562.
FISH—in Routine Diagnostic Settings 69
56. Losada, A.P.; Wessman, M.; Tiainen, M.; Hopman, A.H.; Willard, H.F.; Solé, F.;
Caballín, M.R.; Woessner, S.; Knuutila, S. Trisomy 12 in chronic lymphocytic leukemia:
An interphase cytogenetic study. Blood. 1991, 78, 775–779.
57. Anastasi, J.; Le Beau, M.M.; Vardiman, J.W.; Fernald, A.A.; Larson, R.A.; Rowley,
J.D. Detection of trisomy 12 in chronic lymphocytic leukemia by fuorescence in situ
hybridization to interphase cells: A simple and sensitive method. Blood. 1992, 79,
1796–1801.
58. Jenkins, R.B.; Le Beau, M.M.; Kraker, W.J.; Borell, T.J.; Stalboerger, P.G.; Davis,
E.M.; Penland, L.; Fernald, A.; Espinosa, R 3rd.; Schaid, D.J.; Noel, P.; Dewald, G.W.
Fluorescence in situ hybridization: A sensitive method for trisomy 8 detection in bone
marrow specimens. Blood. 1992, 79, 3307–3315.
59. Dewald, G.; Stallard, R.; Alsaadi, A.; Arnold, S.; Blough, R.; Ceperich, T.M.; Rafael
Elejalde, B.; Fink, J.; Higgins, J.V.; Higgins, R.R.; Hoeltge, G.A.; Hsu, W.T.; Johnson,
E.B.; Kronberger, D.; McCorquodale, D.J.; Meisner, L.F.; Micale, M.A.; Oseth, L.;
Payne, J.S.; Schwartz, S.; Sheldon, S.; Sophian, A.; Storto, P.; Van Tuinen, P.; Wenger,
G.D.; Wiktor, A.; Willis, L.A.; Yung, J.F.; Zenger-Hain, J. A multicenter investigation
with D-FISH BCR/ABL1 probes. Cancer Genet. Cytogenet. 2000, 116, 97–104.
60. Wolff, D.J.; Bagg, A.; Cooley, L.D.; Dewald, G.W.; Hirsch, B.A.; Jacky, P.B.; Rao, K.W.;
Rao, P.N; Association for Molecular Pathology Clinical Practice Committee; American
College of Medical Genetics Laboratory Quality Assurance Committee. Guidance for
fuorescence in situ hybridization testing in hematologic disorders. J. Mol. Diagn. 2007,
9, 134–143.
61. Liehr, T.; Othman, M.A.; Rittscher, K.; Alhourani, E. The current state of molecular
cytogenetics in cancer diagnosis. Expert Rev. Mol. Diagn. 2015, 15, 517–526.
62. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani,
R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; Jaffe, E.S. The 2016 revision of the
World Health Organization classifcation of lymphoid neoplasms. Blood. 2016, 127,
2375–2390.
63. Li, M.M.; Ewton, A.A.; Smith, J.L. Using cytogenetic rearrangements for cancer progno-
sis and treatment (Pharmacogenetics). Curr. Genet. Med. Rep. 2013, 1, 99–112.
64. Gorusu, M.; Benn, P.; Li, Z.; Fang, M. On the genesis and prognosis of variant transloca-
tions in chronic myeloid leukemia. Cancer Genet. Cytogenet. 2007, 173, 97–106.
65. Savage, N.; George, T.I.; Gotlib, J. Myeloid neoplasms associated with eosinophilia and
rearrangement of PDGFRA, PDGFRB, and FGFR1: A review. Int. J. Lab. Hematol.
2013, 35, 491–500.
66. Vance, G.H.; Kim, H.; Hicks, G.A.; Cherry, A.M.; Higgins, R.; Hulshizer, R.L.; Tallman,
M.S.; Fernandez, H.F.; Dewald, G.W. Utility of interphase FISH to stratify patients into
cytogenetic risk categories at diagnosis of AML in an Eastern Cooperative Oncology
Group (ECOG) clinical trial (E1900). Leuk. Res. 2007, 31, 605–609.
67. Moorman, A.V. New and emerging prognostic and predictive genetic biomarkers in
B-cell precursor acute lymphoblastic leukemia. Haematologica. 2016, 101, 407–416.
68. Gesk, S.; Martin-Subero, J.I.; Harder, L.; Luhmann, B.; Schlegelberger, B.; Calasanz,
M.J.; Grote, W.; Siebert, R. Molecular cytogenetic detection of chromosomal break-
points in T-cell receptor gene loci. Leukemia. 2003, 17, 738–745.
69. Smoley, S.A.; Van Dyke, D.L.; Kay, N.E.; Heerema, N.A.; Dell’ Aquila, M.L.; Dal Cin,
P.; Koduru, P.; Aviram, A.; Rassenti, L.; Byrd, J.C.; Rai, K.R.; Brown, J.R.; Greaves,
A.W.; Eckel-Passow, J.; Neuberg, D.; Kipps, T.J.; Dewald, G.W. Standardization of fuo-
rescence in situ hybridization studies on chronic lymphocytic leukemia (CLL) blood and
marrow cells by the CLL Research Consortium. Cancer Genet. Cytogenet. 2010, 203,
141–148.
70 Cytogenetics and Molecular Cytogenetics
70. Döhner, H.; Stilgenbauer, S.; Benner, A.; Leupolt, E.; Kröber, A.; Bullinger, L.; Döhner,
K.; Bentz, M.; Lichter, P. Genomic aberrations and survival in chronic lymphocytic leu-
kemia. N. Engl. J. Med. 2000, 343, 1910–1916.
71. Chen, Z.; Issa, B.; Huang, S.; Aston, E.; Xu, J.; Yu, M.; Brothman, A.R.; Glenn, M. A
practical approach to the detection of prognostically signifcant genomic aberrations in
multiple myeloma. J. Mol. Diagn. 2005, 7, 560–565.
72. Weed, J.; Gibson, J.; Lewis, J.; Carlson, K.; Foss, F.; Choi, J.; Li, P.; Girardi, M. FISH
panel for leukemic CTCL. J. Invest. Dermatol. 2017, 137, 751–753.
73. Van Dekken, H.; Pizzolo, J.G.; Reuter, V.E.; Melamed, M.R. Cytogenetic analysis of
human solid tumors by in situ hybridization with a set of 12 chromosome-specifc DNA
probes. Cytogenet. Cell Genet. 1990, 54, 103–107.
74. Arnoldus, E.P.; Noordermeer, I.A.; Peters, A.C.; Voormolen, J.H.; Bots, G.T.; Raap,
A.K.; van der Ploeg, M. Interphase cytogenetics of brain tumors. Genes Chromosomes
Cancer. 1991, 3, 101–107.
75. Poetsch, M.; Dittberner, T.; Woenckhaus, C.; Kleist, B. Use of interphase cytogenet-
ics in demonstrating specifc chromosomal aberrations in solid tumors: New insights in
the pathogenesis of malignant melanoma and head and neck squamous cell carcinoma.
Histol. Histopathol. 2000, 15, 1225–1231.
76. Arnoldus, E.P.; Dreef, E.J.; Noordermeer, I.A.; Verheggen, M.M.; Thierry, R.F.; Peters,
A.C.; Cornelisse, C.J.; Van der Ploeg, M.; Raap, A.K. Feasibility of in situ hybridisa-
tion with chromosome specifc DNA probes on paraffn wax embedded tissue. J. Clin.
Pathol. 1991, 44, 900–904.
77. El-Naggar, A.K.; van Dekken, H.D.; Ensign, L.G.; Pathak, S. Interphase cytogenetics in
paraffn-embedded sections from renal cortical neoplasms: Correlation with cytogenetic
and fow cytometric DNA ploidy analyses. Cancer Genet. Cytogenet. 1994, 73, 134–141.
78. Jenderny, J.; Koster, E.; Meyer, A.; Borchers, O.; Grote, W.; Harms, D.; Jänig, U.
Detection of chromosome aberrations in paraffn sections of seven gonadal yolk sac
tumors of childhood. Hum. Genet. 1995, 96, 644–650.
79. Nanjangud, G.; Amarillo, I.; Rao, P.N. Solid tumor cytogenetics: Current perspectives.
Clin. Lab. Med. 2011, 31, 785–811.
80. Marino-Enriquez, A.; Li, P.; Samuelson, J.; Rossi, M.R.; Reyes-Mugica, M. Congenital
fbrosarcoma with a novel complex 3-way translocation t(12;15;19) and unusual histo-
logic features. Hum. Pathol. 2008, 39, 1844–1848.
81. Parisi, F.; Ariyan, S.; Narayan, D.; Bacchiocchi, A.; Hoyt, K.; Cheng, E.; Xu, F.; Li, P.;
Halaban, R.; Kluger, Y. Detecting copy number status and uncovering subclonal markers
in heterogeneous tumor biopsies. BMC Genomics. 2011, 12, 230.
82. Nagai, T.; Naiki, T.; Etani, T.; Iida, K.; Noda, Y.; Shimizu, N.; Isobe, T.; Nozaki, S.;
Okamura, T.; Ando, R.; Kawai, N.; Yasui, T. UroVysion fuorescence in situ hybridiza-
tion in urothelial carcinoma: A narrative review and future perspectives. Transl. Androl.
Urol. 2021, 10, 1908–1917.
83. Geiersbach, K.B.; Sill, D.R.; Meyer, R.G.; Yuhas, J.A.; Sukov, W.R.; Mounajjed, T.;
Carter, J.M.; Jenkins, R.B.; Chen, B. HER2 testing for breast cancer in the genomics
laboratory: A sea change for fuorescence in situ hybridization. Arch. Pathol. Lab. Med.
2021, 145, 883–886.
7 FISH—in Leukemia
Diagnostics
Roberto Matos, Mariana de Souza,
Gerson Ferreira, Amanda Figueiredo,
Marcelo Land and Maria Luiza Silva
CONTENTS
Introduction.............................................................................................................. 72
Cytogenetic Analysis ............................................................................................... 73
G-Banding Karyotyping ................................................................................. 73
Molecular Cytogenetics .................................................................................. 74
Chronic Leukemia.................................................................................................... 75
Chronic Myeloid Leukemia............................................................................ 75
Chronic Lymphocytic Leukemia .................................................................... 75
Chronic Myelomonocytic Leukemia (CMML) .................................. 76
Acute Leukemia ....................................................................................................... 76
Acute Myeloid Leukemia ............................................................................... 76
Favorable Prognosis AML .................................................................. 77
Unfavorable Prognosis AML .............................................................. 78
Acute Lymphoblastic Leukemia ..................................................................... 79
Favorable/Intermediate Prognosis ALL.............................................. 80
Unfavorable Prognosis ALL ............................................................... 81
Uncertain Prognosis ALL ................................................................... 82
Recurrent Numerical Abnormalities in Acute Leukemia................................ 82
Complex Karyotypes ...................................................................................... 83
Flexibility in the Application of FISH Probes for Leukemia Diagnosis......... 83
Samples/Tissues ....................................................................................................... 83
Description of Methods............................................................................................ 84
Cell Culture for Chromosome Preparation ..................................................... 84
Cell Extraction Protocol ................................................................................. 85
Slides Preparation Protocol............................................................................. 85
G-Banding Protocol ........................................................................................ 86
Fluorescence In Situ Hybridization (FISH) Protocol ..................................... 87
Pepsin Treatment ................................................................................ 87
FISH Slides Denaturation and Hybridization ..................................... 88
FISH Slides Post Hybridization.......................................................... 89
DOI: 10.1201/9781003223658-7 71
72 Cytogenetics and Molecular Cytogenetics
Yields ....................................................................................................................... 89
FISH Panel and Probes in Leukemia Diagnostics .......................................... 89
FISH in Favorable Prognosis Leukemia ......................................................... 91
RUNX1(21q22)-RUNX1T1(8q22) Probe........................................... 91
CBFb(16q22)-MYH11(16p13) Probe ................................................ 91
PML(15q24)-RARA(17q21) Probe .................................................... 91
KMT2A(11q23)-MLLT3(9p22) Probe ............................................... 92
cMYC(8q24)-IGH(14q32) Probe ....................................................... 92
TCRA/D(14q11) Probe....................................................................... 94
ETV6(12p13)-RUNX1(21q22) Probe ................................................ 94
FISH in Unfavorable Prognosis Leukemia ..................................................... 94
del(5q)/del(7q) Probe.......................................................................... 94
BCR(22q11)-ABL1(9q34) Probe ....................................................... 94
KMT2A(11q23)(MLL) Probe ............................................................ 94
TP53 del(17p13) Probe....................................................................... 94
Conclusions.............................................................................................................. 95
References................................................................................................................ 96
INTRODUCTION
Leukemia comprises a heterogeneous group of hematological neoplasias character-
ized by clonal proliferation of hematopoietic cell lineages that underwent genetic
alterations, leading to be expressed in a specifc differentiation phase. These altera-
tions affect genes with pivotal roles in the control of natural mechanisms of prolif-
eration, differentiation, and apoptosis.[1, 2]
The main clinical symptoms result in accumulation of immature precursors in the
bone marrow (BM). This can harm or even prevent the production of normal blood
cells, such as erythrocytes (causing anemia), or neutrophils (resulting in a suscepti-
bility to infections and platelets, causing hemorrhages). With the excess proliferation
of BM-blast cells, infltration of tissues such as tonsils, lymph nodes, skin, spleen,
kidneys, central nervous system (CNS), and others may occur.[3]
Leukemia cells may be immunophenotypically similar to mature (as observed in
Chronic Lymphocytic Leukemia (CLL)), or precursor cells (originating from several
lineages, as in Acute Leukemia (AL)), or still cells similar to precursor and mature
ones (as in Chronic Myeloid Leukemia (CML)). Leukemia can affect people of any
age. However, there are particular distributions depending on leukemia cell types and
patients’ ages. In this sense, Acute Lymphoblastic Leukemia (ALL) is more common
in early childhood compared with Acute Myeloid Leukemia (AML), which is more
frequently observed in older adults. CML is extremely rare in younger children, and
CLL is almost exclusive to patients over 40 years old, with a median age of 70 years.[4]
Leukemia are a very heterogeneous and vast group of diseases, covering a wide
range of ages.[5] Thus, in this chapter, we will focus on a particular group of leuke-
mia, more specifcally, childhood ALs, to demonstrate the benefts of fuorescence
in situ hybridization (FISH)–based technologies in the daily care of these patients.
Nonetheless, we reiterate that the entire study fowchart detailed here can be applied
to all patients, regardless of age or leukemia type.
FISH—in Leukemia Diagnostics 73
CYTOGENETIC ANALYSIS
The possibility to undertake (banding) cytogenetic analysis of malignant cells repre-
sented the beginning of one of the greatest advances in understanding the biological
nature of neoplasms. This improvement was particularly important in the leukemogen-
esis feld, shedding light on the prognostic implications of chromosomal abnormalities.
Regarding the study of genes involved in disease-specifc translocations, cytogenetics
is considered one of the most powerful tools, leading to a better understanding of chro-
mosomal rearrangements and the mechanisms underlying leukemic transformation.
The precise identifcation of chromosomal breakpoints enabled the cloning of many
genes whose interaction with other genes contributes to the neoplastic process.[6]
For more than 30 years, cytogenetic analysis of patients with leukemia, mainly
AL, led to the discovery of several recurrent chromosome abnormalities, guiding
more effcient treatment regimens. Nonetheless, the prognostic value of rare abnor-
malities is still unknown. In this regard, further cytogenetic studies gathering data
on patients harboring such rare abnormalities are necessary to defne prognosis.[7]
Therefore, an adequate experimental design, individualized for each patient, con-
sidering the knowledge on FISH-based technical possibilities, can help characterize
chromosomal aberrations that in the past would be characterized only as complex/
undefned, thus helping with patient’s risk stratifcation.
G-BANDING K ARYOTYPING
Refnements of chromosome banding techniques over the last 40 years allowed
for improved detection of chromosomal abnormalities in malignant hemopathies.
The advent of these techniques brought the frst revolution in cytogenetic analysis.
Chromosome banding techniques enabled the clear identifcation of individual chro-
mosomes and the description of chromosomic abnormalities.[7]
Pioneers in this feld, Drs. Nowell and Hungerford (1960), described a minute
chromosome in BM samples from patients with CML, which was a game-changer
for hematology. This observation later proved to be the translocation t(9;22)(q34;q11),
generating the breakpoint cluster region protein (BCR)—Abelson tyrosine‑protein
kinase 1 (ABL1) chimeric fusion gene, the product of which was a constitutively
activated tyrosine kinase.[8] Another important step in the feld of tumor cytogenetics
was the description of the reciprocal translocation t(8;21)(q22;q22) by Dr. Rowley
(1973). This translocation is a frequent non-random cytogenetic abnormality in
AML. Dr. Rowley described the translocation t(8;21) from her observation of repeat-
ing pattern of karyotypes analyzed, using banding techniques.[9, 10] Since then, cyto-
genetic analysis has provided remarkable knowledge about the heterogeneity of a
cell population. This approach enabled the detection of several genetic and chromo-
somal abnormalities in different malignancies, making it possible to classify the dis-
ease based on the alteration observed in the patient’s karyotype. For such neoplasms,
74 Cytogenetics and Molecular Cytogenetics
MOLECULAR CYTOGENETICS
In situ hybridization (ISH) is a cytogenetic method used for high-resolution detection
and localization of DNA sequences in tissues and cells.[16] The frst description of
nucleic acid hybridization was made by Gall and Pardue in 1969[17] when DNA dena-
turation was performed in an integrated cytological sample from the ovary of the
frog Xenopus and hybridized with specifc tritium-labeled radioactive RNA. John
and colleagues (1969)[18] also reported in situ hybridization of DNA from the Hela
cell line and Xenopus oocytes with labeled RNA. In 1970, Buongiorno-Nardelli and
Amaldi[19] created a hybridization method using paraffn-embedded hamster brain
and liver tissues.
Immunofuorescence-based protein detection was described in 1941.[20] Until then,
only radioactive-based techniques, which took long exposure times and low resolv-
ing power, were used. One of the frst attempts was the indirect immunofuorescence
procedure using an antiserum against a poly(rA)-poly(dT). This approach was used
to detect RNA-DNA hybrids of Drosophila chromosomes with a rhodamine-conju-
gated secondary antibody.[21] In the early 1980s, Bauman and colleagues[22] devel-
oped the direct detections of Trypanosoma nucleic acids, adenovirus in human tissue
cells, and Polytene chromosomes from the salivary gland of Drosophila. The latter
became possible by using the covalent binding of the fuorochrome rhodamine mol-
ecule to the 3′-terminus of the RNA. The year 1981 was a landmark for molecular
cytogenetics due to the introduction of nonradioactive labeling methods. The use of
fuorescently labeled probes, such as biotin-labeled nucleotides with avidin-based
detection methods, was crucial to improving in situ hybridization technologies.[23]
The advent of FISH-based techniques boosted the diagnosis and study of leuke-
mia. The application of new multicolor karyotyping approaches allowed the com-
plete characterization of complex chromosome rearrangements and the discovery of
new abnormalities and putative genes involved in leukemic transformation.[16]
Therefore, the advent of FISH in the late ’80s paved the way to the second revo-
lution in cytogenetic analysis. Initially, the use of specifc chromosome probes led
to the exact identifcation of chromosomes involved in complex rearrangements.
Moreover, the Human Genome Project prompted the development of several locus-
specifc probes. This technology improved gene mapping strategies and breakpoints
FISH—in Leukemia Diagnostics 75
CHRONIC LEUKEMIA
CHRONIC MYELOID LEUKEMIA
CML is an extensively described malignant disorder of hematopoietic stem cells
(HSCs) that accounts for 15–20% of all adult leukemia cases and about 3% in chil-
dren.[25, 26] This disease is a chronic myeloproliferative neoplasm originating in a
defective pluripotent BM stem cell. The hallmark of this disease is the presence of
the Philadelphia (Ph) chromosome, translocation t(9;22)(q34;q11.2), generating the
BCR‑ABL1 fusion.[8, 27] Pediatric and adult CML share the same molecular features.
However, better outcomes have been observed in pediatric transplant patients due to
the lack of other chronic diseases or organ dysfunction associated with increased age
and lower incidence of graft versus host disease.[26]
ACUTE LEUKEMIA
ALs are a heterogeneous group of diseases that may originate from the interaction
between genetic and epigenetic alterations in hematopoietic progenitors, leading to
dysregulation of multiple important signal transduction pathways, resulting in hema-
topoietic insuffciency due to a clonal proliferation of immature precursors.[34] These
neoplastic precursor cells originate in the BM and can infltrate PB and tissues.[35]
ALs are divided into two main groups, myeloid leukemia, when the myeloblast
lineage is affected, or lymphoid leukemia when the lymphoblastic lineage is affected.
AL groups differ from each other in specifc characteristics regarding cytogenetic
and genomic abnormalities, morphological and immunophenotypic profle, response
to therapy, and the clinical course of the disease. In general, ALs present a rapid fast
clinical course and if not properly treated can lead to death in a few months or even
weeks.[36, 37] AL is the most common form of childhood cancer, and its incidence is
increasing.[38]
90% of pediatric patients with AML have at least one recurrent type I and type II
mutation.[42]
In this sense, molecular cytogenetic analyses have conferred substantial evidence
with regards to the chromosomal architectures in AML and other cancers. Most
importantly, the FISH technique that plays a leading role in diagnostic pathology for
its single-cell analysis has provided crucial information regarding the heterogeneity
of AML malignant cells.[14]
In AML, cytogenetic alterations of numerical and structural types can be found.
Numerical alterations include monosomies and nullisomies, which result in a karyo-
type with less than 46 chromosomes. Trisomies and tetrasomies, among others,
will generate a hyperdiploid karyotype with more than 46 chromosomes. Structural
abnormalities are mainly translocations, insertions, and inversions.[40]
The presence of specifc cytogenetic abnormalities has important prognostic
implications for the disease. Thus, karyotypic alterations in leukemic blasts are con-
sidered one of the most signifcant markers that defne the biology of AML. In addi-
tion to karyotypic changes, gene mutations with clinical relevance associated with
the disease are also part of the classifcation scheme for AML.[5, 39, 40, 43]
COMPLEX K ARYOTYPES
A complex karyotype (CC) is defned as any karyotype with at least three chromosomal
aberrations, regardless of the type of alterations and the chromosomes involved.[94–96]
The frequency of CCs increases with the age of the patients.[41, 55, 60, 97] Presence of CCs
in the BM blast cells represents a possible indicator of a poor prognosis, which may
negatively infuence the patient’s clinical evolution. The literature has also shown that,
in most cases, the disease has been resistant to the initial treatment, with early relapse
of patients.[97–99] CCs have been associated with an unfavorable clinical evolution, and
it is important to emphasize that these karyotypes can carry, among their alterations,
in a cryptic way, chromosomal abnormalities that can be responsible for an adverse
prognosis.[61, 100] Since the literature suggests a possible correlation between the com-
bination of aberrations detected in a CC and the clinical response, the importance of
refnement in the characterization of this karyotypic profle becomes clear.
SAMPLES/TISSUES
For (molecular) cytogenetic analysis, PB and BM samples are processed. The sam-
ples are collected in test tubes with anticoagulant heparin and referred to the labora-
tory by the responsible physicians.
84 Cytogenetics and Molecular Cytogenetics
To assist in the risk stratifcation of patients and in the experimental design based
on FISH panels, clinical diagnoses of cytochemistry, immunophenotyping, and mor-
phology are obtained through the study of medical records, slide review, and patient
fles provided by the responsible physicians in the respective institutions of origin.
The clinical-laboratory data of the diagnosis, as well as the clinical information of
the follow-up of the patients, are also obtained through the study of the medical
records in the institutions of origin.
DESCRIPTION OF METHODS
CELL CULTURE FOR CHROMOSOME PREPARATION
Equipment/Materials
• Class II biological safety cabinet
• Sterile 15ml conic tubes
• Centrifuge
• Sterile transfer pipettes
• Micropipette 50µl (p20–200–1000)
• Pipette tips 50µl (p20–200–1000)
• Neubauer chamber
• Coverslip 24×24mm
• Optic microscope
• Cell culture fasks
• Humidifed 37°C, 5% CO2 incubator
Reagents
• Ready to use growth medium: RPMI 1640 medium at 37ºC
• Fetal bovine serum, previously inactivated in a water bath at 56ºC for
40 minutes, 12ml of penicillin & streptomycin
• L-glutamine (add in RPMI 1640 medium—1:100 proportion)
• Thomas buffer solution: Dilute 10ml of acetic acid and 10 drops of Giemsa
stain in 1l of distilled water. Store in a dark bottle.[105]
Workfow
1. Into the biological safety cabin, transfer the sample (BM/PB) for a 15ml
conic tube;
2. Dilute 50µl of the sample in 1ml Thomas buffer solution using a micropipette;
3. Apply this solution to the surface of the Neubauer chamber and place the
coverslip;
4. Use the optic microscope (20×) to count the cells in the four chamber’s
quadrants;
5. The number of cells is obtained by adding the number of counted cells,
multiplying the result for 21 and afterwards for 2,500;
6. Culture is established using 80% growth medium, 20% fetal bovine serum,
and 5×106 cells of the sample in each cell culture fask;
FISH—in Leukemia Diagnostics 85
7. Cell culture fasks should stay in the humidifed 37°C incubator for 24
hours.
Reagents
• Colcemid solution 10µg/ml
• KCl (potassium chloride): Add 0.6g of KCl to 100ml of distilled water. Mix
by swirling until dissolved.
• Carnoy’s solution/fxative solution (acetic acid and methanol solution—1:3
proportion); combine three parts of methanol to one part of glacial acetic
acid (v/v). Use freshly. Store at room temperature (RT).[106]
Workfow
1. After 23 hours of incubation, 50µl of Colcemid solution should be added
to each of the cell culture fasks;
2. Return the fasks to the humidifed 37°C incubator;
3. One hour later (after completing 24 hours total), transfer the cell culture
in the fasks to 15ml conic tubes. The contents in each fask shall be trans-
ferred to a different conic tube;
4. Centrifuge the material for 6 minutes at 1,500 rpm;
5. Remove the supernatant from the tubes and homogenize the pellet. To
perform the hypotonic shock, add, with the aid of a disposable Pasteur
pipette, the KCl solution until completing 8 ml;
6. Accommodate the tubes in a water bath at 37°C for 20 minutes;
7. Centrifuge for 6 minutes at 1,500rpm;
8. Remove the supernatant from the tubes, homogenize the pellet, and slowly
add the Carnoy’s solution until completing 8ml;
9. Leave the tubes for 20 minutes at RT;
10. Centrifuge for 6 minutes at 1,500rpm;
11. Remove the supernatant, homogenize the pellet, unite the contents of all
the tubes in a single one and add Carnoy’s solution until completing 8ml;
12. Repeat steps 7 and 8 until the material is clear;
13. Store the material at 4°C.
Reagents
• Ethanol 70%
• Giemsa solution (Merck): Dilute 2.5ml of Giemsa stain and 40ml of phos-
phate buffer.[107, 108]
• Phosphate buffer: Dissolve 6.808g of KH2PO4 and 0.882g of NaOH in 1l of
distilled water. Adjust pH to 6.8.
Workfow
1. Centrifuge the tubes containing the chromosome preparation for 6minutes
at 1,500 rpm;
2. Remove the supernatant (until it is twice as large as the pellet) and homog-
enize the content left in the tube;
3. Clean the slides using ethanol 70%;
4. Puff on the cleaned slides;
5. Add three drops from the material onto the slides. Before adding mate-
rial, keep the pipette at arm’s length from the slides for better chromosome
dispersion;
6. Quickly fame the opposite part of the slides over medium heat in a Bunsen
burner;
7. Add the slides in a Coplin jar with Giemsa solution for 12 minutes;
8. Observe the slides under an optical microscope (in phase contrast) to check
for the presence of metaphases;
9. After confrming the presence of metaphases, follow steps 4–6 again to each
new slide, and then store the slides in a microscope slide case for aging.
G-BANDING PROTOCOL
Equipment/Materials
• Spatula
• Precision balance
• Beaker 100ml
• Laboratory water bath
• Disposable Pasteur pipettes
• Glass Coplin jar
Reagents
• Dulbecco’s Phosphate Buffered Saline (PBS): Dilute 8.0 g of NaCl, 0.2 g of
KCl, 0.2 g of KH2PO4, and 1.44g of Na2HPO4H20 in 1l of distilled water.
Adjust pH to 7.8.
• Trypsin solution: Dilute 0.1g of Trypsin from porcine pancreas in 100ml of
Dulbecco’s PBS
FISH—in Leukemia Diagnostics 87
Workfow
1. Heat the trypsin solution (0.1g of trypsin and 100ml of Dulbecco PBS) in
the water bath for 30 minutes;
2. Dip the slide in the trypsin solution for 1–10 seconds;
3. Wash the slide in saline solution;
4. Stain the slide in Giemsa solution for 15 minutes;
5. Wash the slide in saline solution and let it dry at RT;
6. Under the microscope, observe whether the time of exposure to trypsin
was suffcient to generate the chromosomal banding pattern; if the chro-
mosomes are digested too much, decrease the exposure time. If the time is
not enough for the chromosomes to be banded, increase the time until the
desired result;
7. Analyze at least 20 metaphases to confrm the cytogenetic diagnosis.
Reagents
• Ethanol 70%, 85%, and 100%; prepare v/v dilutions of 100% ethanol with
distilled water to make stock solutions of 70% and 85% ethanol. Between
uses, store covered at ambient temperature. Discard stock solutions after
six months.
• 2×SSC (pH 7.0): Dilute stock solution (20×SSC) in distilled water in the
proportion 1:10. Adjust pH for 7. 20×SSC (stock solution): Dilute 175.34g
of NaCl and 88.24g of C6H5Na3O7.2H2O in 500ml of distilled water on
shaker. Add distilled water to bring fnal volume of solution to 1l. Adjust
pH for 5.3.
• Pepsin 0.005%, HCl 0.001M: Dilute 400µl pepsin 0.5% in 39.6ml HCl
0.01M; pepsin 0.5% HCl 0.001M (Stock solution): Dilute 0.5 g pepsin in
100ml HCl 0.01M. Keep in refrigerator.
• 1×PBS: Dilute stock solution (10×PBS) in distilled water the proportion
1:10. Adjust pH for 7.2. 10×PBS (stock solution): Dilute 80g of NaCl, 2g
88 Cytogenetics and Molecular Cytogenetics
Workfow
1. Drop the cell suspension in slides washed in ethanol 70% and leave for dry-
ing in a wet plate for 2–3 minutes. It is recommended to observe the dropped
slides in the optic microscope to pre-select target areas to apply the probes;
2. Incubate the slides in a 2×SSC solution (pH 7.0) at 37°C for 20 minutes;
3. Incubate the slides in pepsin solution (0.005%, 0.001M HCl) at 37ºC for 10
minutes;
4. Wash the slides for 3 minutes in 1×PBS at RT;
5. Incubate the slides in a formaldehyde solution (1% 1×PBS) at RT for 10
minutes;
6. Wash the slides in 1×PBS at RT for 3 minutes;
7. Dehydrate slides in a series of ethanol (70%, 85%, and 100%; 2 minutes
each);
8. Leave the slides for drying at RT.
Reagents
• Pre-selected FISH probe
• Hybridization buffer
• MiliQ (purifed) water
Workfow
Slides Denaturation
1. Add 8µl of the probe hybridization mix (according to the manufactur-
er’s instructions) in the target slides’ areas and cover with the coverslip
20×20mm;
2. Seal the edges of the coverslips with the rubber cement glue;
3. Accommodate the slides in the dry bath at 75ºC for 7 minutes.
FISH—in Leukemia Diagnostics 89
Hybridization
1. Incubate the slides into a humidifed microscope slides case (keep the case
closed to simulate a dark chamber environment). Leave the case in a CO2
incubator at 37ºC overnight (12–16 hours).[107, 109]
Reagents
• 0.4×SSC/3%Tween20 solution (pH=7.0); Dilute 10ml of 20×SSC solution in
distilled water in the proportion 1:10. Adjust pH for 7; keep in refrigerator;
fnally dilute 120µl Tween20 in 40ml 0.4×SSC.
• 2×SSC/0.1%Tween20 solution (pH=7.0): Dilute 40µl Tween20 in 40ml
2×SSC.[109]
• DAPI antifade staining
Workfow
1. Remove the coverslips and wash the slides for 2 minutes in 0.4×SSC, 3%
Tween20 solution (pH=7.0) at 72ºC;
2. Transfer the slides to a 2×SSC 0.1% Tween20 solution (pH=7.0) for 1 minute
at RT;
3. Let the slides dry at RT;
4. Apply 15µl antifade staining e.g. DAPI (Vysis) over the hybridization area
and cover with a coverslip 24×32mm;
5. Incubate the slides into a humidifed microscope slides case (keep the case
closed to simulate a dark chamber environment) for at least 10 minutes;
6. Observe the slides in a fuorescent microscope to detect the FISH signals.
YIELDS
FISH PANEL AND PROBES IN LEUKEMIA DIAGNOSTICS
The WHO recommends an integrated approach for ALs cytogenetic diagnosis.
Chromosomal banding cytogenetic analysis and FISH-based techniques should be
used in the initial diagnosis and monitoring of clonal abnormalities.[5, 101, 102]
The application of a FISH panel enables a quick assessment of the karyotype and
the pattern of abnormalities present in it, including its percentage in BM blast cells.
90 Cytogenetics and Molecular Cytogenetics
In this sense, for hematological emergencies such as APL, a rapid FISH result is
mandatory for the administration of all-trans retinoic acid (ATRA).[101, 110] For cryptic
rearrangements that escape detection by chromosome banding, such as translocation
t(12;21)(p13;q22)/ETV6‑RUNX1, translocation t(10;11)(p12;q23)/KMT2A‑MLLT10,
translocation t(6;11)(q27;q23)/KMT2A‑AFDN, and abnormalities in the 17p13 (TP53)
region, FISH can be considered as a stand-alone diagnostic assay.[42, 101, 103] The FISH
probes and disease-specifc panels most commonly used in clinical cytogenetic labo-
ratories are listed in Table 7.1.
TABLE 7.1
FISH Panel and Probes in Leukemia Diagnostics
Cytogenetic Gene Fusion Prognosis Disease Types Probe Types
Abnormality
t(8;21)(q22;q22) RUNX1‑RUNX1T1 favorable AML DCDF
inv(16)(p13;q22) CBFb‑MYH11 favorable AML DCDF
t(15;17) PML‑RARA favorable AML DCDF, DCSF
(q22;q21)
t(9;11)(p22;q23) KMT2A‑MLLT3 favorable in AML ALL, AML DCDF
patients without
additional
abnormalities or
FAB M5 subtype)
11q23 KMT2A-r intermediate ALL, AML DCBAP
or poor in AML,
Unfavorable in
ALL
del(5q) EGR1(5q31) unfavorable AML DCE
t(6;9)(p23;q34) DEK‑NUP214 often unfavorable AML DCDF
t(6;11)(q27;q23) KMT2A‑AFDN unfavorable in ALL, AML DCDF
AML
t(11;19) KMT2A‑MLLT1 unfavorable in ALL, AML DCDF
(q23;p13.3) AML
t(10;11)(p12;q23) KMT2A‑MLLT10 unfavorable in AML ALL, AML DCDF
t(1;22)(p13;q13) RBM15‑MKL1 unfavorable AMKL DCDF
inv(3) MECOM (3q26) unfavorable AML DCBAP
7q (RELN/ORC5) unfavorable AML, T-ALL DCE, DCBAP
7q22.1-q22.2/
TES(7q31.2)/TCRB(7q34)
t(9;22)(q43;q11) BCR‑ABL1 unfavorable B-ALL, T-ALL, DCDF
AML, CML
t(12;21) ETV6‑RUNX1 favorable B-ALL DCDF
(p13;q22)
+4/+10/+17 D4Z1(4cen),D10Z1 favorable B-ALL TCE
(10cen),D17Z1(17cen)
(Continued)
FISH—in Leukemia Diagnostics 91
TABLE 7.1 (Continued)
FISH Panel and Probes in Leukemia Diagnostics
Cytogenetic Gene Fusion Prognosis Disease Types Probe Types
Abnormality
t(11;14) TCRA/D(14q11) favorable T-ALL DCBAP
(p13;q11)
t(1;19)(q23;p13) PBX1‑E2A controversial, B-ALL DCDF
improved with
therapy
t(4;11)(q21;q23) KMT2A‑AFF1 unfavorable ALL DCDF
iAMP21 RUNX1(21q22) unfavorable B-ALL DCDF, DCE
6q MYB(6q23),D6Z1(6cen) unfavorable ALL DCE
del(9p) CDKN2A(9p21), D9Z3 unfavorable Pre-B-ALL, DCE, DCBAP
(9 cen), PAX5(9p13.2) T-ALL
t(8;14)(q24;q32) cMYC(8q24), favorable Burkitt DCBAP,
IGH(14q32) lymphoma/ DCDF, DCE
leukemia
(BL/L)
del(13q)/+12 DLEU1(13q14), favorable CLL TCE
D13S25 (13q34),
D12Z3 (12 cen)
del(11q)/ ATM(11q22), unfavorable CLL DCE
del(17p) TP53(17p13)
CBFb(16q22)-MYH11(16p13) Probe
Application of FISH-probe as shown in Figure 7.1B is important to stratify the
patient’s risk since inversion inv(16) is related to a good prognosis, with well-defned
treatment protocol guidelines.[5, 40, 68]
PML(15q24)-RARA(17q21) Probe
FISH for rapid detection of PML‑RARA fusion (Figure 7.1C) is mandatory in APL
characterization since a prompt diagnosis and initiation of treatment are critical to
lowering the risk of early mortality.[110]
92 Cytogenetics and Molecular Cytogenetics
KMT2A(11q23)-MLLT3(9p22) Probe
With a combination of FISH probes, it can be possible to delineate the patient’s
karyotypic alterations, confrming, in this case (Figure 7.2A), the recurrent translo-
cation t(9;11) and a trisomy of chromosome 9 with an extra KMT2A‑MLLT3 fusion
gene.[11, 77, 112]
cMYC(8q24)-IGH(14q32) Probe
The combination of IGH‑MYC with a probe for centromere 8 is essential to dis-
close the recurrent translocation t(8;14), enabling rapid detection and diagnosis of
BL (Figure 7.2B). BL is an aggressive disease with a fast clinical course. However,
FISH—in Leukemia Diagnostics 93
FIGURE 7.2 A) Experiment with FISH in interphase nuclei for the translocation t(9;11)
(p22;q23)— 1) Two normal KMT2A gene signals (yellow),
2) One standard KMT2A (yellow) and KMT2A (red and green) split signals with an extra
KMT2A (red) signal—KMT2A dual-color break-apart probe (DCBAP).
3) Standard KMT2A (red) and MLLT3 (green) signals, besides three copies of KMT2A‑
MLLT3 fusion (yellow)—KMT2A-MLLT3 dual color dual fusion.
B) MYC-directed FISH using
1) assay with dual color dual fusion plus centromeric probe (CEP) characterizing the translo-
cation t(8;14)(q24;q32)—IGH (green) cMYC (red) CEP8 (blue);
2) MYC DCBAP showing a cMYC gene rearrangement.
C) FISH results for translocation t(11;14)(p13;q11)/TCRA/D—dual-color break-apart probe in
interphase nuclei showing a TCRA/D gene rearrangement.
D) FISH test with
1) dual color dual fusion on metaphase spread for translocation t(12;21) (p13;q22)—ETV6
(red) RUNX1 (green) and ETV6‑RUNX1 fusion gene (yellow);
2) Flexibility of ETV6-RUNX1 DCDF in interphase nuclei for iAMP21 monitoring—
two ETV6 (red) normal signals and RUNX1 (green) multiple signals revealing the cryptic
iAMP21; G-banding showed normal karyotype (result not shown).
94 Cytogenetics and Molecular Cytogenetics
excellent clinical outcomes can be achieved provided that it is early diagnosed and
properly treated.[71, 74]
TCRA/D(14q11) Probe
Translocation t(11;14)(p13;q11) is associated with an excellent prognosis, so its pre-
cise characterization is fundamental for diagnosis and therapy (Figure 7.2C). The
translocation t(11;14)(p13;q11) and translocation t(7;11)(q35;p13)/TCRB are variant
translocations of each other.[41, 113]
ETV6(12p13)-RUNX1(21q22) Probe
The translocation t(12;21) is subtle to the G-banding technique and is associated
with a favorable prognosis once adequate treatment is administered.[78, 114] On the
other hand, iAMP21 is considered to confer a poor prognosis, including a high risk
of relapse. In this example (Figure 7.2D), the ETV6‑RUNX1 probe can be used for a
prompt diagnosis of this hematological urgency. Besides, the same probe could help
detect the cryptic iAMP21 even with the G-banding karyotype being apparently
normal.[104]
BCR(22q11)-ABL1(9q34) Probe
The application of a BCR‑ABL1 probe is important to stratify the patient’s risk indi-
cating targeted therapy against BCR‑ABL1 fusion (Figure 7.3B) by treatment with
tyrosine kinase inhibitors.[5, 101]
KMT2A(11q23)(MLL) Probe
KMT2A—specifc probe (11q23) is essential to disclose KMT2A-r, a group of aber-
rations mainly associated with infant leukemia and related to intermediate/adverse
prognoses.[51] In this example (Figure 7.3C), the probe helped identify a cryptic inser-
tion in a translocation t(10;11), with chromosomal material between the green and
red signals.[61]
CONCLUSIONS
Hematological neoplasms are classifed according to their differences in clinical and
biological characteristics. Thus, acknowledging the association between specifc
cytogenetic abnormalities and clinical-morphological features is fundamental.[121]
In this sense, FISH-based techniques can be applied to metaphase chromosomes
96 Cytogenetics and Molecular Cytogenetics
REFERENCES
1. Greaves, M.F. Biological models for leukaemia and lymphoma. IARC Sci. Publ. 2004,
157, 351–372.
2. Brown, C.M.S.; Larsen, S.R.; Iland, H.J.; Joshua, D.E.; Gibson, J. Leukaemias into the
21st century: Part 1: The acute leukaemias. Intern. Med. J. 2012, 42(11), 1179–1186.
3. Bloomfeld, C.D., Foon, K.A., Levine, E.G. Leukemias. In: Medical Oncology; Calabresi,
P., Schein, P.S., Eds. McGraw Hill, Inc, New York, NY, 1993, pp. 459–501.
4. Juliusson, G.; Hough, R. Leukemia. In: Tumors in Adolescents and Young Adults; Stark,
D.P., Vassal, G., Eds. Prog. Tumor. Res., Karger, Basel, 2016, pp. 43, 87–100.
5. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.;
Bloomfeld, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health
Organization classifcation of myeloid neoplasms and acute leukemia. Blood. 2016, 127,
2391–2405.
6. Silva, M.L.M.; Land, M. Anomalias Citogenéticas e Moleculares nas Leucemias da
Infância. In: Doenças genéticas em pediatria; Carakushansky, G., Ed. Guanabara
Koogan S.A., Rio de Janeiro, RJ, 2001, pp. 36, 350–362.
7. Wan, T.S.K. Cancer cytogenetics: Methodology revisited. Ann. Lab. Med. 2014, 34(6),
413–425.
8. Cortes, J.; Pavlovsky, C.; Saußele, S. Chronic myeloid leukaemia. Lancet Sem. 2021,
398, 1914–1926.
9. Udayakumar, A.M.; Alkindi, S.; Pathare, A.V.; Raeburn, J.A. Complex t(8;13;21)
(q22;q14;q22)-A novel variant of t(8;21) in a patient with acute myeloid leukemia (AML-
M2). Arch. Med. Res. 2008, 39, 252–256.
10. Mangan, J.K.; Speck, N.A. RUNX1 mutations in clonal myeloid disorders: From conven-
tional cytogenetics to next generation sequencing, a story 40 years in the making. Crit.
Rev. Oncog. 2011, 16, 77–91.
11. Braoudaki, M.; Tzortzatou-Stathopoulou, F. Clinical cytogenetics in pediatric acute leu-
kemia: An update. Clin. Lymph., Myel. Leuk. 2012, 12, 230–237.
12. Bint, S.; Davies, A.; Ogilvie, C. Multicolor banding remains an important adjunct to
array CGH and conventional karyotyping. Mol. Cytogenet. 2013, 6, 55.
FISH—in Leukemia Diagnostics 97
13. Bochtler, T.; Stölzel, F.; Heilig, C.E.; Kunz, C.; Mohr, B.; Jauch, A.; Janssen, J.W.;
Kramer, M.; Benner, A.; Bornhäuser, M.; Ho, A.D.; Ehninger, G.; Schaich, M.; Krämer,
A. Clonal heterogeneity as detected by metaphase karyotyping is an indicator of poor
prognosis in acute myeloid leukemia. J. Clin. Oncol. 2013, 31, 3898–3905.
14. Das, K.; Tan, P. Molecular cytogenetics: Recent developments and applications in can-
cer. Clin. Genet. 2013, 84, 315–325.
15. Gozzetti, A.; Le Beau, M.M. Fluorescence in situ hybridization: Uses and limitations.
Semin. Hematol. 2000, 37, 320–333.
16. Wan, T.S.K. Cancer cytogenetics: An introduction. In: Cancer Cytogenetics Methods
and Protocols; Wan, T.S.K. Ed. Humana Press, New York, NY, 2017, 5–7.
17. Gall, G.; Pardue, M.L. Formation and detection of RNA-DNA hybrid molecules in cyto-
genetical preparations. Proc. Natl. Acad. Sci. USA. 1969, 63, 378–381.
18. John, H.A.; Birnstiel, M.L.; Jones, K.W. RNA-DNA hybrids at the cytological level.
Nature. 1969, 223, 582–587.
19. Buongiorno-Nardelli, M.; Amaldi, F. Autoradiographic detection of molecular hybrids
between rRNA and DNA in tissue sections. Nature. 1970, 225, 946–948.
20. Coons, A.H.; Creech, H.J.; Jones, R.N. Immunological properties of an antibody con-
taining a fuorescent group. Proc. Soc. Exp. Biol. Med. 1941, 47, 200–202.
21. Rudkin, G.T.; Stollar, B.D. High resolution detection of DNA: RNA hybrids in situ by
indirect immunofuorescence. Nature. 1977, 265, 472–473.
22. Bauman, J.G.J.; Wiegant, J.; Borst, P.; Van Duijn, P. A new method for fuorescence
microscopical localization of specifc DNA sequences by in situ hybridization of fuoro-
chrome-labelled RNA. Experimental Cell Research. 1980, 128, 485–490.
23. Langer, P.R.; Waldrop, A.A.; Ward, D.C. Enzymatic synthesis of biotin-labeled poly-
nucleotides: Novel nucleic acid affnity probes. Proc. Natl. Acad. Sci USA. 1981, 78,
6633–6637.
24. Wolff, D.J.; Bagg, A.; Cooley, L.D.; Dewald, G.W.; Hirsch, B.A.; Jacky, P.B.; Rao, K.W.;
Rao, P.N.; Association for Molecular Pathology Clinical Practice Committee; American
College of Medical Genetics Laboratory Quality Assurance Committee. Guidance for
fuorescence in situ hybridization testing in hematologic disorders. J. Mol. Diagn. 2007,
9, 134–143.
25. Flis, S.; Chojnacki, T. Chronic myelogenous leukemia, a still unsolved problem: Pitfalls
and new therapeutic possibilities. Drug Des. Devel. Ther. 2019, 8, 825–843.
26. Andolina, J.R.; Neudorf, S.M.; Corey, S.J. How I treat childhood CML. Blood. 2012,
119, 1821–1830.
27. Khemka, R.; Gupta, M.; Jena, N.K. CML with megakaryocytic blast crisis: Report of 3
cases. Pathol. Oncol. Res. 2018, 25, 1253–1258.
28. Rozman, C.; Montserrat, E. Chronic lymphocytic leukemia. N. Engl. J. Med. 1995, 333,
1052–1057.
29. Kikushige, Y.; Ishikawa, F.; Miyamoto, T.; Shima, T.; Urata, S.; Yoshimoto, G.; Mori,
Y.; Iino, T.; Yamauchi, T.; Eto, T.; Niiro, H.; Iwasaki, H.; Takenaka, K.; Akashi, K. Self-
renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic
lymphocytic leukemia. Cancer Cell. 2011, 20, 246–259.
30. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.;
Rattan, S.; Keating, M.; Rai, K.; Rassenti, L.; Kipps, T.; Negrini, M.; Bullrich, F.;
Croce, C.M. Frequent deletions and down-regulation of micro- RNA genes miR15 and
miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA. 2002, 99,
15524–15529.
31. Klein, U.; Lia, M.; Crespo, M.; Siegel, R.; Shen, Q.; Mo, T.; Ambesi-Impiombato, A.;
Califano, A.; Migliazza, A.; Bhagat, G.; Dalla-Favera, R. The DLEU2/miR-15a/16-1
98 Cytogenetics and Molecular Cytogenetics
cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leuke-
mia. Cancer Cell. 2010, 17, 28–40.
32. Onida, F.; Kantarjian, H.M.; Smith, T.L.; Ball, G.; Keating, M.J.; Estey, E.H.; Glassman,
A.B.; Albitar, M.; Kwari, M.I.; Beran, M. Prognostic factors and scoring systems in
chronic myelomonocytic leukemia: A retrospective analysis of 213 patients. Blood.
2002, 99, 840–849.
33. Such, E.; Cervera, J.; Costa, D.; Sole, F.; Vallespí, T.; Luño, E.; Collado, R.; Calasanz,
M.J.; Hernández-Rivas, J.M.; Cigudosa, J.C.; Nomdedeu, B.; Mallo, M.; Carbonell,
F.; Bueno, J.; Ardanaz, M.T.; Ramos, F.; Tormo, M.; Sancho-Tello, R.; del Cañizo,
C.; Gómez, V.; Marco, V.; Xicoy, B.; Bonanad, S.; Pedro, C.; Bernal, T.; Sanz, G.F.
Cytogenetic risk stratifcation in chronic myelomonocytic leukemia. Haematologica.
2011, 96, 375–383.
34. Ross, J.A.; Johnson, K.J.; Spector, G.L.; Kersey, J.H. Epidemiology of acute childhood
leukemia. In: Childhood Leukemia: A Practical Handbook; Reaman, G.H.; Smith, F.O.,
Eds. Springer, Berlin, Heidelberg, 2011, pp. 3–21.
35. Gilliland, D.; Jordan, C.; Felix, C. The molecular basis of leukemia. Am. Soc. Hematol.
2004, 80–97.
36. Lo-Coco, F.; Ammatuna, E. The biology of acute promyelocytic leukemia and its impact
on diagnosis and treatment. Am. Soc. Hematol. 2006, 514, 156–161.
37. Arceci, R., Aplenc, R. Acute myelogenous leukemia in children. In: Wintrobe’s Clinical
Hematology; Greer, J.P. Ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2009,
pp. 1919–1932.
38. Bolouri, H.; Farrar, J.E.; Triche, T.Jr.; Ries, R.E.; Lim, E.L.; Alonzo, T.A.; Ma, Y.;
Moore, R.; Mungall, A.J.; Marra, M.A.; Zhang, J.; Ma, X.; Liu, Y.; Liu, Y.; Auvil, J.M.G.;
Davidsen, T.M.; Gesuwan, P.; Hermida, L.C.; Salhia, B.; Capone, S.; Ramsingh, G.;
Zwaan, C.M.; Noort, S.; Piccolo, S.R.; Kolb, E.A.; Gamis, A.S.; Smith, M.A.; Gerhard,
D.S.; Meshinchi, S. The molecular landscape of pediatric acute myeloid leukemia reveals
recurrent structural alterations and age-specifc mutational interactions. Nat. Med. 2018,
24, 103–112.
39. Raimondi, S.; Chang, M.N.; Ravindranath, Y.; Behm, F.G.; Gresik, M.V.; Steuber, C.P.;
Weinstein, H.J.; Carroll, A.J. Chromosomal abnormalities in 478 children with acute
myeloid leukemia: Clinical characteristics and treatment outcome in a cooperative
Pediatric Oncology Group Study: POG 8821. Blood. 1999, 94, 3707–3716.
40. Manola, K. Cytogenetics of pediatric acute myeloid leukemia. Europ J Haematol. 2009,
83, 391–405.
41. Mrozek, K.; Heerema, N.; Bloomfeld, C. Cytogenetics in acute leukemia. Blood
Reviews. 2004, 18, 115–134.
42. Pui, C.H.; Carroll, W.L.; Meshinchi, S.; Arceci, R.J. Biology, risk stratifcation, and
therapy of pediatric acute leukemias: An update. J Clin Oncol. 2011, 29, 551–565.
43. Arceci, R., Meshinchi, S. Biology of acute myeloid Leukemia. In: Childhood Leukemia:
A Practical Handbook; Reaman, G.H.; Smith, F.O., Eds. Springer, Berlin, Heidelberg,
2011, pp. 63–74.
44. Creutzig, U.; Zimmermann, M.; Ritter, J.; Reinhardt, D.; Hermann, J.; Henze, G.;
Jürgens, H.; Kabisch, H.; Reiter, A.; Riehm, H.; Gadner, H.; Schellong, G. Treatment
strategy and long term results in pediatric patients treated in four consecutive AML-
BFM trials. Leukemia. 2005, 19, 2030–2042.
45. Gamerdinger, U.; Teigler-Schlegel, A.; Pils, S.; Bruch, J.; Viehmann, S.; Keller, M.;
Jauch, A.; Harbott, J. Cryptic chromosomal aberrations leading to an AML1/ETO rear-
rangement are frequently caused by small insertions. Genes Chromosomes Cancer.
2003, 36, 261–272.
FISH—in Leukemia Diagnostics 99
46. Creutzig, U.; Kutny, M.A.; Barr, R.; Schlenk, R.F.; Ribeiro, R.C. Acute myelogenous
leukemia in adolescents and young adults. Pediatr. Blood Cancer. 2018, 65, e27089.
47. De Figueiredo, A.F.; Liehr, T.; Bath, S.; Binato, R.; De Souza, M.T.; De Matos, R.R.;
Salles, T.de, J.; Jordy, F.C.; Ribeiro, R.C.; Abdelhay, E.; Silva, M.L. A complex karyo-
type masked a cryptic variant t(8;21)(q22;q22) in a child with acute myeloid leukemia.
Leuk. Lymphoma. 2011, 52, 1593–1596.
48. Silva, M.L.M.; Raimondi, S.C.; Abdelhay, E.; Gross, M.; Mkrtchyan, H.; de Figueiredo,
A.F.; Ribeiro, R.C.; de Jesus Marques-Salles, T.; Sobral, E.S.; Gerardin Land, M.P.;
Liehr, T. Banding and molecular cytogenetic studies detected a CBFB-MYH11 fusion
gene that appeared as abnormal chromosomes 1 and 16 in a baby with acute myeloid
leukemia FAB M4-Eo. Cancer Genet. Cytogenet. 2008, 182, 56–60.
49. Nakase, K.; Wakita, Y.; Minamikawa, K.; Yamaguchi, T.; Shiku, H. Acute promyelo-
cytic leukemia with del(6)(p23). Leuk. Res. 2000, 24, 79–81.
50. Matos, R.R.C.; Mkrtchyan, H.; Amaral, B.A.S.; Liehr, T.; de Souza, M.T.; Ney-Garcia,
D.R.; Santos, N.; Marques-Salles, T.J.; Ribeiro, R.C.; Figueiredo, A.F.; Silva, M.L. An
unusual cytogenetic rearrangement originating from two different abnormalities in
chromosome 6 in a child with acute promyelocytic leukemia. Acta Haematol. 2013,
130, 23–26.
51. Meyer, C.; Burmeister, T.; Gröger, D.; Tsaur, G.; Fechina, L.; Renneville, A.; Sutton, R.;
Venn, N.C.; Emerenciano, M.; Pombo-de-Oliveira, M.S.; Barbieri Blunck, C.; Almeida
Lopes, B.; Zuna, J.; Trka, J.; Ballerini, P.; Lapillonne, H.; De Braekeleer, M.; Cazzaniga,
G.; Corral Abascal, L.; van der Velden, V.H.J.; Delabesse, E.; Park, T.S.; Oh, S.H.;
Silva, M.L.M.; Lund-Aho, T.; Juvonen, V.; Moore, A.S.; Heidenreich, O.; Vormoor,
J.; Zerkalenkova, E.; Olshanskaya, Y.; Bueno, C.; Menendez, P.; Teigler-Schlegel,
A.; Zur Stadt, U.; Lentes, J.; Göhring, G.; Kustanovich, A.; Aleinikova, O.; Schäfer,
B.W.; Kubetzko, S.; Madsen, H.O.; Gruhn, B.; Duarte, X.; Gameiro, P.; Lippert, E.;
Bidet, A.; Cayuela, J.M.; Clappier, E.; Alonso, C.N.; Zwaan, C.M.; van den Heuvel-
Eibrink, M.M.; Izraeli, S.; Trakhtenbrot, L.; Archer, P.; Hancock, J.; Möricke, A.;
Alten, J.; Schrappe, M.; Stanulla, M.; Strehl, S.; Attarbaschi, A.; Dworzak, M.; Haas,
O.A.; Panzer-Grümayer, R.; Sedék, L.; Szczepański, T.; Caye, A.; Suarez, L.; Cavé, H.;
Marschalek, R. The MLL recombinome of acute leukemias in 2017. Leukemia. 2018,
32, 273–284.
52. De Figueiredo, A.F.; Liehr, T.; Bath, S.; Binato, R.; Ventura, E.M.; de Souza, M.T.;
de Matos, R.R.; Ribeiro, R.C.; Abdelhay, E.; Silva, M.L. A new cryptic ins(11;1)
(q23;q21q31) detected in a t(1;8;11)(q21;p21;q23) in a baby with acute myeloid leukemia
FAB AML-M5. Blood Cells Mol. Dis. 2010, 45, 197–198.
53. Soupir, C.P.; Vergilio, J.A.; Dal Cin, P.; Muzikansky, A.; Kantarjian, H.; Jones, D.;
Hasserjian, R.P. Philadelphia chromosome-positive acute myeloid leukemia: A rare
aggressive leukemia with clinicopathologic features distinct from chronic myeloid leu-
kemia in myeloid blast crisis. Am. J. Clin. Pathol. 2007, 127, 642–650.
54. Cuneo, A.; Ferrant, A.; Michaux, J.L.; Demuynck, H.; Boogaerts, M.; Louwagie,
A.; Doyen, C.; Stul, M.; Cassiman, J.J.; Dal Cin, P.; Castoldi, G.; Van den Berghe,
H. Philadelphia chromosome-positive acute myeloid leukemia: Cytoimmunologic and
cytogenetic features. Haematologica. 1996, 81, 423–427.
55. Meshinchi, S.; Arceci, R.J. Prognostic factors and risk-based therapy in pediatric acute
myeloid leukemia. The Oncologist. 2007, 12, 341–355.
56. Von Lindern, M.; Fornerod, M.; van Baal, S.; Jaegle, M.; de Wit, T.; Buijs, A.; Grosveld,
G. The translocation t(6;9), associated with a specifc subtype of acute myeloid leuke-
mia, results in the fusion of two genes, DEK and CAN, and the expression of a chime-
ric, leukemiaspecifc DEK-CAN mRNA. Mol. Cell. Biol. 1992, 12, 1687–1697.
100 Cytogenetics and Molecular Cytogenetics
57. Martineau, M.; Berger, R.; Lillington, D.M.; Moorman, A.V.; Secker-Walker, L.M.
The t(6;11)(q27;q23) translocation in acute leukemia: A laboratory and clinical study
of 30 cases: EU concerted action 11q23 Workshop participants. Leukemia. 1998, 12,
788–791.
58. Balgobind, B.V.; Raimondi, S.C.; Harbott, J.; Zimmermann, M.; Alonzo, T.A.;
Auvrignon, A.; Beverloo, H.B.; Chang, M.; Creutzig, U.; Dworzak, M.N.; Forestier,
E.; Gibson, B.; Hasle, H.; Harrison, C.J.; Heerema, N.A.; Kaspers, G.J.; Leszl, A.;
Litvinko, N.; Nigro, L.L.; Morimoto, A.; Perot, C.; Pieters, R.; Reinhardt, D.; Rubnitz,
J.E.; Smith, F.O.; Stary, J.; Stasevich, I.; Strehl, S.; Taga, T.; Tomizawa, D.; Webb, D.;
Zemanova, Z.; Zwaan, C.M.; van den Heuvel-Eibrink, M.M. Novel prognostic sub-
groups in childhood 11q23/MLL-rearranged acute myeloid leukemia: Results of an
international retrospective study. Blood. 2009, 114, 2489–2496.
59. de Matos, R.R.C.; Ferreira, G.M.; Meyer, C.; Marschalek, R.; Larghero, P.; Ribeiro,
R.C.; Liehr, T.; Othman, M.; Bizarro, M.T.S.M.; Sobral da Costa, E.; Land, M.G.P.;
Abdelhay, E.; Binato, R.; Silva, M.L.M. KMT2A-MLLT1 and the novel SEC16A-
KMT2A in a cryptic 3-way translocation t(9;11;19) present in an infant with acute lym-
phoblastic leukemia. J. Pediatr. Hematol. Oncol. 2022, 44, e719–e722.
60. De Braekeleer, E.; Meyer, C.; Douet-Guilbert, N.; Morel, F.; Le Bris, M.J.; Berthou,
C.; Arnaud, B.; Marschalek, R.; Férec, C.; De Braekeleer, M. Complex and cryptic
chromosomal rearrangements involving the MLL gene in acute leukemia: A study of 7
patients and review of the literature. Blood Cells Mol. Dis. 2010, 44, 268–274.
61. De Figueiredo, A.F.; Vieira, T.P.; Liehr, T.; Bhatt, S.; de Souza, M.T.; Binato, R.;
Marques-Salles, T.deJ.; Carboni, E.; Ribeiro, R.C.; Silva, M.L.; Abdelhay, E. A rare
cryptic and complex rearrangement leading to MLL-MLLT10 gene fusion masked by
del(10)(p12) in a child with acute monoblastic leukemia (AML-M5). Leuk. Res. 2012,
36, e74–e77.
62. Ma, Z.; Morris, S.W.; Valentine, V.; Li, M.; Herbrick, J.A.; Cui, X.; Bouman, D.; Li, Y.;
Mehta, P.K.; Nizetic, D.; Kaneko, Y.; Chan, G.C.; Chan, L.C.; Squire, J.; Scherer, S.W.;
Hitzler, J.K. Fusion of two novel genes, RBM15 and MKL1, in the t(1;22)(p13q13) of
acute megakaryoblastic leukemia. Nat. Genet. 2001, 28, 220–221.
63. Carroll, A.; Civin, C.; Schneider, N.; Dahl, G.; Pappo, A.; Bowman, P.; Emami, A.;
Gross, S.; Alvarado, C.; Phillips, C.; Krischer, J.; Crist, W.; Head, D.; Gresik, M.;
Ravindranath, Y.; Weinstein, H. The t(1;22)(p13;q13) is nonrandom and restricted to
infants with acute megakaryoblastic leukemia: A Pediatric Oncology Group study.
Blood. 1991, 78, 748–752.
64. Dastugue, N.; Lafage-Pochitaloff, M.; Pagès, M.P.; Radford, I.; Bastard, C.; Talmant,
P.; Mozziconacci, M.J.; Léonard, C.; Bilhou-Nabéra, C.; Cabrol, C.; Capodano,
A.M.; Cornillet-Lefebvre, P.; Lessard, M.; Mugneret, F.; Pérot, C.; Taviaux, S.;
Fenneteaux, O.; Duchayne, E.; Berger, R.; Groupe Français d’Hematologie Cellulaire.
Cytogenetic profle of childhood and adult megakaryoblastic leukemia (M7): A study
of the Group Francais de Cytogenetique Hematologique (GFCH). Blood. 2002, 100,
618–626.
65. Capela de Matos, R.R.; Othman, M.A.K.; Ferreira, G.M.; Costa, E.S.; Melo, J.B.;
Carreira, I.M.; de Souza, M.T.; Lopes, B.A.; Emerenciano, M.; Land, M.G.P.; Liehr, T.;
Ribeiro, R.C.; Silva, M.L.M. Molecular approaches identify a cryptic MECOM rear-
rangement in a child with a rapidly progressive myeloid neoplasm. Cancer Genet. 2018,
221, 25–30.
66. Onciu, M. Acute lymphoblastic leukemia. Hematol. Oncol. Clin. North Am. 2009, 23,
655–674.
FISH—in Leukemia Diagnostics 101
67. Bhojwani, D.; Yang, J.J.; Pui, C.H. Biology of childhood acute lymphoblastic leukemia.
Pediatr. Clin. North. Am. 2015, 62, 47–60.
68. Pui, C.H. Recent research advances in childhood acute lymphoblastic leukemia. J.
Formos. Med. Assoc. 2010, 109, 777–787.
69. Vrooman, L.M.; Silverman, L.B. Treatment of childhood acute lymphoblastic leuke-
mia: Prognostic factors and clinical advances. Curr. Hematol. Malig. Rep. 2016, 11,
385–394.
70. Shago, M. Chromosome preparation for acute lymphoblastic leukemia. In: Cancer
Cytogenetics Methods and Protocols; Wan, T.S.K., Ed. Humana Press, New York, NY,
2017, pp. 19–31.
71. Greenought, A.; Dave, S.S. New clues to the molecular pathogenesis of Burkitt lym-
phoma revealed through next-generation sequencing. Curr. Opin. Hematol. 2014, 21,
326–332.
72. Swerdlow, S.H., Campo, E., Harris, N.L., Jaffe, E.S., Pileri, S.A.; Stein, H.; Thiele,
J.; Vardiman, J.W. (eds.). WHO Classifcation of Tumours of Haematopoietic and
Lymphoid Tissues. 4th Edition. IARC: Lyon; 2008.
73. Petit, B.; Mele, L.; Rack, K.; Camera, A.; Vekemans, M.C.; Bassan, R.; Pulsoni, A.;
Delannoy, A.; Pagano, L. Characteristics of secondary acute lymphoblastic leukemia
with L3 morphology in adult patients. Leuk Lymphoma. 2002, 43, 1599–1604.
74. De Souza, M.T.; Hassan, R.; Liehr, T.; Marques-Salles, T.J.; Boulhosa, A.M.; Abdelhay,
E.; Ribeiro, R.C.; Silva, M.L. Conventional and molecular cytogenetic characterization
of Burkitt lymphoma with bone marrow involvement in Brazilian children and adoles-
cents. Pediatr. Blood Cancer. 2014, 61, 1422–1426.
75. Chan, K.W. Acute lymphoblastic leukemia. Curr. Probl. Pediatr. Adolesc. Health
Care. 2002, 32, 40–49.
76. Kimura, S.; Mullighan, C.H. Molecular markers in ALL: Clinical implications. Best.
Pract. Res. Clin. Haematol. 2020, 33, 101193.
77. Ney-Garcia, D.R.; Liehr, T.; Emerenciano, M.; Meyer, C.; Marschalek, R.; Pombo-de-
Oliveira, M.do, S.; Ribeiro, R.C.; Poirot Land, M.G.; Macedo Silva, M.L. Molecular
studies reveal a MLL-MLLT3 gene fusion displaced in a case of childhood acute lym-
phoblastic leukemia with complex karyotype. Cancer Genet. 2015, 15, 1–4.
78. Garcia, D.R.; Arancibia, A.M.; Ribeiro, R.C.; Land, M.G.; Silva, M.L. Intrachromosomal
amplifcation of chromosome 21 (iAMP21) detected by ETV6/RUNX1 FISH screen-
ing in childhood acute lymphoblastic leukemia: A case report. Rev. Bras. Hematol.
Hemoter. 2013, 35, 2–4.
79. Ney-Garcia, D.R.; Liehr, T.; Bhatt, S.; de Souza, M.T.; de Matos, R.R.; Binato, R.;
Jordy, F.C.; Abdelhay, E.; Ribeiro, R.C.; Silva, M.L. Molecular cytogenetics stud-
ies reveal unexpected chromosomal inversion as variant of t(12;21)(p13;q22) in child
with B-cell precursor acute lymphoblastic leukemia. Leuk Lymphoma. 2012, 53,
342–344.
80. Mrozek, K.; Harper, D.P.; Aplan, P.D. Cytogenetics and molecular genetics of acute
lymphoblastic leukemia. Hematol. Oncol. Clin. N. Am. 2009, 23, 991–1010.
81. Sundaresh, A.; Williams, O. Mechanism of ETV6-RUNX1 leukemia. Adv. Exp. Med.
Biol. 2017, 962, 201–216.
82. Lange, B.J.; Smith, F.O.; Feusner, J.; Barnard, D.R.; Dinndorf, P.; Feig, S.; Heerema,
N.A.; Arndt, C.; Arceci, R.J.; Seibel, N.; Weiman, M.; Dusenbery, K.; Shannon, K.;
Luna-Fineman, S.; Gerbing, R.B.; Alonzo, T.A. Outcomes in CCG-2961, a Children’s
Oncology Group Phase 3 trial for untreated pediatric acute myeloid leukemia: A report
from the Children’s Oncology Group. Blood. 2008, 111, 1044–1053.
102 Cytogenetics and Molecular Cytogenetics
97. Ney Garcia, D.R.; de Souza, M.T.; de Figueiredo, A.F.; Othman, M.A.K.; Rittscher, K.;
Abdelhay, E.; Capela de Matos, R.R.; Meyer, C.; Marschalek, R.; Land, M.G.P.; Liehr,
T.; Ribeiro, R.C.; Silva, M.L.M. Molecular characterization of KMT2A fusion partner
genes in 13 cases of pediatric leukemia with complex or cryptic karyotypes. Hematol.
Oncol. 2017, 35, 760–768.
98. Betts, D.; Ammann, R.A.; Hirt, A.; Hengartner, H.; Beck-Popovic, M.; Kuhne, T.;
Nobile, L.; Cafisch, U.; Wacker, P.; Niggli, F.K. The prognostic signifcance of cytoge-
netic aberrations in childhood acute myeloid leukaemia: A study of the Swiss Paediatric
Oncology Group (SPOG). European Journal of Haematology. 2007, 78, 468–476.
99. Marchesi, F.; Annibali, O.; Cerchiara, E.; Tirindelli, M.C.; Avvisati, G. Cytogenetic
abnormalities in adult non-promyelocytic acute myeloid leukemia: A concise review.
Crit. Rev. Oncol. Hemat. 2011, 80, 331–346.
100. Ney-Garcia, D.; Vieira, T.P.; Liehr, T.; Bhatt, S.; de Souza, M.T.; de Figueiredo, A.F.;
Ribeiro, R.C.; Silva, M.L. A case of childhood T cell acute lymphoblastic leukemia
with a complex t(9;9) and homozygous deletion of CDKN2A gene associated with a
Philadelphia-positive minor subclone. Blood Cells, Mol. Dis. 2012, 50, 131–133.
101. Cui, C.; Shu, W.; Li, P. Fluorescence in situ hybridization: Cell-based genetic diagnos-
tic and research applications. Front. Cell. Dev. Biol. 2016, 4, 89.
102. Bishop, R. Applications of fuorescence in situ hybridization (FISH) in detecting
genetic aberrations of medical signifcance. Bioscience Horizons: The International
Journal of Student Research. 2010, 3, 85–95.
103. Nordgren, A. Hidden aberrations diagnosed by interphase fuorescence in situ hybrid-
ization and spectral karyotyping in childhood acute lymphoblastic leukaemia. Leuk.
Lymphoma. 2003, 44, 2039–2053.
104. Capela de Matos, R.R.; Othman, M.A.K.; Ferreira, G.M.; Monteso, K.; de Souza, M.T.;
Rouxinol, M.; Melo, J.B.; Carreira, I.M.; Abdelhay, E.; Liehr, T.; Ribeiro, R.C.; Silva,
M. Somatic homozygous loss of SH2B3, and a non-Robertsonian translocation t(15;21)
(q25.3;q22.1) with NTRK3 rearrangement, in an adolescent with progenitor B-cell
acute lymphoblastic leukemia with the iAMP21. Cancer Genet. 2021, 262–263, 16–22.
105. Testa, J.R.; Misawa, S.; Ogrema, N.; van Slaten, K.; Wiernik, P.H. Chromosomal altera-
tions in acute leukemia patients: Studies with improved culture methods. Cancer Res.
1985, 45, 430–434.
106. Hungerford, D.A. Leukocytes cultures from small in occula of whole blood and the
preparation of metaphase chromosomes by treatament with hypotonic (KCl). Stain
Technol., 1965, 40, 333–338.
107. Gisselsson, D. Cytogenetic methods. In: Cancer Genetics; Heim, S.; Mitelman, F., Eds.
John Wiley & Sons, Inc, New Jersey, NY, 2009, p. 18.
108. Seabright, M. A rapid banding technique for human chromosomes. Lancet. 1971, 2,
971–972.
109. Guerra, M., Ed. FISH Conceito e aplicações na citogenética. Sociedade Brasileira de
Genética, Ribeirão Preto, SP; 2004.
110. Tallman, M.S.; Altman, J.K. How I treat acute promyelocytic leukemia. Blood. 2009,
114, 5126–5135.
111. Capela de Matos, R.R.; de Figueiredo, A.F.; Liehr, T.; Alhourani, E.; De Souza, M.T.;
Binato, R.; Ribeiro, R.C.; Silva, M.L. A novel three-way variant t(8;13;21)(q22;q33;q22)
in a child with acute myeloid leukemia with RUNX1/RUNX1T1: The contribution
of molecular approaches for revealing t(8;21) variants. Acta Haematol. 2015, 134,
243–245.
112. Capela de Matos, R.R.; Ney-Garcia, D.R.; Cifoni, E.; Othman, M.A.K.; Tavares de
Souza, M.; Carboni, E.K.; Ferreira, G.M.; Liehr, T.; Ribeiro, R.C.; Macedo Silva, M.L.
GAS6 oncogene and reverse MLLT3-KMT2A duplications in an infant with acute
104 Cytogenetics and Molecular Cytogenetics
CONTENTS
Introduction ............................................................................................................ 105
Samples/Tissues ..................................................................................................... 106
Description of Methods.......................................................................................... 106
FISH on FFPE-Tissue Sections (No Nuclear Extraction) ............................ 106
How to Extract Nuclei from FFPE-Tissue Sections ..................................... 107
Yields ..................................................................................................................... 108
Conclusions ............................................................................................................ 108
Acknowledgments.................................................................................................. 108
References .............................................................................................................. 108
INTRODUCTION
Application of fuorescence in situ hybridization (FISH) in tissues refers here to stud-
ies done in interphase cells derived from various kinds of solid tissues from the
human body. These tissues are normally fxed in one of two ways: (i) formalin-fxed/
paraffn-embedded (FFPE) tissue,[1] or (ii) cryofxed ones.[2] Especially the pathology
of solid tumors,[3–5] and more exceptionally postmortem analysis of aborted fetus-
esare done by FISH.[6] As in FFPE sections embedded after long fxations times in
unbuffered formalin may contain highly degraded DNA not suited for FISH in most
clinics now FFPE and cryofxation are done in parallel.[7]
Of the four types of FISH-probes, whole chromosome paints, partial chromo-
some paints, centromeric probes (CPs) and locus-specifc probes (LSPs) only the
latter two are suited for routine interphase-FISH studies.[8] This is as only CPs and
LSPs lead to well-defned and relatively small signals in the interphase nucleus.
Thus, such signals can be counted to determine copy numbers of chromosomes or
chromosomal regions. Also a smart use of several LSPs or LSPs with CPs labeled
by different fuorochromes enables not only proof of deletions, duplications or even
amplifcations, and translocations or inversions may be picked up on the single-cell
level.[9]
Most FISH-studies done on tissues in pathology stick to sectioned and mounted
FFPE sections[1–5]; this has the disadvantage of partially overlapping, as well as
incomplete, cut nuclei, which cannot be evaluated properly and lead to artifcial loss
of signals.[10–11] However, it is argued that invasive and small tumor pieces could
be identifed better on sections.[12] Thus, the alternative approach where a nuclear
extraction technique[1, 2, 13] is applied is not used much in routine pathology, but has
been shown to lead to same results with lower cut-off rates; after nuclear extraction,
these are non-overlapping, and all incomplete nuclei have been discarded during the
extraction procedure.[1–2, 6, 14–16]
Here a commercial standard protocol for handling of FFPE sections is described;
also, a procedure how to extract nuclei from a cryosection, which can be used in
FISH later. Protocols for nuclear extraction from FFPE and a block of cyrofxed
material were previously described.[16]
SAMPLES/TISSUES
Each kind of solid human tissue, which can be either cryo- or FFPE-fxed, is suited
for this kind of FISH-study. It is not part of this protocol to describe the production
of a cryo- or FFPE-block or how to cut the slices and mount on glass-slides. Slices
may be differently thick—normally between from 3 to 8 µm. Also, after cutting
and thawing of cryo-sections in most cases a fxation with 3% paraformaldehyde is
necessary.[16]
DESCRIPTION OF METHODS
FISH ON FFPE-TISSUE SECTIONS (NO NUCLEAR EXTRACTION)
(Here a modifcation of the ZytoVision protocol is described—www.zytovision.
com/)
FIGURE 8.1 Typical results as obtained after FISH on FFPE-tissue sections (without
nuclear extraction) are shown.
A) A sarcoma cell with MDM2 amplifcation and a normal cell from same section are shown
after application of ZytoLight ® SPEC MDM2/CEN 12 Dual Color Probe (ZytoVision,
Bremerhaven, Germany, # Z-2013–50).
B) A Dermatofbrosarcoma protuberans (DFPS) cell with t(17;22)(q21.33;q13.1) (one red, one
green and two fusion signals) and a normal cell are visible—probe applied was ZytoLight
® SPEC COL1A1/PDGFB Dual Color Dual Fusion Probe (ZytoVision, Bremerhaven,
Germany, # Z-2116–50).
YIELDS
Results of FISH on FFPE sections fxed on slides are shown in Figure 8.1. As stan-
dard in interphase FISH analyses, it is not suffcient to evaluate 1 to 5 nuclei only. In
pathology, 50 nuclei of tumor tissue is taken into account.[17–18]
CONCLUSIONS
Even though it has been proven that FISH results from archived, cryo- or FFPE-
fxed tissues are at least as reliable, if not even better, when instead of tissue-sections
including cut and overlaying nuclei, extracted nuclei are studied, most labs stick to
sections. This is most likely due to habit and practice but also the argument that after
nuclear extraction it is no longer reproducible where exactly the evaluated nuclei are
derived from in the original section.
ACKNOWLEDGMENTS
Figure 8.1 was provided by Stefanie Kankel, Institute of Human Genetics, Jena
University Hospital, Jena, Germany; sections were provided by the Institute for
Forensic Medicine FSU, Pathology Section, Jena University Hospital, Jena, Germany.
REFERENCES
1. Liehr, T.; Grehl, H.; Rautenstrauss, B. FISH analysis of interphase nuclei extracted from
paraffn-embedded tissue. Trends Genet. 1995, 11, 377–378.
FISH—in Tissues 109
2. Liehr, T.; Grehl, H.; Rautenstrauss, B. A rapid method for FISH analysis on interphase
nuclei extracted from cryofxed tissue. Trends Genet. 1996, 12, 505–506.
3. Wolfe, K.Q.; Herrington, C.S. Interphase cytogenetics and pathology: A tool for diagno-
sis and research. J. Pathol. 1997, 181, 359–361.
4. Fuller, C.E.; Perry, A. Fluorescence in situ hybridization (FISH) in diagnostic and inves-
tigative neuropathology. Brain Pathol. 2002, 12, 67–86.
5. Lim, A.S.; Lim, T.H. Fluorescence in situ hybridization on tissue sections. Methods Mol.
Biol. 2017, 1541, 119–125.
6. Fickelscher, I.; Starke, H.; Schulze, E.; Ernst, G.; Kosyakova, N.; Mkrtchyan, H.;
MacDermont, K.; Sebire, N.; Liehr, T. A further case with a small supernumerary
marker chromosome (sSMC) derived from chromosome 1: Evidence for high variability
in mosaicism in different tissues of sSMC carriers. Prenat. Diagn. 2007, 27, 783–785.
7. Long, A.A.; Komminoth, P.; Lee, E.; Wolfe, H.J. Comparison of indirect and direct in-
situ polymerase chain reaction in cell preparations and tissue sections: Detection of viral
DNA, gene rearrangements and chromosomal translocations. Histochemistry. 1993, 99,
151–162.
8. Liehr, T. Molecular cytogenetics in the era of chromosomics and cytogenomic
approaches. Front Genet. 2021, 12, 720507.
9. Iourov, I.; Vorsanova, S.; Yurov, Y. (Eds.). Human Interphase Chromosomes: Biomedical
Aspects. Springer, Berlin; 2020.
10. Dhingra, K.; Sahin, A.; Supak, J.; Kim, S.Y.; Hortobagyi, G.; Hittelman, W.N.
Chromosome in situ hybridization on formalin-fxed mammary tissue using non-
isotopic: Non-fuorescent probes: Technical considerations and biological implications.
Breast Cancer Res. Treat. 1992, 23, 201–210.
11. Liehr, T.; Stübinger, A.; Thoma, K.; Tulusan, H.A.; Gebhart, E. Comparative interphase
cytogenetics using FISH on human ovarian carcinomas. Anticancer Res. 1994, 14,
183–188.
12. Köpf, I.; Hanson, C.; Delle, U.; Verbiené, I.; Weimarck, A. A rapid and simplifed tech-
nique for analysis of archival formalin-fxed, paraffn-embedded tissue by fuorescence
in situ hybridization (FISH). Anticancer Res. 1996, 16, 2533–2536.
13. Hedley, D.W.; Friedlander, M.L.; Taylor, I.W.; Rugg, C.A.; Musgrove, E.A. Method for
analysis of cellular DNA content of paraffn-embedded pathological material using fow
cytometry. J. Histochem. Cytochem. 1983, 31, 1333–1335.
14. Qian, J.; Bostwick, D.G.; Takahashi, S.; Borell, T.J.; Brown, J.A.; Lieber, M.M.; Jenkins,
R.B. Comparison of fuorescence in situ hybridization analysis of isolated nuclei and rou-
tine histological sections from paraffn-embedded prostatic adenocarcinoma specimens.
Am. J. Pathol. 1996, 149, 1193–1199.
15. Köpf, I.; Hanson, C.; Delle, U.; Verbiené, I.; Weimarck, A. A rapid and simplifed tech-
nique for analysis of archival formalin-fxed, paraffn-embedded tissue by fuorescence
in situ hybridization (FISH). Anticancer Res. 1996, 16, 2533–2536.
16. Liehr, T. Characterization of archived formalin-fxed/paraffn-embedded or cryofxed
tissue, including nucleus extraction. In: Fluorescence In Situ Hybridization (FISH):
Application Guide. 2nd Edition. Springer, Berlin, 2017, pp. 201–208.
17. Wayne, P.A. Fluorescence In Situ Hybridization Methods for Clinical Laboratories‑
Aproved Guidelines. 2nd Edition. Clinical Laboratory Standards Institute MM07-A2;
2013.
18. Wiktor, A.E.; Van Dyke, D.L.; Stupca, P.J.; Ketterling, R.P.; Thorland, E.C.; Shearer,
B.M.; Fink, S.R.; Stockero, K.J.; Majorowicz, J.R.; Dewald, G.W. Preclinical validation
of fuorescence in situ hybridization assays for clinical practice. Genet. Med. 2006, 8,
16–23.
9 FISH—in Human
Sperm and Infertility
Martina Rincic and Thomas Liehr
CONTENTS
Introduction ............................................................................................................ 111
Indications .............................................................................................................. 112
Description of Methods.......................................................................................... 112
Sample Preparation ....................................................................................... 112
Results and Interpretation ...................................................................................... 113
Limitations and Pitfalls .......................................................................................... 114
Troubleshooting ..................................................................................................... 114
Conclusions ............................................................................................................ 115
References .............................................................................................................. 115
INTRODUCTION
According to a generally agreed defnition, infertility is the inability to have off-
spring despite active sexual intercourse of a couple for at least one year. This
problem affects ~12% of the population in reproductive age; one in six couples
needs the help of assisted reproduction techniques (ARTs) to conceive a child.[1]
Pre-implantation genetic testing (PGT) as an alternative to prenatal diagnosis was
introduced over 30 years ago. PGT performed together with ARTs aims to reduce
the transmission of genetic or chromosomal abnormality. Recently, the European
Society of Human Reproduction and Embryology (ESHRE) has subdivided PGT
into (i) PGT-M (for the monogenic disorder), (ii) PGT-SR (for structural rearrange-
ments), which causes miscarriage, and (iii) PGT-A (for aneuploidy screening).[2]
Despite all technical advancements in the feld of molecular genetics, according to
the latest ESHRE recommendations fuorescence in situ hybridization (FISH) and
single nucleotide polymorphism (SNP) array are recommended as techniques of
choice for PGT-SR.[3]
Infertility is a complex issue where male infertility factors are relatively com-
mon conditions, affecting at least 6% of men of reproductive age. There are many
causes known from changes in gross semen parameters (for instance, low sperm
counts or compromised motility) to more complex and severe factors. In general,
gametes of infertile individuals tend to display higher rates of chromosomal abnor-
malities than those seen in fertile persons, resulting fnally in aneuploidy embryos.
Ideally, clinical diagnosis of gamete aneuploidy should include analysis of all
human autosomes and sex chromosomes. On the other hand, that type of analysis
is time-consuming and with a high cost, and is often unfeasible. Since most aneu-
ploid embryos do not survive the early stages of development, the aneuploidies that
are of major clinical importance are those that can be non-lethal, and thus compat-
ible with survival. They include autosomal trisomies, such as Trisomy 13 (Pätau
syndrome), Trisomy 18 (Edwards syndrome), and Trisomy 21 (Down syndrome),
and aneuploidies of the sex chromosomes, such as e.g. Turner- and Klinefelter-
syndrome (45,X and 47,XXY).
In 1978, direct cytogenetic analysis of the human sperm cells was introduced.[4]
Given, that this original technique was diffcult to perform, time-consuming, expen-
sive, and above all yields only a small number of karyotypes, interphase FISH was
an elegant solution to replace it. Nowadays FISH on human sperm is most commonly
used to determine the proportion of aneuploidy present in autosomes and sex chro-
mosomes of infertile men.[1]
INDICATIONS
Generally, FISH analysis of human sperm is used to perform a basic study of
aneuploidy. According to literature, a FISH sperm analysis is indicated in cases of
(i) recurrent pregnancy loss, (ii) repeated in vitro fertilization failure, (iii) abnor-
mal seminal parameters including count, motility, and morphology, and (iv) genetic
aberrations (such as but not limited to, Robertsonian and reciprocal translocations,
chromosome inversions, ring chromosomes, and numerical sex chromosome aberra-
tions).[5, 6] Additionally, FISH analysis of human sperm is indicated in men exposed
to potential mutagens, and to evaluate the impact of lifestyle factors on sperm aneu-
ploidy rate.[7–9]
DESCRIPTION OF METHODS
Considering the diverse causes of man infertility and referral reasons starting sam-
ple can be ejaculated, epididymal, or testicular-derived sperm. As already men-
tioned, the chromosomes that are generally considered for analyses are 13, 18, 21,
X, and Y, given that those aneuploidies are compatible with life. Nevertheless, all
chromosomes can be studied by us of probes specifc for each individual chromo-
some. The technical procedure of interphase FISH on human sperm nuclei has
several phases: A) sample preparation, B) hybridization, C) post-hybridization
washes, and D) visualization.[10, 11] As steps B–D are following standard conditions
(as e.g. described by the provider of corresponding FISH probes), they are not
further detailed here.
SAMPLE PREPARATION
As sperms (including their DNA) are extremely condensed, being caused by exten-
sive intermolecular disulfde cross-links, they must be decondensed prior to the
FISH procedure. To allow DNA probes access to the chromatin Otto’s solution is
FISH—in Human Sperm and Infertility 113
applied {100 ml Tris-HCl buffer 10 mmol/1, pH 7.4, 220 mg KCl (30 mmol/1), 100
mg MgCl2×6H2O (5 mmol/l), and 46 mg dithioerythritol (3 mmol/1)}.
interprets FISH results as a numerical value (a score, essentially) that would indicate
the patient’s “degree of risk”. This approach has some shortcomings that are the fea-
tures intrinsic to this technique. It is very diffcult (if not impossible) to study simul-
taneously all the chromosomes of the karyotype (normally only X, Y, 13, 18, and
21 are analyzed), and also not all possible chromosomal abnormalities can be regis-
tered. Accordingly, this method tends to underestimate the extent of anomaly rates.
In the qualitative approach, signifcant increases in aneuploidy count would have
to be interpreted as evidence that there are meiotic defects, thus indicating that the
quality of the spermatogenesis is not at optimum. Such an approach has the potential
to identify most “at-risk” patients and should be favored over the quantitative one.[15]
TROUBLESHOOTING
The most common technical points that can interfere with the quality of FISH in
human sperm are the following: no or weak signals, too much background, and
diffuse signals and cells. Reasons for signal loss may be too short denaturation or
hybridization time. Another possibility could be too stringent washing conditions
after hybridization. An increase in salt concentration or decrease of temperature
could resolve this, while higher dextran sulfate concentration and increase in probe
concentration could lead to better signals.
Usually, too much background is the result of some kind of “dirtiness” on slide
surface and can be reduced during hybridization or post-hybridization washing by
increasing the temperature or decreasing salt concentration. Additionally, the appli-
cation of directly labeled probes reduces background fuorescence. During the wash-
ing steps, it is important to prevent the slide surfaces from drying out; otherwise
background problems may arise. In addition, slides can be dirty at delivery and may
need some cleaning before use in FISH-procedure/before dropping cell suspension
on them.
To resolve diffuse signals and cells usually decreasing denaturation time and
temperature by 2°C, and/or shortening decondensation time will help to settle this
issue. Pepsin pretreatment conditions, as well as the denaturation time of the target
DNA, should be tested in each laboratory at the introduction of this kind of test.
FISH—in Human Sperm and Infertility 115
If pepsin concentration is too stringent, it can result in clean slides without any
remaining nuclei.
CONCLUSIONS
FISH is the fastest and most accurate test to assess the aneuploidy of chromosomes in
sperm nuclei on a single-cell level. Clinically, the result of FISH sperm analysis can
be used in counseling couples affected by male infertility, to come to an informed
and consented choice regarding their further reproductive plans.
REFERENCES
1. Chandra, A.; Martinez, G.M.; Mosher, W.D.; Abma, J.C.; Jones, J. Fertility, family plan-
ning, and reproductive health of U.S. women: Data from the 2002 National Survey of
Family Growth. Vital Health Stat. 2005, 23, 1–160.
2. Zegers-Hochschild, F.; Adamson, G.D.; Dyer, S.; Racowsky, C.; de Mouzon, J.; Sokol,
R.; Rienzi, L.; Sunde, A.; Schmidt, L.; Cooke, I.D.; Simpson, J.L.; van der Poel, S. The
international glossary on infertility and fertility care, 2017. Hum. Reprod. 2017, 32,
1786–1801.
3. ESHRE PGT-SR/PGT-A Working Group; Coonen, E.; Rubio, C.; Christopikou, D.;
Dimitriadou, E.; Gontar, J.; Goossens, V.; Maurer, M.; Spinella, F.; Vermeulen, N.; De
Rycke, M. ESHRE PGT Consortium good practice recommendations for the detection
of structural and numerical chromosomal aberrations. Hum. Reprod. Open. 2020, 2020,
hoaa017.
4. Rudak, E.; Jacobs, P.A.; Yanagimachi, R. Direct analysis of the chromosome constitu-
tion of human spermatozoa. Nature. 1978, 274, 911–913.
5. Ramasamy, R.; Besada, S.; Lamb, D.J. Fluorescent in situ hybridization of human
sperm-diagnostics.; indications.; and therapeutic implications. Fertil. Steril. 2014, 102,
1534–1539.
6. Fakhrabadi, M.P.; Kalantar, S.M.; Montazeri, F.; Ashkezari, M.D.; Fakhrabadi, M.P.;
Yazd, S.S.N. FISH-based sperm aneuploidy screening in male partner of women with a
history of recurrent pregnancy loss. Middle East Fert. Soc. J. 2020, 25, 23.
7. Saad, A.A.; Hussein, T.; El-Sikaily, A.; Abdel-Mohsen, M.A.; Mokhamer, E.H.; Youssef,
A.I.; Mohammed, J. Effect of polycyclic aromatic hydrocarbons exposure on sperm
DNA in idiopathic male infertility. J. Health Pollut. 2019, 9, 190309.
8. Srám RJ.; Binková, B.; Rössner, P.; Rubes, J.; Topinka, J.; Dejmek, J. Adverse repro-
ductive outcomes from exposure to environmental mutagens. Mutat. Res. 1999, 428,
203–215.
9. Robbins, W.A.; Vine, M.F.; Truong, K.Y.; Everson, R.B. Use of fuorescence in situ
hybridization (FISH) to assess effects of smoking, caffeine, and alcohol on aneuploidy
load in sperm of healthy men. Environ. Mol. Mutagen. 1997, 30, 175–183.
10. Sarrate, Z.; Anton, E. Fluorescence in situ hybridization (FISH) protocol in human
sperm. J. Vis. Exp. 2009, (31), 1405.
11. Rauch, A. Human sperm cells. In: FISH Technology; Rautenstrauss, B.W.; Liehr, T. Eds.
Springer, Berlin, Heidelberg, 2002, pp. 127–137.
12. García-Mengual, E.; Triviño, J.C.; Sáez-Cuevas, A.; Bataller, J.; Ruíz-Jorro, M.; Vendrell,
X. Male infertility: Establishing sperm aneuploidy thresholds in the laboratory. J. Assist.
Reprod. Genet. 2019, 36, 371–381.
116 Cytogenetics and Molecular Cytogenetics
13. Schultz, H.; Mennicke, K.; Schlieker, H.; Al-Hasani, S.; Bals-Pratsch, M.; Diedrich, K.;
Schwinger, E. Comparative study of disomy and diploidy rates in spermatozoa of fertile
and infertile men: A donor-adapted protocol for multi-colour fuorescence in situ hybrid-
ization (FISH). Int. J. Androl. 2000, 23, 300–308.
14. Blanco, J.; Egozcue, J.; Vidal, F. Incidence of chromosome 21 disomy in human sper-
matozoa as determined by fuorescent in-situ hybridization. Hum. Reprod. 1996, 11,
722–726.
15. Sarrate, Z.; Vidal, F.; Blanco, J. Role of sperm fuorescent in situ hybridization studies
in infertile patients: Indications.; study approach.; and clinical relevance. Fertil. Steril.
2010, 93, 1892–1902.
10 FISH—in Spontaneously
Aborted Products
of Conception
Thomas Liehr
CONTENTS
Introduction ............................................................................................................ 117
Samples/Tissues ..................................................................................................... 118
Description of Methods.......................................................................................... 118
Yields ..................................................................................................................... 119
Conclusions ............................................................................................................ 119
References .............................................................................................................. 119
INTRODUCTION
Infertility is a major problem in Western countries, but also—either less expressed
or less registered—in all human societies around the world. Besides the inability to
conceive, some couples experience (repeated) abortions at early, middle or late preg-
nancy stages.[1] Such couples should attract gynecologist’s attention during routine
patient admission, and these couples should be offered (cyto)genetic studies for both
partners.[2] In a subset of these cases, a gonosomal mosaic, a small supernumerary
marker chromosome or a balanced rearrangement can be traced, as being causative
for the problem.[3, 4] Besides, and especially in cases where no such conclusive results
can be found, the spontaneously aborted products of conception (PoCs) themselves
should be studied genetically.[5] Chromosomal aberrations identifed in these PoCs
can be important (i) to make such couples better understand why an abortion has
happened,[6] and (ii) also as they can be a hint on a cryptic gonadal mosaicism being
present in male or female partner.[7]
In some settings, DNA extraction from PoCs and subsequent chromosome-
microarray,[8] multiplex ligation-dependent probe amplifcation (MLPA),[9] or simple
STR (= short tandem repeat DNA) -analyses (own unpublished data) may be used
to fnd genomic imbalances, that have being causative for the abortion. Another
straightforward method is tissue cultivation from PoCs and subsequent chromosome
analyses by GTG-banding[5, 10, 11]; alternatively and if available, also formalin-fxed
formalin embedded material of PoCs may be analyzed by molecular cytogenetics.[12]
However, the latter is missing the opportunity to possibly get an informative banding
cytogenetic result. Furthermore, as PoC-cell-cultivation is prone to culture failure,
it has been shown that interphase molecular cytogenetics may be a way out here to
obtain nonetheless informative results.[5]
SAMPLES/TISSUES
Cytogenetic preparations with and without metaphase spreads, but at least contain-
ing suffcient interphase nuclei from PoCs are used for the here described analyses.
Preparation is done using standard techniques.[13]
DESCRIPTION OF METHODS
The method applied here is a standard fuorescence in situ hybridization (FISH)
approach—see e.g.[14] Commercially available probes, either centromere- or locus-
specifc ones, are applied. Preferentially used are centromeric probes, still, for
chromosomes 13 and 21, and 14 and 22 only the probes D13/21Z1 and D14/22Z1
staining both corresponding chromosome pairs are available. In addition, problems
may arise with centromeric probe D1Z7 in 1p11.1–1q11 being identical to D5Z2 in
5p11-q11.1 and D19Z3 in 19q11; chromosome 1 may be identifed by D1Z5 in 1q11-
q12, but additional (derivatives of) chromosomes 5 and 19 cannot be indubitably
distinguished in metaphase and not at all in interphase.[15, 16] Accordingly, com-
mercially available locus-specifc probes can be selected if these chromosomes
shall be studied.
As the available amount of PoC-material normally is restricted, at least two- to
multicolor-FISH settings are recommended. According to available commercially
available probes applied multicolor-FISH probe sets may be put together (e.g.
Table 10.1). In a study from 2017 we had an ~37% detection rate of abortion causing
chromosomal aneuploidies when concentrating on chromosomes 13/21, 14/22, 15,
16, 17, 18, X and Y. Suited probe sets which are composed of commercially avail-
able probes from Abbott/Vysis and Cytocell are listed in Table 10.1. Probe sets 1
to 3 (Table 10.1) should be applied subsequently to samples to be tested. In case of
fve signals for D13/21Z1 or D14/22Z1 probes, the result needs to be checked by
locus-specifc (LSI) probes, as listed in Table 10.1. The latter is necessary, as it can
easily be that instead of a trisomy 13, 14, 21 or 22 suited chromosome-specifc LSI
probes do not reveal an aberrant signal pattern. This may be due to chromosomal
heteromorphisms, which can lead to additional signals in the centromeric regions
of another chromosome.[17] Additionally, such a result can be due to a partial tri-
somy of one of the four chromosomes in the sense of a small supernumerary marker
chromosome.[15]
For the here-suggested set-up and composition of probe sets (Table 10.1), com-
mercially available probes are suggested. Thus, the established lab internal standard
FISH-procedure may be used, or the protocol as suggested by the provider of the
commercial probes can be applied. It has to be tested under each individual lab-con-
dition if a pretreatment with RNAse and/or pepsin is necessary or can be skipped;
this is dependent on the plasma load of interphase nuclei after cytogenetic work-up
of PoCs.
FISH—in Spontaneously Aborted Products of Conception 119
TABLE 10.1
Probe Sets Adapted from[5] Consisting of Commercially Available Probes Are
Listed; With This Setting Polyploidies, Gonsomal Aberrations and Numerical
Aberrations of Chromosomes 13, 14, 15, 16, 17, 21 and 22 Can Be Accessed.
Probe Set Probes Location Fluorochrome
1 D13/21Z1 13p11.1-q11 and SpectrumGreen (SG)
D15Z3 21p11.1-q11.1 TexasRed (TR)
D18Z1 15q11 Diethylaminocoumarin
18p11.1-q11.1 (DEAC)
2 D14/22Z1 14p11.1-q11.1 and SG
D16Z2 22p11.1-q11.1 TR
16p11.1-q11.1
3 D17Z1 17p11.1-q11.1 SpectrumOrange (SO)
DXZ1 Xp11.1-q11.1 DEAC
Yq12 Yq12 SG
if D13/21Z1 gives 5 signals LSI 13/21 13q14 and 21q22.13-q22.2 SG and SO
if D14/22Z1 gives 5 signals LSI D22S75/ 22q11.2 and 22q13.3 SO and SG
ARSA
YIELDS
Studying PoCs with the suggested probe sets (Table 10.1) identifed in a pilot study
of 286 PoCs chromosomal abnormalities in 106 cases. Polyploidy was present in
23/106 cases, sex-chromosome abnormalities in 37/106 cases and trisomies in the
remainder 46 cases. Twenty-two of the latter 46 cases were trisomy 16.[5]
CONCLUSIONS
Taking advantage of interphase FISH to unravel reasons of (repeated) abortions is
still only scarcely used in human genetic labs. The possibilities to obtain a diagnosis
on the reasons for an abortion are still by far not exhausted in routine settings; not
only the study referred to here,[5] but also other authors highlighted this option,[5, 8–12]
which among others provides also the advantage of a single cell specifc evaluation,
and thus to detect mosaic conditions, too.
REFERENCES
1. Flyckt, R.; Falcone, T. Infertility: A practical framework. Cleve Clin. J. Med. 2019, 86,
473–482.
2. Larsen, E.C.; Christiansen, O.B.; Kolte, A.M.; Macklon, N. New insights into mecha-
nisms behind miscarriage. BMC Med. 2013, 11, 154.
3. Liehr, T. Small supernumerary marker chromosomes detected in connection with infer-
tility. Zhonghua Nan. Ke. Xue. 2014, 20, 771–780.
120 Cytogenetics and Molecular Cytogenetics
4. Manvelyan, M.; Schreyer, I.; Höls-Herpertz, I.; Köhler, S.; Niemann, R.; Hehr, U.; Belitz,
B.; Bartels, I.; Götz, J.; Huhle, D.; Kossakiewicz, M.; Tittelbach, H.; Neubauer, S.;
Polityko, A.; Mazauric, M.L.; Wegner, R.; Stumm, M.; Küpferling, P.; Süss, F.; Kunze,
H.; Weise, A.; Liehr, T.; Mrasek, K. Forty-eight new cases with infertility due to bal-
anced chromosomal rearrangements: Detailed molecular cytogenetic analysis of the 90
involved breakpoints. Int. J. Mol. Med. 2007, 19, 855–864.
5. Tkach, I.R.; Huleyuk, N.L.; Zastavna, D.V.; Weise, A.; Liehr, T.; Ciszkowicz, E.; Tyrka,
M. Chromosomal aberrations in spontaneously aborted products of conception from
Ukraine. Biopolymers Cell. 2017, 33, 424–433.
6. Griebel, C.P.; Halvorsen, J.; Golemon, T.B.; Day, A.A. Management of spontaneous
abortion. Am. Fam. Physician. 2005, 72, 1243–1250.
7. Kamel, A.K.; Abd El-Ghany, H.M.; Mekkawy, M.K.; Makhlouf, M.M.; Mazen, I.M.; El
Dessouky, N.; Mahmoud, W.; Abd El Kader, S.A. Sex chromosome mosaicism in the
gonads of DSD patients: A karyotype/phenotype correlation. Sex. Dev. 2015, 9, 279–288.
8. Gao, J.; Liu, C.; Yao, F.; Hao, N.; Zhou, J.; Zhou, Q.; Zhang, L.; Liu, X.; Bian, X.; Liu, J.
Array-based comparative genomic hybridization is more informative than conventional
karyotyping and fuorescence in situ hybridization in the analysis of frst-trimester spon-
taneous abortion. Mol. Cytogenet. 2012, 5, 33.
9. Kim, J.W.; Lyu, S.W.; Sung, S.R.; Park, J.E.; Cha, D.H.; Yoon, T.K.; Ko, J.J.; Shim, S.H.
Molecular analysis of miscarriage products using multiplex ligation-dependent probe
amplifcation (MLPA): Alternative to conventional karyotype analysis. Arch. Gynecol.
Obstet. 2015, 291, 347–354.
10. Vorsanova, S.G.; Kolotii, A.D.; Iourov, I.Y.; Monakhov, V.V.; Kirillova, E.A.; Soloviev,
I.V.; Yurov, Y.B. Evidence for high frequency of chromosomal mosaicism in spontane-
ous abortions revealed by interphase FISH analysis. J. Histochem. Cytochem. 2005, 53,
375–380.
11. Jobanputra, V.; Sobrino, A.; Kinney, A.; Kline, J.; Warburton, D. Multiplex interphase
FISH as a screen for common aneuploidies in spontaneous abortions. Hum. Reprod.
2002, 17, 1166–1170.
12. Russo, R.; Sessa, A.M.; Fumo, R.; Gaeta, S. Chromosomal anomalies in early spontane-
ous abortions: Interphase FISH analysis on 855 FFPE frst trimester abortions. Prenat.
Diagn. 2016, 36, 186–191.
13. Weise, A.; Liehr, T. Pre- and postnatal diagnostics and research on peripheral
blood, bone marrow chorion, amniocytes, and fbroblasts. In: Fluorescence In Situ
Hybridization (FISH): Application Guide; Liehr, T., Ed. 2nd Edition. Springer, Berlin,
2017, pp. 171–180.
14. Weise, A.; Liehr, T. Background. In: Fluorescence In Situ Hybridization (FISH):
Application Guide; Liehr, T., Ed. 2nd Edition. Springer, Berlin, 2017, pp. 1–14.
15. Nietzel, A.; Rocchi, M.; Starke, H.; Heller, A.; Fiedler, W.; Wlodarska, I.; Loncarevic,
I.F.; Beensen, V.; Claussen, U.; Liehr, T. A new multicolor-FISH approach for the charac-
terization of marker chromosomes: Centromere-specifc multicolor-FISH (cenM-FISH).
Hum. Genet. 2001, 108, 199–204.
16. Liehr, T. Benign & Pathological Chromosomal Imbalances Microscopic and
Submicroscopic Copy Number Variations (CNVs) in Genetics and Counseling. Academic
Press, New York, 2014.
17. Liehr, T.; Ziegler, M. Rapid prenatal diagnostics in the interphase nucleus: Procedure
and cut-off rates. J. Histochem. Cytochem. 2005, 53, 289–291.
11 FISH—Characterization
of Chromosomal
Alterations,
Recombination, and
Outcomes after
Segregation
Ilda Patrícia Ribeiro, Eunice Matoso,
Joana Barbosa Melo, Ana Jardim,
Thomas Liehr and Isabel Marques Carreira
CONTENTS
Introduction............................................................................................................ 122
Chromosomal Alterations ...................................................................................... 124
Numerical Alterations................................................................................... 125
Unbalanced Structural Rearrangements ....................................................... 129
Balanced Structural Rearrangements............................................................ 135
Case Problems........................................................................................................ 138
Clinical Case 1 ....................................................................................................... 138
Clinical Information...................................................................................... 138
Techniques Performed .................................................................................. 138
Results........................................................................................................... 138
Questions ...................................................................................................... 138
Curiosity ....................................................................................................... 139
Conclusion .................................................................................................... 140
Clinical Case 2 ....................................................................................................... 140
Clinical Information...................................................................................... 140
Techniques Performed .................................................................................. 140
Results Twin 1............................................................................................... 140
Results Twin 2............................................................................................... 140
Questions ...................................................................................................... 140
Conclusion .................................................................................................... 142
INTRODUCTION
The process leading to the formation of haploid gametes from diploid cells through
one round of DNA replication and two rounds of chromosome segregation is called
meiosis, or more precisely Meiosis I (MI) and Meiosis II (MII).[1] Both meiosis and
mitosis begin with replication of DNA in order to originate a cell with four chroma-
tids of each type of chromosome, two from the mother and two from the father.[2]
During the frst meiotic division (MI), the separation of maternal and paternal copies
of each chromosome occurs: this is the so-called reductional division. It is in the sec-
ond meiotic division (MII) that the separation of sister chromatids occurs: this is the
equational division (very similar to mitosis). The resulting four chromatids are dis-
tributed to four different nuclei, which occurs simultaneously for all chromosomes
in the two rounds of chromosome segregation.[2] During the meiosis process, there
is not only a reduction of chromosome number to a haploid set but also the aleatoric
segregation of maternal and paternal chromosomes, and genetic combinations in the
offspring due to exchange of genetic material in maternal and paternal homologues
through recombination.[3] The reduction in chromosome number is pivotal to restore
diploid chromosome complement in the subsequent generation through the union of
two gametes during sexual reproduction, ensuring continuity of the species.[4]
There are three signifcant deviations in MI in comparison to mitosis:
tissue distribution of the aneuploid cells. A distinct event is the chimerism, where
different cell lines are originated from more than one zygote.
CHROMOSOMAL ALTERATIONS
In 1956, a new era of clinical cytogenetics started with the identifcation of the exact
chromosome number in humans, and therefore the description of several chromo-
somal syndromes with altered chromosome numbers.[15] Chromosomal alterations
could be categorized as numerical or structural and could involve more than one
chromosome. Numerical chromosomal alterations are more common than struc-
tural ones and represent a deviation from the normal diploid number for a given
species. This category can be subdivided in two groups: (i) aneuploidy (a group in
which individual chromosomes may be either missing or present in one or more
additional copies) and (ii) polyploidy (here whole haploid sets of chromosomes may
be added or lost, as triploidy (3n) in human with 69 chromosomes, or tetraploidy
(4n) in human 92 chromosomes).[16] Triploidy or tetraploidy are lethal disorders often
observed in spontaneous abortions and rarely in newborns with a reduced survival
time. Triploidy is one of the most common chromosome abnormalities occurring in
~1% of recognized conceptions, and 10% of recognized miscarriages.[17] Triploidy
can refect the double contribution coming from the father (di-andry) or the mother
(di-gyny). Diandry is usually the consequence of two sperm simultaneously fertil-
izing the ovum.[18–20] Digyny could be the result of nondisjunction of the entire set at
either the MI or MII in oogenesis, or a polar body may fail to be extruded.[21]
Structural rearrangements result from chromosome breakage and reunion within a
single chromosome or between two or more non-homologous chromosomes originat-
ing either balanced or unbalanced complements.[15] Balanced rearrangement means
that there is usually no alteration in chromosome content, while unbalanced ones lead
to gains or losses of chromosomal material. At birth, structural rearrangements (bal-
anced and unbalanced) were reported in about 1/400 infants.[22] Signifcant clinical
phenotypes were observed in carriers of unbalanced rearrangements, as a consequence
of loss and/or duplication of genetic material. Some examples of unbalanced rearrange-
ments are deletions, duplications, rings and isochromosomes, and of balanced rear-
rangements are inversions (paracentric and pericentric), insertions (direct or inverted),
and translocations (reciprocal and Robertsonian).[15] Additionally, we also have small
marker chromosomes (SMCs), which are usually supernumerary (sSMCs).
Balanced as well as unbalanced structural abnormalities can be inherited from
a carrier parent or could arise as de novo. For inherited balanced structural rear-
rangements, the risk for a clinical phenotype is low; however, for de novo rearrange-
ments, this risk is higher, even if the rearrangement looks balanced: submicroscopic
deletions/duplications at the breakpoints and alterations in the genes or regulatory
elements near to the breakpoints cannot be excluded.[22] So, the management of bal-
anced rearrangements identifed in prenatal cases is diffcult, especially with a de
novo origin. It is important to highlight that a balanced carrier (heterozygote) could
present with diffculties in reproduction, namely infertility, spontaneous abortions,
or abnormal offspring, which is a result of anomalous pairing and segregation at
prophase I.[22]
FISH—Characterization of Chromosomal Alterations 125
NUMERICAL ALTERATIONS
Aneuploidy can affect any one of the 23 pairs of chromosomes. In humans, aneu-
ploidy leads to about 35% of miscarriages and 4% of stillbirths.[10] Aneuploidy is
also present in around 30–60% of embryos, in 30–70% of oocytes and in 0.3% of
newborns (with the most common abnormalities being trisomy 21 and sex chro-
mosome trisomies), facing developmental disabilities and intellectual disability.[10]
Regarding human aneuploidy, examples of trisomic conditions compatible with
126 Cytogenetics and Molecular Cytogenetics
full-term pregnancies are the Down (trisomy 21), Edwards (trisomy 18), and Pätau
(trisomy 13) syndromes (Figure 11.1).
Down syndrome is one of the best-described and most frequent chromosome
disorders that causes intellectual disability. The incidence of Down syndrome
is approximately 1/600 newborns, being originated in almost 94% of cases from
meiotic nondisjunction.[15, 20] In approximately 95% of cases, the extra chromo-
some 21 is of maternal origin, and around 80% of those are due to an error during
MI.[15] Nevertheless, almost 4% of Down syndrome patients present an unbalanced
Robertsonian translocation involving chromosome 21 and the long arm of chromo-
somes 13, 14, 15, 21, or 22.[15] It is important to highlight that if a parent is a balanced
21/21 isochromosome carrier, this represents a 100% risk of having offspring with
Down syndrome. Female carriers of balanced 14/21 or 21/22 Robertsonian translo-
cations have a 10–15% risk for unbalanced Down syndrome offspring in comparison
to 5% for male carriers.[15] These translocations could be de novo or inherited from a
balanced carrier parent (usually the mother). Additionally, mosaicism for trisomy 21
is also the cause of Down syndrome in almost 2% of the cases.[15]
FISH—Characterization of Chromosomal Alterations 127
Edwards syndrome has a frequency of about 1/6000 live births and a poor post-
natal survival estimate (~90% die in the frst 6 months).[20] Few cases of trisomy 18
mosaicism were reported, but some patients with unbalanced translocations involv-
ing all or most of chromosome 18 long arm were also described presenting with
trisomy 18 features.[15]
Pätau syndrome has an estimated incidence of about 1/12,000 live births, and
90% of these patients rarely survive the newborn period.[20] The origin of the extra
chromosome 13 is frequently from a maternal meiotic non-disjunction error, but may
also arise from an unbalanced Robertsonian translocation involving chromosome
13 (in ~20% of the cases) or yet from trisomy 13 mosaicism, which is a rare event
and presents a less severe clinical phenotype.[15]
Likewise, aneuploidies involving sex chromosomes are also a possible consequence
of errors in chromosome segregation but present less severe phenotypes in comparison
with somatic trisomies. The gene dosage imbalances for genes encoded in sex chro-
mosomes are lower than for the genes encoded on somatic chromosomes, and besides
that there is the inactivation process of the extra X chromosome(s), if there is more than
one present per cell.[25] Examples of sex chromosome abnormalities are Turner (45,X),
triple X (XXX), Klinefelter (47,XXY) and 47,XYY syndromes (Figure 11.2).[25]
of the deleted acentric fragment and consequently the gene content and also due to
ring chromosome instability, but usually overlaps that of the syndromes associated
with deletion of both ends of the corresponding chromosome.[34] So, besides dele-
tions that occur by ring formation, ring instability can also lead to other genomic
imbalances with gain or loss of genetic material and consequently to distinct phe-
notypes. Sister chromatid exchanges happening during mitosis in patients with ring
chromosomes result in secondary chromosomal abnormalities like dicentric rings,
interlocked rings, and other structural alterations or even ring chromosome loss lead-
ing to monosomic cells.[30]
Isochromosomes are composed of two copies of the same arm of one chromo-
some (mirror image of one of the arms of the chromosome). Some mechanisms
have been suggested for their formation, like transversal mis-division of the
134 Cytogenetics and Molecular Cytogenetics
FIGURE 11.9 A case with mosaicism involving a cell line 45,X and fve cell lines present-
ing different dicentric Y chromosomes with the breakpoints in the terminal segment of the
short arm, and another cell line with two derivative Y chromosomes is characterized here.
A) Karyogram with a large dicentric Y chromosome (circle).
B) Enlarged sex-chromosomes of the case (GTG-banded).
C) CBG banding staining Yq12 two times on the der(Y)—arrow.
D–I) FISH analysis using different probes as shown in the pictures.
centromere during MII, sister chromatid breakage and reunion near the centro-
mere, or exchange between homologues during meiosis.[15] Isochromosomes X are
often observed in Turner syndrome individuals, and represent the most frequent
isochromosomes observed, that are, in fact, in the majority of cases dicentric.[15]
Isochromosome Yp, that results in duplication of the short arm and loss of the long
arm, has been reported to be linked to azoospermia (Figure 11.9).[35] Autosomal
isochromosomes occur more often in acrocentric chromosomes with loss of the
short arms.
Small supernumerary marker chromosomes (sSMCs) are structurally abnor-
mal chromosomes that cannot be unambiguously identifed or characterized by
conventional banding cytogenetics alone.[23] These sSMCs are equal to or smaller
in size than a chromosome 20 of the same metaphase spread.[36] The origin and
composition of an sSMC is recognizable by molecular cytogenetics analysis. They
are seen in different shapes like ring, centric minute, and inverted duplication,
and could be centric, or acentric, complex (= deriving from more than one chro-
mosome), being discontinuous due to formation via chromothripsis and being
present in mosaic.
FISH—Characterization of Chromosomal Alterations 135
FIGURE 11.10 Karyograms of cases with structural chromosomal aberrations (in parts
with schematic depictions):
A) pericentric inversion: 46,XX,inv(1)(p22.1q44).
B) paracentric inversion: 46,XY,inv(1)(q23.1q32.1).
C) reciprocal translocation: 46,XY,t(7;12)(p13;q22).
D) Robertsonian translocation: 45,XX,der(13;14)(q10;q10).
E) Insertion: 46,XX,ins(12;18)(q24.31;q21.3q23).
FISH—Characterization of Chromosomal Alterations 137
CASE PROBLEMS
Selected clinical cases will be presented as examples of karyotyping and FISH appli-
cations in postnatal and in prenatal diagnosis.
For all clinical post and prenatal cases, three questions will be answered:
CLINICAL CASE 1
CLINICAL INFORMATION
A 6-year-old girl with microcephaly, moderate to severe psychomotor developmental
delay, and prenatal history of intrauterine growth restriction was studied.
TECHNIQUES PERFORMED
G banding cytogenetics;
FISH for DiGeorge critical region and for the subtelomeric regions.
RESULTS
The combined result for this case was 46,XX.ish del(22)(q13.33)(D22S1726-).
QUESTIONS
i) What kind of chromosomal alteration was found? How to interpret the
results? What is the most probable mechanism of origin of the alteration?
The high resolution karyotype was normal (Figure 11.12 A) as well as the FISH
results for the DiGeorge critical region in 22q11.2 (Figure 11.12 B).
FISH—Characterization of Chromosomal Alterations 139
CURIOSITY
Deletion 22q13 has been accidentally diagnosed by FISH in several individuals pri-
marily analyzed for 22q11.3 deletion syndrome. Numerous commercially available
FISH assays for 22q11.3 deletion syndrome use arylsulfatase A (ARSA) gene as the
control probe. ARSA maps to 22q13 and therefore detects deletions of this region.
The ARSA probe successfully detects the majority of 22q13.3 deletions; however,
microdeletions distal to ARSA may require FISH analysis using the 22q subtelomere
probes,[42] as happened in our case.
In order to establish the most probable mechanism of origin of this alteration, both
parents were also analyzed by karyotyping and FISH techniques. This did not reveal
any numerical or structural alteration (data not shown), and therefore this terminal
deletion was classifed as de novo.
This couple was referred for genetic counselling. Monitoring and testing future
pregnancies was recommended.
140 Cytogenetics and Molecular Cytogenetics
CONCLUSION
This terminal deletion of the long arm of chromosome 22 (22q13.33-qter) seems to
explain the clinical phenotype of the girl. The deletion 22q13.3 syndrome or Phelan-
McDermid syndrome is characterized by neonatal hypotonia, global developmental
delay, normal to accelerated growth, absent to severely delayed speech, and minor
dysmorphic features. The loss of 22q13.3 can result from simple deletion, transloca-
tion, ring chromosome formation, and less commonly structural changes affecting
the long arm of chromosome 22, specifcally the region containing the SHANK3
gene.[43] Almost 80% of individuals with Phelan-McDermid syndrome have de novo,
simple deletions of 22q13,[44] as observed in our case.
CLINICAL CASE 2
CLINICAL INFORMATION
A woman in the 15th week of twin pregnancy did trophoblast biopsy as sonography
diagnosed hygroma in one of fetus and a borderline nuchal translucency in the other
fetus.
TECHNIQUES PERFORMED
QF-PCR for aneuploidy screening;
G banding cytogenetics;
FISH technique using subtelomeric probes for chromosomes 11 and 18.
RESULTS TWIN 1
QF-PCR for aneuploidy screening was performed with a normal result (data not
shown).
The combined result (Figure 11.13 A and B) for this case was:
46,X Y,der(18)t(11;18)(p15.3;p11.2).ish der(18)t(11;18)(p15.5;p11.32)
(D11S2071+,D18S552-)
RESULTS TWIN 2
QF-PCR for aneuploidy screening was performed with a normal result (data not
shown).
The combined result (Figure 11.13 C—FISH not shown) for this case was:
46,XX,t(11;18)(p15.5;p11.23).ish t(11;18)(p15.5-,p11.32+;p11.32-,p15.5+)(D11S2071-,
D18S552+; D18S552-, D11S2071+)
QUESTIONS
i) What kind of chromosomal alteration was found? How to interpret the
results? What is the most probable mechanism of origin of the alteration?
FISH—Characterization of Chromosomal Alterations 141
The G-banded metaphases of the trophoblast biopsy cells revealed, in twin 1, a male
fetus with a chromosome 18 derivative from a translocation involving chromosomes
11 and 18 (Figure 11.13 A) and, in twin 2, a female fetus with an apparently bal-
anced reciprocal translocation involving the same chromosomes (Figure 11.13 C).
The structural alterations identifed in both twins were confrmed by FISH technique
using subtelomeric probes, for 11p15.5 and 18p11.32 (twin 1—Figure 11.13 B; twin
2—not shown). It is important to stress that the result for twin 1 does not contradict
that obtained with the screening for aneuploidies by the QF-PCR technique, since
this technique is not suitable for the detection of partial aneuploidies in the tested
chromosomes, such as this one identifed in chromosome 18.
In order to establish the most probable mechanism of origin of the alteration, the
study of the parents was recommended.
The study (G-banding from peripheral blood) of both parents revealed a normal
karyotype for the mother, and the presence of a reciprocal translocation involving
chromosomes 11 and 18 in the father, 46,XY,t(11;18)(p15.3;p11.2) (data not shown).
Regarding the most probable biological mechanism, it can be concluded that the
male progenitor is a carrier of a structural chromosomic rearrangement, a reciprocal
translocation with breakpoints in 11p15.3 and 18p11.2, which has no implication in
the phenotype of the carrier. However, during spermatogenesis there is a risk for the
offspring to inherited imbalances as a result of anomalous segregations. In this case,
the identifed structural chromosomal rearrangement in fetus 1 is likely to be due
to an adjacent I segregation, having the fetus a derivative chromosome 18 inherited
from the paternal translocation. Fetus 2 is the result of an alternate segregation as it
carries the same translocation as the father.
This couple was offered genetic counselling where it was well explained the conse-
quence of this reciprocal translocation in this phenotypically normal father for future
pregnancies, and the need of family studies (parents, brothers, sisters, and other fam-
ily members at risk). To all carriers, invasive prenatal diagnosis should be offered.
As chromosome 11 is involved in the rearrangement also the hint should be given,
that this imprinting associated chromosome is slightly more likely to be involved in
uniparental disomy (UPD) due to the t(11;18). Thus, prenatal UPD test may also be
considered in future pregnancies.
CONCLUSION
This clinical case shows the implications for the offspring of a balanced structural
rearrangement in one of the progenitors and the need to offer to this couple prena-
tal diagnosis in future gestations and genetic counselling to explain the implica-
tions of these genetic fndings, the risk of recurrence, and the need to offer family
FISH—Characterization of Chromosomal Alterations 143
studies to other relatives. In this clinical case, the male progenitor is a carrier of a
reciprocal translocation between chromosomes 11 and 18, and the unbalanced struc-
tural chromosomal rearrangement involving partial monosomy of the short arm of
chromosome 18 (p11.2-pter) and partial trisomy of the short arm of chromosome 11
(p15.3-pter) observed in twin 1 was originated by an adjacent 1 segregation during
spermatogenesis. Twin 2, on the other hand, was a balanced carrier like the male
progenitor, and resulted from an alternate type of segregation. The imbalances iden-
tifed in twin 1 are most probably the cause of the ultrasound alterations detected and
consequently are correlated to the clinical phenotype.
CLINICAL CASE 3
CLINICAL INFORMATION
A pregnant woman in 13th week of gestation had a sonographic diagnosis of holo-
prosencephaly in the fetus, and thus amniotic fuid was collected.
TECHNIQUES PERFORMED
FISH for the rapid detection of chromosome aneuploidies in uncultured amniocytes;
G banding cytogenetics;
FISH technique using whole chromosome painting probe for chromosome 21
(WCP21).
RESULTS
FISH for the rapid detection of chromosome aneuploidies in uncultured amniocytes
was performed with a normal result (Figure 11.14 A-3). The combined result of G
banding and molecular cytogenetics revealed two partial monosomies involving
the terminal region of chromosome 13q32-qter and the proximal region of chro-
mosome 21pter-q22.1. Karyotype: 45,XY,der(13)t(13;21)(q32;q22.1),−21.ish t(13;21)
(q32;q22.1)(wcp21+;wcp21+).
QUESTIONS
i) What kind of chromosomal alteration was found? How to interpret the
results? What is the most probable mechanism of origin of the alteration?
G-banding in amniotic fuid cells revealed a male fetus with 45 chromosomes due
to a monosomy of chromosome 21 and also a structurally altered chromosome 13
(Figure 11.14 A-1). By FISH technique using WCP21, it was possible to characterize
the derivative chromosome 13 as an unbalanced structural chromosomal rearrange-
ment involving chromosome 13 and 21 (Figure 11.14 A-1 and 11.14 A-2). This structural
rearrangement was also confrmed in fbroblast cells of the fetus (results not shown)
and seems to be the origin of the detected ultrasound anomaly, being consequently
144 Cytogenetics and Molecular Cytogenetics
correlated with the clinical phenotype. In order to ascertain the origin of this structural
alteration, both parents were also analyzed by conventional cytogenetics and FISH.
CURIOSITY
Monosomy 21 as seemingly present in GTG-banding is not compatible with life,
and such cases abort spontaneously in very early embryogenesis; thus, here a cryptic
rearrangement should be suspected by the clinical laboratory geneticists.
This couple accepted genetic counselling so that the consequences of this translo-
cation in this phenotypically normal father for future pregnancies and the need for
family studies (parents, brothers, sisters, and other family members at risk) were out-
lined. Also all carriers in the family should be offered invasive prenatal diagnosis,
when expecting a child.
CONCLUSION
It was possible to infer that the alteration of the fetus was inherited from the male
progenitor (Figure 11.14 C). The explanation for the normal result obtained using
FISH for the rapid detection of aneuploidies is because this fetus carries an unbal-
anced structural chromosomal rearrangement involving chromosomes 13 and 21 and
the FISH probe used for rapid detection of aneuploidies is located more distally at
21q22.13-q22.2; and since this region is present in the derivative chromosome 13, the
FISH study showed two signals for chromosome 21.
CLINICAL CASE 4
CLINICAL INFORMATION
In a 14-month-old girl, hypotonia due to damage to the 1st neuron, agenesis of the
corpus callosum, and macrocephaly were diagnosed. Peripheral blood was taken for
cytogenetic analyses.
TECHNIQUES PERFORMED
G banding cytogenetics;
146 Cytogenetics and Molecular Cytogenetics
RESULTS
Karyotyping showed a normal result, 46,XX (data not shown).
FISH using subtelomeric probes for 7q36.3 and 22q13.3 revealed a partial mono-
somy of the terminal region of the long arm of chromosome 22 and a partial trisomy
of the terminal region of the long arm of chromosome 7, 46,XX.ish der(22)t(7;22)
(q36.3;q13.3)(MS607-,VIJyRM2000+) (Figure 11.15).
QUESTIONS
i) What kind of chromosomal alteration was found? How to interpret the
results? What is the most probable mechanism of origin of the alteration?
FIGURE 11.15 Family with t(7;22): Banding and molecular cytogenetics results obtained
are shown for
A) the affected daughter.
B) the affected brother.
C) the carrier mother.
FISH—Characterization of Chromosomal Alterations 147
which was also referred for G banding and molecular cytogenetic study. These
studies revealed a partial monosomy of the terminal region of chromosome 7q
and a partial trisomy of the terminal region of the chromosome 22q, 46,XY.ish
der(7)t(7;22)(q36.3;q13.3)(VIJyRM2000-,MS607+). The presence of this deriva-
tive 7 from a translocation involving chromosomes 7 and 22 seems to explain the
phenotype of this boy. These alterations are the exact opposite of those observed
in the sister (Figure 11.15 B).
Once again, genetic counselling was offered and all implications for the specifc
situation explained further, including prenatal diagnosis for future pregnancies and
the need for additional studies offered to relatives of the carrier. As chromosome 7
is involved in the rearrangement, also the hint should be given that this imprinting
associated chromosome is slightly more likely to be involved in UPD due to the
t(7;22). Thus, prenatal UPD test may also be considered in future pregnancies.
CONCLUSION
These two different unbalanced chromosomal structural rearrangements in these
siblings involving the same two chromosomes (7 and 22) were inherited from the
female progenitor and seem to explain their respective phenotypes.
CONCLUSIONS
The complex behavior of chromosomes during the two meiotic divisions is error-
prone. Aneuploidy can affect any chromosome; however, only trisomies of the sex
chromosomes, of autosomes 13, 18, or 21, and monosomy of the X chromosome,
are compatible with survival up to the end of pregnancy, the remainder having been
eliminated by natural selection. Structural alterations originated from chromosomal
breakage and rejoining could affect the number, distribution, and expression of
genes with a signifcant impact in the offspring phenotype. Couples at particularly
increased risk of having offspring with a numerical or structural alteration are those:
• where one partner has gonadal or germinal mosaic for a trisomic cell line.
Couples with various conceptions involving the same trisomy give a clear
evidence of a gonadal mosaicism for a trisomic cell line affecting the pri-
mordial germ cells.
DEDICATION
To all our patients and their families, to everyone our laboratory (Cytogenetics and
Genomics Laboratory of the Faculty of Medicine of the University of Coimbra) and
clinical colleagues, to our students
REFERENCES
1. Pezza, R.J. Mechanisms of chromosome segregation in meiosis: New views on the old
problem of aneuploidy. FEBS J. 2015, 282, 2424–2425.
2. Petronczki, M.; Siomos, M.F.; Nasmyth, K. Un ménage à quatre: The molecular biology
of chromosome segregation in meiosis. Cell. 2003, 112, 423–440.
3. Greaney, J.; Wei, Z.; Homer, H. Regulation of chromosome segregation in oocytes and
the cellular basis for female meiotic errors. Hum. Reprod. Update. 2018, 24, 135–161.
4. Villeneuve, A.M.; Hillers, K.J. Whence meiosis? Cell. 2001, 106, 647–650.
5. Duro, E.; Marston, A.L. From equator to pole: Splitting chromosomes in mitosis and
meiosis. Genes Dev. 2015, 29, 109–122.
6. Storchova, Z. Chromosomal numerical aberrations. In: eLS. John Wiley & Sons, Ltd,
Chichester, 2016.
7. Queremel Milani, D.A.; Tadi, P. Genetics, Chromosome Abnormalities. Treasure Island,
FL, StatPearls Publishing; 2022.
8. Delhanty, J.D. Origins of human aneuploidy. In: eLS. John Wiley & Sons, Ltd, Chichester,
2018.
9. Mahdieh, N.; Rabbani, B. An overview of mutation detection methods in genetic disor-
ders. Iran J. Pediatr. 2013, 23, 375–388.
10. Hassold, T.; Hunt, P. To err (meiotically) is human: The genesis of human aneuploidy.
Nat. Rev. Genet. 2001, 2, 280–291.
11. McFadden, D.E.; Friedman, J.M. Chromosome abnormalities in human beings. Mutat.
Res. 1997, 396, 129–140.
12. Nagaoka, S.I.; Hassold, T.J.; Hunt, P.A. Human aneuploidy: Mechanisms and new
insights into an age-old problem. Nat. Rev. Genet. 2012, 13, 493–504.
13. Ottolini, C.S.; Newnham, L.; Capalbo, A.; Natesan, S.A.; Joshi, H.A.; Cimadomo, D.;
Griffn, D.K.; Sage, K.; Summers, M.C.; Thornhill, A.R.; Housworth, E.; Herbert, A.D.;
FISH—Characterization of Chromosomal Alterations 149
Rienzi, L.; Ubaldi, F.M.; Handyside, A.H.; Hoffmann, E.R. Genome-wide maps of
recombination and chromosome segregation in human oocytes and embryos show selec-
tion for maternal recombination rates. Nat. Genet. 2015, 47, 727–735.
14. Holubcová, Z.; Blayney, M.; Elder, K.; Schuh, M. Human oocytes: Error-prone chro-
mosome-mediated spindle assembly favors chromosome segregation defects in human
oocytes. Science. 2015, 348, 1143–1147.
15. Luthardt, F.W.; Keitges, E. Chromosomal syndromes and genetic disease. In: eLS. John
Wiley & Sons, Ltd, Chichester, 2001.
16. McFeely, R.A. Chromosome abnormalities. Vet. Clin. North Am. Food Anim. Pract.
1993, 9, 11–22.
17. McFadden, D.E.; Robinson, W.P. Phenotype of triploid embryos. J. Med. Genet. 2006,
43, 609–612.
18. Zaragoza, M.V.; Surti, U.; Redline, R.W.; Millie, E.; Chakravarti, A.; Hassold, T.J.
Parental origin and phenotype of triploidy in spontaneous abortions: Predominance of
diandry and association with the partial hydatidiform mole. Am. J. Hum. Genet. 2000,
66, 1807–1820.
19. McFadden, D.E.; Langlois, S. Parental and meiotic origin of triploidy in the embryonic
and fetal periods. Clin. Genet. 2000, 58, 192–200.
20. Gardner, R.J.M.; Amor, D.J. Gardner and Sutherland’s Chromosome Abnormalities and
Genetic Counseling. 5th Edition. Oxford University Press, Oxford; 2018.
21. Filges, I.; Manokhina, I.; Peñaherrera, M.S.; McFadden, D.E.; Louie, K.; Nosova, E.;
Friedman, J.M.; Robinson, W.P. Recurrent triploidy due to a failure to complete maternal
meiosis II: Whole-exome sequencing reveals candidate variants. Mol. Hum. Reprod.
2015, 21, 339–346.
22. Moore, C.M.; Best, R.G. Chromosomal genetic disease: Structural aberrations. In eLS.
John Wiley & Sons, Ltd, Chichester, 2001.
23. ISCN 2020. An International System for Human Cytogenomic Nomenclature (2020);
McGowan-Jordan J.; Hastings, R.J.; Moore, S., Eds. Karger, Basel; 2020.
24. Speicher, M.R.; Carter, N.P. The new cytogenetics: Blurring the boundaries with molec-
ular biology. Nat. Rev. Genet. 2005, 6, 782–792.
25. Potapova, T.; Gorbsky, G.J. The consequences of chromosome segregation errors in
mitosis and meiosis. Biology (Basel). 2017, 6, 12.
26. Hook, E.B.; Cross, P.K. Rates of mutant and inherited structural cytogenetic abnormali-
ties detected at amniocentesis: Results on about 63,000 fetuses. Ann. Hum. Genet. 1987,
51, 27–55.
27. Clancy, S.; Shaw, K. DNA deletion and duplication and the associated genetic disorders.
Nat. Educ 2008, 1, 23.
28. Sharp, A.J.; Locke, D.P.; McGrath, S.D.; Cheng, Z.; Bailey, J.A.; Vallente, R.U.; Pertz,
L.M.; Clark, R.A.; Schwartz, S.; Segraves, R.; Oseroff, V.V.; Albertson, D.G.; Pinkel, D.;
Eichler, E.E. Segmental duplications and copy-number variation in the human genome.
Am. J. Hum Genet. 2005, 77, 78–88.
29. Sigurdardottir, S.; Goodman, B.K.; Rutberg, J.; Thomas, G.H.; Jabs, E.W.; Geraghty,
M.T. Clinical, cytogenetic, and fuorescence in situ hybridization fndings in two cases of
“complete ring” syndrome. Am. J. Med. Genet. 1999, 87, 384–390.
30. Guilherme, R.S.; Meloni, V.F.; Kim, C.A.; Pellegrino, R.; Takeno, S.S.; Spinner, N.B.;
Conlin, L.K.; Christofolini, D.M.; Kulikowski, L.D.; Melaragno, M.I. Mechanisms of
ring chromosome formation, ring instability and clinical consequences. BMC Med.
Genet. 2011, 12, 171.
31. Caba, L.; Rusu, C.; Plăiaşu, 5th.; Gug, G.; Grămescu, M.; Bujoran, C.; Ochiană, D.;
Voloşciuc, M.; Popescu, R.; Braha, E.; Pânzaru, M.; Butnariu, L.; Sireteanu, A.; Covic,
150 Cytogenetics and Molecular Cytogenetics
M.; Gorduza, E. Ring autosomes: Some unexpected fndings. Balkan J. Med. Genet.
2012, 15, 35–46.
32. Knijnenburg, J.; van Haeringen, A.; Hansson, K.B.; Lankester, A.; Smit, M.J.; Belfroid,
R.D.; Bakker, E.; Rosenberg, C.; Tanke, H.J.; Szuhai, K. Ring chromosome formation as
a novel escape mechanism in patients with inverted duplication and terminal deletion.
Eur. J. Hum. Genet. 2007, 15, 548–545.
33. Yip, M.Y. Autosomal ring chromosomes in human genetic disorders. Transl Pediatr.
2015, 4, 164–174.
34. Schinzel, A. Catalogue of Unbalanced Chromosome Aberrations in Man Berlin. Walter
de Gruyter. Basel; 2001.
35. Hemmat, M.; Hemmat, O.; Boyar, F.Z. Isochromosome Yp and jumping translocation of
Yq resulting in fve cell lines in an infertile male: A case report and review of the litera-
ture. Mol. Cytogenet. 2013, 6, 36.
36. Liehr, T.; Claussen, U.; Starke, H. Small supernumerary marker chromosomes (sSMC)
in humans. Cytogenet Genome Res. 2004, 107, 55–67.
37. Therman, E.; Susman, B.; Denniston, C. The nonrandom participation of human acro-
centric chromosomes in Robertsonian translocations. Ann. Hum. Genet. 1989, 53,
49–65.
38. Van Hemel, J.O.; Eussen, H.J. Interchromosomal insertions: Identifcation of fve cases
and a review. Hum Genet. 2000, 107, 415–432.
39. Neill, N.J.; Ballif, B.C.; Lamb, A.N.; Parikh, S.; Ravnan, J.B.; Schultz, R.A.; Torchia,
B.S.; Rosenfeld, J.A.; Shaffer, L.G. Recurrence, submicroscopic complexity, and poten-
tial clinical relevance of copy gains detected by array CGH that are shown to be unbal-
anced insertions by FISH. Genome Res. 2011, 21, 535–544.
40. Kang, S.H.; Shaw, C.; Ou, Z.; Eng, P.A.; Cooper, M.L.; Pursley, A.N.; Sahoo, T.; Bacino,
C.A.; Chinault, A.C.; Stankiewicz, P.; Patel, A.; Lupski, J.R.; Cheung, S.W. Insertional
translocation detected using FISH confrmation of array-comparative genomic hybrid-
ization (aCGH) results. Am. J. Med. Genet. A. 2010, 152, 1111–1126.
41. Nowakowska, B.A.; de Leeuw, N.; Ruivenkamp, C.A.; Sikkema-Raddatz, B.; Crolla,
J.A.; Thoelen, R.; Koopmans, M.; den Hollander, N.; van Haeringen, A.; van der Kevie-
Kersemaekers, A.M.; Pfundt, R.; Mieloo, H.; van Essen, T.; de Vries, B.B.; Green, A.;
Reardon, W.; Fryns, J.P.; Vermeesch, J.R. Parental insertional balanced translocations
are an important cause of apparently de novo CNVs in patients with developmental
anomalies. Eur. J. Hum. Genet. 2012, 20, 166–170.
42. Durand, C.M.; Betancur, C.; Boeckers, T.M.; Bockmann, J.; Chaste, P.; Fauchereau, F.;
Nygren, G.; Rastam, M.; Gillberg, I.C.; Anckarsäter, H.; Sponheim, E.; Goubran-Botros,
H.; Delorme, R.; Chabane, N.; Mouren-Simeoni, M.C.; de Mas, P.; Bieth, E.; Rogé, B.;
Héron, D.; Burglen, L.; Gillberg, C.; Leboyer, M.; Bourgeron, T. Mutations in the gene
encoding the synaptic scaffolding protein SHANK3 are associated with autism spec-
trum disorders. Nat. Genet. 2007, 39, 25–27.
43. Phelan, M.C. Deletion 22q13.3 syndrome. Orphanet. J. Rare Dis. 2008, 3, 14.
44. Cammarata-Scalisi, F.; Callea, M.; Martinelli, D.; Willoughby, C.E.; Tadich, A.C.;
Araya Castillo, M.; Lacruz-Rengel, M.A.; Medina, M.; Grimaldi, P.; Bertini, E.; Nevado,
J. Clinical and genetic aspects of Phelan-McDermid syndrome: An interdisciplinary
approach to management. Genes (Basel). 2022, 13, 504.
12 Multicolor-FISH—
Methods and
Applications
Thomas Liehr
CONTENTS
Introduction ............................................................................................................ 151
Samples/Tissues ..................................................................................................... 152
Description of Methods.......................................................................................... 152
Yields ..................................................................................................................... 154
Conclusions ............................................................................................................ 154
References .............................................................................................................. 154
INTRODUCTION
Since their establishment in routine (cyto-)genetic diagnostics in the mid-1990s, mul-
ticolor fuorescence in situ hybridization (mFISH) approaches have become indis-
pensable for the identifcation and description of chromosomal rearrangements. The
subsequent exact characterization of chromosomal breakpoints is of superior clinical
impact and is the requisite condition for further molecular investigations aimed at
the identifcation of disease-related genes.[1, 2] Thus, various approaches for a dif-
ferentiation of chromosomal subbands based on mFISH assays were established.[3]
Here an overview is presented on available mFISH approaches, with special focus
on mFISH-banding methods including their advantages, limitations, and possible
applications.
Although the G-bands by trypsin using Giemsa (GTG) banding technique is still
the starting point of most (molecular) cytogenetic test requests, its technical restric-
tions are well known (e.g., only chromosome morphology combined with a black and
white banding pattern can be evaluated). Nonetheless, a quick and not costly over-
view on eventual gross changes within the human whole genome can be achieved by
that cytogenetic banding approach.[2]
mFISH methods using all 24 human whole chromosome painting probes simulta-
neously, such as multiplex-FISH (M-FISH), spectral karyotyping (SKY), combined
ratio labeling FISH (COBRA-FISH), and others,[3] reach their limits in the identi-
fcation of intrachromosomal rearrangements (such as small interstitial deletions,
duplications, and inversions without change of the centromeric index) and when
exact characterization of breakpoints is required. These limitations were overcome
by the development of new mFISH probe sets during the last decades. There were
many tumor-[3, 4] and clinical-cytogenetics[2, 3] -oriented probe sets established for the
human genome, all of them being suited for any kind of imaginative research appli-
cation,[2, 5] but also mFISH-probe sets for animal,[3] plant,[3] or even bacteria.[3, 5] Here
we concentrate on human mFISH probe sets suited for FISH-banding.[3, 6]
FISH banding methods are defned as “any kind of FISH technique, which pro-
vides the possibility to characterize simultaneously several chromosomal subregions
smaller than a chromosome arm (excluding the short arms of the acrocentric chro-
mosomes)”.[6] In contrast to the standard cytogenetic chromosome banding tech-
niques, giving a protein-related banding pattern,[2] the FISH banding techniques are
DNA-mediated.
SAMPLES/TISSUES
In routine settings, mFISH-banding is applicable in each cytogenetic preparation
derived from dividing cells on metaphase spreads. These are normally tumor cells,
cells derived from bone marrow, blood or fbroblasts (normally limited to fetal tis-
sues including chorion, amnion, and abortion material or skin fbroblasts in postna-
tal cases).[5] In research, also interphase cells from each tissue, including archival
formalin-fxed paraffn embedded or cryofxed tissues.[3, 5]
DESCRIPTION OF METHODS
Five different FISH banding methods are available at present, which differ in their
probe composition as well as in their banding resolution:
FIGURE 12.1 MCB pseudo-color pattern for all 24 human chromosomes in a ~380-band
level. Two homologue autosomes and one gonosome each are presented. The chromosomes
depicted here have been put together from 24 different MCB experiments.
154 Cytogenetics and Molecular Cytogenetics
FISH banding approaches such as the CBC-technique using locus specifc probes,
region-specifc human–hamster somatic cell hybrids, or nonoverlapping microdis-
section libraries (SCAN) have, per defnition, the disadvantage that unstained and
non-informative gaps are left along the chromosome. Such gaps can cause problems,
as breakpoints within the unstained gaps cannot exactly be described. Conversely,
techniques based on locus-specifc probes would theoretically provide the advan-
tage that chromosomal breakpoints could be defned very exactly by the correspond-
ing breakpoint-spanning or fanking clones. The IPM-FISH approach, the Rx-FISH
technique, the Rx-FISH combined with somatic cell hybrids, and the MCB methods
provide the advantage of leaving no non-informative gaps.
The MCB probe sets are commercially available as m-banding (MetaSystems,
Altlussheim, Germany) or are available on request at [email protected].
de. Besides, probes can be prepared as previously described using chromosome
microdissection.[15]
YIELDS
Only above mentioned FISH banding probe sets (1), (4), and (5) are fnished for
the whole human genome and can be applied to achieve comprehensive informa-
tion in one single hybridization step. Concerning banding resolution, MCB has
the highest and most fexible one. Additionally, according to the question to be
studied, it can be chosen if only selected chromosomes or the whole genome shall
be hybridized.
CONCLUSIONS
The introduction of FISH banding methods was a great step forward for molec-
ular cytogenetic diagnostics. However, none of the mentioned new methods can,
for technical reasons, ever be fully informative for itself. The cytogeneticist always
has to double check the results obtained in FISH banding with those achieved by
other approaches, such as GTG banding, M-FISH or SKY, chromosomal microarray
analyses (CMA), sequencing or others. Examples for that modus operandi can be
found in the literature.[4] Nevertheless, the goal must be to achieve fully informative
cytogenetic results in a minimum of time and with a minimum of FISH experiments.
Thus, further developments with respect to probe combinations (such as M-FISH/
SKY with FISH banding methods) will be necessary and are on the way.[19] To per-
form and evaluate such complex experiments, which then will have to be based on up
to nine fuorochromes, further technical developments in microscopy and computer
software would be needed.
REFERENCES
1. Liehr, T. Molecular cytogenetics in the era of chromosomics and cytogenomic
approaches. Front. Genet. 2021, 12, 720507.
2. Liehr, T. Cytogenomics. Academic Press, New York; 2021.
Multicolor-FISH—Methods and Applications 155
CONTENTS
Introduction ............................................................................................................ 157
Samples to Be Characterized ................................................................................. 158
Centromere- and Heterochromatin-Specifc FISH Approaches ............................. 158
Yields ..................................................................................................................... 160
Conclusions ............................................................................................................ 160
Acknowledgments.................................................................................................. 160
References .............................................................................................................. 160
INTRODUCTION
The human genome project has been declared to be fnished by 2003.[1] However, in
the almost two decades since then, it has been a well-known truism that there was
almost 10% of the DNA-sequence still not accessed, yet.[2] This affects especially
the highly repetitive DNA-stretches, mainly being concentrated in the highly variant
heterochromatic parts of the human genome.[3] This gap was just recently closed by
Evan Eichler and colleagues for a single genome.[2] Nonetheless, heterochromatic
regions of the human genome are understudied and cannot be accessed by standard
molecular genetic approaches, including sequencing, as of now.[4]
Thus, it is also not surprising that since 1996 different fuorescence in situ hybrid-
ization (FISH) probe sets were available to characterize euchromatic material along
marker or derivative chromosomes.[5] However until 2001, whole genome–oriented
heterochromatic FISH-probe sets were not established; the frst then available one
was specifc for all human centromeres.[6] It even lasted until 2012 until a probe set
for all large heterochromatic regions in the human genome was developed.[7] The
two mentioned probe sets are suited for the one-step-characterization of the chro-
mosomal origin in small supernumerary marker chromosomes (sSMCs),[8, 9] or larger
derivative chromosomes with rearrangements including human hererochromatin.[10]
SAMPLES TO BE CHARACTERIZED
In about 50–70% of cases with sSMCs, the derivatives are known to originate from
chromosome 15.[8, 9] Among the remaining cases, there is: 1) a great variation in chro-
mosomal and parental origin, 2) possibly a mosaicism, 3) the possibility of genomic
imprinting effects, and 4) of homozygosity of autosomal recessively inherited muta-
tions in uniparental isodisomy (UPD). As great dissimilarities in their clinical out-
comes are reported, too, the characterization of prenatally detected, particularly de
novo sSMCs, is of special interest for genetic counseling and appropriate medical
care. Characterization of one or more sSMCs in a patient should be followed by test-
ing for UPD, as the latter can cause clinical signs and symptoms even in heterochro-
matic sSMC cases, as well.[8, 9, 11, 12]
Many sSMCs consist only of heterochromatic material, and here it is important to
distinguish those from such cases with potentially deleterious euchromatic material
on them.[8, 9] Besides, there are other incidences where larger derivatives may either
include enlarged heterochromatin (chromosomal heteromorphisms), which may be
cytogenetically similar to deleterious unbalanced rearrangements.[3, 10]
YIELDS
In Figure 13.1, the possible yield of application of (sub-)cenM-FISH probe sets is
shown—i.e. the characterization of origin and content of an sSMC. As summarized
elsewhere, only molecular cytogenetics can reliably characterized sSMCs, especially
in infertile patients.[15]
CONCLUSIONS
In summary, nowadays the detection of an sSMC does not stop after GTG banding,
NOR staining, and exclusion of a chromosome 15 origin. It can be exactly described
after (acro)cenM-FISH and/or HCM-FISH, subcenM-FISH provides information on
additional euchromatin, and molecular genetics uncovers eventually present UPD.
CMA can only be applied in euchromatic sSMCs without mosaicism.[15] Meanwhile,
a relatively good correlation of sSMC content and clinics is available,[8] being
extremely helpful in genetic counseling.
ACKNOWLEDGMENTS
The case shown in Figure 13.1 was provided by Drs. Cramer and Hickmann, Essen,
Germany.
REFERENCES
1. https://en.wikipedia.org/wiki/Human_Genome_Project [accessed on 01/12/2022].
2. Nurk, S.; Koren, S.; Rhie, A.; Rautiainen, M.; Bzikadze, A.V.; Mikheenko, A.; Vollger,
M.R.; Altemose, N.; Uralsky, L.; Gershman, A.; Aganezov, S.; Hoyt, S.J.; Diekhans, M.;
Logsdon, G.A.; Alonge, M.; Antonarakis, S.E.; Borchers, M.; Bouffard, G.G.; Brooks,
S.Y.; Caldas, G.V.; Cheng, H.; Chin, C.-S.; Chow, W.; de Lima, L.G.; Dishuck, P.C.;
Durbin, R.; Dvorkina, T.; Fiddes, I.T.; Formenti, G.; Fulton, R.S.; Fungtammasan, A.;
Garrison, E.; Grady, P.G.S.; Graves-Lindsay, T.-A.; Hall, I.M.; Hansen, N.F.; Hartley,
G.A.; Haukness, M.; Howe, K.; Hunkapiller, M.W.; Jain, C.; Jain, M.; Jarvis, E.D.;
Kerpedjiev, P.; Kirsche, M.; Kolmogorov, M.; Korlach, J.; Kremitzki, M.; Li, H.; Maduro,
V.V.; Marschall, T.; McCartney, A.M.; McDaniel, J.; Miller, D.E.; Mullikin, J.C.; Myers,
E.W.; Olson, N.D.; Paten, B.; Peluso, P.; Pevzner, P.A.; Porubsky, D.; Potapova, T.; Rogaev,
E.I.; Rosenfeld, J.A.; Salzberg, S.L.; Schneider, V.A.; Sedlazeck, F.J.; Shafn, K.; Shew,
C.J.; Shumate, A.; Sims, Y.; Smit, A.F.A.; Soto, D.C.; Sović, I.; Storer, J.M.; Streets, A.;
Sullivan, B.A.; Thibaud-Nissen, F.; Torrance, J.; Wagner, J.; Walenz, P.P.; Wenger, A.;
Wood, J.M.D.; Xiao, C.; Yan, S.M.; Young, A.C.; Zarate, S.; Surti, U.; McCoy, R.C.;
Dennis, M.Y.; Alexandrov, I.A.; Gerton, J.L.; O’Neill, R.J.; Timp, W.; Zook, J.M.;
Schatz, M.C.; Eichler, E.E.; Miga, K.H.; Phillippy, A.M. The complete sequence of a
human genome. bioRxiv. 2021. preprint doi: https://doi.org/10.1101/2021.05.26.445798.
3. Liehr, T. Benign & Pathological Chromosomal Imbalances, 1st Edition Microscopic
and Submicroscopic Copy Number Variations (CNVs) in Genetics and Counseling.
Academic Press, New York; 2014.
4. Liehr, T. Repetitive elements in humans. Int. J. Mol. Sci. 2021, 22, 2072.
5. Liehr, T. Molecular cytogenetics in the era of chromosomics and cytogenomic
approaches. Front. Genet. 2021, 12, 720507.
Multicolor-FISH—Centromere- and Heterochromatin-Specifc 161
6. Nietzel, A.; Rocchi, M.; Starke, H.; Heller, A.; Fiedler, W.; Wlodarska, I.; Loncarevic,
I.F.; Beensen, V.; Claussen, U.; Liehr, T. A new multicolor-FISH approach for the charac-
terization of marker chromosomes: Centromere-specifc multicolor-FISH (cenM-FISH).
Hum. Genet. 2001, 108, 199–204.
7. Bucksch, M.; Ziegler, M.; Kosayakova, N.; Mulatinho, M.V.; Llerena, J.C. Jr.; Morlot,
S.; Fischer, W.; Polityko, A.D.; Kulpanovich, A.I.; Petersen, M.B.; Belitz, B.; Trifonov,
V.; Weise, A.; Liehr, T.; Hamid, A.B. A new multicolor fuorescence in situ hybridiza-
tion probe set directed against human heterochromatin: HCM-FISH. J. Histochem.
Cytochem. 2012, 60, 530–536.
8. Liehr, T. Small supernumerary marker chromosomes. http://cs-tl.de/DB/CA/sSMC/0-
Start.html [accessed on 01/12/2022].
9. Liehr, T. Small Supernumerary Marker Chromosomes (sSMC): A Guide for Human
Geneticists and Clinicians: With Contributions by UNIQUE (The Rare Chromosome
Disorder Support Group). Springer, Berlin; 2012.
10. Liehr, T. Cases with heteromorphisms. http://cs-tl.de/DB/CA/HCM/0-Start.html
[accessed on 01/12/2022].
11. Liehr, T. Uniparental Disomy (UPD) in Clinical Genetics: A Guide for Clinicians and
Patients. Springer, Berlin; 2014.
12. Liehr, T. Cases with uniparental disomy. http://cs-tl.de/DB/CA/UPD/0-Start.html
[accessed on 01/12/2022].
13. Trifonov, V.; Seidel, J.; Starke, H.; Prechtel, M.; Beensen, V.; Ziegler, M.; Hartmann, I.;
Heller, A.; Nietzel, A.; Claussen, U.; Liehr, T. Enlarged chromosome 13 p-arm hiding a
cryptic partial trisomy 6p22.2-pter. Prenat. Diagn. 2003, 23, 427–430.
14. Weise, A.; Liehr, T. Subtelomeric and/or subcentromeric probe sets. In: Fluorescence
In Situ Hybridization (FISH): Application Guide; Liehr, T., Ed. 2nd Edition. Springer,
Berlin, 2017, pp. 261–269.
15. Liehr, T.; Hamid Al-Rikabi, A.B. Impaired spermatogenesis due to small supernumerary
marker chromosomes: The reason for infertility is only reliably ascertainable by cytoge-
netics. Sex. Dev. 2018, 12, 281–287.
14 FISH—Detection
of Individual Radio
Sensitivity
Thomas Liehr
CONTENTS
Introduction ............................................................................................................ 163
Samples/Tissues ..................................................................................................... 164
Description of Methods.......................................................................................... 165
One- to Three-Color FISH ............................................................................ 165
24-Color FISH .............................................................................................. 166
Multicolor Banding ....................................................................................... 166
Specifc Methodological Details for M-FISH Evaluation...................................... 166
Yields ..................................................................................................................... 167
Conclusions ............................................................................................................ 167
References .............................................................................................................. 167
INTRODUCTION
In radiotherapy, alongside tumor control, the prevention of severe treatment-related
side effects is a major concern. Although the majority of radio-oncological patients
tolerate a standard treatment protocol, toxic side effects can be detected in 0.2–10%
of the cases.[1, 2] The latter can be a result of increased individual radio sensitiv-
ity, caused by a combination of exogenous and endogenous factors including genetic
reasons. However, most of the exact underlying still mechanisms remain unknown.
Only in few cases is increased radio sensitivity a result of an identifed single gene
mutation, e.g., in ataxia telangiectasia (AT) or Nijmegen breakage syndrome (NBS)
patients, OMIM #208900 and #251260,[3] but in the majority, it is supposed to be
modulated by a mixture of different genes.[4]
To have a chance to detect a possible overreaction prior to therapy, the availabil-
ity of a predictive test system has been the aim for some decades. Although different
approaches were tested (e.g., clonogenic cell survival,[5] G2-assay,[6] comet assay[7]),
there is still no reliable routine assay to identify hypersensitive or less sensitive patients
to adjust their therapeutic dose. It is known that chromosomal aberrations 1) are indica-
tors of a previous exposure to irradiation and 2) can be used to estimate radio sensitiv-
ity. Previous studies demonstrated that molecular cytogenetic methods are superior to
conventional cytogenetic analysis in detection and characterization of aberrations. At
this, the frequency of breaks and the occurrence of specifc aberration types refect
individual sensitivity to radiation. Especially complex chromosomal aberrations (CCR)
were identifed as indicators for increased individual radio sensitivity.
To detect individual radio sensitivity by fuorescence in situ hybridization (FISH),
three approaches are presently available: i) painting of one up to three different chro-
mosomes in one or different colors, ii) painting of all 24 human chromosomes in
different colors (24-color FISH), and iii) FISH-based chromosome banding for the
detection of intra-chromosomal rearrangements.
SAMPLES/TISSUES
Our own studies have focused on the analysis of in vitro-induced chromosomal
aberrations in NBS and AT homozygote and heterozygote individuals compared
with normal reacting controls (Figures 14.1 and 14.2).[8, unpublished data] The aim was to
FIGURE 14.2 For the graphic depicted here, four normal controls, two Nijmegen breakage
syndrome (NBS), and two ataxia telangiectasia (AT) heterozygotes (NBS-het, AT-het) plus
one NBS and one AT homozygote (NBS-hm, AT-hm) were included. (A) shows the average
number of breaks per mitosis, while (B) presents the average number of CCR occurring in
each mitosis for each of the fve groups. Three different in vitro irradiation doses were ana-
lyzed per patient. The applied doses were 0.0, 0.7, and 2.0 Gy; in the AT-hm patient, instead
of 2.0 Gy, 1.4 Gy was used. A clear differentiation was possible in both diagrams between
normal controls and the homozygote patients, as well as between controls and AT-het.
demonstrate that a difference in radio sensitivity which was already known between
these individuals is also detectable using the 24-color FISH technique. Thus, the
increased radio sensitivity of NBS and AT patients can serve as a positive control in
a predictive assay.
DESCRIPTION OF METHODS
ONE- TO THREE-COLOR FISH
FISH applying one up to three whole-chromosome painting (wcp) probes simulta-
neously was used to detect radiation-induced chromosome instability in peripheral
166 Cytogenetics and Molecular Cytogenetics
blood lymphocytes and fbroblasts.[9–16] However, in the analyses, the used chromo-
somes are selected by chance, although a random distribution of chromosomal rear-
rangements along the chromosomes is up to now still discussed controversially.[17–18]
To avoid this problem and to obtain an overview of the whole karyotype, 24-color
FISH can be applied using a probe mix of the 24 different human wcp probes.[8, 9, 19, 20]
24-COLOR FISH
This method, frst described in 1996,[21, 22] allows the simultaneous visualization of
all chromosomes within a metaphase in different specifc colors. Nearly all inter-
chromosomal aberrations (as reciprocal and nonreciprocal translocations, complex
rearrangements, ring chromosomes, acentric fragments, dicentric fragments, or
insertions) can be detected and defned in more detail.
24-color FISH has been used, for example, for studies on normal peripheral blood
lymphocytes irradiated in vitro,[8, 19] on radiotherapy-induced residual chromosomal
damage in peripheral lymphocytes,[9] on bone marrow cells of Chernobyl victims,[19]
and for analysis of chromosomal aberrations after irradiation with ionizing alpha
particles. The latter was also used for investigations on the formation of CCR.[23]
MULTICOLOR BANDING
FISH methods using all 24 human whole-chromosome painting probes simultane-
ously reach their limits in identifcation of intra-chromosomal rearrangements (such
as small interstitial deletions, duplications, and inversions without change of the cen-
tromeric index) and when exact characterization of breakpoints is required. These
limitations have been overcome by the development of FISH-banding methods dur-
ing the last decades.[25] In the meantime, one of these approaches, the multicolor
banding (MCB) technique (or mBAND),[26] was also used for the analysis of X-ray-
induced aberrations.[27, 28] However, only a probe set for chromosome 5 was applied,
demonstrating that intra chromosomal aberrations are present in a considerable por-
tion in radiation-induced changes.
YIELDS
The 24-color FISH is a highly informative approach concerning the characterization
of radiation-induced chromosomal rearrangements, but is based on a very sophisti-
cated and time-consuming evaluation procedure. A typical result for one metaphase
and a summary of obtained data are shown in Figures 14.1 and 14.2.
CONCLUSIONS
In summary, genetically determined intrinsic radiosensitivity can be detected and
quantifed by FISH approaches. Twenty-four-color FISH can be used as well as the
three-color FISH. FISH is as reliable but more informative than conventional cytoge-
netic data.[10, 11] As stated before, the 24-color FISH approach requires a very sophis-
ticated evaluation. However, data obtainable by this technique are indispensable to
decide whether there exist any hot spots for radiation-induced breakpoints. If so,
such hot spots could be characterized in detail by FISH-banding methods,[25] and
in the following, locus-specifc breakpoint-spanning probes could be applied in the
future for quick detection of individual radio sensitivity, possibly even in interphase
cytogenetics.
REFERENCES
1. Roberts, S.A.; Spreadborough, A.R.; Bulman, B.; Barber, J.B.; Evans, D.G.; Scott, D.
Heritability of cellular radiosensitivity: A marker of low-penetrance predisposition
genes in breast cancer? Am. J. Hum. Genet. 1999, 65, 784–794.
2. Rogers, P.B.; Plowman, P.N.; Harris, S.J.; Arlett, C.F. Four radiation hypersensitiv-
ity cases and their implications for clinical radiotherapy. Radiother. Oncol. 2000, 57,
143–154.
3. OMIM. www.ncbi.nlm.nih.gov/omim/ [accessed on 01/12/2022].
4. Turesson, I.; Nyman, J.; Holmberg, E.; Oden, A. Prognostic factors for acute and late skin
reactions in radiotherapy patients. Int. J. Radiat. Oncol. Biol. Phys. 1996, 36, 1065–1075.
168 Cytogenetics and Molecular Cytogenetics
5. Cole, J.; Arlett, C.F.; Green, M.H.; Harcourt, S.A.; Priestley, A.; Henderson, L.; Cole, H.;
James, S.E.; Richmond, F. Comparative human cellular radiosensitivity: II. The survival
following gamma-irradiation of unstimulated (G0) T-lymphocytes, T-lymphocyte lines,
lymphoblastoid cell lines and fbroblasts from normal donors, from ataxia-telangiectasia
patients and from ataxia-telangiectasia heterozygotes. Int. J. Radiat. Biol. 1988, 54,
929–943.
6. Sanford, K.K.; Parshad, R.; Price, F.M.; Jones, G.M.; Tarone, R.E.; Eierman, L.; Hale,
P.; Waldmann, T.A. Enhanced chromatid damage in blood lymphocytes after G2 phase
x irradiation, a marker of the ataxia-telangiectasia gene. J. Natl. Cancer Inst. 1990, 8,
1050–1054.
7. Hovhannisyan, G.G. Fluorescence in situ hybridization in combination with the comet
assay and micronucleus test in genetic toxicology. Mol. Cytogenet. 2010, 3, 17.
8. Kuechler, A.; Neubauer, S.; Grabenbauer, G.G.; Claussen, U.; Liehr, T.; Sauer, R.; Wendt,
T.G. Is 24-color FISH detection of in-vitro radiation-induced chromosomal aberrations
suited to determine individual intrinsic radiosensitivity? Strahlenther. Onkol. 2002, 178,
209–215.
9. Kuechler, A.; Dreidax, M.; Pigorsch, S.U.; Liehr, T.; Claussen, U.; Wendt, T.G.; Dunst,
J. Residual chromosomal damage after radiochemotherapy with and without amifostine
detected by 24-color FISH. Strahlenther. Onkol. 2003, 179, 493–498.
10. Neubauer, S.; Arutyunyan, R.; Stumm, M.; Dörk, T.; Bendix, R.; Bremer, M.; Varon, R.;
Sauer, R.; Gebhart, E. Radiosensitivity of ataxia telangiectasia and Nijmegen breakage
syndrome homozygotes and heterozygotes as determined by three-color FISH chromo-
some painting. Radiat. Res. 2002, 157, 312–321.
11. Gebhart, E.; Neubauer, S.; Schmitt, G.; Birkenhake, S.; Dunst, J. Use of three-color
chromosome in situ suppression technique for the detection of past radiation exposure.
Radiat. Res. 1996, 145, 47–52.
12. Neubauer, S.; Dunst, J.; Gebhart, E. The impact of complex chromosomal rearrange-
ments on the detection of radiosensitivity in cancer patients. Radiother. Oncol. 1997, 43,
189–195.
13. Stritzelberger, J.; Lainer, J.; Gollwitzer, S.; Graf, W.; Jost, T.; Lang, J.D.; Mueller, T.M.;
Schwab, S.; Fietkau, R.; Hamer, H.M.; Distel, L. Ex vivo radiosensitivity is increased in
non-cancer patients taking valproate. BMC Neurol. 2020, 20, 390.
14. Schuster, B.; Ellmann, A.; Mayo, T.; Auer, J.; Haas, M.; Hecht, M.; Fietkau, R.; Distel,
L.V. Rate of individuals with clearly increased radiosensitivity rise with age both in
healthy individuals and in cancer patients. BMC Geriatr. 2018, 18, 105.
15. Gryc, T.; Putz, F.; Goerig, N.; Ziegler, S.; Fietkau, R.; Distel, L.V.; Schuster, B. Idelalisib
may have the potential to increase radiotherapy side effects. Radiat. Oncol. 2017, 12,
109.
16. Rave-Fränk, M.; Virsik-Kopp, P.; Pradier, O.; Nitsche, M.; Grunefeld, S.; Schmidberger,
H. In vitro response of human dermal fbroblasts to X-irradiation: Relationship between
radiation-induced clonogenic cell death, chromosome aberrations and markers of prolif-
erative senescence or differentiation. Int. J. Radiat. Biol. 2001, 77, 1163–1174.
17. Barrios, L.; Miro, R.; Caballin, M.R.; Fuster, C.; Guedea, F.; Subias, A.; Egozcue, J.
Cytogenetic effects of radiotherapy. Breakpoint distribution in induced chromosome
aberrations. Cancer Genet. Cytogenet. 1989, 41, 61–70.
18. Cigarran, S.; Barrios, L.; Barquinero, J.F.; Caballin, M.R.; Ribas, M.; Egozcue, J.
Relationship between the DNA content of human chromosomes and their involvement in
radiation-induced structural aberrations, analysed by painting. Int. J. Radiat. Biol. 1998,
74, 449–455.
FISH—Detection of Individual Radio Sensitivity 169
19. Greulich, K.M.; Kreja, L.; Heinze, B.; Rhein, A.P.; Weier, H.G.; Bruckner, M.; Fuchs,
P.; Molls, M. Rapid detection of radiation-induced chromosomal aberrations in lympho-
cytes and hematopoietic progenitor cells by mFISH. Mutat. Res. 2000, 452, 73–81.
20. Liehr, T. Basics and literature on multicolor fuorescence in situ hybridization applica-
tion. http://cs-tl.de/DB/TC/mFISH/5-wcp-oth.html#2 [accessed on 01/12/2022].
21. Speicher, M.R.; Gwyn Ballard, S.; Ward, D.C. Karyotyping human chromosomes by
combinatorial multi-fuor FISH. Nat. Genet. 1996, 12, 368–375.
22. Schröck, E.; du Manoir, S.; Veldman, T.; Schoell, B.; Wienberg, J.; Ferguson-Smith,
M.A.; Ning, Y.; Ledbetter, D.H.; Bar-Am, I.; Soenksen, D.; Garini, Y.; Ried, T. Multicolor
spectral karyotyping of human chromosomes. Science. 1996, 273, 494–497.
23. Anderson, R.M.; Stevens, D.L.; Goodhead, D.T. M-FISH analysis shows that complex
chromosome aberrations induced by {alpha}-particle tracks are cumulative products of
localized rearrangements. Proc. Natl. Acad. Sci. U. S. A. 2002, 99, 12167–12172.
24. Savage, J.R.K.; Simpson, P.J. FISH “painting” pattern resulting from complex exchanges.
Mutat. Res. 1994, 312, 51–60.
25. Liehr, T. Molecular cytogenetics in the era of chromosomics and cytogenomic
approaches. Front. Genet. 2021, 12, 720507.
26. Liehr, T.; Heller, A.; Starke, H.; Rubtsov, N.; Trifonov, V.; Mrasek, K.; Weise, A.;
Kuechler, A.; Claussen, U. Microdissection based high resolution multicolor banding for
all 24 human chromosomes. Int. J. Mol. Med. 2002, 9, 335–339.
27. Johannes, C.; Chudoba, I.; Obe, G. Analysis of X-ray-induced aberrations in human
chromosome 5 using high-resolution multicolour banding FISH (mBAND). Chromosom.
Res. 1999, 7, 625–633.
28. Hande, M.P.; Azizova, T.V.; Geard, C.R.; Burak, L.E.; Mitchell, C.R.; Khokhryakov,
V.F.; Vasilenko, E.K.; Brenner, D.J. Past exposure to densely ionizing radiation leaves a
unique permanent signature in the genome. Am. J. Hum. Genet. 2003, 72, 1162–1170.
29. ISCN (2020). An International System for Human Cytogenomic Nomenclature;
McGowan-Jordan, J.; Hastings R.J.; Moore, S., Eds. S Karger, Basel; 1995.
15 FISH—Detection of CNVs
Tigran Harutyunyan, Anzhela Sargsyan
and Rouben Aroutiounian
CONTENTS
Introduction............................................................................................................ 171
Samples/Tissues/Materials..................................................................................... 173
Human Whole Blood Culture ....................................................................... 174
Slides with Metaphase Chromosomes .......................................................... 174
Pod-FISH Probes .......................................................................................... 174
Pretreatment and Postfxation ....................................................................... 174
Pod-FISH and Washing................................................................................. 175
Description of Methods.......................................................................................... 175
Human Whole Blood Cultivation ................................................................. 175
Preparation of Metaphase Spreads................................................................ 175
Homemade Pod-FISH Probes....................................................................... 176
Slide Pretreatment......................................................................................... 176
Pod-FISH Procedure..................................................................................... 176
Washing and Counterstaining of Slides ........................................................ 177
Yields ..................................................................................................................... 177
Analysis of CNVs Using ImageJ Program ................................................... 177
Conclusions............................................................................................................ 178
Acknowledgments.................................................................................................. 179
References.............................................................................................................. 179
INTRODUCTION
DNA copy number variations (CNVs) occur in the genome due to spontaneous and
induced gains, losses, and/or insertions of 50 bp (as repeats) to several Mb long DNA
segments leading to copy number changes of a particular DNA sequence within the
chromosomes.[1] Thus, CNVs are structural changes within chromosomes and are
distinguished from whole chromosome gains and losses which are a separate class
of cytogenetic alterations (i.e. aneuploidy). When the frequency of a given CNV in
a population is greater than 1%, it is referred to as a copy number polymorphism.
Mounting evidence demonstrates the importance of de novo CNVs as a source of
genomic variation and in development of pathogenic conditions. Therefore, the
identifcation of CNVs in human chromosomes is of crucial task from clinical and
research perspectives. However, their detection remains challenging.
Zarrei et al.[2] constructed a high-resolution CNV map of the human genome that
includes 2,057,368 variants (195,084 gains and 1,862,284 losses). It was shown that
CNVs have unequal frequency among chromosomes. The highest frequencies of
gains were observed in chromosomes 22 and Y, followed by chromosomes 16, 9, and
15; however, the highest proportions of variable sequences with losses were detected
in chromosomes 19, 22, and Y. In addition, higher proportions of CNVs were iden-
tifed in pericentromeric and subtelomeric regions of all chromosomes. Moreover,
approximately for 100 genes it has been shown that they may be completely deleted
without apparent phenotypic consequences.[2] Overall, it was shown that CNVs con-
tribute to 4.8–9.7% of the variability in the human genome.
There are several databases for CNVs of which the “Database of Genomic
Variants” is the largest host for data of structural variations in healthy subjects from
worldwide populations.[3] The recently developed database CNVIntegrate is hosting
data from both healthy persons and cancer patients, and enables comparisons of
CNV frequencies within multiple ethnic populations.[4]
Based on differences in breakpoint positions during genomic rearrangements,
CNVs are classifed as recurrent and non-recurrent. Recurrent CNVs typically
occur via non-allelic homologous recombination between region-specifc low-copy
repeats with >95% nucleotide sequence identity.[5] Therefore, recurrent CNVs have
the same DNA sequence at the rearrangement breakpoints and similar size in unre-
lated individuals. In contrast, non-recurrent CNVs are thought to originate due to
inappropriate repair of accidental DNA double-strand breaks in proliferating cells
via microhomology-mediated pathways. Thus, non-recurrent CNVs have unique
sequences at the breakpoints in each individual.[6] Localization of recurrent and non-
recurrent CNVs in the genome is of high importance for distinguishing benign and
pathogenic variants, as well as for the identifcation of environmental factors that
potentially are capable of inducing CNVs.
Mounting evidence demonstrates the role of CNVs in various pathological condi-
tions including severe intellectual disability, autism, schizophrenia, cardiovascular
diseases, and cancer.[7–10] However, clinical interpretation of CNVs is still a challeng-
ing task. Fortin et al.[11] identifed CNVs in patients suffering from inherited epilepsy
with febrile seizures plus (GEFS+) syndrome. Deletions in 15q11.2, 19p13.3, and
22q11.2 and duplication in 10q11.22 were detected in four patients from 12 fami-
lies. Nevertheless, the clinical signifcance of these CNVs is diffcult to determine
considering the small cohort and the absence of strong evidence demonstrating the
causative impact of the identifed CNVs. Here intrachromosomal interactions may be
suggested as an underlying cause, as recently shown to play a role in 2q37-deletion
syndrome.[12]
Very large (> 500 kb) and rare (<1%) CNVs are more likely to be detected in
pathological conditions than small and common ones. Nevertheless, the number
of genes within the CNV, their dosage sensitivity, and functions (e.g. protein-
coding or non-coding) are likely the determinants of the clinical signifcance of
CNVs.[13]
Recently, the American College of Medical Genetics and Genomics and the
Clinical Genome Resource have developed guideline for the classifcation and quan-
titative interpretation of constitutional CNVs (inherited and de novo). Thus, there
FISH—Detection of CNVs 173
are fve main categories for postnatal CNVs according to their fnal point values of
evidence scoring metrics: pathogenic (≥0.99), likely pathogenic (0.90–0.98), variant
of uncertain signifcance (−0.89–0.89), likely benign (−0.90 and −0.98), and benign
(≤−0.99).[14] The authors developed a publicly available CNV classifcation calculator
(http://cnvcalc.clinicalgenome.org/cnvcalc/) to facilitate the use of this semi-quanti-
tative system. This tool allows applying points for individual evidence categories for
a given CNV and automatically determines the CNV category. However, the authors
emphasize that these standards cannot be implemented for the acquired CNVs in
neoplasia while CNVs are nearly ubiquitous in cancers. Therefore, identifcation of
clinically relevant CNVs and analysis of their frequency are important issues in can-
cer diagnostics.
Accumulating evidence demonstrates the potential of the environmental muta-
gens and carcinogens to induce de novo CNVs. In particular, DNA damage and/
or DNA replication stress-inducing agents, such as ionizing radiation, asbes-
tos, benzo(a)pyrene, and hydroxyurea are known to cause non-recurrent CNVs in
humans and animal cells.[1] Considering the mechanistic differences between recur-
rent and non-recurrent CNVs it can be assumed that different groups of environmen-
tal mutagens are likely to induce a specifc type of CNVs. Therefore, currently used
genetic endpoints of mutagenicity testing should also include CNVs as a marker of
mutagenic impact. However, the methods of the identifcation of CNVs and their loci
that should be analyzed in the standard genotoxicity testing are frontiers of modern
mutagenesis and clinical genetics.
Currently, fuorescence in situ hybridization (FISH) is the most commonly
used diagnostic tool and the gold standard for the detection of CNVs of differ-
ent genes of clinical importance. Namely, in patients with invasive breast cancer,
FISH analysis for HER2/NEU1 typically demonstrates the clear presence or lack
of HER2/NEU1 amplifcation.[15] However, CNVs in homologous chromosomes
can be used to distinguish otherwise cytogenetically similar homologs from each
other. Moreover, the application of BAC (bacterial artifcial chromosomes) probes
specifc for CNV loci and allows identifcation of the parental origin of the homol-
ogous chromosomes in the offspring. Accordingly, parental-origin-determination-
FISH (pod-FISH) analysis was developed.[16] Analysis of variations in fuorescence
intensities between treated and untreated groups enables the detection of altera-
tions in CNV loci. In afatoxin B1 treated human peripheral blood lymphocytes,
application of pod-FISH revealed losses in 8p21.2 and 15q11.2 chromosome loci,[17]
while in lymphocytes irradiated with AREAL laser-driven electron accelerator
(CANDLE, Armenia), gains were identifed in 1p31.1, 7q11.22, 9q21.3, 10q21.1,
and 16q23.1 chromosome loci.[18]
In this chapter, the semi-automatic quantitative analysis of CNVs by pod-FISH is
described, which can be used for CNV-studies of clinical importance and the inves-
tigation of alterations induced by mutagens in by pod-FISH accessible CNV loci.
SAMPLES/TISSUES/MATERIALS
Pod-FISH allows the detection of CNVs on a single-cell level in any cells; how-
ever, whole blood lymphocytes are most suitable for the analysis of spontaneous or
174 Cytogenetics and Molecular Cytogenetics
induced CNVs, as not only interphases but also metaphases can be evaluated. Slides
for the pod-FISH analysis can be prepared from heparinized whole blood or blood
collected in EDTA containing vacutainers.
POD-FISH PROBES
• BACs for the specifc CNV loci[16]
• Nick translation kit (Cat. No. 11745808910, Roche)
• EDTA 0.5 M (Cat. No. 324506, Sigma-Aldrich)
• Ethanol 100 % (Cat. No. 459836, Sigma-Aldrich)
• tRNA (Cat. No. 10109541001, Sigma-Aldrich)
• Sodium acetate buffer solution 3 M, pH 5.2 (Cat. No. S7899, Sigma-Aldrich)
• Dextran sulfate buffer: 2 g + 10 ml 50 % deionized formamide/2×SSC/50
mM phosphate buffer for 3 h, 70°C. Store at −20°C
DESCRIPTION OF METHODS
Although pod-FISH is applicable for both interphase nuclei and metaphase chro-
mosomes, we recommend analyzing CNV signals in metaphase chromosomes,
which decreases the background noise and provides better resolution for the detec-
tion of small changes in fuorescence intensities of FISH signals. For the analysis of
radiation-induced CNVs, human whole blood should be exposed to radiation imme-
diately after sampling. For the analysis of chemical mutagen-induced CNVs, whole
blood should be cultured for 24 h before exposure.
SLIDE PRETREATMENT
1. Dehydrate slides in an ethanol series (70 %, 90 %, 100 %, 3 min each) and
air-dry at room temperature.
2. Add 0.5 ml of pepsin to 100 ml of pepsin pretreatment solution, and put
slides for 3–5 min in a Coplin jar at 37°C in the water bath.
3. Wash slides in the PBS for 5 min at room temperature.
4. Add 100 µl of postfx solution, and cover with coverslips for 10 min at room
temperature.
5. Remove coverslips; repeat step 3 and step 1.
POD-FISH PROCEDURE
1. Mix 1 µl of the probe with 7 µl dextran sulfate in the Eppendorf tube con-
taining COT DNA. The optimal concentration of the COT DNA should be
determined experimentally.
FISH—Detection of CNVs 177
YIELDS
ANALYSIS OF CNVS USING IMAGEJ PROGRAM
1. The fuorescence intensities of CNV signals can be measured using e.g.
the publically available ImageJ program.[19]
2. Capture CNV signals for each fuorochrome and separately save in “.TIF”
image.
3. Activate ImageJ and upload each image in the program by pressing
“Ctrl+O” and selecting images for evaluation.
4. Zoom in the area containing CNV signals by holding “Ctrl” and scrolling
the mouse (Figure 15.1A).
5. Select CNV signals using the “Freehand selection” tool (Figure 15.1B).
6. To obtain a numerical value of the CNV signal, select “Analyze” from the
command bar and select “Measure”, or simply press “Ctrl+M” after the
signal selection step. A “Results table” will occur with numerical values
of mean fuorescence intensity or mean area.
178 Cytogenetics and Molecular Cytogenetics
FIGURE 15.1 Example of the analysis of CNVs using pod-FISH. (A) CNV signals in
14q11.2 chromosome loci (Spectrum Green) are detected by pod-FISH. (B) In the ImageJ
program, CNV signals are selected using the “Freehand selection tool”, and the fuorescence
measurement values are presented in the “Results table”.
7. Repeat steps 5–7 for the second signal from a homologous chromosome.
Thus, the frst pair of signals will be measured, and so on (Figure 15.1B).
8. After measuring signals in the selected group of CNVs (e.g. control), go to
the “File” in the “Results table” and select “Save as” or press “Ctrl+S”.
9. After measuring signals in the control and the treatment groups, statistical
analysis can be performed to compare signal intensities within groups and
between groups.
10. This approach will allow the detection of even those CNVs that are not
detectable by fuorescence microscopy. In addition, the bias by the subjec-
tivity of the researcher visually analyzing the difference between CNV
signal intensities will signifcantly decrease.
CONCLUSIONS
Spontaneous CNVs are an important source of genomic variations and polymor-
phisms. However, de novo CNVs can have pathogenic consequences such as cancer
or neurological disorders. Moreover, induction of DNA strand breaks or inhibition
of DNA replication with environmental mutagens can also induce de novo CNVs
in human and animal cells. Therefore, we recommend including CNVs as a novel
genetic endpoint in mutagenicity testing of environmental factors. Alterations in
molecular-cytogenetically visible CNVs can be measured with the application of
the ImageJ program. Further studies with the application of standard mutagens are
required to establish threshold values of fuorescence intensities of CNVs, as well as
to determine CNV loci that can be useful for the determination of recurrent or non-
recurrent CNV-inducing agents.
FISH—Detection of CNVs 179
ACKNOWLEDGMENTS
This work was supported by the RA MESCS and BMBF (project #AG-01/20), and
RA MESCS (project #21AG-1F068).
REFERENCES
1. Hovhannisyan, G.; Harutyunyan, T.; Aroutiounian, R.; Liehr, T. DNA Copy number
variations as markers of mutagenic impact. Int. J. Mol. Sci. 2019, 20, 4723.
2. Zarrei, M.; MacDonald, J.R.; Merico, D.; Scherer, S.W. A copy number variation map of
the human genome. Nat. Rev. Genet. 2015, 16, 172–183.
3. MacDonald, J.R.; Ziman, R.; Yuen, R.K.; Feuk, L.; Scherer, S.W. The database of
genomic variants: A curated collection of structural variation in the human genome.
Nucleic Acids Res. 2014, 42, D986–D992.
4. Chattopadhyay, A.; Teoh, Z.H.; Wu, C.Y.; Juang, J.J.; Lai, L.C.; Tsai, M.H.; Wu, C.H.;
Lu, T.P.; Chuang, E.Y. CNVIntegrate: The frst multi-ethnic database for identifying
copy number variations associated with cancer. Database (Oxford). 2021, 2021, baab044.
5. Smajlagić, D.; Lavrichenko, K.; Berland, S.; Helgeland, Ø.; Knudsen, G.P.; Vaudel, M.;
Haavik, J.; Knappskog, P.M.; Njølstad, P.R.; Houge, G.; Johansson, S. Population preva-
lence and inheritance pattern of recurrent CNVs associated with neurodevelopmental
disorders in 12,252 newborns and their parents. Eur. J. Hum. Genet. 2021, 29, 205–215.
6. Conover, H.N.; Argueso, J.L. Contrasting mechanisms of de novo copy number muta-
genesis suggest the existence of different classes of environmental copy number muta-
gens. Environ. Mol. Mutagen. 2016, 57, 3–9.
7. Lew, A.R.; Kellermayer, T.R.; Sule, B.P.; Szigeti, K. Copy number variations in adult-
onset neuropsychiatric diseases. Curr. Genomics. 2018, 19, 420–430.
8. Velinov, M. Genomic copy number variations in the autism clinic: Work in progress.
Front. Cell Neurosci. 2019, 13, 57.
9. Prestes, P.R.; Maier, M.C.; Charchar, F.J. DNA copy number variations: Do these big
mutations have a big effect on cardiovascular risk? Int. J. Cardiol. 2020, 298, 116–117.
10. Brezina, S.; Feigl, M.; Gumpenberger, T.; Staudinger, R.; Baierl, A.; Gsur, A. Genome-
wide association study of germline copy number variations reveals an association with
prostate cancer aggressiveness. Mutagenesis. 2020, 35, 283–290.
11. Fortin, O.; Vincelette, C.; Chénier, S.; Ghais, A.; Shevell, M.I.; Simard-Tremblay, E.;
Myers, K.A. Copy number variation in genetic epilepsy with febrile seizures plus. Eur.
J. Paediatr. Neurol. 2020, 27, 111–115.
12. Maass, P.G.; Weise, A.; Rittscher, K.; Lichtenwald, J.; Barutcu, A.R.; Liehr, T.; Aydin,
A.; Wefeld-Neuenfeld, Y.; Pölsler, L.; Tinschert, S.; Rinn, J.L.; Luft, F.C.; Bähring,
S. Reorganization of inter-chromosomal interactions in the 2q37-deletion syndrome.
EMBO J. 2018, 37, e96257.
13. Rice, A.M.; McLysaght, A. Dosage sensitivity is a major determinant of human copy
number variant pathogenicity. Nat. Commun. 2017, 8, 14366.
14. Riggs, E.R.; Andersen, E.F.; Cherry, A.M.; Kantarci, S.; Kearney, H.; Patel, A.; Raca,
G.; Ritter, D.I.; South, S.T.; Thorland, E.C.; Pineda-Alvarez, D.; Aradhya, S.; Martin,
C.L. Technical standards for the interpretation and reporting of constitutional copy-num-
ber variants: A joint consensus recommendation of the American College of Medical
Genetics and Genomics (ACMG) and the Clinical Genome Resource (ClinGen). Genet
Med. 2020, 22, 245–257.
180 Cytogenetics and Molecular Cytogenetics
15. Liu, Y.; Wu, S.; Shi, X.; Mao, F.; Zeng, X. Cancer with a HER2 IHC2+ and FISH HER2/
CEP17 ratio ≥2.0 and an average HER2 gene copy number <4.0 per tumor cell: HER2
mRNA overexpression is a rare event. Front. Oncol. 2020, 10, 985.
16. Weise, A.; Gross, M.; Mrasek, K.; Mkrtchyan, H.; Horsthemke, B.; Jonsrud, C.; Von
Eggeling, F.; Hinreiner, S.; Witthuhn, V.; Claussen, U.; Liehr, T. Parental-origin-
determination fuorescence in situ hybridization distinguishes homologous human chro-
mosomes on a single-cell level. Int. J. Mol. Med. 2008, 21, 189–200.
17. Harutyunyan, T.; Hovhannisyan, G.; Babayan, N.; Othman, M.A.; Liehr, T.; Aroutiounian,
R. Infuence of afatoxin B1 on copy number variants in human leukocytes in vitro. Mol.
Cytogenet. 2015, 8, 25.
18. Harutyunyan, T.; Hovhannisyan, G.; Sargsyan, A.; Grigoryan, B.; Al-Rikabi, A.H.;
Weise, A.; Liehr, T.; Aroutiounian, R. Analysis of copy number variations induced by
ultrashort electron beam radiation in human leukocytes in vitro. Mol. Cytogenet. 2019,
12, 18.
19. Iourov, I.Y. Quantitative fuorescence in situ hybridization (QFISH). In: Methods in
Molecular Biology: Cancer Cytogenetics Methods and Protocols; Wan, T.S.K., Ed.
Springer, New York, 2017, pp. 143–149.
16 FISH—Interphase
Applications
Including Detection
of Chromosome
Instability (CIN)
Ivan Y. Iourov, Svetlana G. Vorsanova
and Yuri B. Yurov
CONTENTS
Introduction ............................................................................................................ 181
Interphase FISH and CIN....................................................................................... 182
Interphase Chromosome-Specifc Multicolor Banding ......................................... 184
Diagnostic Issues ................................................................................................... 184
Conclusions ............................................................................................................ 184
Acknowledgments.................................................................................................. 185
References .............................................................................................................. 185
INTRODUCTION
Fluorescence in situ hybridization (FISH) is a technological basis of interphase
molecular cytogenetics. Interphase FISH is found useful for a wide spectrum of
applications from molecular cytogenetic diagnosis to interphase chromosome biol-
ogy.[1–3] Since a kind of a decrease in interest in interphase cytogenetics is observed
in the postgenomic era,[4, 5] a brief overview of interphase FISH applications seems
to be required.
Molecular cytogenetic analysis is an integral part of the research and medical care
in clinical/medical genetics, reproduction, oncology, neurology, psychiatry, pediatrics.
The signifcance of FISH applications in biomedicine has been consistently reported.[2, 3]
Interphase FISH techniques are currently suggested to be an important technological
element of research in somatic cell genetics/genomics, aging, single-cell biology, chro-
mosome/chromatin biology (i.e. genome organization in interphase).[1, 5]
A more specifc application of FISH is the analysis of chromosome instability
(CIN) in interphase nuclei. The growing importance of CIN analysis in current bio-
medicine results from observations on CIN involvement in genetic intercellular/
DOI: 10.1201/9781003223658-16 181
182 Cytogenetics and Molecular Cytogenetics
FIGURE 16.1 Interphase molecular cytogenetic analysis CIN in the fetal human brain.
Interphase FISH with chromosome-enumeration DNA probes:
(A) two nuclei characterized by additional chromosomes Y and X and a normal nucleus;
(B) a nucleus with monosomy of chromosome 15 and a normal nucleus;
(C) a nucleus with monosomy of chromosome 18 and a normal nucleus.
(D to G) interphase chromosome-specifc MCB: nuclei with monosomy, disomy, trisomy,
and G-banding ideograms with MCB color-code labeling of a chromosome (from left to
right), (D)—chromosome 9, (E)—chromosome 16, and (F)—chromosome 18. (G) inter-
phase QFISH: (1) a nucleus with two signals for chromosomes 18 (relative intensities: 2058
and 1772 pixels), (2) a nucleus with one paired signal mimics monosomy of chromosome 18
(relative intensity: 4012 pixels), (3) a nucleus with two signals for chromosomes 15 (relative
intensities: 1562 and 1622 pixels), (4) a nucleus with one signal showing monosomy of chro-
mosome 15 (relative intensity: 1678 pixels).
Source: From Yurov et al.,[16] an open-access article distributed under the terms of the Creative Commons
Attribution License.
184 Cytogenetics and Molecular Cytogenetics
solve this problem, quantitative FISH may be applied (Figure 16.1G). This technique
offers an opportunity to differ between chromosomal associations and chromosome
loss (aneuploidy/monosomy).[22–24]
DIAGNOSTIC ISSUES
In the diagnostic context, the availability of single-cell molecular cytogenetic analy-
sis via visualization is a striking advantage offered by interphase FISH. Additional
advantage offered by FISH are the highest possible cell scoring potential by the
visualization of chromosomal changes in interphase nuclei. Since chromosomal
imbalances and CIN cause a wide spectrum of human morbid conditions, inter-
phase FISH-based methods are to be recognized as a valuable part of molecular
diagnosis.[1–3] It is noteworthy that a diagnosis is referred to as a knowledge about
molecular and/or cellular mechanism of a disease. Consequently, interphase FISH-
based techniques could be an important part of a diagnostic research (diagnosis
and monitoring) of CIN-mediated diseases. Interphase FISH-based study of chro-
mosomal changes may be an addition to pathway-based analysis of genome varia-
tions to model the functional consequences of chromosome imbalances and CIN.
Chromosomal biomarkers (specifc chromosome imbalances or reproducible pat-
terns of CIN) detectable by interphase FISH are to be addressed by systems biology
(bioinformatic) methodology for determining causes and consequences of CIN and
for developing algorithms of unraveling disease mechanisms.[8, 34–37] Finally, com-
bining immunohistochemical detection of proteins and interphase FISH (Immuno-
FISH) offers an opportunity to analyze interphase chromosomes (interphase CIN)
in nuclei of specifc cell types. This is found useful to detect chromosome instability
in post-mitotic tissues and in cancer cells.[31, 32, 38]
CONCLUSIONS
In the postgenomic era, collecting genomic data often lacks chromosomal context.
As a result, such important genetic phenomena/processes as chromosomal abnor-
malities and CIN are still poorly understood.[5, 39] Interphase FISH-based technolo-
gies may help to gain further understanding of causes and consequences of CIN and
FISH—Interphase Applications and CIN 185
ACKNOWLEDGMENTS
The chapter is dedicated to Dr. Ilia V Soloviev. Interphase FISH studies in authors’
labs are supported by the Government Assignment of the Russian Ministry of
Science and Higher Education, Assignment no. AAAA-A19–119040490101–6, and
by the Government Assignment of the Russian Ministry of Health, Assignment no.
121031000238–1.
REFERENCES
1. Vorsanova, S.G.; Yurov, Y.B.; Iourov, I.Y. Human interphase chromosomes: A review of
available molecular cytogenetic technologies. Mol. Cytogenet. 2010, 3, 1.
2. Liehr, T. Fluorescence In Situ Hybridization (FISH): Application Guide. Springer,
Berlin; 2017.
3. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B. Human Interphase Chromosomes: Biomedical
Aspects. 2nd Edition. Springer, Berlin; 2020.
4. Liehr, T. From human cytogenetics to human chromosomics. Int. J. Mol. Sci. 2019, 20,
826.
5. Iourov, I.Y.; Yurov, Y.B.; Vorsanova, S.G. Chromosome-centric look at the genome. In:
Human Interphase Chromosomes: Biomedical Aspects; Iourov, I.Y.; Vorsanova, S.G.;
Yurov, Y.B., Eds. Springer, Berlin, 2020, pp. 157–170.
6. Heng, H.H.; Bremer, S.W.; Stevens, J.B.; Horne, S.D.; Liu, G.; Abdallah, B.Y.; Ye, K.J.;
Ye, C.J. Chromosomal instability (CIN): What it is and why it is crucial to cancer evolu-
tion. Cancer Metastasis Rev. 2013, 32, 325–340.
7. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B.; Kutsev, S.I. Ontogenetic and pathogenetic
views on somatic chromosomal mosaicism. Genes. 2019, 10, 379.
8. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B.; Zelenova, M.A.; Kurinnaia, O.S.; Vasin, K.S.;
Kutsev, S.I. The cytogenomic “theory of everything”: Chromohelkosis may underlie
chromosomal instability and mosaicism in disease and aging. Int. J. Mol. Sci. 2020, 21,
8328.
9. Ye, C.J.; Sharpe, Z.; Heng, H.H. Origins and consequences of chromosomal instability:
From cellular adaptation to genome chaos-mediated system survival. Genes. 2020, 11,
1162.
186 Cytogenetics and Molecular Cytogenetics
10. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B. Recent patents on molecular cytogenetics.
Recent Pat. DNA Gene Seq. 2008, 2, 6–15.
11. Liehr, T.; Othman, M.A.; Rittscher, K.; Alhourani, E. The current state of molecular
cytogenetics in cancer diagnosis. Expert Rev. Mol. Diagn. 2015, 15, 517–526.
12. Iourov, I.Y.; Vorsanova, S.G.; Yurov, Y.B. Single cell genomics of the brain: Focus on
neuronal diversity and neuropsychiatric diseases. Curr. Genomics. 2012, 13, 477–488.
13. Bakker, B.; van den Bos, H.; Lansdorp, P.M.; Foijer, F. How to count chromosomes in a
cell: An overview of current and novel technologies. Bioessays. 2015, 37, 570–577.
14. Manvelyan, M.; Hunstig, F.; Bhatt, S.; Mrasek, K.; Pellestor, F.; Weise, A.; Simonyan, I.;
Aroutiounian, R.; Liehr, T. Chromosome distribution in human sperm: A 3D multicolor
banding-study. Mol. Cytogenet. 2008, 1, 25.
15. McClelland, S.E. Single-cell approaches to understand genome organisation throughout
the cell cycle. Essays Biochem. 2019, 63, 209–216.
16. Yurov, Y.B.; Iourov, I.Y.; Vorsanova, S.G.; Liehr, T.; Kolotii, A.D.; Kutsev, S.I.; Pellestor,
F.; Beresheva, A.K.; Demidova, I.A.; Kravets, V.S.; Monakhov, V.V.; Soloviev, I.V.
Aneuploidy and confned chromosomal mosaicism in the developing human brain. PLoS
One. 2007, 2, e558.
17. Iourov, I.Y.; Yurov, Y.B.; Vorsanova, S.G.; Kutsev, S.I. Chromosome instability, aging
and brain diseases. Cells. 2021, 10, 1256.
18. Liehr, T.; Starke, H.; Weise, A.; Lehrer, H.; Claussen, U. Multicolor FISH probe sets and
their applications. Histol. Histopathol. 2004, 19, 229–237.
19. Devadhasan, J.P.; Kim, S.; An, J. Fish-on-a-chip: A sensitive detection microfuidic sys-
tem for Alzheimer’s disease. J. Biomed. Sci. 2011, 18, 33.
20. Hu, Q.; Maurais, E.G.; Ly, P. Cellular and genomic approaches for exploring structural
chromosomal rearrangements. Chromosome Res. 2020, 28, 19–30.
21. Vorsanova, S.G.; Yurov, Y.B.; Kolotii, A.D.; Soloviev, I.V. FISH analysis of replication
and transcription of chromosome X loci: New approach for genetic analysis of Rett syn-
drome. Brain Dev. 2001, 23, S191–S195.
22. Iourov, I.Y.; Soloviev, I.V.; Vorsanova, S.G.; Monakhov, V.V.; Yurov, Y.B. An approach
for quantitative assessment of fuorescence in situ hybridization (FISH) signals for
applied human molecular cytogenetics. J. Histochem. Cytochem. 2005, 53, 401–408.
23. Vorsanova, S.G.; Iourov, I.Y.; Beresheva, A.K.; Demidova, I.A.; Monakhov, V.V.;
Kravets, V.S.; Bartseva, O.B.; Goyko, E.A.; Soloviev, I.V.; Yurov, Y.B. Non-disjunction
of chromosome 21, alphoid DNA variation, and sociogenetic features of Down syn-
drome. Tsitol. Genet. 2005, 39, 30–36.
24. Iourov, I.Y. Quantitative fuorescence in situ hybridization (QFISH). Methods Mol. Biol.
2017, 1541, 143–149.
25. Liehr, T.; Heller, A.; Starke, H.; Rubtsov, N.; Trifonov, V.; Mrasek, K.; Weise, A.;
Kuechler, A.; Claussen, U. Microdissection based high resolution multicolor banding for
all 24 human chromosomes. Int. J. Mol. Med. 2002, 9, 335–339.
26. Liehr, T.; Othman, M.A.; Rittscher, K. Multicolor karyotyping and fuorescence in situ
hybridization-banding (MCB/mBAND). Methods Mol. Biol. 2017, 1541, 181–187.
27. Iourov, I.Y.; Liehr, T.; Vorsanova, S.G.; Kolotii, A.D.; Yurov, Y.B. Visualization of inter-
phase chromosomes in postmitotic cells of the human brain by multicolour banding
(MCB). Chromosome Res. 2006, 14, 223–229.
28. Iourov, I.Y.; Liehr, T.; Vorsanova, S.G.; Yurov, Y.B. Interphase chromosome-specifc
multicolor banding (ICS-MCB): A new tool for analysis of interphase chromosomes in
their integrity. Biomol. Eng. 2007, 24, 415–417.
29. Lemke, J.; Claussen, J.; Michel, S.; Chudoba, I.; Mühlig, P.; Westermann, M.; Sperling,
K.; Rubtsov, N.; Grummt, U.W.; Ullmann, P.; Kromeyer-Hauschild, K.; Liehr, T.;
FISH—Interphase Applications and CIN 187
CONTENTS
Introduction............................................................................................................ 189
Samples/Tissues ..................................................................................................... 191
Description Q-FISH Method.................................................................................. 191
Reagents........................................................................................................ 192
Procedure ...................................................................................................... 192
Description CO-FISH Method............................................................................... 194
Reagents........................................................................................................ 195
Procedure ...................................................................................................... 195
Yields ..................................................................................................................... 196
Q-FISH for Telomere Length ....................................................................... 196
Q-FISH for Telomere Fragility ..................................................................... 196
Co-FISH for Detection of Telomeric Sisterchromatid Exchanges
(T-SCEs) ........................................................................................... 196
Conclusions............................................................................................................ 198
References.............................................................................................................. 198
INTRODUCTION
Telomeres are specialized nucleoprotein structures at the ends of linear chromo-
somes. They consist of a shelterin protein complex and a long assembly of hexameric
repeats (TTAGGG) oriented in 5′ to 3′ direction, ending as a single strand 3′ over-
hang. This 3′ overhang invades 5′ double-stranded telomeric duplex and is housed
deep inside by clipping with shelterin heteroduplex POT1/TTP1. Shelterin sub-
units TRF1 and TRF2 (= telomere binding factors 1 and 2) directly bind the double
stranded TTAGGG, while TIN2 (= TERF1 interacting nuclear factor) links TPP1/
POT1 (= tripeptidyl peptidase 1/protection of telomeres 1) heterodimer and stabilizes
its association with chromosome ends. Shelterin assembly creates a telomere-specifc
structure T-loop (Figure 17.1) that caps the chromosomes and protects their termini
from being recognized as breakpoints by DNA repair mechanisms.[1]
SAMPLES/TISSUES
Telomere length can be determined in human and other vertebrate cells and should
be done in metaphases. The method is particularly convenient for species that
contain interstitial telomeric sites in their genome, as well as for species that have
ultra-long and heterogeneous telomeres, such as mice. A slide-based method can be
applied to all cells that can be cultivated (primary cells as lymphocytes, fbroblast,
and cell lines).
REAGENTS
• 1×TBS (Tris-Buffered Saline, pH 7.5)—500 ml per experiment
• Pre-treatment stock solution: Dilute 2,000× concentrated proteinase K in
1×PBS
• Pre-treatment solution (working solution): mix 40 µl pre-treatment stock
solution with 80 ml TBS buffer at RT. Prepare fresh for each experiment.
• Telomere and centromere PNA probes ready to use probe (Cy3 conjugated
PNA probes in hybridization solution = 70% formamide) available on
request from manufacturer (DAKO/Agilent)
• Rinse solution ×1—Dilute 50× stock from DAKO/Agilent in pure water and
store at RT
• Wash solution ×1—Dilute 50× stock from DAKO/Agilent in pure water and
warm to 65°C ~2 h before experiment; solution might be milky, but it does
not infuence the results.
• Pure water (deionized or double-distilled water)
• 37% formaldehyde (e.g. Merck)
• Formaldehyde in TBS (working solution): add 8 ml of 37% formaldehyde to
72 ml 1×TBS buffer. Do not use more than four weeks.
• Cold ethanol series; 70%, 85%, 96% (e.g. 4°C in refrigerator)
• Mounting solution: Antifade containing DAPI as a counterstain. To pre-
serve signal intensity, the best is to use Vectashield (VectorLabs) supple-
mented with 0.1 µg/ml of DAPI.
PROCEDURE
1. Prepare metaphase spreads on a glass microscope slides employing stan-
dard cytogenetic procedure. Slide quality is the most important factor for
good hybridization results. Cells should be well fxed and well spread.
2. All following incubations are at RT if not otherwise specifed.
3. Heat working solution of Wash solution to 65oC in a water bath and
pre-worm heating incubator (e.g. Plate Shaker Thermostat PST-100 HL,
Biosan, Riga, Latvia, LV-1067) on 80oC.
4. Prepare six glass Hellendahl jars (100 ml), and fll in ~80 ml each of (i)
TBS, (ii) Formaldehyde in TBS (working solution), (iii) TBS, (iv) Pre-
treatment solution (working solution), (v) TBS, (vi) TBS
5. Immerse slides in TBS (i) for 2 min
6. Transfer and incubate slides in jar (ii) for 2 min exactly
7. Wash slides in TBS (i) and then in TBS (iii) 5 min, each
8. Incubate slides in jar (iv) 10 min
FISH—Determination of Telomere Length (Q-FISH/CO-FISH) 193
9. Wash slides in TBS (v) and then in TBS (vi) 5 min, each
10. Immerse slides in cold ethanol series (70%, 85% and 96%) 3×2 min
11. Airdry slides in a vertical position
12. Add 10 µl of PNA probes/Cy3 to carefully chosen area on the slide
13. Cover the area with a 18×18 mm coverslip
14. Place the slides in the pre-heated incubator to 80°C for 5 min
15. Place the slides in moist chamber (in the dark) at RT for 2 hours
16. Briefy immerse slides in Rinse solution ×1 to remove coverslip
17. Immerse slides in the pre-heated Wash solution ×1 for 5 min at 65°C
18. Repeat steps 11 and 12
19. Apply 2×10 µl of mounting solution onto each slide and put 24×50 mm
coverslip. For the counterstain color to develop, leave the slides 15 min
in the dark. Slides are ready for microscopy. Since a variety of micro-
scopes and image acquisitions systems are used in different laboratories,
the manufacturer manual for use should be followed. Figure 17.2 presents
captured metaphase for telomere length analysis.
20. For analysis of telomere length, using measurement tool for ISIS software
(MetaSystems, Altlussheim, Germany) with centromere fuorescence of
chromosome 2 set to value of 100% is described. Upon capturing, con-
trast and threshold of signals are adjusted; using option of inverse DAPI
staining chromosomes are arranged in karyogram form. Afterwards, the
karyogram is reverted to fuorescence mode. The telomere measurement
software displays two horizontal lines overlaid to each chromosome that
defne the centromere signal on chromosome 2 (red lines) and telomere
signals on p and q arms (green lines) (Figure 17.3).
FIGURE 17.2 Captured metaphase hybridized with Cy3-labeled PNA probe for measure-
ment of telomere length.
194 Cytogenetics and Molecular Cytogenetics
FIGURE 17.3 Fluorescence intensity of centromere signal is set at value of 100%. Each
chromosome arm is bordered with green software line.
REAGENTS
• BrdU/BrdC: Take 15 µl of BrdU and 5 µl of BrdC from stock (50mM conc.)
to make a 3:1 ratio, mix well and add 1 µl of the mixture into 5 ml of T25
fask. N.B.: for a T75 add 3 µl of the mixture to make a fnal concentration
of 10 µM of Brdu/BrdC.
• Exonuclease III (e.g Thermofsher): Take 1.5 µl of stock (200 U/µl) and mix
it with 100 µl of buffer to make a fnal concentration of (3 U/µl)
• 70% formamide: 70ml formamide + 10 ml 20×SSC + 20 ml of ddH2O to
make a fnal volume of 100 ml
• Hoechst 33258: Take 50 µl of stock (0.5 µg/ml) and mix it with 50 ml of
ddH2O in a glass Hellendahl jar.
• Phosphate buffered saline: 1×PBS (e.g. Merck)
• 2×SSC: Dilute 20×SSC (commercially available, e.g. Merck) with ddH2O
PROCEDURE
1. Apply metaphase spread protocol and make slides and “age” them for few
days under room temperature.
2. Put the slides in glass Hellendahl jar comprising working solution of
Hoechst 33258 for 15 minutes at RT.
3. Remove and mount the slide with 2×SSC, and put the glass coverslip over
the each slide. The thin layer of 2×SSC between slide and glass coverslip
should be present during exposure to 365 nm UV light. Expose slides for
30 minutes.
4. Briefy wash the slides with 1×PBS by dipping to remove coverslips.
5. Add 20 µl of Exonuclease III (3 U/µl) onto each slide, and cover them with
paraflm for 10 minutes.
6. Wash the slide in 1×PBS by dipping into a glass Hellendahl jar.
7. Dehydrate the slide by adding 1 ml of 100% ethanol onto slide surface for
1 minute.
8. Airdry the slide.
9. Add 20 µl/slide of PNA probe (Cy3 or FITC- labelled)—and leave it to
hybridize for two minutes at 70ºC in the pre-heated incubator. Keep in wet
and dark containment for two hours.
10. Wash in 70% formamaide for 15 min twice.
11. Wash in 1×PBS for 10 minutes.
12. Dehydrate by serial dehydration (70%, 90% and 100%) with ethanol for
5 minutes.
13. Add 20 µl of DAPI Vectashield onto each slide, and put on 24x50mm
coverslip. For the counterstain color to develop, leave the slides 15 min in
the dark. Slides are ready for microscopy.
196 Cytogenetics and Molecular Cytogenetics
FIGURE 17.4 A) T-SCE staining using CO-FISH. Red arrows indicate for sister chromatid
exchanges at telomeres. B) Metaphase stained using CO-FISH without T-SCE.
14. Since a variety of microscopes and image acquisitions systems are used
in different laboratories, the manufacturer manual for use should be fol-
lowed. Upon capturing, contrast and threshold of signals are adjusted.
Cells that pass two rounds of replication will display one telomeric signal
per chromosome ends. Telomeric sisterchromatid exchanges (T-SCEs) are
seen as two signals on each chromatid (Figure 17.4).
YIELDS
Q-FISH FOR TELOMERE LENGTH
Employing measurement tools, intensity of each telomere signals results are dis-
played graphically and numerically. Results of measurement are expressed as T/C
ratio, i.e. percentage vs. centromere signal for each chromosome arm (Figure 17.5).
FIGURE 17.5 Telomere length for each chromosome arm expressed as T/C ratio.
CONCLUSIONS
Telomeres are complex nucleoprotein structures whose integrity determines the sta-
bility of the genome. Many DNA damage repair proteins are located on telomeres.
They are activated upon early DNA damage or directly participate in its repair.
Telomeres shortening during each cell division, having the function of biological
“clock”, leads to elimination of cells with critically shortened telomeres. Rich in
guanine, telomeres are susceptible to guanine oxidative damage. While oxidized
bases are supplemented, break-induced repair can make disruption and elongation of
telomeres inducing genomic instability, which is known as a hallmark of tumor cells.
Molecular biology methods in combination with molecular cytogenetic methods will
certainly in the near future clarify the mechanisms by which telomeres maintain
genomic stability. This will certainly be important for the prevention and treatment
of many diseases.
REFERENCES
1. Blackburn, E.H.; Greider, C.W.; Jack, W.; Szostak, J.W. The nobel prize in physiology or
medicine 2009. Nobel Found. 2009, 1, 10–105.
2. de Lange, T. How telomeres solve the end-protection problem. Science. 2009, 326,
948–952.
3. de Lange, T. Shelterin-mediated telomere protection. Annu. Rev. Genet. 2018, 52,
223–247.
4. Sfeir, A.; de Lange, T. Removal of shelterin reveals the telomere end-protection problem.
Science 2012, 336, 593–597.
5. Deng, Z.; Wang, Z.; Stong, N.; Plasschaert, R.; Moczan, A.; Chen, H.-S.; Hu, S.;
Wikramasinghe, P.; Davuluri, R.V.; Bartolomei, M.S.; Riethman, H.; Lieberman, P.M.
A role for CTCF and cohesin in subtelomere chromatin organization, TERRA transcrip-
tion, and telomere end protection. EMBO J. 2012; 31, 4165–4178.
6. Bettin, N.; Pegorar, C.O.; Cusanelli, E. The emerging roles of TERRA in telomere main-
tenance and genome stability. Cells. 2019, 8, 246.
7. Muraki, K.; Nyhan, K.; Han, L.; Murnane, J. Mechanisms of telomere loss and their
consequences for chromosome instability. Front. Oncol. 2012, 2, 135.
8. Hayfick, L. Living forever and dying in attempt. Exp. Gerontol. 2003, 38, 1231–1241.
9. Tomáška, Ľ.; Cesare, A.J.; AlTurki, T.M.; Griffth, J.D. Twenty years of t-loops: A case
study for the importance of collaboration in molecular biology. DNA Repair (Amst).
2020, 94, 102901.
10. Liu, H.; Xie, Y.; Zhang, Z.; Mao, P.; Liu, J.; Ma, W.; Zhao, Y. Telomerric recombina-
tion induced by DNA damage results in telomere extension and length heterogeneity.
Neoplasia. 2018, 20, 905–916.
11. Martínez, P.; Thanasoula, M.; Muñoz, P.; Liao, C.; Tejera, A.; McNees, C.; McNees, C.;
Flores, J.M.; Fernández-Capetillo, O.; Tarsounas, M.; Blasco, M.A. Increased telomere
fragility and fusions resulting from TRF1 defciency lead to degenerative pathologies
and increased cancer in mice. Genes Dev. 2009, 23, 2060–2075.
12. Kychygina, A.; Dall’Osto, M.; Joshua, A.M.; Cadoret, J.C.; Piras, V.; Picket, H.A.;
Crabbe, L. Progerin impairs 3D genome organization and induces fragile telomeres by
limiting the dNTP pools. Sci. Rep. 2012, 11, 13195.
13. von Zglinicki, T. Oxidative stress shortens telomeres. Trends Biochem. Sci. 2002, 27,
339–344.
FISH—Determination of Telomere Length (Q-FISH/CO-FISH) 199
14. Barnes, R.P.; Fouquerel, E.; Opresko, P.L. The impact of oxidative DNA damage and
stress on telomere homeostasis. Mech. Ageing Dev. 2019, 177, 37–45.
15. Blackburn, E.H. Switching and signaling at the telomere. Cell. 2001, 106, 661–673.
16. Nielsen, P.E.; Egholm, M.; Berg, R.H.; Buchardt, O. Sequence-selective recognition of
DNA by strand displacement with a thymine-substituted polyamide. Science. 1991, 254,
1497–1500.
17. Perner, S.; Brüderlein, S.; Hasel, C.; Waibel, I.; Holdenried, A.; Ciloglu, N.; Chopurian,
H.; Nielsen, K.V.; Plesch, A.; Högel, J.; Möller, P. Quantifying telomere lengths of human
individual chromosome arms by centromere-calibrated fuorescence in situ hybridiza-
tion and digital imaging. Am. J. Pathol. 2003, 163, 1751–1756.
18. Bailey, S.M.; Williams, E.S.; Cornforth, M.N.; Goodwin, E.H. Chromosome orientation
fuorescence in situ hybridization or strand-specifc FISH. Methods Mol. Biol. 2010, 659,
173–183.
19. Giunta, S. Centromere chromosome orientation fuorescent in situ hybridization (Cen-
CO-FISH) detects sister chromatid exchange at the centromere in human cells. Bio.
Protoc. 2018, 8, e2792.
18 FISH—in Three
Dimensions—3D-FISH
Thomas Liehr
CONTENTS
Introduction ............................................................................................................ 201
Samples/Tissues ..................................................................................................... 202
Description of Methods.......................................................................................... 202
How to Proceed ............................................................................................. 202
Yields ..................................................................................................................... 203
Conclusions ............................................................................................................ 203
References .............................................................................................................. 203
INTRODUCTION
Molecular cytogenetics, particularly fuorescence in situ hybridization (FISH), can
be applied as one of nowadays three major approaches to study the nuclear architec-
ture (= 3D-FISH)[1]; the two other ways to access this problem are based on life-cell
imaging,[2] and long read sequencing,[3] and are not further covered here. However,
all studies applying such methods with the goal to unravel the peculiarities of inter-
phases can be summarized as nucleomics.[1]
Besides high-resolution laser scanning microscopes, it is also possible to use stan-
dard fuorescence microscopes for 3D-FISH studies. Even though such studies can
be performed in normally prepared, fattened nuclei,[4] there is a simple approach to
retain original shape and size of interphases before, during and after FISH-procedure,
as well as during microscopic evaluation, called suspension-FISH (S-FISH).[5]
As summarized elsewhere,[1] nucleomics can be traced back to 1885, when Carl
Rabl suggested that interphase chromosomes are threadlike but separated into indi-
vidual chromosomes.[6] Thus, he already suggested “chromosome territories”, a
wording shaped in 1909 by Theodor Boveri, and Eduard Strasburger at about the
same time.[1] In the 1950s, the inactive X-chromosome in females has been char-
acterized as a stainable Barr-body[7] within the nucleus. And since 1985, there was
evidence that chromosome territories and/or subdomains of them are able to change
shapes and structures during time in cell differentiation.[8] In the last decade fnally,
the next generation and long-read sequencing approaches could identify the topologi-
cally associating domains (TADs) and show that genes do interact in cis and trans
on DNA-level.[4, 9, 10]
According to data obtained during the last decades, in the interphase nucleus the
DNA is protein-covered, and there is never-ending transcription, de- and refolding,
and repair going on.[11] Even though the nucleus is imagined nowadays as fuent,
DNA-compartments of chromosomes, being subunits of the nucleus, are never really
separated,[10 –12] which surprisingly also includes a connection of maternal and pater-
nal genomes via spindles and spindle-pole.[1, 13] Besides, chromosomes are arranged
in a nucleus normally according to size and gene-density; an exception has yet been
seen only in rod cells in retina of nocturnal mammals, where an “inverted” arrange-
ment is needed to support night vision.[1, 14, 15]
Here the S-FISH approach is outlined.[5]
SAMPLES/TISSUES
Any kind of tissue can be used for 3D-FISH-anaylses, as long as intact interphase
nuclei can be accessed in it. However, to keep interphase cells in their original spher-
ical shape, to the best of our knowledge only the S-FISH approach has been reported
yet. For this technique, only cells fxed in Carnoy’s fxative from standard cytoge-
netic preparations are suited.[5]
DESCRIPTION OF METHODS
S-FISH was frst reported in 2002 and was e.g. successfully applied for 3D-FISH
studies of human sperm,[14] or in studied of great ape cells using human probes,[16]
or others like positioning-characterization of small supernumerary marker chro-
mosomes, (derivative) X-chromosomes, or chromosomes involved in chromosomal
rearrangements in leukemia cases.[17]
HOW TO PROCEED
A) Transfer prepared interphase cells from Carnoy’s fxative into hybridiza‑
tion buffer:
1. Centrifuge ~50–100 µl of a concentrated cell suspension diluted in Car-
noy’s fxative (1,500 rpm/4°C/10 min).
2. Wash in 500 μl methanol, incubate for 2 min and centrifuge as in step 1.
3. Wash in 500 μl 0.9% NaCl, incubate for 2 min and centrifuge as in step 1.
4. Incubate in 500 µl pepsin solution (950 μl of distilled water + 50 μl 0.2
N HCl + 5 μl pepsin stock solution) at 37°C for 5 min, and centrifuge as
in step 1.
5. Incubate in 500 μl 0.9% NaCl solution for 2 min at room temperature
(RT), and repeat step 1. Leave 50 μl suspension in the tube.
6. Denature at 95°C/5 min, pellet the cells by centrifugation for 10 min at
1,500 rpm, and repeat step 1. Leave 20 μl suspension in the tube.
7. Prepare a 3× concentrated probe (compared to normal FISH-experiment) in
25 µl hybridization buffer; if necessary add 5–50 μg of COT1 DNA, dena-
ture at 95°C for 5 min, and (if needed) prehybridize at 37°C for 30–60 min.
8. Now add this pre-hybridized probe (step 7) to suspension from step 6.
FISH—in Three Dimensions 203
YIELDS
By including data from electron microscopy not much considered up to now, Joan-
Ramon Daban could do the following statement recently:
CONCLUSIONS
Based on data from nucleomics, some basic puzzle stones of chromosome and
interphase structure seem to be solved. Still, this just opens manifold doors of new
research directions, specifcally in clinical genetics and tumor genetics. Yet there are
just few such examples,[19–21] but more are expected to come.
REFERENCES
1. Liehr, T. Nuclear architecture. In: Cytogenomics; Liehr, T., Ed. Academic Press, New
York, 2021, pp. 297–305.
2. Potlapalli, B.P.; Schubert, V.; Metje-Sprink, J.; Liehr, T.; Houben, A. Application of Tris-
HCl allows the specifc labeling of regularly prepared chromosomes by CRISPR-FISH.
Cytogenett. Genome Res. 2020, 160, 156–165.
204 Cytogenetics and Molecular Cytogenetics
19. Manvelyan, M.; Kempf, P.; Weise, A.; Mrasek, K.; Heller, A.; Lier, A.; Höffken, K.;
Fricke, H.J.; Sayer, H.G.; Liehr, T.; Mkrtcyhan, H. Preferred co-localization of chromo-
some 8 and 21 in myeloid bone marrow cells detected by three dimensional molecular
cytogenetics. Int. J. Mol. Med. 2009, 24, 335–341.
20. Klonisch, T.; Wark, L.; Hombach-Klonisch, S.; Mai, S. Nuclear imaging in three dimen-
sions: A unique tool in cancer research. Ann. Anat. 2010, 192, 292–301.
21. Timme, S.; Schmitt, E.; Stein, S.; Schwarz-Finsterle, J.; Wagner, J.; Walch, A.; Werner,
M.; Hausmann, M.; Wiech, T. Nuclear position and shape deformation of chromosome 8
territories in pancreatic ductal adenocarcinoma. Analyt. Cell. Pathol. 2011, 34, 21–33.
19 FISH—On Fibers
Thomas Liehr
CONTENTS
Introduction ............................................................................................................ 207
Samples/Tissues ..................................................................................................... 207
Description of Methods ................................................................................ 208
Yields ..................................................................................................................... 209
Conclusions ............................................................................................................ 209
References .............................................................................................................. 209
INTRODUCTION
Fluorescence in situ hybridization (FISH) on stretched or highly extended chromo-
some fbers is known as Fiber-FISH,[1] or molecular combing.[2] In Figure 19.1, it is
visualized that FISH has a principal resolution from a few dozen to hundred base pairs
(in molecular combing), a few ten- to a few hundred-thousand base pairs (in Fiber-
FISH), a few hundred-thousand base pairs to mega base pairs in interphase-FISH
and mega base pair to chromosome size when accessing metaphases in FISH. Both
high-resolution approaches of FISH—Fiber-FISH,[1] and molecular combing[2]—were
described in the 1990s, and later on used in some specialized labs.[3–5] According to a
PubMed search,[6] molecular combing was applied in research at about the same rates
between 2001 and 2021 (Figure 19.2); on the other hand Fiber-FISH applications in
human samples had a kind of hype between 1994 and 2000 and afterwards declined
(Figure 19.2). In non-human-oriented research, Fiber-FISH has been used in about
the same rates since 1994 (Figure 19.2). While molecular combing has recently been
rediscovered and even commercialized,[7] for Fiber-FISH such a renaissance has not
happened yet.
As Fiber-FISH, also sometimes called nuclear chromatin release, neither needs
sophisticated equipment nor is complicated to perform, here the protocol for this
approach shall be brought back to molecular cytogeneticists’ minds.
SAMPLES/TISSUES
Each cell suspension prepared in Carnoy’s fxative (methanol/acetic acid = 3:1) is
suited as starting material to produce stretched DNA-fbers suited for Fiber-FISH.
FIGURE 19.2 A Pubmed search[6] revealed the number of research papers published
between 1994 and 2021 using molecular combing or Fiber-FISH (broken down in human and
non-human oriented studies).
DESCRIPTION OF METHODS
The Fiber-FISH approach is a modifcation from Fidlerová et al.,[3] and was pre-
viously published elsewhere as a modifed version.[8] Cytogenetically worked up
material in Carnoy’s fxative (methanol/acetic acid = 3:1), previously being stored at
−20°C for up to several years, may be used.
FISH—On Fibers 209
YIELDS
The author of this paper used Fiber-FISH to clearly map the CMT1A-REP elements
being ~1.4 Mb apart from each other and the in-between localized PMP22 gene.[5, 8]
Alterations of regions being in range of 100,000 to 1,000,000 base pairs can be opti-
mally accessed by Fiber-FISH using locus-specifc probes.
CONCLUSIONS
In case of necessity to study chromosomal changes at a resolution not accessible any
more by interphase-FISH and being too large to be visualized in molecular comb-
ing, Fiber-FISH is the optimal alternative to correspondingly prepare cytogenetic
material.
REFERENCES
1. Heng, H.H.; Squire, J.; Tsui, L.C. High-resolution mapping of mammalian genes
by in situ hybridization to free chromatin. Proc. Natl. Acad. Sci. U. S. A. 1992, 89,
9509–9513.
210 Cytogenetics and Molecular Cytogenetics
2. Bensimon, A.; Simon, A.; Chiffaudel, A.; Croquette, V.; Heslot, F.; Bensimon, D.
Alignment and sensitive detection of DNA by a moving interface. Science. 1994, 265,
2096–2098.
3. Fidlerová, H.; Senger, G.; Kost, M.; Sanseau, P.; Sheer, D. Two simple procedures for
releasing chromatin from routinely fxed cells for fuorescence in situ hybridization.
Cytogenet. Cell Genet. 1994, 65, 203–205.
4. Heiskanen, M.; Peltonen, L.; Palotif, A. Visual mapping by high resolution FISH. Trends
Genet. 1996, 12, 379–382.
5. Rautenstrauss, B.; Fuchs, C.; Liehr, T.; Grehl, H.; Murakami, T.; Lupski, J.R. Visualization
of the CMT1A duplication and HNPP deletion by FISH on stretched chromosome fbers.
J. Peripher. Nerv. Syst. 1997, 2, 319–322.
6. Pubmed. https://pubmed.ncbi.nlm.nih.gov [accessed on 03/2022].
7. Bisht, P.; Avarello, M.D.M. Molecular combing solutions to characterize replication kinet-
ics and genome rearrangements. In: Cytogenomics; Liehr, T., Ed. Academic Press, New
York, 2021, pp. 47–72.
8. Fuchs, C.; Liehr, T.; Rautenstrauss, B. High-resolution FISH of stretched chromosome
fbers. Trends Genet. 1997, 13, 287.
20 FISH—and Single-Cell
Gel Electrophoresis
Assay (Comet Assay)
Galina Hovhannisyan, Tigran Harutyunyan
and Rouben Aroutiounian
CONTENTS
Introduction............................................................................................................ 212
Comet-FISH.................................................................................................. 212
Main Achievements of Comet-FISH Application......................................... 213
Comet-FISH in Studies for Gene Damage and Repair In Vitro........ 213
Comet-FISH in Studies of Chromosomes and Specifc
Chromosomal Regions In Vitro ......................................... 214
Comet-FISH In Vitro with Padlock and Strand-Specifc Probes ...... 214
Comet-FISH for In Vivo Studies....................................................... 214
Samples/Tissues ..................................................................................................... 215
Description of Methods.......................................................................................... 216
Materials ....................................................................................................... 216
Equipment and Supplies (CA and Comet-FISH).............................. 216
Reagents and Solutions (CA)............................................................ 216
Reagents and Solutions (FISH) ........................................................ 216
Method.......................................................................................................... 216
Cell Samples Preparation.................................................................. 216
Comet Assay ..................................................................................... 217
Comet-FISH...................................................................................... 217
Evaluation ......................................................................................... 217
Yields ..................................................................................................................... 218
Important Notes ............................................................................................ 219
Conclusions............................................................................................................ 219
Acknowledgments.................................................................................................. 219
References.............................................................................................................. 219
INTRODUCTION
The Comet Assay (= CA, also called single cell gel electrophoresis) was developed
in the 1980s as a relatively simple and fast way of detecting DNA damage and
repair at the level of individual cells.[1, 2] The main steps of the CA include fxa-
tion of cell suspension with agarose onto glass microscope slides, lysis of cells to
disrupt membranes and remove histones, and electrophoresis. Negatively charged,
fragmented DNA migrates out of the nucleus in the electric feld more rapidly than
intact DNA, forming a structure resembling the tail of a comet, whereas undam-
aged DNA forms the head of the comet.[3] Images of comets stained with fuores-
cent dyes are analyzed under a microscope using image analysis software. The
percentage of DNA in the tail refects the level of DNA damage in individual
cells.[4] The range of detection is between a few hundred DNA breaks per cell and
a few thousand, encompassing levels of damage that can be repaired and tolerated
by human cells.[5]
CA can be conducted under neutral or alkaline electrophoresis conditions. The
neutral version of CA allows the detection of double-strand breaks. Currently, the
most commonly used is the alkaline CA version, which detects a wider range of dam-
age, including single- and double-strand breaks and alkali-labile sites.[3] The alkaline
CA also identifes DNA lesions that are converted into strand breaks with various
lesion-specifc enzymes.[6] Up to now, twelve enzymes have been used; however, only
the bacterial formamidopyrimidine DNA glycosylase (Fpg) and endonuclease III
(EndoIII), which induce breaks at sites of oxidized purines and pyrimidines, respec-
tively, are used extensively.[6, 7]
The ability to detect DNA damage, induced by genotoxic agents at subtoxic,
physiologically relevant exposures,[8] has contributed to the widespread use of the
CA in environmental biomonitoring and in vitro and in vivo genotoxicity testing in
animals,[9–11] plants,[12] and humans.[5, 13–15]
COMET-FISH
The alkaline CA was combined with fuorescence in situ hybridization (FISH)
to detect DNA damage and repair of specifc DNA sequences. According to Glei
et al.,[16]
the nature of the measured Comet-FISH endpoint precludes us from stating basically
that damage and repair are occurring within the specifc gene, it is at least possible to
evaluate whether the damage and repair are occurring within the vicinity of the gene
of interest.
The FISH protocol has been adapted to the experimental limitations of CA and
was frst applied to comet preparations to determine the spatial distribution of telo-
meres, centromeres, and segments of the O6-methylguanine–DNA methyltransfer-
ase (MGMT) gene in human lymphocytes.[17] To avoid damaging of agarose layer
with cells, the thermal denaturation of DNA in standard FISH was replaced with a
chemical denaturation in Comet-FISH.[17] Protocols of the Comet-FISH technique
FISH—and Comet Assay 213
and their modifcations have been developed for different DNA probes application to
a wide spectrum of biological material.[18–27] Several reviews provide technical and
theoretical aspects of Comet-FISH, limitations as well as advantages of the assay
and examples of its applications.[8, 28–32]
SAMPLES/TISSUES
A major advantage of CA is that cells from various tissues of a wide variety of
eukaryotic organisms can be studied.[3, 10, 11, 26, 63, 64] CA slides are typically prepared
from fresh tissue; however, frozen tissue samples can also be applied[65]; in pre-
frozen cells, the background DNA level does not increase and the outcome of the
assay remains unchanged.[66, 67]
In principle, FISH can be applied to any comet slides. To date, Comet-FISH stud-
ies have been done on primary cells, including human,[17, 38–40, 45, 48–50, 54, 57] and mouse
blood,[56] human oropharyngeal mucosa cells,[43, 44, 47] human colon cells from surgi-
cal tissue,[42] and skin fbroblasts from Xeroderma pigmentosum patients.[55]
Furthermore, Comet-FISH studies have been carried out on cell lines, includ-
ing RT4 and RT112 (bladder carcinoma),[34–36] CHO (Chinese hamster ovary),[38, 51]
MCF-7, MDA-MB468, and CRL2336 (breast cancer), GM1310B (lymphoblastoma)
and AG11134 (normal mammary epithelial),[37] HT1080 (human fbrosarcoma) and
CCRF-CEM (human T lymphoblastoma),[51] TK6 (human lymphoblastoma),[46]
GM38 (normal fbroblast) and CSA and CSB (Cockayne syndrome fbroblast),[36] Raji
(human B lymphoblastoma),[41] and HeLa (human breast cancer cells).[52, 53] Moreover,
Comet-FISH was applied to the hemocytes of the Pacifc oyster Crassostrea gigas,[58]
and plant tissues, including seeds,[61] roots,[59] and leaves.[60, 62, 63]
The following protocol provides a detailed description of a standard Comet-FISH
experiment suitable for human blood cells and cell lines. Blood samples can be col-
lected and directly used or cultivated according to the experimental design. Cell
lines should be cultivated and harvested according to standard procedures.
216 Cytogenetics and Molecular Cytogenetics
DESCRIPTION OF METHODS
MATERIALS
Equipment and Supplies (CA and Comet-FISH)
• Glass slides.
• Glass cover slips.
• Incubator (37°C).
• Water bath
• Staining jars.
• Moist chamber.
• Electrophoresis tank (horizontal) and power supply (500 mA, 25 V).
• Fluorescence microscope.
• Image analysis system (e.g., Comet Assay IV, Perceptive Instruments,
Suffolk, UK).
METHOD
Cell Samples Preparation
1. Collect human blood samples, use heparin as an anticoagulant. Use any cell
line, depending on the design of the experiment.
FISH—and Comet Assay 217
Comet Assay
1. Cover the glass with a layer of 1% normal melting point agarose by dipping
it in a vertical staining jar with melted agarose solution.
2. Dry slides at 37°C about 24 h in cell culture incubator to solidify agarose.
3. Drop 100 µl of cell/low–melting point agarose suspension (containing 10
µl of whole blood or cell line culture with 90 µl 0.9% low melting point
agarose in 1× phosphate buffered saline = PBS) onto microscope slide.
4. Cover slides with cover slips to allow a homogeneous distribution of mix-
ture of cells with low melting point agaroses on microscope slide.
5. Cool slides for 10 min at 4°C to solidify agarose.
6. Remove cover slips and immerse slides in cold lysis solution for 60 min at 4°C.
7. Place slides into an electrophoresis chamber containing cold (4°C) electro-
phoresis solution for 20 min.
8. Connect the electrophoresis tank to the power supply, and perform electro-
phoresis at 1.25 V/cm and 300 mA for 20 min at 4°C.
9. Remove slides from the electrophoresis tank, and wash them once for 20
min in neutralization buffer at room temperature (RT).
Comet-FISH
1. Dehydrate slides in absolute ethanol for at least three days.
2. Rehydrate slides in double-distilled H2O for 15 min.
3. Denature DNA in 0.5 M NaOH for 25 min at RT.
4. Dehydrate slides in an ethanol series (70, 80, and 95%, each for 5 min).
5. Dry slides at RT.
6. Denature telomere PNA probes by preheating to 80°C in water bath for 3
min.
7. Pipette 10 µl denatured probe to an area of approximately 20×20 mm,
cover with coverslip of appropriate size, and seal with rubber cement.
8. Incubate overnight at 37°C in a humid box.
9. Wash slides in prewarmed post-hybridization washing solution (from the
PNA FISH Kit/Cy3) at 65°C in water bath, without agitation for 2.5 min,
and cool the slides immediately in cold 1× phosphate-buffered detergent
(PBD).
10. Follow the supplier’s instructions for denaturation, hybridization, and
post-hybridization washing when using other DNA probes.
11. Counterstain the slides with SYBR Green including 50% antifade (30 μl
per slide), cover with a coverslip 24×60mm, and evaluate the results under
fuorescence microscope.
Evaluation
1. Apply comet analysis software (e.g. Comet Assay IV analysis system,
Perceptive Instruments, Suffolk, UK) for comet images.
218 Cytogenetics and Molecular Cytogenetics
2. Analyze visually the number of FISH signals per comet and the distribution
of FISH signals between head and tail for Comet-FISH images.
YIELDS
Comet-FISH allows the analysis of damage and repair at the level of the overall
DNA, and the localization of damage in specifc loci of the genome on the same
preparations (Figure 20.1). Methodological differences between Comet-FISH and
FISH are aimed at preserving the gel layer, and practice shows that this goal is quite
achievable. The following outputs are expected to be achieved:
IMPORTANT NOTES
• The FISH signal is able to migrate to the comet’s tail if the break occurs
not only within a specifc loci of the genome, but also in the vicinity of the
area of interest.[16]
• Certain genes or parts of genes or other loci of the genome remain in the
head of comet even when there is a DNA break nearby, and it is likely that
this localization within the head refects the presence nearby of scaffold- or
matrix-associated areas.[16]
• Cells immersed in gel retain their minimally deformed three-dimensional
structure. However, the location of cells at different depths in the gel sug-
gests special attention when analyzing images,[28] and even does not exclude
the possibility that some signals immersed deep in the gel may be invisible.
CONCLUSIONS
FISH allows detecting genomic regions of interest immediately on comet prepara-
tions. Application of FISH on comets is based on replacing the thermal denaturation
of DNA with an alkaline one to preserve the structure of the gel in which the cells
are embedded. In that way, global genomic and sequence-specifc DNA damage and
repair of cells from almost any tissue can be comparatively investigated.
Comet-FISH studies were carried out using DNA probes for whole chromosomes,
specifc chromosome regions, centromeres, telomeres, genes, and any other genome
loci. Various examples of the application of the method on human, animal, and plant
cells confrmed sensitivity and the usefulness of this approach in genotoxicity testing
and biomonitoring.
ACKNOWLEDGMENTS
This work was supported by the RA MESCS and BMBF (project #AG-01/20), and
RA MESCS (project #21AG-1F068).
REFERENCES
1. Ostling, O.; Johanson, K.J. Microelectrophoretic study of radiation-induced DNA dam-
ages in individual mammalian cells. Biochem Biophys Res Commun. 1984, 123(1),
291–298.
2. Singh, N.P.; McCoy, M.T.; Tice, R.R.; Schneider, E.L. A simple technique for quan-
titation of low levels of DNA damage in individual cells. Exp Cell Res. 1988, 175(1),
184–191.
3. Cordelli, E.; Bignami, M.; Pacchierotti, F. Comet assay: A versatile but complex tool in
genotoxicity testing. Toxicol Res (Camb). 2021, 10(1), 68–78.
4. Karbaschi, M.; Ji, Y.; Abdulwahed, A.M.S.; Alohaly, A.; Bedoya, J.F.; Burke, S.L.;
Boulos, T.M.; Tempest, H.G.; Cooke, M.S. Evaluation of the major steps in the conven-
tional protocol for the alkaline comet assay. Int J Mol Sci. 2019, 20(23), 6072.
5. Azqueta, A.; Muruzabal, D.; Boutet-Robinet, E.; Milic, M.; Dusinska, M.; Brunborg, G.;
Møller, P.; Collins, A.R. Technical recommendations to perform the alkaline standard
220 Cytogenetics and Molecular Cytogenetics
and enzyme-modifed comet assay in human biomonitoring studies. Mutat Res Genet
Toxicol Environ Mutagen. 2019, 43, 24–32.
6. Muruzabal, D.; Collins, A.; Azqueta, A. The enzyme-modifed comet assay: Past, pres-
ent and future. Food Chem Toxicol. 2021, 147, 111865.
7. Muruzabal, D.; Sanz-Serrano, J.; Sauvaigo, S.; Treillard, B.; Olsen, A.K.; López de
Cerain, A.; Vettorazzi, A.; Azqueta, A. Validation of the in vitro comet assay for DNA
cross-links and altered bases detection. Arch Toxicol. 2021, 95(8), 2825–2838.
8. Spivak, G.; Cox, R.A.; Hanawalt, P.C. New applications of the Comet assay: Comet-
FISH and transcription-coupled DNA repair. Mutat Res. 2009, 681(1), 44–50.
9. de Lapuente, J.; Lourenço, J.; Mendo, S.A.; Borràs, M.; Martins, M.G.; Costa, P.M.;
Pacheco, M. The Comet assay and its applications in the feld of ecotoxicology: A mature
tool that continues to expand its perspectives. Front Genet. 2015, 4(6), 180.
10. Gajski, G.; Žegura, B.; Ladeira, C.; Pourrut, B.; Del Bo’, C.; Novak, M.; Sramkova, M.;
Milić, M.; Gutzkow, K.B.; Costa, S.; Dusinska, M.; Brunborg, G.; Collins, A. The comet
assay in animal models: From bugs to whales: (Part 1 Invertebrates). Mutat Res. 2019,
779, 82–113.
11. Gajski, G.; Žegura, B.; Ladeira, C.; Novak, M.; Sramkova, M.; Pourrut, B.; Del Bo’, C.;
Milić, M.; Gutzkow, K.B.; Costa, S.; Dusinska, M.; Brunborg, G.; Collins, A. The comet
assay in animal models: From bugs to whales: (Part 2 Vertebrates). Mutat Res Rev Mutat
Res. 2019, 781, 130–164.
12. Pietrini, F.; Iannilli, V.; Passatore, L.; Carloni, S.; Sciacca, G.; Cerasa, M.; Zacchini, M.
Ecotoxicological and genotoxic effects of dimethyl phthalate (DMP) on Lemna minor
L. and Spirodela polyrhiza (L.) Schleid. plants under a short-term laboratory assay. Sci
Total Environ. 2022, 1;806(Pt 4), 150972.
13. Azqueta, A.; Ladeira, C.; Giovannelli, L.; Boutet-Robinet, E.; Bonassi, S.; Neri, M.;
Gajski, G.; Duthie, S.; Del Bo’, C.; Riso, P.; Koppen, G.; Basaran, N.; Collins, A.; Møller,
P. Application of the comet assay in human biomonitoring: An hCOMET perspective.
Mutat Res Rev Mutat Res. 2020, 783, 108288.
14. Bonassi, S.; Ceppi, M.; Møller, P.; Azqueta, A.; Milić, M.; Neri, M.; Brunborg, G.;
Godschalk, R.; Koppen, G.; Langie, S.A.S.; Teixeira, J.P.; Bruzzone, M.; Da Silva,
J.; Benedetti, D.; Cavallo, D.; Ursini, C.L.; Giovannelli, L.; Moretti, S.; Riso, P.; Del
Bo’, C.; Russo, P.; Dobrzyńska, M.; Goroshinskaya, I.A.; Surikova, E.I.; Staruchova,
M.; Barančokova, M.; Volkovova, K.; Kažimirova, A.; Smolkova, B.; Laffon, B.;
Valdiglesias, V.; Pastor, S.; Marcos, R.; Hernández, A.; Gajski, G.; Spremo-Potparević,
B.; Živković, L.; Boutet-Robinet, E.; Perdry, H.; Lebailly, P.; Perez, C.L.; Basaran, N.;
Nemeth, Z.; Safar, A.; Dusinska, M.; Collins, A. hCOMET project. DNA damage in
circulating leukocytes measured with the comet assay may predict the risk of death. Sci
Rep. 2021, 11(1), 16793.
15. Milić, M.; Ceppi, M.; Bruzzone, M.; Azqueta, A.; Brunborg, G.; Godschalk, R.; Koppen,
G.; Langie, S.; Møller, P.; Teixeira, J.P.; Alija, A.; Anderson, D.; Andrade, V.; Andreoli,
C.; Asllani, F.; Bangkoglu, E.E.; Barančoková, M.; Basaran, N.; Boutet-Robinet, E.;
Buschini, A.; Cavallo, D.; Costa Pereira, C.; Costa, C.; Costa, S.; Da Silva, J.; Del Boˊ, C.;
Dimitrijević Srećković, V.; Djelić, N.; Dobrzyńska, M.; Duračková, Z.; Dvořáková, M.;
Gajski, G.; Galati, S.; García Lima, O.; Giovannelli, L.; Goroshinskaya, I.A.; Grindel,
A.; Gutzkow, K.B.; Hernández, A.; Hernández, C.; Holven, K.B.; Ibero-Baraibar,
I.; Ottestad, I.; Kadioglu, E.; Kažimirová, A.; Kuznetsova, E, Ladeira, C, Laffon, B,
Lamonaca, P, Lebailly, P, Louro, H, Mandina Cardoso, T, Marcon, F.; Marcos, R.;
Moretti, M.; Moretti, S.; Najafzadeh, M.; Nemeth, Z.; Neri, M.; Novotna, B.; Orlow, I.;
Paduchova, Z.; Pastor, S.; Perdry, H.; Spremo-Potparević, B.; Ramadhani, D.; Riso, P.;
Rohr, P.; Rojas, E.; Rossner, P.; Safar, A.; Sardas, S.; Silva, M.J.; Sirota, N.; Smolkova,
FISH—and Comet Assay 221
B.; Staruchova, M.; Stetina, R.; Stopper, H.; Surikova, E.I.; Ulven, S.M.; Ursini, C.L.;
Valdiglesias, V.; Valverde, M.; Vodicka, P.; Volkovova, K.; Wagner, K.H.; Živković, L.;
Dušinská, M.; Collins, A.R.; Bonassi, S. The hCOMET project: International database
comparison of results with the comet assay in human biomonitoring. Baseline frequency
of DNA damage and effect of main confounders. Mutat Res Rev Mutat Res. 2021, 787,
108371.
16. Glei, M.; Hovhannisyan, G.; Pool-Zobel, B.L. Use of Comet-FISH in the study of DNA
damage and repair: Review. Mutat Res. 2009, 681(1), 33–43.
17. Santos, S.J.; Singh, N.P.; Natarajan, A.T. Fluorescence in situ hybridization with comets.
Exp Cell Res. 1997, 232(2), 407–411.
18. Rapp, A.; Hausmann, M.; Greulich, K.O. The comet-FISH technique: A tool for detec-
tion of specifc DNA damage and repair. Methods Mol Biol. 2005, 291, 107–119.
19. Spivak, G. The Comet-FISH assay for the analysis of DNA damage and repair. Methods
Mol Biol. 2010, 659, 129–145.
20. Shaposhnikov, S.; Thomsen, P.D.; Collins, A.R. Combining fuorescent in situ hybridiza-
tion with the comet assay for targeted examination of DNA damage and repair. Methods
Mol Biol. 2011, 682, 115–132.
21. Henriksson, S.; Nilsson, M. Padlock probes and rolling circle amplifcation for detection
of repeats and single-copy genes in the single-cell comet assay. Methods Mol Biol. 2012,
853, 95–103.
22. Schlörmann, W.; Glei, M. Detection of DNA damage by comet fuorescence in situ
hybridization. Methods Mol Biol. 2012, 920, 91–100.
23. Laubenthal, J.; Anderson, D. Fluorescence in situ hybridization on electrophoresed cells
to detect sequence specifc DNA damage. Methods Mol Biol. 2013, 1054, 219–235.
24. Glei, M.; Schlörmann, W. Analysis of DNA damage and repair by comet fuorescence in
situ hybridization (Comet-FISH). Methods Mol Biol. 2014, 1094, 39–48.
25. Shaposhnikov, S.; El Yamani, N.; Collins, A.R. Fluorescent in situ hybridization on com-
ets: FISH comet. Methods Mol Biol. 2015, 1288, 363–373.
26. Mondal, M.; Guo, J. Comet-FISH for ultrasensitive strand-specifc detection of DNA
damage in single cells. Methods Enzymol. 2017, 591, 83–95.
27. Hovhannisyan, G.; Aroutiounian, R. Comet-FISH In: Fluorescence In Situ Hybridization
(FISH), Application Guide; Liehr, T., Ed. 2nd Edition. Springer, Berlin, 2017,
pp. 373–378.
28. Baugh, E.H.; Ke, H.; Levine, A.J.; Bonneau, R.A.; Chan, C.S. Why are there hotspot
mutations in the TP53 gene in human cancers? Cell Death Differ. 2018, 25(1),
154–160.
29. McKenna, D.J.; Rajab, N.F.; McKeown, S.R.; McKerr, G.; McKelvey-Martin, V.J. Use
of the comet-FISH assay to demonstrate repair of the TP53 gene region in two human
bladder carcinoma cell lines. Radiat Res. 2003, 159(1), 49–56.
30. McKenna, D.J.; Gallus, M.; McKeown, S.R.; Downes, C.S.; McKelvey-Martin, V.J.
Modifcation of the alkaline Comet assay to allow simultaneous evaluation of mitomycin
C-induced DNA cross-link damage and repair of specifc DNA sequences in RT4 cells.
DNA Repair (Amst). 2003, 2(8), 879–890.
31. McKenna, D.J.; Doherty, B.A.; Downes, C.S.; McKeown, S.R.; McKelvey-Martin, V.J.
Use of the comet-FISH assay to compare DNA damage and repair in p53 and hTERT
genes following ionizing radiation. PLoS One. 2012, 7(11), e49364.
32. Shaposhnikov, S.; Frengen, E.; Collins, A.R. Increasing the resolution of the comet assay
using fuorescent in situ hybridization: A review. Mutagenesis. 2009, 24(5), 383–389.
33. Glei, M.; Hovhannisyan, G.; Pool-Zobel, B.L. Use of Comet-FISH in the study of DNA
damage and repair: Review. Mutat Res. 2009, 681(1), 33–43.
222 Cytogenetics and Molecular Cytogenetics
34. Hovhannisyan, G.G. Fluorescence in situ hybridization in combination with the comet
assay and micronucleus test in genetic toxicology. Mol Cytogenet. 2010, 3, 17.
35. Azqueta, A.; Collins, A.R. The essential comet assay: A comprehensive guide to measur-
ing DNA damage and repair. Arch Toxicol. 2013, 87(6), 949–968.
36. Spivak, G. New developments in comet-FISH. Mutagenesis. 2015, 30(1), 5–9.
37. Kumaravel, T.S.; Bristow, R.G. Detection of genetic instability at HER-2/neu and p53
loci in breast cancer cells sing Comet-FISH. Breast Cancer Res Treat. 2005, 91(1), 89–93.
38. Horváthová, E.; Dusinská, M.; Shaposhnikov, S.; Collins, A.R. DNA damage and repair
measured in different genomic regions using the comet assay with fuorescent in situ
hybridization. Mutagenesis. 2004, 19(4), 269–276.
39. Mladinic, M.; Zeljezic, D.; Shaposhnikov, S.A.; Collins, A.R. The use of FISH-comet
to detect c-Myc and TP 53 damage in extended-term lymphocyte cultures treated with
terbuthylazine and carbofuran. Toxicol Lett. 2012, 211(1), 62–69.
40. Zeljezic, D.; Vinkovic, B.; Kasuba, V.; Kopjar, N.; Milic, M.; Mladinic, M. The effect of
insecticides chlorpyrifos, α-cypermethrin and imidacloprid on primary DNA damage,
TP 53 and c-Myc structural integrity by comet-FISH assay. Chemosphere. 2017, 182,
332–338.
41. McAllister, K.A.; Yasseen, A.A.; McKerr, G.; Downes, C.S.; McKelvey-Martin, V.J.
FISH comets show that the salvage enzyme TK1 contributes to gene-specifc DNA
repair. Front Genet. 2014, 5, 233.
42. Glei, M.; Schaeferhenrich, A.; Claussen, U.; Kuechler, A.; Liehr, T.; Weise, A.; Marian,
B.; Sendt, W.; Pool-Zobel, B.L. Comet fuorescence in situ hybridization analysis for
oxidative stress-induced DNA damage in colon cancer relevant genes. Toxicol Sci. 2007,
96(2), 279–284.
43. Reiter, M.; Welz, C.; Baumeister, P.; Schwenk-Zieger, S.; Harréus, U. Mutagen sensitivity
and DNA repair of the EGFR gene in oropharyngeal cancer. Oral Oncol. 2010, 46(7),
519–524.
44. Reiter, M.; Baumeister, P.; Hartmann, M.; Schwenk-Zieger, S.; Harréus, U.
Chemoprevention by celecoxib and mutagen sensitivity of cyclin d1 in patients with oro-
pharyngeal carcinoma. In Vivo. 2014, 28(1), 49–53.
45. Razo-Aguilera, G.; Baez-Reyes, R.; Alvarez-González, I.; Paniagua-Pérez, R.; Madrigal-
Bujaidar, E. Inhibitory effect of grapefruit juice on the genotoxicity induced by hydrogen
peroxide in human lymphocytes. Food Chem Toxicol. 2011, 49(11), 2947–2953.
46. Escobar, P.A.; Smith, M.T.; Vasishta, A.; Hubbard, A.E.; Zhang, L. Leukaemia-specifc
chromosome damage detected by comet with fuorescence in situ hybridization (comet-
FISH). Mutagenesis. 2007, 22(5), 321–327.
47. Harréus, U.A.; Kleinsasser, N.H.; Zieger, S.; Wallner, B.; Reiter, M.; Schuller, P.;
Berghaus, A. Sensitivity to DNA-damage induction and chromosomal alterations in
mucosa cells from patients with and without cancer of the oropharynx detected by a
combination of Comet assay and fuorescence in situ hybridization. Mutat Res. 2004,
563(2), 131–138.
48. Mohseni Meybodi, A.; Mozdarani, H. DNA damage in leukocytes from Fanconi ane-
mia (FA) patients and heterozygotes induced by mitomycin C and ionizing radiation as
assessed by the comet and comet-FISH assay. Iran Biomed J. 2009, 13(1), 1–8.
49. Arutyunyan, R.; Gebhart, E.; Hovhannisyan, G.; Greulich, K.O.; Rapp, A. Comet-FISH
using peptide nucleic acid probes detects telomeric repeats in DNA damaged by bleomy-
cin and mitomycin C proportional to general DNA damage. Mutagenesis. 2004, 19(5),
403–408.
50. Arutyunyan, R.; Rapp, A.; Greulich, K.O.; Hovhannisyan, G.; Haroutiunian, S.; Gebhart,
E. Fragility of telomeres after bleomycin and cisplatin combined treatment measured in
human leukocytes with the Comet-FISH technique. Exp Oncol. 2005, 27(1), 38–42.
FISH—and Comet Assay 223
51. Hovhannisyan, G.; Rapp, A.; Arutyunyan, R.; Greulich, K.O.; Gebhart, E. Comet-assay
in combination with PNA-FISH detects mutagen-induced DNA damage and specifc
repeat sequences in the damaged DNA of transformed cells. Int J Mol Med. 2005, 15(3),
437–442.
52. Shaposhnikov, S.; Azqueta, A.; Henriksson, S.; Meier, S.; Gaivão, I.; Huskisson, N.H.;
Smart, A.; Brunborg, G.; Nilsson, M.; Collins, A.R. Twelve-gel slide format optimised
for comet assay and fuorescent in situ hybridisation. Toxicol Lett. 2010, 195(1), 31–34.
53. Shaposhnikov, S.; Larsson, C.; Henriksson, S.; Collins, A.; Nilsson, M. Detection of
Alu sequences and mtDNA in comets using padlock probes. Mutagenesis. 2006, 21(4),
243–247.
54. Henriksson, S.; Shaposhnikov, S.; Nilsson, M.; Collins, A. Study of gene-specifc DNA
repair in the comet assay with padlock probes and rolling circle amplifcation. Toxicol
Lett. 2011, 202(2), 142–147.
55. Guo, J.; Hanawalt, P.C.; Spivak, G. Comet-FISH with strand-specifc probes reveals
transcription-coupled repair of 8-oxoGuanine in human cells. Nucleic Acids Res. 2013,
41(16), 7700–7712.
56. Amendola, R.; Basso, E.; Pacifci, P.G.; Piras, E.; Giovanetti, A.; Volpato, C.; Romeo, G.
Ret, Abl1 (cAbl) and Trp53 gene fragmentations in comet-FISH assay act as in vivo bio-
markers of radiation exposure in C57BL/6 and CBA/J mice. Radiat Res. 2006, 165(5),
553–5561.
57. Zeljezic, D.; Mladinic, M.; Kopjar, N.; Radulovic, A.H. Evaluation of genome damage
in subjects occupationally exposed to possible carcinogens. Toxicol Ind Health. 2016,
32(9), 1570–1580.
58. Pérez-García, C.; Rouxel, J.; Akcha, F. Development of a comet-FISH assay for the
detection of DNA damage in hemocytes of Crassostrea gigas. Aquat Toxicol. 2015, 161,
189–195.
59. Menke, M.; Angelis, K.J.; Schubert, I. Detection of specifc DNA lesions by a combina-
tion of comet assay and FISH in plants. Environ Mol Mutagen. 2000, 35(2), 132–138.
60. Kwasniewska, J.; Grabowska, M.; Kwasniewski, M.; Kolano, B. Comet-FISH with rDNA
probes for the analysis of mutagen-induced DNA damage in plant cells. Environ Mol
Mutagen. 2012, 53(5), 369–375.
61. Kwasniewska, J.; Kwasniewski, M. Comet-FISH for the evaluation of plant DNA damage
after mutagenic treatments. J Appl Genet. 2013, 54(4), 407–415.
62. Kwasniewska, J.; Mikolajczyk, A. Infuence of the presence of B chromosomes on DNA
damage in Crepis capillaris. PLoS One. 2014, 9(1), e87337.
63. Castro, C.; Carvalho, A.; Gaivão, I.; Lima-Brito, J. Evaluation of copper-induced DNA
damage in Vitis vinifera L. using Comet-FISH. Environ Sci Pollut Res Int. 2021, 28(6),
6600–6610.
64. Russo, C.; Acito, M.; Fatigoni, C.; Villarini, M.; Moretti, M. B-Comet Assay (Comet
Assay on buccal cells) for the evaluation of primary DNA damage in human biomonitor-
ing studies. Int J Environ Res Public Health. 2020, 17(24), 9234.
65. Hobbs, C.A.; Recio, L.; Winters, J.; Witt, K.L. Use of frozen tissue in the Comet Assay
for the evaluation of DNA damage. J Vis Exp. 2020, 157.
66. Pant, K.; Springer, S.; Bruce, S.; Lawlor, T.; Hewitt, N.; Aardema, M.J. Vehicle and posi-
tive control values from the in vivo rodent comet assay and biomonitoring studies using
human lymphocytes: Historical database and infuence of technical aspects. Environ
Mol Mutagen. 2014, 55(8), 633–642.
67. Ladeira, C.; Koppen, G.; Scavone, F.; Giovannelli, L. The comet assay for human bio-
monitoring: Effect of cryopreservation on DNA damage in different blood cell prepara-
tions. Mutat Res Genet Toxicol Environ Mutagen. 2019, 843, 11–17.
21 Molecular Karyotyping
Ilda Patrícia Ribeiro, Luís Miguel Pires,
Susana Isabel Ferreira, Mariana Val,
Joana Barbosa Melo and Isabel Marques Carreira
CONTENTS
Introduction............................................................................................................ 225
The aCGH Technology and Data Interpretation .................................................... 226
Different Platforms: Past and at Present ................................................................ 229
Advantages and Limitations................................................................................... 232
Applications ........................................................................................................... 232
Molecular Karyotyping Approach in Human Genetic Diagnostics.............. 233
Molecular Karyotyping Approach in Prenatal Diagnostics .......................... 233
Molecular Karyotyping Approach in Cancer Research ................................ 233
Clinical Cases......................................................................................................... 233
Clinical Case 1.............................................................................................. 234
Clinical Case 2.............................................................................................. 236
Conclusion for Clinical Cases 1 and 2.......................................................... 237
Clinical Cases 3 and 4................................................................................... 238
Clinical Case 3.............................................................................................. 238
Clinical Case 4.............................................................................................. 240
Clinical Cases 5 and 6................................................................................... 243
Clinical Case 5.................................................................................. 243
Clinical Case 6.................................................................................. 243
The Impact of Molecular Karyotyping in Clinical Practice................................... 246
Conclusions............................................................................................................ 246
References.............................................................................................................. 247
INTRODUCTION
Nowadays, several diseases are linked to copy-number variants (CNVs), and various
genome-wide methods can be used for its detection. However, it is important to high-
light that the presence of CNVs by itself does not mean an abnormal or pathogenic
phenotype. The clinical relevance of CNVs depends on its functional impact, having
a high probability of a phenotypic effect if the region of imbalance maps critical
gene/s or an important regulatory region.
The most commonly used method for CNVs detection has been array-based
comparative genomic hybridization (aCGH). This technique was frst described
in 1997 by Solinas Toldo,[1] and its frst applications were published by the groups
of Pinkel, Snidjers and Buckley in the following years.[2–4] Initially, this technique
was introduced as matrix-CGH, and later coined as array-CGH (aCGH).[3] In 2005,
Vermeesch and colleagues proposed to call this technology as molecular karyo-
typing.[5] Currently, the use of aCGH technology is massifed worldwide either at
diagnostic or investigation point of view. Many reports were published concerning
chromosome imbalances associated with cancer as well as in constitutional cytoge-
netics, with postnatal and prenatal applications. The aCGH technology use whole
genome probes immobilized on glass slides (a microarray) that allow, due to the
deep resolution, the diagnosis of smaller genomic imbalances, being the method
of choice to small genomic imbalances identifcation with a wide range of clinical
applications. It was estimated that the introduction of aCGH technique in the clinical
practice allowed increasing the detection of apparently pathogenic genomic imbal-
ances in as much as 20% of cases that have had normal karyotyping tests.[6]
DNA is washed off the slide. The reference sample should be gender-matched with
the sample in study. After the hybridization, the slides are scanned into TIFF image
fles by a microarray scanner, which uses laser light to measure the relative intensity
of emission, usually at green (Cy3—for control) and red (Cy5—for biological sam-
ple in study) wavelengths being all targets’ intensities measured, and the image fles
are quantifed in a specifc feature extraction software. The green to red fuorescence
log2 ratio is calculated for each spot. The copy number alterations are calculated by
differences in fuorescence intensities along any given spot(s), enabling the detection
of gains and losses throughout the genome with high resolution. Therefore, equal
amount of green and red fuorescence for a specifc spot signifes that this region of
the genome to which that spot is mapped harbors an equal copy-number in the tested
and control samples. In the other hand, a ratio favoring red fuorescence implies
an excess of that genomic segment in the test genome (i.e., a copy-number gain)
in comparison with the control, whereas a green signal shows a copy number loss
(Figure 21.1).
The principle of aCGH technique can be demonstrated with the following sim-
ple example (Figure 21.2A). DNA from a male with 9p trisomy and a partial 9q
(9q13-q31.2) trisomy, labeled in red, is co-hybridized with DNA from a normal male
(46,XY) labeled in green against an array slide. At each target sequence of the array
slide without copy number variants, a balanced, yellow color is observed. Since at
chromosome 9 (specifcally 9p and 9q13-q31.2) there is more DNA labeled with red
due to trisomy 9, this means that in the competitive hybridization higher quantity of
‘DNA red’ will be linked in comparison with DNA labelled with green (from con-
trol) for this chromosome, and consequently a gain of chromosome 9 material will
be visualized after this experiment (represented in a positive scale, by a right devia-
tion of the blue color in the Figure 21.2A).
A genotype–phenotype correlation is crucial to interpret the CNVs detected. Not
all CNVs are associated with unfavorable clinical phenotypes, being in some cases
classifed as benign—without impact in the phenotype. In other cases, CNVs are
linked to a spectrum of clinical phenotypes that may range from benign to pathologi-
cal, including a very challenging class of CNVs named VUS (variant of unknown/
uncertain clinical signifcance). Therefore, CNVs could be phenotypically benign,
because of phenotypic differences created by evolution, or could be responsible for
disease or for disease susceptibility. Additionally, the CNVs could be categorized
as germ line (constitutional), where every cell of the body is altered, or somatic,
where only a subgroup of cells in the organism harbors the alteration, such as in
cancer or in mosaics. The interpretation of the results and consequently the assess-
ment of the pathogenicity of a CNV could be challenging and needs to consider the
size and location of the copy number alteration, breakpoint information, whether it
has occurred de novo, and gene content, namely whether the region contains genes
of known importance. Determining whether a specifc CNV is present in affected
(or unaffected) relatives can also be important as well as verify in databases if other
unrelated but affected individuals with similar clinical phenotypes are described,
namely the use of CNV databases such as DECIPHER (www.deciphergenomics.org/)
and if those CNVs are also reported among healthy individuals, using CNV data-
bases such as the Database of Genomic Variants (http://dgv.tcag.ca/dgv/app/home).
228
Cytogenetics and Molecular Cytogenetics
FIGURE 21.1 aCGH results of chromosome 16 from three different samples, using CytoGenomics software for analysis.
A) Chromosome view.
B) Gene view, presenting the gene content in this chromosome region. Sample 1 presents a deletion of 541 kb at 16p11.2, involving 30 genes arr[GRCh37]
16p11.2(29656684_30197341)x1; Sample 2 without CNVs at 16p11.2; and Sample 3 presents a duplication of 544 kb at 16p11.2, involving 34 genes
arr[GRCh37] 16p11.2(29652999_30197341)x3.
Molecular Karyotyping 229
FIGURE 21.2 A) Chromosome view and respective gene content of a 9p trisomy and a
partial 9q trisomy. Results obtained using aCGH technique and analyzed with Cytogenomics
software. In orange are represented the OMIM Morbid Map genes, in blue the OMIM genes
and in grey the RefSeq genes. Trisomy is represented as a positive scale. B) Plot of chro-
mosome 22 displaying a gain at 22q11.21, detected by aCGH technique and analyzed with
Cytogenomics software. The duplicated region is shown in the UCSC Genome Browser
(https://genome.ucsc.edu/) with some selected tracks to assist in the genotype–phenotype
correlation and consequently in the clinical relevance determination. Example of these tracks
are chromosome band, RefSeq genes, OMIM genes, DECIPHER CNVs, ClinGen CNVs and
DGV.
There are also several public databases and sources such as PubMed (https://
pubmed.ncbi.nlm.nih.gov/), OMIM (www.omim.org/) and human genome browsers
(UCSC—https://genome.ucsc.edu/, Ensembl—www.ensembl.org/index.html) that
provide useful tracks of genome-oriented data (Figure 21.2B).[15]
231
232 Cytogenetics and Molecular Cytogenetics
APPLICATIONS
The application of aCGH into the clinical practice has introduced a paradigm shift
in the diagnostic workup and accelerated the identifcation of the molecular basis of
several genetic diseases.[22] Initially, CGH was developed as a research tool for the
investigation of CNVs in cancer. However, nowadays, aCGH technique is mainly
used as a routine diagnostic tool, being the frst-tier clinical diagnostic test for
patients with intellectual disability, congenital anomalies and autistic spectrum dis-
order, and more recently has also been applied for the detection of genomic imbal-
ances in prenatal genetic diagnosis. Since CNVs are common in the genome, the
clinical signifcance and the genotype-phenotype correlation of aCGH results can be
Molecular Karyotyping 233
challenging, parental analysis and the use of several internet-based databases being
pivotal. The application of aCGH technique in three main areas will be discussed:
CLINICAL CASES
Selected clinical cases will be presented as example of aCGH applications in post-
natal human genetic diagnosis, in prenatal diagnosis and in cancer characterization.
All cases were analyzed by aCGH technique using Agilent SurePrint G3 Human
Genome microarray 180 K (Agilent Technologies, Santa Clara, CA, USA).
234 Cytogenetics and Molecular Cytogenetics
For all clinical post and prenatal cases, three questions are to be answered:
i) How to interpret this result? How to classify this CNV? What is the most
probable mechanism of origin of the alteration?
ii) What is the origin of this CNV?
iii) What can we offer to this couple? What is the future strategy for the family?
Clinical cases 1 and 2 are two examples of aCGH application in postnatal human
genetic diagnosis (using DNA extracted from peripheral blood), with different
outcomes.
CLINICAL CASE 1
A 11-year-old boy with renal insuffciency, retinopathy and ataxia was referred for
aCGH that identifed a 105 kb homozygous deletion at chromosome 2, arr[GRCh37]
2q13(110862477_110967912)x0 (Figure 21.4A).
i) How to interpret this result? How to classify this CNV? What is the most
probable mechanism of origin of the alteration?
There are several CNVs described in this region at DECIPHER database. Two genes,
MALL and NPHP1, are mapped in this region. NPHP1 gene is present in OMIM
Morbid Map and is associated with nephronophthisis 1, juvenile, an autosomal reces-
sive cystic kidney disease that leads to renal failure. Almost 15% of nephronophthi-
sis patients present extrarenal symptoms affecting other organs, such as eyes, liver,
bones and central nervous system.[36] Around two thirds of the patients with familial
juvenile nephronophthisis have a homozygous deletion at the 2q13.[37]
This homozygous deletion seems to explain the clinical phenotype of the boy. In
order to establish the most probable mechanism of origin of the alteration, the aCGH
study of the progenitors were recommended.
iii) What can we offer to this couple? What is the future strategy for the
family?
This couple received genetic counselling, where the implication of the molecular
fndings was explained, and prenatal diagnosis was offered for future pregnancies.
Later on, this couple had a new pregnancy and requested an invasive prenatal diag-
nosis. DNA from amniotic fuid was analyzed by aCGH technique, and it was verifed
that the fetus has inherited the deletion from one of the parents (Figure 21.4C). Thus,
it is expected that the fetus did not present any phenotype like the carrier progenitors.
FIGURE 21.4 A) Chromosomal view and corresponding gene content of a 2q13 homo-
zygous deletion identifed in the 11-year-old boy. Results obtained using aCGH technique
and analyzed using Agilent Genomic Workbench v6.5 software. The results are according
to Human Genome build 19 and include imbalances with at least three consecutive probes
with abnormal log2 ratios. Deletion is represented in a negative scale. B) Chromosomal view
and respective gene content of the 2q13 deletion identifed in the mother and the father of
the 11-year-old boy. Results obtained using aCGH technique and analyzed using Agilent
Genomic Workbench v6.5 software. The results are according to Human Genome build 19
and include imbalances with at least three consecutive probes with abnormal log2 ratios.
Deletion is represented in a negative scale. C) Chromosomal view and respective gene content
of the 2q13 deletion identifed in a sample of amniotic fuid during pregnancy of this couple
that have a heterozygous deletion in arr[GRCh37] 2q13(110862477_110967912)x1 and a son
that inherited from both parents this deletion. Results obtained using aCGH technique and
analyzed with Cytogenomics software. Deletion is represented in a negative scale.
236 Cytogenetics and Molecular Cytogenetics
CLINICAL CASE 2
A newborn with several congenital anomalies, such as axial hypotonia, retrog-
nathism, fetal growth restriction, high palate, and hypogonadism was referred
for aCGH that identifed two genomic anomalies (Figure 21.5): arr[GRCh37]
10p12.1(26182512_26815179)x3,15q11.2q13.1(22765628_28940098)x4. The mother
has a brother with global psychomotor developmental delay and a son with hyperac-
tivity and global psychomotor developmental delay.
Results obtained using aCGH technique and analyzed with CytoGenomics soft-
ware. Gain of genetic material is represented in a positive scale.
i) How to interpret this result? How to classify this CNV? What is the most
probable mechanism of origin of the alteration?
• duplication of 632 Kb at 10p12.1 includes three genes described at OMIM
database (OMIM ID: 606808-MYO3A, 138275‑GAD2, 609036-APB‑
B1IP). The MYO3A gene is described at OMIM Morbid Map and related
to autosomal recessive deafness. CNVs in duplication are not reported
for this region in healthy individuals at Database of Genomic Variants.
In the DECIPHER database, there is one patient with cognitive defcit
that has a similar duplication (ID: 300716) with unknown origin, which
was classifed as likely pathogenic. However, this reported patient also
presented another genomic alteration, a 657Kb deletion at 1p36.23 of
unknown origin, also classifed as likely pathogenic. At the ClinGen data-
base, there are bigger CNVs in duplication than those of our case, which
were classifed either as benign or pathogenic or even likely pathogenic.
• triplication of 6.1Mb at 15q11.2q13.1 includes several genes, 24 reported
at OMIM database and 10 at OMIM Morbid Map database (OMIM ID:
608145-NIPA1, 603856-MKRN3, 605283-MAGEL2, 602117-NDN,
182279-SNRPN, 601623-UBE3A, 137192-GABRB3, 137142-GABRA5,
611409-OCA2, 605837-HERC2). In the literature, triplication of
15q11.2q13.1 is related with hypotonia, motor delay, intellectual disabil-
ity and autism.[38] These disabilities ft with the clinical phenotype of our
patient.
ii) What is the origin of this CNV?
Both parents were also analyzed by aCGH. It was possible to conclude that the
10p12.1 duplication of our patient was inherited from the mother (phenotypi-
cally normal) and was classifed, considering the present knowledge as a CNV
of unknown clinical signifcance. The tetrasomy of 15q11.2q13.1 is de novo and
classifed as pathogenic. arr[GRCh37]10p12.1(26182512_26815179)×3mat,15q11
.2q13.1(22765628_28940098)×4 dn.
iii) What can we offer to this couple? What is the future strategy for the
family?
This couple was referred to genetic counselling. Monitoring and testing future preg-
nancies were recommended.
about the clinical signifcance of some CNVs, which is even more challenging in
prenatal cases. It is important, whenever possible to revisit some of these VUS, spe-
cifcally when a new pregnancy occurs in that family.
CLINICAL CASE 3
A 29 weeks and 5 days’ pregnancy, showed by ultrasound a fetus with cleft lip, cleft
palate and defect in the ventricular septum. Amniotic fuid was collected and conven-
tional cytogenetic and aCGH techniques requested. Both techniques identifed two
genomic anomalies in this female fetus: 46,XX,rec(4)dup(4p)inv(4)(p15.33q31.3)
a r r[GRCh37]4p16.3p1533(45882 _11887210)×3,4q313q35.2(155125113_
190469337)×1.
i) How to interpret this result? How to classify this CNV? What is the most
probable mechanism of origin of the alteration?
Regarding the most probable biological mechanism, it can be concluded that the
male progenitor is a carrier of a pericentric inversion of chromosome 4 (Figure 21.6C
and 21.6D). Thus, from the conventional cytogenetic analysis of the fetus and the
progenitors, it was demonstrated that the fetus inherited a recombinant chromosome
4 resulting from the crossing over in the pericentric inversion loop of the paternal
inverted chromosome 4 during spermatogenesis.
FIGURE 21.6 A) Chromosome view of the 4pter duplication and 4qter deletion identi-
fed in a sample of amniotic fuid (Fetus 1) and B) of the 4pter deletion and 4qter duplica-
tion identifed in fetus 2 and 3. Results obtained using aCGH technique and analyzed with
Cytogenomics software. Duplication is represented in a positive scale and deletion in a nega-
tive scale. C) Pair of chromosomes 4 obtained from representative G-banded metaphases of
the amniotic cells from the fetus D) and of the peripheral blood from the father.
240 Cytogenetics and Molecular Cytogenetics
These two genomic imbalances identifed in the fetus seem to be in the origin of the
ultrasound malformations detected and consequently are correlated to the clinical
phenotype.
iii) What can we offer to this couple? What is the future strategy for the
family?
This couple was offered genetic counselling where it was well explained the conse-
quence of this pericentric inversion in this phenotypically normal father for future
pregnancies and the need of family studies (parents, brothers, sisters and other
family members at risk). To all carriers, invasive prenatal diagnosis should be
offered.
This couple had two more pregnancies. Trophoblastic cells from fetuses 2 and
3 were collected and aCGH revealed two genomic imbalances in mirror compared
with fetus 1: 4p16.3p15.33 deletion and 4q31.3q35.2 duplication.
Fetuses 2 and 3 had similar alterations but different from fetus 1 (Figure 21.6B)
that resulted from the meiotic recombination in the spermatogenesis of the male
progenitor.
Fetus 2 and 3: 46,XX,rec(4)dup(4q)inv(4)(p15.33q31.3)pat—arr[GRCh37]
4p16.3p15.33(72447_1,887210)x1,4q31.3q35.2(155125113_190976439)x3.
Although in fetus 2 and 3 the genomic imbalances in chromosome 4 are in oppo-
site way to that observed in fetus 1 (mirror, i.e., fetus 1 presented 4pter duplication
and fetus 2 and 3 deletion and fetus 1 presented 4qter duplication and fetus 2 and 3
a deletion), the mechanism of origin is the same: inheritance of a recombinant chro-
mosome 4 as a result of a paternal pericentric inversion in chromosome 4.
Deletions in the 4p16.3 region observed in the fetus are the main cause of Wolf-
Hirschhorn syndrome, which presents as clinical manifestations growth delay, sei-
zures, intellectual disability and distinct craniofacial features. In prenatal, 4pter
deletion/4qter duplication were already reported with clinical manifestations like
our patient.[39]
This case shows the clinical implications for the offspring of a balanced structural
rearrangement in one of the progenitors and the need to offer to this couple prenatal
diagnosis in future gestations and genetic counselling to explain the clinical implica-
tions of these genetic fndings, the risk of recurrence and the need to offer family
studies to other relatives.
CLINICAL CASE 4
Cells cultured from amniotic fuid of a fetus were analyzed by conventional cytoge-
netics, being the clinical indication for the invasive prenatal diagnosis a positive bio-
chemical screening. The G-banded metaphases of the amniotic cells revealed a male
fetus with two structural alterations: a paracentric inversion in chromosome 3 and a
reciprocal translocation between chromosomes 2 and 5 (Figure 21.7A). 46,XY,t(2;5)
(q33;q22)dn,inv(3)(p26.2p23).
i) How to interpret this result? How to classify this CNV? What is the most
probable mechanism of origin of the alteration?
Molecular Karyotyping 241
In order to ascertain the origin of these structural alterations, both parents were also
analyzed by conventional cytogenetics, and it was possible to infer that these altera-
tions of the fetus were de novo.
In order to further characterize the inversion, molecular cytogenetics (FISH tech-
nique) (Figure 21.7B) was also performed in the amniotic cells from the fetus using
a whole chromosome painting (WCP) probe for chromosome 3 (WCP3, Cambio)
labelled with SpectrumOrange that showed a continuum pattern in the chromosome
3; e.g. the inversion in the chromosome 3 did not change the pattern observed in this
or any chromosome with this probe. A subtelomeric probe for 3p26.3 (D3S4559,
TotelVysion, Vysis) labelled with SpectrumGreen was needed to confrm that this
terminal region in the short arm of the chromosome 3 (3p26.3) was not involved in
the inversion.
With these two techniques we can concluded that the fetus carries two structural
chromosomal alterations that are de novo and seem to be balanced. Being de novo,
the risk for the fetus of intellectual disability would be 6–10%.
In order to ascertain whether there were micro imbalances aCGH technique
was performed and revealed losses of genetic material in two of the chromosomes
involved (chromosomes 3 and 5) in the structural rearrangements observed by
conventional cytogenetics arr[GRCh37]3q22.2q22.3(135247918_136673227)×1
,5p13.2(33825339_35133937)×1 (Figure 21.7C):
DNA from the peripheral blood of the progenitors were also analyzed by aCGH and
did not reveal these imbalances (data not shown), which were therefore classifed as
de novo in the fetus.
iii) What can we offer to this couple? What is the future strategy for the
family?
Clinical Case 5
DNA from tumor tissue of a 47 years-old woman diagnosed with foor of the mouth
tumor, stage I, human papilloma virus (HPV) negative and with tobacco consump-
tion, was analyzed by aCGH. This whole genome approach revealed imbalances
in several chromosomes, such as, chromosomes 1, 2, 3, 7 and 9, where there are
mapped several genes with key roles for the carcinogenesis process (Figure 21.8).
We observed that tumor cells of this patient displayed numerous rearrangements on
several chromosomes simultaneously, while showing a small number of chromo-
somes without or with few aberrations. The interpretation of the large-scale genomic
profling of cancer samples has been challenging due to the intra- and inter-tumor
heterogeneity displayed by solid tumors. The aCGH technique has helped in the
identifcation and translation to clinical practice, of specifc diagnostic and prognos-
tic cancer biomarkers, which are of utmost importance in order to improve patient’s
treatment selection, prophylactic screening strategies and ultimately the patients
quality of life.
Clinical Case 6
A primary cell culture established from a surgical tongue tumor resected sample was
characterized by aCGH and banding cytogenetics. This primary culture presented
a huge karyotypic heterogeneity with different numerical and structural chromo-
somal abnormalities identifed in the analyzed metaphases (Figure 21.9A). Results
from aCGH helped to establish the specifc chromosomal bands involved in copy
number gains and losses, assisting in the delimitation of the size of the imbalances
and the establishment of specifc breakpoints as well as to identify additional small
copy number gains and losses not detected by conventional cytogenetics due to their
reduced size. With the integration of these two techniques, we can observe aneuploi-
dies and also to infer and characterize some structural rearrangements that are in the
origin of the CNVs and aneuploidy (Figure 21.9 B–C).
244
Cytogenetics and Molecular Cytogenetics
FIGURE 21.8 Chromosome view of chromosomes 1, 2, 3, 7 and 9 displaying several CNVs (gains and losses). The results were obtained through the
analysis of DNA from a foor of the mouth tumor using aCGH technique and Agilent Genomic Workbench v6.5 software. The results are according to
Human Genome build 19 (GRCh37) and include imbalances with at least three consecutive probes with abnormal log2 ratios. Deletion is represented in
a negative scale and amplifcation in a positive scale.
Molecular Karyotyping 245
CONCLUSIONS
Molecular karyotyping is a powerful tool to identify with high resolution chromo-
somal and gene alterations for diagnostic and prognostic purposes. It is important to
stress that aCGH cannot detect all genetic anomalies and that a normal aCGH result
does not reduce the risk for other genetic conditions or even, in prenatal context, for
birth defects, related to balanced rearrangements or single gene mutations, which are
the detection limitations of this technique. Therefore, the results of molecular karyo-
typing are frequently complementary to those obtained with conventional karyotyp-
ing and Next Generation Sequencing (NGS) mutation studies.
The implementation of aCGH technique in the post and prenatal feld has been a
signifcant impact on the diagnosis of human constitutional diseases, since allowed
to detect genomic copy number alterations undetectable by other cytogenetic and
molecular technologies. This technique allows not only identifying numerous
well-characterized genetic syndromes but also to fnd new genomic disorders and
disease-causing genes, improving the knowledge of human genetic disorders and
consequently the capability to diagnosis and treat the patients. The implementation
of this technique in the clinical practice can help therapeutic selection and novel
molecular targets identifcation, improving precision medicine in a wide range of
human genetic disorders and cancer.
Molecular Karyotyping 247
REFERENCES
1. Solinas-Toldo, S.; Lampel, S.; Stilgenbauer, S.; Nickolenko, J.; Benner, A.; Dohner, H.;
Cremer, T.; Lichter, P.; Matrix-based comparative genomic hybridization: Biochips to
screen for genomic imbalances. Genes Chromosomes Cancer. 1997, 20, 399–407.
2. Snijders, A.M.; Nowak, N.; Segraves, R.; Blackwood, S.; Brown, N.; Conroy, J.;
Hamilton, G.; Hindle, A.K.; Huey, B.; Kimura, K.; Law, S.; Myambo, K.; Palmer, J.;
Ylstra, B.; Yue, J.P.; Gray, J.W.; Jain, A.N.; Pinkel, D.; Albertson, D.G. Assembly of
microarrays for genome-wide measurement of DNA copy number. Nat. Genet. 2001, 29,
263–264.
3. Pinkel, D.; Segraves, R.; Sudar, D.; Clark, S.; Poole, I.; Kowbel, D.; Collins, C.; Kuo,
W.L.; Chen, C.; Zhai, Y.; Dairkee, S.H.; Ljung, B.M.; Gray, J.W.; Albertson, D.G. High
resolution analysis of DNA copy number variation using comparative genomic hybrid-
ization to microarrays. Nat. Genet. 1998, 20, 207–211.
4. Buckley, P.G.; Mantripragada, K.K.; Benetkiewicz, M.; Tapia-Paez, I.; Diaz De Stahl, T.;
Rosenquist, M.; Ali, H.; Jarbo, C.; De Bustos, C.; Hirvela, C.; Sinder Wilen, B.; Fransson,
I.; Thyr, C.; Johnsson, B.I.; Bruder, C.E.; Menzel, U.; Hergersberg, M.; Mandahl, N.;
Blennow, E.; Wedell, A.; Beare, D.M.; Collins, J.E.; Dunham, I.; Albertson, D.; Pinkel,
D.; Bastian, B.C.; Faruqi, A.F.; Lasken, R.S.; Ichimura, K.; Collins, V.P.; Dumanski, J.P.
A full-coverage, high-resolution human chromosome 22 genomic microarray for clinical
and research applications. Hum. Mol. Genet. 2002, 11, 3221–3229.
5. Vermeesch, J.R.; Melotte, C.; Froyen, G.; Van Vooren, S.; Dutta, B.; Maas, N.; Vermeulen,
S.; Menten, B.; Speleman, F.; De Moor, B.; Van Hummelen, P.; Marynen, P.; Fryns,
J.P.; Devriendt, K. Molecular karyotyping: Array CGH quality criteria for constitutional
genetic diagnosis. J. Histochem. Cytochem. 2005, 53, 413–422.
6. Lee, C.; Hyland, C.; Lee, A.S.; Hislop, S.; Ihm, C. Copy number variation and human
health. In: Genomic and Personalized Medicineed; Willard, P.D.H.F.; Ginsburg, G.S.,
Eds. Academic Press, New York, 2009, pp. 108–119.
7. Kallioniemi, A.; Kallioniemi, O.P.; Sudar, D.; Rutovitz, D.; Gray, J.W.; Waldman, F.;
Pinkel, D. Comparative genomic hybridization for molecular cytogenetic analysis of
solid tumors. Science. 1992, 258, 818–821.
8. du Manoir, S.; Speicher, M.R.; Joos, S.; Schröck, E.; Popp, S.; Döhner, H.; Kovacs, G.;
Robert-Nicoud, M.; Lichter, P.; Cremer, T. Detection of complete and partial chromo-
some gains and losses by comparative genomic in situ hybridization. Hum. Genet. 1993,
90, 590–610.
9. Lichter, P.; Joos, S.; Bentz, M.; Lampel, S. Comparative genomic hybridization: Uses and
limitations. Semin Hematol. 2000, 37, 348–357.
10. Gebhart, E.; Liehr, T. Patterns of genomic imbalances in human solid tumors (Review).
Int J Oncol 2000, 16, 383–399.
11. Spangenberg, V.; Kolomiets, O.; Stepanyan, I.; Galoyan, E.; de Bello Cioff, M.;
Martynova, E.; Martirosyan, I.; Grishaeva, T.; Danielyan, F.; Al-Rikabi, A.; Liehr,
T.; Arakelyan, M. Evolution of the parthenogenetic rock lizard hybrid karyotype:
Robertsonian translocation between two maternal chromosomes in Darevskia ros-
tombekowi. Chromosoma. 2020, 129, 275–283.
12. Deon, G.A.; Glugoski, L.; Vicari, M.R.; Nogaroto, V.; Sassi, F.M.C.; Cioff, M.B.;
Liehr, T.; Bertollo, L.A.C.; Moreira-Filho, O. Highly rearranged karyotypes and mul-
tiple sex chromosome systems in armored catfshes from the genus Harttia (Teleostei.;
Siluriformes). Genes. 2020, 11, 1366.
13. Szczaluba, K.; Demkow, U. Array comparative genomic hybridization and genomic
sequencing in the diagnostics of the causes of congenital anomalies. J. Appl. Genet.
2017, 58, 185–198.
248 Cytogenetics and Molecular Cytogenetics
30. Tavares, I.; Martins, R.; Ribeiro, I.P.; Esteves, L.; Caramelo, F.; Abrantes, A.M.; Neves,
R.; Caetano-Oliveira, R.; Botelho, M.F.; Barbosa de Melo, J.; Diogo, D.; Tralhao, J.G.;
Carreira, I.M. Development of a genomic predictive model for cholangiocarcinoma
using copy number alteration data. J. Clin. Patho. 2022, 75, 274–278.
31. Couto Oliveira, A.; Ribeiro, I.P.; Pires, L.M.; Goncalves, A.C.; Paiva, A.; Geraldes, C.;
Roque, A.; Sarmento-Ribeiro, A.B.; Barbosa de Melo, J.; Carreira, I.M. Genomic char-
acterisation of multiple myeloma: Study of a Portuguese cohort. J. Clin Pathol, 2022, 75,
422–425.
32. Pinto-Leite, R.; Carreira, I.; Melo, J.; Ferreira, S.I.; Ribeiro, I.; Ferreira, J.; Filipe, M.;
Bernardo, C.; Arantes-Rodrigues, R.; Oliveira, P.; Santos, L. Genomic characterization
of three urinary bladder cancer cell lines: Understanding genomic types of urinary blad-
der cancer. Tumour Biol. 2014, 35, 4599–4617.
33. Spasova, V.; Mladenov, B.; Rangelov, S.; Hammoudeh, Z.; Nesheva, D.; Serbezov, D.;
Staneva, R.; Hadjidekova, S.; Ganev, M.; Balabanski, L.; Vazharova, R.; Slavov, C.;
Toncheva, D.; Antonova, O. Clinical impact of copy number variation changes in blad-
der cancer samples. Exp. Ther. Med. 2021, 22, 901.
34. Capela de Matos, R.R.; Ney Garcia, D.R.; Othman, M.A.K.; Moura Ferreira, G.; Melo,
J.B.; Carreira, I.M.; Meyer, C.; Marschalek, R.; Costa, E.S.; Land, M.G.P.; Liehr, T.;
Ribeiro RC.; Silva, M.L.M. A new complex karyotype involving a KMT2A-r variant
three-way translocation in a rare clinical presentation of a pediatric patient with acute
myeloid leukemia. Cytogenet. Genome Res. 2019, 157, 213–219.
35. Ronaghy, A.; Yang, R.K.; Khoury, J.D.; Kanagal-Shamanna, R. Clinical applications of
chromosomal microarray testing in myeloid malignancies. Curr. Hematol. Malign. Rep.
2020, 15, 194–202.
36. Wolf, M.T.F. Nephronophthisis and related syndromes. Curr. Opin. Pediatr. 2015, 27,
201–211.
37. Ala-Mello, S.; Koskimies, O.; Rapola, J.; Kääriäinen, H. Nephronophthisis in Finland:
Epidemiology and comparison of genetically classifed subgroups. Europ. J. Hum.
Genet. 1999, 7, 205–211.
38. Lu, Y.; Liang, Y.; Ning, S.; Deng, G.; Xie, Y.; Song, J.; Zuo, N.; Feng, C.; Qin, Y. Rare
partial trisomy and tetrasomy of 15q11-q13 associated with developmental delay and
autism spectrum disorder. Mol. Cytogenet. 2020, 13, 21.
39. Bi, W.; Cheung, S.-W.; Breman, A.M.; Bacino, C.A. 4p16.3 microdeletions and microdu-
plications detected by chromosomal microarray analysis: New insights into mechanisms
and critical regions. Am. J. Med. Genet. A. 2016, 170, 2540–2550.
40. Wapner, R.J.; Martin, C.L.; Levy, B.; Ballif, B.C.; Eng, C.M.; Zachary, J.M.; Savage,
M.; Platt, L.D.; Saltzman, D.; Grobman, W.A.; Klugman, S.; Scholl, T.; Simpson, J.L.;
McCall, K.; Aggarwal, V.S.; Bunke, B.; Nahum, O.; Patel, A.; Lamb, A.N.; Thom, E.A.;
Beaudet, A.L.; Ledbetter, D.H.; Shaffer, L.G.; Jackson, L. Chromosomal microarray ver-
sus karyotyping for prenatal diagnosis. N. Engl. J. Med. 2012, 367, 2175–2184.
41. Gao, J.; Liu, C.; Yao, F.; Hao, N.; Zhou, J.; Zhou, Q.; Zhang, L.; Liu, X.; Bian, X.; Liu, J.
Array-based comparative genomic hybridization is more informative than conventional
karyotyping and fuorescence in situ hybridization in the analysis of frst-trimester spon-
taneous abortion. Mol. Cytogenet. 2012, 5, 33.
22 FISH—Mitochondrial
DNA
Tigran Harutyunyan
CONTENTS
Introduction............................................................................................................ 251
Samples/Tissues/Materials..................................................................................... 254
Human Whole Blood Culture ....................................................................... 254
Slides with Metaphase Chromosomes .......................................................... 254
FISH Probe for mtDNA................................................................................ 254
Pretreatment and Postfxation ....................................................................... 255
mtDNA-FISH and Washing .......................................................................... 255
Description of Methods.......................................................................................... 255
Human Whole Blood Cultivation ................................................................. 255
Preparation of Metaphase Spreads................................................................ 256
Homemade mtDNA-FISH Probes ................................................................ 256
Slide Pretreatment......................................................................................... 257
mtDNA-FISH Procedure .............................................................................. 257
Washing and Counterstaining of Slides ........................................................ 258
Yields ..................................................................................................................... 258
Analysis of mtDNA Insertions in Metaphase Chromosomes....................... 258
Conclusions............................................................................................................ 258
Acknowledgments.................................................................................................. 259
References.............................................................................................................. 259
INTRODUCTION
Mitochondria are the powerhouses of cells, generating over 90% of energy in the
form of adenosine triphosphate (ATP) required for the normal cell functioning.[1]
Moreover, mitochondria are involved in cell cycle regulation and intrinsic apopto-
sis. In addition, each mitochondrion contains up to several thousand copies of its
own DNA molecule. In human cells, the mitogenome consists of a circular molecule
called mitochondrial chromosome, or better mitochondrial DNA (mtDNA), that har-
bors 16,569 bp and 37 genes, including 13 proteins (for the OXPHOS system), 22
tRNAs, and 2 rRNAs. In addition, at least eight mitochondrial-derived peptides have
been identifed which are encoded by 12S and 16S rRNA genes.[2]
Recent data indicates horizontal transfer of mitochondria as a mechanism of inter-
cellular signaling in normal and pathological conditions.[3] In addition, numtogenesis,
a de novo insertion of mtDNA in the nuclear genome, was observed at high fre-
quency in cancer cell, which can also result in genetic diseases.[4] It was shown that
environmental mutagens can increase the frequency of de novo mtDNA insertions
in human chromosomes after exposure to DNA double-strand break (DSB) induc-
ing agents.[5] Also, Lutz-Bonengel et al.[6] and Wei et al.[7] demonstrated the pres-
ence of amplifed NUMTs (nuclear mtDNA sequences) or mega-NUMTs in nuclear
genome of healthy individuals, resembling paternal mtDNA. In addition, implica-
tion of mtDNA in the innate immunity and COVID-19 pathogenesis was recently
shown.[8] Therefore, availability of tools for identifcation of mitochondrial exchange
and integration of mtDNA in the nuclear genome has clinical, forensic, phylogenetic
and research importance.
Sequencing of eukaryotic genomes revealed the presence of NUMTs in nuclear
DNA (nDNA) which occurred by insertion of corresponding sequences during evo-
lution, with variant frequencies within different species.[9] In the human genome,
over 1,000 NUMTs have been identifed, with the sizes ranging from 39 to 16,106
bp and encompassing ~400 kb of DNA stretches. Interestingly, the number of these
insertions in chromosomes positively correlate with the relative chromosome length
indicating random insertion of mtDNA. However, there is also evidence that inser-
tion loci of NUMTs in (human) genome are non-random, since enrichment with
repetitive elements in 2 kb of both fanking regions were identifed.[9] In recent years,
it was also shown that NUMTs might cause bias in the interpretation of nDNA
sequencing data due to high homology with mtDNA sequences. In particular, bipa-
rental inheritance of mtDNA was suggested after sequencing and analysis for het-
eroplasmy in the offspring of three heteroplasmic fathers, which indicated for the
presence of maternally and paternally inherited mtDNA haplotypes.[10] However,
the authors did not completely rule out NUMT contamination of the sequencing
data. Recently, the results of Lutz-Bonengel et al.[6] have challenged this hypothesis
by demonstrating the presence of heteroplasmy in the offspring carrying paternal
U4c1 mitogenome haplogroup along with maternal V haplogroup. Sequencing stud-
ies revealed the presence of maternal V mitotype only in cells devoid of nucleus (e.g.
thrombocytes and hair shafts), while cells depleted from mtDNA had only U4c1
mitotype. Fluorescence in situ hybridization (FISH) analysis confrmed the inser-
tion of mtDNA in 14q31 chromosome loci, proving the presence of mega-NUMT in
nDNA.[6] Thus, NUMTs should be considered in phylogenetic, medical genetic, and
forensic studies to avoid false-positive results.
While the biological role of inherited NUMTs is not determined, the occurrence
of de novo NUMTs and mtDNA insertions in the nDNA, defned as numtogenesis,
can be observed in pathological conditions, too.[4] The analysis of whole-genome
sequencing data from over 2,600 cancers demonstrated elevation in frequencies of
somatically acquired mtDNA insertions being highest in HER2+ breast cancer cells
and squamous cell lung cancers.[11] Elevation of de novo NUMTs was also observed
in colorectal adenocarcinoma genomes, which contained up to 4.2-fold higher
NUMTs compared to blood-derived normal genomes.[12]
Furthermore, human diseases caused by de novo insertions of mtDNA in
nDNA are described, although in a limited number of cases. Such insertions
were identifed in single patients with severe plasma factor VII defciency,[13]
FISH—Mitochondrial DNA 253
SAMPLES/TISSUES/MATERIALS
FISH enables the detection of mtDNA on a single-cell level in any cells. Slides for
FISH analysis of numtomutagenesis can be prepared from both heparinized whole
blood or blood collected in EDTA containing vacutainers. Nevertheless, if meta-
phase chromosomes should be used in the analysis the heparinized blood is more
suitable in contrast to EDTA.
DESCRIPTION OF METHODS
Although mtDNA-FISH is applicable for both interphase nuclei and metaphase chro-
mosomes, the analysis of insertions of mtDNA in nDNA using metaphase chromo-
somes is recommended. This approach permits the identifcation of chromosomes
with mtDNA insertion(s) and insertion loci, enabling the analysis of the distribution of
mtDNA insertions in the genome. For the analysis of numtomutagenesis, human whole
blood should be exposed to the mutagen and cultivated under standard conditions.
SLIDE PRETREATMENT
1. Dehydrate slides in an ethanol series (70 %, 90 %, 100 %, 3 min each) and
air-dry at room temperature.
2. Add 0.5 ml of pepsin to 100 ml of pepsin pretreatment solution, and put
slides for 3–5 min in a Coplin jar at 37°C in the water bath.
3. Wash slides in the PBS for 5 min at room temperature.
4. Add 100 µl of postfx solution and cover with coverslips for 10 min at room
temperature.
5. Remove coverslips; repeat step 3 and step 1.
MTDNA-FISH PROCEDURE
1. Mix 1 µl of the probe with 7 µl dextran sulfate in the Eppendorf tube con-
taining COT DNA. The optimal concentration of the COT DNA should be
determined experimentally.
2. Prewarm the mixture at 45°C for 20 min.
258 Cytogenetics and Molecular Cytogenetics
3. Add 100 µl of denaturation buffer to each slide, cover with coverslip, and
incubate on a warming plate at 81°C for 3 min and in parallel begin the
denaturation of the probe at 75°C in PCR machine.
4. Remove the coverslip and put slides in a 70 % ethanol at −20°C for 3 min.
5. Dehydrate slides in ethanol series of 90 % and 100 % at room temperature
for 3 min each and air-dry.
6. Add 8 µl of the probe onto half of the slide, and cover with 24×24 coverslip
and seal with rubber cement.
7. Place the slides in the humid chamber and incubate at 37°C overnight.
YIELDS
ANALYSIS OF MTDNA INSERTIONS IN METAPHASE CHROMOSOMES
1. For the detection of mtDNA insertions in metaphase chromosomes by
FISH, at least 1,800 metaphase spreads are recommended to be analyzed.
2. Capture mtDNA signals using fuorescent microscope quipped with 100×
objective and the camera with high sensitivity.
3. Statistical analysis can be performed using t-test for the comparison of
mtDNA insertion frequencies between untreated and treated variants.
4. Construction of the map with mtDNA insertions in each chromosome
according to Harutyunyan et al.[5] is recommended for the description of
the distribution of mtDNA insertions in the genome.
5. For evaluation of the quantitative changes in mitochondrial network,
the application of free-to-use program ImageJ (developed at the NIH) is
recommended.
CONCLUSIONS
Functional mitochondria are important players of intracellular and intercellular sig-
naling. However, exchange of mitochondria between cells, mutations of mtDNA, or
translocation of mtDNA in the nuclear genome can have deleterious effects for the
organism. Moreover, DSB-inducing environmental agents are capable to increase
numtomutagenic events in the cells. Thus, methods for rapid detection of aberrant
FISH—Mitochondrial DNA 259
ACKNOWLEDGMENTS
This work was supported by the RA MESCS and BMBF (project #AG-01/20), and
RA MESCS (project #21AG-1F068).
REFERENCES
1. Yan, C.; Duanmu, X.; Zeng, L.; Liu, B.; Song, Z. Mitochondrial DNA: Distribution,
mutations, and elimination. Cells. 2019, 8(4), 379.
260 Cytogenetics and Molecular Cytogenetics
2. Merry, T.L.; Chan, A.; Woodhead, J.; Reynolds, J.C.; Kumagai, H.; Kim, S.J.; Lee, C.
Mitochondrial-derived peptides in energy metabolism. Am J Physiol Endocrinol Metab.
2020, 319(4), E659–E666.
3. Valenti, D.; Vacca, R.A.; Moro, L.; Atlante, A. Mitochondria can cross cell boundaries:
An overview of the biological relevance, pathophysiological implications and therapeutic
perspectives of intercellular mitochondrial transfer. Int J Mol Sci. 2021, 22(15), 8312.
4. Singh, K.K.; Choudhury, A.R.; Tiwari, H.K. Numtogenesis as a mechanism for develop-
ment of cancer. Semin Cancer Biol. 2017, 47, 101–109.
5. Harutyunyan, T.; Al-Rikabi, A.; Sargsyan, A.; Hovhannisyan, G.; Aroutiounian, R.;
Liehr, T. Doxorubicin-induced translocation of mtDNA into the nuclear genome of
human lymphocytes detected using a molecular-cytogenetic approach. Int J Mol Sci.
2020, 21(20), 7690.
6. Lutz-Bonengel, S.; Niederstätter, H.; Naue, J.; Koziel, R.; Yang, F.; Sänger, T.; Huber, G.;
Berger, C.; Pfugradt, R.; Strobl, C.; et al. Evidence for multi-copy Mega-NUMTs in the
human genome. Nucleic Acids Research. 2021, 49(3), 1517–1531.
7. Wei, W.; Pagnamenta, A.T.; Gleadall, N.; Sanchis-Juan, A.; Stephens, J.; Broxholme, J.;
Tuna, S.; Odhams, C.A.; Genomics England Research Consortium; NIHR BioResource;
et al. Nuclear-mitochondrial DNA segments resemble paternally inherited mitochon-
drial DNA in humans. Nat Commun. 2020, 11(1), 1740.
8. Singh, K.K.; Chaubey, G.; Chen, J.Y.; Suravajhala, P. Decoding SARS-CoV-2 hijack-
ing of host mitochondria in COVID-19 pathogenesis. Am J Physiol Cell Physiol. 2020,
319(2), C258–C267.
9. Calabrese, F.M.; Balacco, D.L.; Preste, R.; Diroma, M.A.; Forino, R.; Ventura, M.;
Attimonelli, M. NumtS colonization in mammalian genomes. Sci Rep. 2017, 7(1), 16357.
10. Luo, S.; Valencia, C.A.; Zhang, J.; Lee, N.C.; Slone, J.; Gui, B.; Wang, X.; Li, Z.; Dell,
S.; Brown, J.; et al. Biparental inheritance of mitochondrial DNA in humans. Proc Natl
Acad Sci U S A. 2018, 115(51), 13039–13044.
11. Yuan, Y.; Ju, Y.S.; Kim, Y.; Li, J.; Wang, Y.; Yoon, C.J.; Yang, Y.; Martincorena, I.;
Creighton, C.J.; Weinstein, J.N.; et al. Comprehensive molecular characterization of
mitochondrial genomes in human cancers. Nat Genet. 2020, 52(3), 342–352.
12. Srinivasainagendra, V.; Sandel, M.W.; Singh, B.; Sundaresan, A.; Mooga, V.P.; Bajpai,
P.; Tiwari, H. K.; Singh, K.K. Migration of mitochondrial DNA in the nuclear genome of
colorectal adenocarcinoma. Genome Med. 2017, 9(1), 31.
13. Borensztajn, K.; Chafa, O.; Alhenc-Gelas, M.; Salha, S.; Reghis, A.; Fischer, A.M.;
Tapon-Bretaudière, J. Characterization of two novel splice site mutations in human fac-
tor VII gene causing severe plasma factor VII defciency and bleeding diathesis. Br J
Haematol. 2002, 117(1), 168–171.
14. Ahmed, Z.M.; Smith, T.N.; Riazuddin, S.; Makishima, T.; Ghosh, M.; Bokhari, S.;
Menon, P.S.; Deshmukh, D.; Griffth, A.J.; Riazuddin, S.; et al. Nonsyndromic recessive
deafness DFNB18 and Usher syndrome type IC are allelic mutations of USHIC. Hum
Genet. 2002, 110(6), 527–531.
15. Turner, C.; Killoran, C.; Thomas, N.S.; Rosenberg, M.; Chuzhanova, N.A;, Johnston,
J.; Kemel, Y.; Cooper, D.N.; Biesecker, L.G. Human genetic disease caused by de novo
mitochondrial-nuclear DNA transfer. Hum Genet. 2003, 112(3), 303–309.
16. Goldin, E.; Stahl, S.; Cooney, A.M.; Kaneski, C.R.; Gupta, S.; Brady, R.O.; Ellis, J.R.;
Schiffmann, R. Transfer of a mitochondrial DNA fragment to MCOLN1 causes an
inherited case of mucolipidosis IV. Hum Mutat. 2004, 24(6), 460–465.
17. Millar, D.S.; Tysoe, C.; Lazarou, L.P.; Pilz, D.T.; Mohammed, S.; Anderson, K.;
Chuzhanova, N.; Cooper, D.N.; Butler, R. An isolated case of lissencephaly caused by
FISH—Mitochondrial DNA 261
inside nuclear DNA in rat tissues and increase with age. Mitochondrion. 2010, 10(5),
479–486.
34. Koo, D.H.; Singh, B.; Jiang, J.; Friebe, B.; Gill, B.S.; Chastain, P.D.; Manne, U.; Tiwari,
H.K.; Singh, K.K. Single molecule mtDNA fber FISH for analyzing numtogenesis. Anal
Biochem. 2018, 552, 45–49.
35. Ramos, A.; Santos, C.; Barbena, E.; Mateiu, L.; Alvarez, L.; Nogués, R.; Aluja, M.P.
Validated primer set that prevents nuclear DNA sequences of mitochondrial origin
co-amplifcation: A revision based on the New Human Genome Reference Sequence
(GRCh37). Electrophoresis. 2011, 32(6–7), 782–783.
23 FISH—in Birds
Rafael Kretschmer, Michelly da Silva dos Santos,
Ivanete de Oliveira Furo, Edivaldo Herculano
Correa de Oliveira and Marcelo de Bello Cioff
CONTENTS
Introduction............................................................................................................ 263
Material .................................................................................................................. 264
FISH Probe Generation and Labeling........................................................... 265
Slide Pretreatment......................................................................................... 265
FISH Procedure ............................................................................................ 266
Methods.................................................................................................................. 266
Slide Preparation: Dropping the Cell Suspension ........................................ 266
FISH Probes.................................................................................................. 267
Commercially Available Probes........................................................ 267
Homemade Probes ............................................................................ 267
Probes Obtained by Flow Sorting................................................................. 270
Probe Labeling.............................................................................................. 272
Nick Translation................................................................................ 272
DOP-PCR Labeling .......................................................................... 272
Precipitation of DNA Probe.......................................................................... 273
Fluorescence In Situ Hybridization (FISH).................................................. 273
FISH with rDNA and Telomeric Sequences ..................................... 274
Slide Pretreatment with Pepsin..................................................................... 274
Chromosome Painting................................................................................... 274
FISH with Microsatellite Sequences ............................................................ 275
Important Notes for Proceeding FISH.......................................................... 276
Acknowledgments.................................................................................................. 276
References.............................................................................................................. 276
INTRODUCTION
Birds are the most diverse biological group among tetrapods, with more than
10,900 existing species.[1] Molecular and morphological studies support the division
of living birds into three monophyletic groups: Palaeognathae, Galloanserae, and
Neoaves.[2] Due to this wide variation, birds have been used as model species in
numerous biological studies, focusing on ecology, behavior, genetics, and cytogenet-
ics, and others.[3–6]
Cytogenetics is a useful tool in bird cytotaxonomy, especially for groups with a great
chromosome variability. Most of the karyological data in birds are based on classical
cytogenetic techniques, involving conventional staining and chromosomal banding
methods, such as C-banding, (used to detect constitutive heterochromatin), G-banding
(to identify regions rich in adenine and thymine (AT), and cytosine and guanine (CG)),
and AgNOR silver staining method applied to reveal nucleolar organizer regions.[7–9]
Despite its signifcant contribution to the characterization of karyotypes and provision
of basic genomic information used in cytotaxonomy, banding resolution stands as a
limitation in the identifcation of chromosome homology and comparative studies.
In this sense, the emergence of the fuorescence in situ hybridization (FISH) technique
allowed an advance towards more accurate cytogenetic studies, enabling the identifca-
tion and localization of DNA sequences or specifc regions along the chromosomes.[10]
Therefore, FISH helped to overcome limitations of comparative chromosomal studies
based on banding methods, especially in birds, with typically high 2n and many micro-
chromosomes (i.e. punctiform elements not easily distinguishable from each other).[6]
Chromosome painting data are available for 117 species, which represent less than
1% of all known bird species.[11] So far, whole chromosome painting probes (WCP)
from only six different bird species have been used in avian comparative cytoge-
netics: chicken (Gallus gallus—GGA, Galliformes, 2n=78), stone-curlew (Burhinus
oedicnemus—BOE, Charadriiformes, 2n=42), white hawk (Leucopternis albicollis—
LAL, Falconiformes, 2n= 68), griffon vulture (Gyps fulvus—GFU, Falconiformes,
2n=66), eared dove (Zenaida auriculata—ZAU, Columbiformes, 2n=76), and monk
parakeet (Myiopsitta monachus—MMO, Psittaciformes, 2n=48).[12–17] Among them,
G. gallus probes stand out as the most used ones due to the economic importance
of chickens, its well-known genome, and by having a karyotype very similar to the
putative avian ancestral one.[6] Overall, comparative chromosome studies using WCP
in birds have detected numerous inter- and intrachromosomal rearrangements, even
in species with apparently conserved karyotypes.[6, 11, 18]
In addition to WCP, the development of probes using specifc DNA sequences for
FISH has allowed cytogenetic mapping of different repetitive sequences in distinct
birds species, such as telomeric sequences, 18S/28SrDNA, 5S rDNA, and microsat-
ellites, establishing chromosomal landmarks and providing information concerning
different aspects such as karyotype organization and evolution and, besides, allow-
ing insights on origin and differentiation of sex chromosomes.[19–26]
For all these reasons, FISH has become an indispensable tool for cytogenomic
investigations in birds, and in this chapter, we present the current FISH protocol used
in our laboratories. We also include details about the isolation and preparation of the
most commonly used probes in avian cytogenetics. In addition, we highlight some
steps related to the quality of avian chromosome preparations, which is a crucial
condition for successful hybridizations.
MATERIAL
Besides the standard cell biological and molecular cytogenetic equipment, including
standard reagents (e.g., colchicine, methanol, acid acetic, ethanol, among others),
specialized items required are listed next.
FISH—in Birds 265
SLIDE PRETREATMENT
• 1×PBS (phosphate-buffered saline—Cat. No.: L1825, Biochrom, Germany;
store at room temperature)
• Pepsin stock solution (Cat. No.: P-7012, Sigma, USA)
• Pepsin solution (0.005%): 99 µl H2O, 10µl HCl and 2.5µl Pepsin (20 mg/ml)
• RNase A (Cat. No.: R4642, Sigma, USA)
• RNase solution (10µg/ml): 1.5 µl RNase A (10 mg/ml) and 1.5 ml 2×SSC.
266 Cytogenetics and Molecular Cytogenetics
FISH PROCEDURE
• Avidin-FITC (Cat. No.: A2901, Sigma, USA)
• Anti-digoxigenin fuorescein (Cat. No.: 11207741910, Roche Diagnostics,
Switzerland)
• Anti-digoxigenin rhodamin (Cat. No.: 11207750910, Roche Diagnostics,
Switzerland)
• Denaturation buffer: 70% (v/v) deionized formamide (See Note 1), 20% (v/v)
fltered double distilled water, 10% (v/v) 20×SSC; make fresh as required
• Detection solution 1: anti-biotin-labeled probes using a layer of Cy3-a or
Cy5-avidin (1:1000)
• Detection solution 2: anti-digoxigenin-labeled probes using monoclonal
anti-digoxin antibody produced in mice (1:500)
• Detection solution 3: FITC-labeled probes were detected with a layer of
rabbit anti-FITC (1:200) followed by a layer of goatanti-rabbit-FITC anti-
bodies (1:100)
• Formamide solution: 2×SSC/50% deionized formamide, pH 7.0 (See
Note 1)
• Hybridization buffer: Dissolve 2g of dextran sodium sulfate in 10 ml of 50%
formamide/2×SSC/50 mM phosphate buffer for 1 hour at 70°C. Aliquot
and stock at −20°C
• 1×PBS(phosphate-buffered saline—Cat. No.: L1825, Biochrom, Germany;
store at room temperature).
• Phosphate buffer: prepare 0.5 M Na2HPO4 and 0.5 M NaH2PO4, mix these
two solutions (1:1) to get pH 7.0, and then aliquot and store at −20°C
• 20×SSC = saline sodium citrate (Cat. No.: 15557–036; Invitrogen, USA;
store at room temperature); set up 1× and 2× SSC before use
• Streptavidin-Cy5 (Cat. No.: 25800881, GE Healthcare Life Sciences, USA)
• Streptavidin-Cy3 (Cat. No.: S6402, Sigma, USA)
• Tween 20 = polyoxyethylene-sorbitan monolaurate (Cat. No.: 10670–1000,
Sigma, Germany, store at room temperature)
• Vectashield Mounting Medium with DAPI FR/10 ml (Cat. No.: H-1200,
Vector, USA)
• Washing buffer (4×SCCT): 4×SSC, 0.05% Tween 20; make fresh as
required.
METHODS
SLIDE PREPARATION: DROPPING THE CELL SUSPENSION
To obtain chromosome preparations from birds demands biological samples; hence,
it is important to notice that such procedures usually require previous approval by
an Animal Experimentation Ethics Committee and permission to sample the ani-
mals, either for in situ or ex situ studies in birds. Avian mitotic chromosomes can
be directly obtained from samples of different tissues, including kidney, liver, bulb
FISH—in Birds 267
1. Wash and thoroughly clean a glass slide, and prepare a fresh fxative solu-
tion (3:1 methanol: acetic acid).
2. Place a coupling jar with distillate water in a microwave to heat. Hold the
slide over boiling water to create a flm of condensation. Alternatively,
place the slide under a rack in a water bath at 55°C.
3. Pipette about 20 μl of the cell suspension on the slide.
4. Pipette immediately 20 μl fxative on the slide.
5. Dry the preparation directly on the air.
FISH PROBES
Commercially Available Probes
We have used two kinds of commercially available probes for FISH in bird species:
Homemade Probes
Several methods are available to obtain probes for FISH experiments. Usually, the
probes for FISH experiments in birds can be obtained through PCR amplifcation
(rDNA and telomeric) or isolated by fow cytometry followed by PCR amplifca-
tion (whole chromosome probes). Next, we list the methods for obtaining the most
used probes in our laboratories.
268 Cytogenetics and Molecular Cytogenetics
• rDNAs: The distribution of the 45S gene has been widely investigated in
different groups of birds, being a useful marker for evolutionary studies.[26]
These genes are highly conserved and organized in multigene families con-
sisting of many copies, in which the 45S rDNA encodes for 18S, 5.8S, and
28S rRNAs.[30] The 5S ribosomal gene, which encodes the 5S rRNA′,[30] has
been investigated only in a few species.
1. For sequence amplifcation, mix the reagents listed next in a 0.6 ml tube
(DNAse free).
2. Perform amplifcation in a thermocycler according to the program
described next.
3. Check 2 μl of amplifcation product on a 1% agarose gel with appro-
priate size markers. The product should yield a visible smear between
~1,400bp.
4. Store the amplifed DNA at −20°C.
• 18S-rDNA directed PCR amplifcation can be obtained using 18SF
(5′CCGAGGACCTCACTAAACCA-3′) and 18SR (5′-CCGCTTTGG
TGACTCTTGAT-3′) as primers to generate a fragment of approximately
1,400 base pairs. The primers were developed from the fsh Hoplias mala‑
baricus,[31] and due to the high sequence conservation, it has been applied
successfully in several birds species.[26] Here, we amplifed the 18S rDNA
gene using the Emu (Dromaius novaehollandiae) DNA as a template. It is
known that the Emu has this multigene family in a pair of microchromo-
somes,[32] and we confrmed this result (Figure 23.1 A). Reagents and details
for the reaction are shown in Table 23.1.
• 5S-rDNA directed PCR: The probes were obtained from the DNA
of the fsh Leporinus obtusidens by PCR using the primers 5SF
(5′-TACGCCCGATCTCGTCCGATC-3′) and 5SR (5′-CAGGCTGGTAT
GGCCGTAAGC-3′), according to.[33] These fragments of approximately
200 bp were obtained and successfully applied in FISH experiments in
FIGURE 23.1 18S (A, green) and 5S (B, red) rDNA distribution in the Emu (Dromaius
novaehollandiae).
FISH—in Birds 269
TABLE 23.1
a) How to Prepare 25 μl Reaction Master Mix (per Sample) for 18S-rDNA
Reagent Amount Final Concentration
Genomic DNA 25 ng -
Buffer 10× 2.5 µl 1×
MgCl2 (50 mM) 0.75 µl 1.5 mM
Primer reverse (10 mM) 0.5 µl 0.2 mM
Primer forward (10 mM) 0.5 µl 0.2 mM
dNTP Mix (10mM each) 3.1 µl 2.5mM each (dATP, dCTP, dGTP, dTTP)
Adjust with ddH2O 16.45 µl -
Taq polymerase 5U/µl 0.2 µl 1.0 U
birds.[34] Reagents and details for the reaction are shown in Tables 23.1 and
23.2. As an example, here we amplifed the 5S rDNA gene using the Emu
DNA as a template. This gene was found in a small macrochromosome,
probably the ninth pair in the Emu (Figure 23.1 B).
1. For the sequence amplifcation, mix the reagents listed next in a 0.6 ml
tube (DNAse free).
2. Perform amplifcation in a thermocycler according to the program
described next.
3. Check 2 μl of amplifcation product on a 1% agarose gel with appropriate
size markers. The product should yield a visible smear between ~200bp.
4. Store the amplifed DNA at −20°C.
• Telomere directed PCR amplifcation can be obtained using Primer: F
(5′ TTAGGG-3′)5 e R (5′ CCCTAA-3′)5.[35] In this case, the amplifcation
reaction can be placed with (Table 23.3) or without DNA as a template
(Table 23.4).
1. For the sequence amplifcation, mix the reagents listed next in a 0.6 ml
tube (DNAse free).
2. Perform amplifcation in a thermocycler according to the program
described next (Table 23.2).
3. Check 5 μl of amplifcation product on a 1% agarose gel with appropriate
size markers. The product size should be up to 25,000bp or 25Kb.
4. Store the amplifed DNA at −20°C.
270 Cytogenetics and Molecular Cytogenetics
TABLE 23.2
Thermocycler Program to Produce 5S-rDNA Probes Are Shown
Number of cycles Reaction Temperature, Time
1 Initial denaturation 94°C—5′
Denaturation 94°C—45′′
35 cycles Annealing 59°C—45′′
Extension 72°C—1′00′′
1 Final extension 72°C—10′
- Hold 4º∞
For reagents for this reaction, see Table 23.1.
TABLE 23.3
a) Reagents for the Telomere Directed PCR Amplifcation Reaction with
DNA (50 μl Reaction Master Mix per Sample)
Reagent Amount Final Concentration
Genomic DNA Variable 1–100 ng DNA
Buffer 10× 5.0 µl 1×
MgCl2 (50 mM) 2.0 µl 2.0 mM
Primer 18SR (10 mM) 1.5 µl 0.3 mM
Primer 18SF (10 mM) 1.5 µl 0.3 mM
2.5 mM each (dATP, dCTP, dGTP)
dNTP Mix 3.7 µl
1.25 mM (dTTP)
Adjust with ddH2O 33.9 µl -
Dye 1.0 µl -
Taq polymerase 5U/µl 0.4 µl 1.0 U
TABLE 23.4
a) Reagents for the Telomere Directed PCR Amplifcation Reaction without
DNA (50 μl Reaction Master Mix per Sample)
Reagent Amount Final Concentration
Buffer 10× 5.0 µl 1×
MgCl2 (50 mM) 2.5µl 2.5 mM
Primer 18SR (10 mM) 0.5 µl 0.1 mM
Primer 18SF (10 mM) 0.5 µl 0.1 mM
2.5mM each (dATP, dCTP, dGTP)
dNTP Mix 3.7 µl
1.25 mM (dTTP)
Adjust with ddH2O 36.8 µl -
Dye 0.6 µl -
Taq polymerase 5U/µl 0.4 µl 1.0 U
FIGURE 23.2 Examples of FISH experiments with fow sorted whole chromosome probes
derived from Gallus gallus (GGA) (A) and Leucopternis albicollis (LAL) (B) onto white-
tipped dove Leptotila verreauxi (LVE). The probes were labeled with biotin and detected
with Cy3-streptavidin. Both signals are on LVE 1.
272 Cytogenetics and Molecular Cytogenetics
PROBE LABELING
Probe labeling is an essential step for a successful FISH experiment and usually,
labeling methods such as Nick translation and DOP-PCR are effective. The Nick
translation procedure follows the manufacturer’s instructions and is appropriate
when the DNA sequence of interest was already isolated by fow sorting. In addi-
tion, DOP-PCR labeling is also applied after the fow sorting procedure to label
sequences of whole chromosomes.[36, 38] By these methods, probes can be directly or
indirectly labeled. In the frst case, nucleotides carrying a specifc fuorochrome are
incorporated into the probe-DNA. In the second case, nucleotides carrying haptens
(like biotin and digoxigenin) are incorporated, requiring a further detection step
with appropriate fuorochrome-conjugated antibodies to enable the visualization of
the hybridization sites.
Nick Translation
1. Add 1 µg of probe DNA to 4 µl biotin- or digoxigenin nick translation mix
and fll up to 20 µl with ddH2O; continue with step 3.
2. OR: add 1 µg of probe DNA to 4 µl of Nick translation mix and 4 µl of 5×
concentrated fuorophore labeling mixture* and fll up to 20 µl with ddH2O.
– *Fluorophore labeling mixture:
– 5 µl 2.5 mM dATP
– 5 µl 2.5 mM dCTP
– 5 µl 2.5 mM dGTP
– 3.4 µl 2.5 mM dTTP
– 4 µl of either 1mM of Cy5 dUTP/Spectrum-Green dUTP/Spectrum-
Orange dUTP/Texas Red-dUTP
– 27.6 µl ddH2O
3. Mix carefully using the tip of a 20 µl pipette, and incubate at 15°C for
90 min.
4. Stop the reaction by adding 2 µl of EDTA 0.5M (pH 8.0) and incubating at
65°C for 10 min.
DOP-PCR Labeling
DOP-PCR is particularly used for the amplifcation of chromosome painting
probes, which are usually generated by fow-sorted chromosomes. The amplifca-
tion can be obtained using the degenerate 6 MW primer sequence: CCGACTCGAG
NNNNNNATGTGG.
TABLE 23.5
a) Reagents for the DOP-PCR Amplifcation Reaction (25 μl Reaction Master
Mix per Sample)
Reagent Amount Final Concentration
Genomic DNA Variable 1–100 ng DNA
Buffer 10× 2.5 µl 1×
MgCl2 (50 mM) 1.25 µl 2.5 mM
6MW primer (20 mM) 2 µl 2 mM
dNTP Mix 2.5 µl 2.5mM each (dATP, dCTP, dGTP)
1.25 mM (dTTP)
Adjust with ddH2O 12.5 µl -
Dye 2,5 µl -
Taq polymerase 5U/µl 0.25 µl 1.25 U
1. Mix carefully and incubate at −80°C for 30 min or at −20°C for at least 2 h.
2. Centrifugate at 13,000 rpm for 20 min at −4°C, and remove the supernatant
with a micropipette without touching the pellet.
3. Dry the pellet in a vacuum centrifuge or let it at room temperature (RT)
until it dries.
4. Add 1 µl of ddH20, spin, and let at 37°C for 5 min to help dissolve.
5. Add 20 µl of the hybridization buffer, vortex it, and let it shake at 65°C
for 5 min. In the end, a fnal concentration of 50 ng/µl of labeled probe is
obtained.
with pepsin, to remove nuclear RNA and prevent nonspecifc hybridization between
probes and single-strand sequences.
1. Add 200 µl of the RNase A solution to slide and incubate at 37°C for 25 min
(1 µl of the RNase 10mg/ml solution in 500 µl of the 2×SSC).
2. Followed by treatment with pepsin (protocol described next).
3. Follow the steps for FISH described next.
1. Add 100 µl of a pepsin solution 0.01% on the slides, cover with a 22×50 mm
coverslip, and keep at 37°C for 5 min.
2. Remove the coverslip and incubate the slides in a coupling jar containing
2×SSC for 5 min at RT.
3. Repeat three times step 2.
4. Dehydrate the slides in ethanol series (70, 90%) for 2 min in each and 100%
for 4 min (RT).
5. Air-dry and incubate the slides at 60°C for 1 h or 37°C overnight.
CHROMOSOME PAINTING
1. Incubate the slides in a formamide solution at 72°C, 1 min and 20 seconds,
for DNA chromosomal denaturation.
2. Transfer the slides to a coupling jar flled with cold 70% ethanol (−20°C;
2 min) to conserve target DNA as single strands. Proceed slides into the
ethanol series (90 and 100%, RT, 2 min each) and air-dry.
3. Denature the hybridization mix (containing 20 µl of the hybridization buf-
fer and 100 ng of the labeled probe) in a thermocycler at 75°C for 10 min.
(this step can be done while ethanol series is performed).
4. Add 20 µl of the hybridization mix to the slides, cover with a 22×50 mm
coverslip, and incubate at 37°C for 72 h in a darkened moist chamber.
5. Remove the coverslip and wash the slides twice in 50% formamide and
twice in 2×SSC at a temperature ranging from 40 to 44°C, depending on
the applied probes and the phylogenetic distance between the species inves-
tigated (Note 3).
6. Add 100 µl of the appropriated detection solution (see Section 2.3), to each
slide, cover with a 22×50 mm coverslip and incubate at 37°C for 1h in a
darkened moist chamber. Detection steps 6 are necessary if the probes were
indirectly labeled. In the case of directly labeled probes, proceed to step 7.
FISH—in Birds 275
7. Remove the coverslip and wash 5 min for 3 times in 4×SSC/Tween = wash-
ing buffer (RT), under agitation (shaker).
8. Add 20 µl of Vectashield Mounting Medium with DAPI on each slide, cover
with a 22×50 mm coverslip, and press gently. The slides are ready to be
analyzed in a fuorescence microscope. For long-term storage, slides can be
kept at 4°C to prolong the fuorescence intensity.
ACKNOWLEDGMENTS
We would like to thank the Brazilian funding agencies FAPESP (Fundação de Amparo
à Pesquisa do Estado de São Paulo, Process 2020/11669–2) and CNPq (Conselho
Nacional de Desenvolvimento Científco e Tecnológico, Process 307382/2019–2 and
302449/2018–3) for the fnancial support.
REFERENCES
1. Gill, F.; Donsker, D.; Rasmussen, P. (Eds.). IOC world bird list; 2021. www.worldbird-
names.org/new/ [accessed on 08/31/2021].
2. Hackett, S.J.; Kimball, R.T.; Reddy, S.; Bowie, R.C.; Braun, E.L.; Braun, M.J.;
Chojnowski, J.L.; Cox, W.A.; Han, K.L.; Harshman, J.; Huddleston, C.J.; Marks,
B.D.; Miglia, K.J.; Moore, W.S.; Sheldon, F.H.; Steadman, D.W.; Witt, C.C.; Yuri, T.
A phylogenomic study of birds reveals their evolutionary history. Science 2008, 320,
1763–1768.
3. Isaksson, C.; Rodewald, A.D.; Gil, D. Editorial: Behavioural and ecological conse-
quences of urban life in birds. Front. Ecol. Evol. 2018, 6, 50.
4. Jarvis, E.D.; Mirarab, S.; Aberer, A.J.; Li, B.; Houde, P.; Li, C.; Ho, S.Y.; Faircloth, B.C.;
Nabholz, B.; Howard, J.T.; Suh, A.; Weber, C.C.; da Fonseca, R.R.; Li, J.; Zhang, F.; Li,
H.; Zhou, L.; Narula, N.; Liu, L.; Ganapathy, G.; Boussau, B.; Bayzid, M.S.; Zavidovych,
V.; Subramanian, S.; Gabaldón, T.; Capella-Gutiérrez, S.; Huerta-Cepas, J.; Rekepalli,
B.; Munch, K.; Schierup, M.; Lindow, B.; Warren, W.C.; Ray, D.; Green, R.E.; Bruford,
M.W.; Zhan, X.; Dixon, A.; Li, S.; Li, N.; Huang, Y.; Derryberry, E.P.; Bertelsen, M.F.;
Sheldon, F.H.; Brumfeld, R.T.; Mello, C.V.; Lovell, P.V.; Wirthlin, M.; Schneider, M.P.;
Prosdocimi, F.; Samaniego, J.A.; Vargas Velazquez, A.M.; Alfaro-Núñez, A.; Campos,
P.F.; Petersen, B.; Sicheritz-Ponten, T.; Pas, A.; Bailey, T.; Scofeld, P.; Bunce, M.;
Lambert, D.M.; Zhou, Q.; Perelman, P.; Driskell, A.C.; Shapiro, B.; Xiong, Z.; Zeng, Y.;
Liu, S.; Li, Z.; Liu, B.; Wu, K.; Xiao, J.; Yinqi, X.; Zheng, Q.; Zhang, Y.; Yang, H.; Wang,
J.; Smeds, L.; Rheindt, F.E.; Braun, M.; Fjeldsa, J.; Orlando, L.; Barker, F.K.; Jønsson,
K.A.; Johnson, W.; Koepfi, K.P.; O’Brien, S.; Haussler, D.; Ryder, O.A.; Rahbek, C.;
Willerslev, E.; Graves, G.R.; Glenn, T.C.; McCormack, J.; Burt, D.; Ellegren, H.; Alström,
P.; Edwards, S.V.; Stamatakis, A.; Mindell, D.P.; Cracraft, J.; Braun, E.L.; Warnow, T.;
Jun, W.; Gilbert, M.T.; Zhang, G. Whole-genome analyses resolve early branches in the
tree of life of modern birds. Science 2014, 346, 1320–1331.
5. Prum, R.O.; Berv, J.S.; Dornburg, A.; Field, D.J.; Townsend, J.P.; Lemmon, E.M.;
Lemmon, A.R. A comprehensive phylogeny of birds (Aves) using targeted next-genera-
tion DNA sequencing. Nature 2015, 526, 569–577.
6. Kretschmer, R.; Ferguson-Smith, M.A.; de Oliveira, E.H.C. Karyotype evolution in
birds: From conventional staining to chromosome painting. Genes 2018, 9, 181.
FISH—in Birds 277
7. Howell, W.M.; Black, D.A. Controlled silver-staining of nucleolus organizer regions with
a protective colloidal developer: A 1-step method. Experientia 1980, 36, 1014–1015.
8. Seabright, M. A rapid banding technique for human chromosomes. Lancet. 1971, 2,
971–972.
9. Sumner, A.T. A simple technique for demonstrating centromeric heterochromatin. Exp.
Cell Res. 1972, 75, 304–306.
10. Pinkel, D.; Straume, T.; Gray, J.W. Cytogenetic analysis using quantitative, high-sensitiv-
ity, fuorescence hybridization. Proc. Natl. Acad. Sci. U. S. A. 1986, 83, 2934–2938.
11. Degrandi, T.M.; Barcellos, A.S.; Costa, A.L.; Garnero, A.D.V.; Hass, I.; Gunski, R.J.
Introducing the bird chromosome database: An overview of cytogenetic studies in birds.
Cytogenet. Genome Res. 2020, 160, 199–205.
12. Griffn, D.K.; Haberman, F.; Masabanda, J.; O’Brien, P.; Bagga, M.; Sazanov, A.;
Smith, J.; Burt, D.W.; Ferguson-Smith, M.; Wienberg, J. Micro- and macrochromosome
paints generated by fow cytometry and microdissection: Tools for mapping the chicken
genome. Cytogenet. Cell Genet. 1999, 87, 278–281.
13. Nie, W.; O’Brien, P.C.M.; Ng, B.L.; Fu, B.; Volobouev, V.; Carter, N.P.; Ferguson-Smith,
M.A.; Yang, F. Avian comparative genomics: 112 reciprocal chromosome painting
between domestic chicken (Gallus gallus) and the stone curlew (Burhinus oedicnemus,
Charadriiformes): An atypical species with low diploid number. Chromosome Res. 2009,
17, 99–113.
14. de Oliveira, E.H.C.; Tagliarini, M.M.; Rissino, J.D.; Rissino, J.D.; Pieczarka, J.C.;
Nagamachi, C.Y.; O’Brien, P.C.; Ferguson-Smith, M.A. Reciprocal chromosome painting
between white hawk (Leucopternis albicollis) and chicken reveals extensive fusions and
fssions during karyotype evolution of accipitridae (Aves, Falconiformes). Chromosome
Research. 2010, 18, 349–355.
15. Nie, W.; O’Brien, P.C.M.; Fu, B.; Wang, J.; Su, W.; He, K.; Bed’Hom, B.; Volobouev, V.;
Ferguson-Smith, M.A.; Dobigny, G.; Yang, F. Multidirectional chromosome painting
substantiates the occurrence of extensive genomic reshuffing within Accipitriformes.
BMC Evol. Biol. 2015, 15, 205.
16. Kretschmer, R.; Furo, I.O.; Gunski, R.J.; Del Valle Garnero, A.; Pereira, J.C.; O’Brien,
P.C.M.; Ferguson-Smith, M.A.; de Oliveira, E.H.C.; de Freitas, T.R.O. Comparative
chromosome painting in Columbidae (Columbiformes) reinforces divergence in Passerea
and Columbea. Chromosome Res. 2018, 26, 211–223.
17. Furo, I.O.; Kretschmer, R.; O’Brien, P.C.; Pereira, J.C.; Garnero, A.D.V.; Gunski, R.J.;
O’Connor, R.E.; Griffn, D.K.; Gomes, A.J.B.; Ferguson-Smith, M.A.; de Oliveira,
E.H.C. Chromosomal evolution in the phylogenetic context: A remarkable karyotype
reorganization in neotropical parrot myiopsitta monachus (Psittacidae). Front. Genet.
2020, 11, 721.
18. Griffn, D.K.; Robertson, L.B.; Tempest, H.G.; Skinner, B.M. The evolution of the avian
genome as revealed by comparative molecular cytogenetics. Cytogenet. Genome Res.
2007, 117, 64, 77.
19. dos Santos, M.S.; Kretschmer, R.; Silva, F.A.O.; Silva, F.A.; Ledesma, M.A.; O’Brien,
P.C.; Ferguson-Smith, M.A.; Del Valle Garnero, A.; de Oliveira, E.H.; Gunski, R.J.
Intrachromosomal rearrangements in two representatives of the genus Saltator
(Thraupidae, Passeriformes) and a case of polymorphism in Z chromosome. Genetica
2015, 143, 535–543.
20. dos Santos, M.S.; Kretschmer, R.; Frankl-Vilches, C.; Bakker, A.; Gahr, M.; O’Brien,
P.C.; Ferguson-Smith, M.A.; de Oliveira, E.H. Comparative cytogenetics between
two important sonbird, models: The zebra fnch and the canary. PLoS One 2017, 12,
e0170997.
278 Cytogenetics and Molecular Cytogenetics
21. dos Santos, M.S.; Furo, I.O.; Tagliarini, M.M.; Kretschmer, R.; O’Brien, P.C.M.;
Ferguson-Smith, M.A.; de Oliveira, E.H.C. The karyotype of the hoatzin (Opisthocomus
hoazin): A phylogenetic enigma of the neornithes. Cytogenet. Genome Res. 2018, 156,
158–164.
22. de Oliveira, T.D.; Kretschmer, R.; Bertocchi, N.A.; Degrandi, T.M.; de Oliveira, E.H.;
Cioff, M.B.; Garnero, A.D.; Gunski RJ. Genomic organization of repetitive DNA in
woodpeckers (Aves, Piciformes): Implications for karyotype and ZW sex chromosome
differentiation. PLoS One 2017, 12, e0169987.
23. Kretschmer, R.; de Oliveira, T.D.; Furo, I.O.; Oliveira Silva, F.A.; Gunski, R.J.; Del
Valle Garnero, A.; de Bello Cioff, M.; de Oliveira, E.H.C.; de Freitas, T.R.O. Repetitive
DNAs and shrink genomes: A chromosomal analysis in nine Columbidae species (Aves,
Columbiformes). Genet. Mol. Biol. 2018, 41, 1, 98–106.
24. Furo, I.O.; Kretschmer, R.; dos Santos, M.S.; de Lima Carvalho, C.A.; Gunski, R.J.;
O’Brien, P.C.M.; Ferguson-Smith, M.A.; Cioff, M.B.; de Oliveira, E.H.C. Chromosomal
mapping of repetitive DNAs in Myiopsitta monachus and Amazona aestiva
(Psittaciformes, Psittacidae: Psittaciformes), with emphasis on the sex chromosomes.
Cytogenet. Genome Res. 2017, 151, 151–160.
25. Barcellos, S.A.; Kretschmer, R.; Souza, M.S.; Costa, A.L.; Degrandi, T.M.; Dos Santos,
M.S.; de Oliveira, E.H.C.; Cioff, M.B.; Gunski, R.J.; Garnero, A.D.V. Karyotype evo-
lution and distinct evolutionary history of the W chromosomes in swallows (Aves,
Passeriformes). Cytogenet. Genome Res. 2019, 158 (2), 98–105.
26. Degrandi, T.M.; Gunski, R.J.; Garnero, A.D.V.; Oliveira, E.H.C.; Kretschmer, R.; Souza,
M.S.; Barcellos, S.A.; Hass, I. The distribution of 45S rDNA sites in bird chromosomes
suggests multiple evolutionary histories. Genet. Mol. Biol., 2020, 43, 2, e20180331.
27. Garnero, A.V.; Gunski, R.J. Comparative analysis of the karyotype of Nothura maculosa
and Rynchotus rufescens (Aves: Tinamidae). A case of chromosomal polymorphism.
The Nucleus, 2000, 43, 64–70.
28. Barcellos, S.A.; de Souza, M.S.; Tura, V.; Pereira, L.R.; Kretschmer, R.; Gunski, R.J.;
Del Valle Garnero, A. Direct chromosome preparation method in Avian embryos for
cytogenetic studies: Quick, easy and cheap. DNA 2022, 2, 22–29.
29. Moorhead, P.S.; Nowell, P.C.; Mellman, W.J.; Battips, D.M.; Hungerford, D.A.
Chromosome preparations of leukocytes cultured from human peripheral blood. Exp
Cell Res. 1960, 20, 613–616.
30. López-Flores, I.; Garrido-Ramos, M.A. The repetitive DNA content of eukaryotic
genomes. Genome Dyn, 2012, 7, 1–28.
31. Cioff, M.B.; Martins, C.; Centofante, L.; Jacobina, U.; Bertollo, L.A. Chromosomal vari-
ability among allopatric populations of Erythrinidae fsh Hoplias malabaricus: Mapping
of three classes of repetitive DNAs. Cytogenet. Genome Res. 2009, 125, 132–141.
32. Nishida-Umehara, C.; Tsuda, Y.; Ishijima, J.; Ishijima, J.; Ando, J.; Fujiwara, A.; Matsuda,
Y.; Griffn, D.K. The molecular basis of chromosome orthologies and sex chromosomal
differentiation in palaeognathous birds. Chromosome Res. 2007, 15, 721–734.
33. Martins, C.; Galleti, P.M. Chromosomal localization of 5S rDNA genes in Leporinus fsh
(Anostomidae, Characiformes). Chromosome Res. 1999, 7, 363–367.
34. Gunski, R.J.; Kretschmer, R.; de Souza, M.S.; de Oliveira Furo, I.; Barcellos, S.A.; Costa,
A.L.; Cioff, M.B.; de Oliveira, E.H.C.; Del Valle Garnero, A. Evolution of bird sex
chromosomes narrated by repetitive sequences: Unusual W chromosome enlargement in
Gallinula melanops (Aves: Gruiformes: Rallidae). Cytogenet. Genome Res. 2019, 158,
152–159.
35. Ijdo, J.W.; Wells, R.A.; Baldini, A.; Reeders, S.T. Improved telomere detection using a
telomere repeat probe (TTAGGG)n generated by PCR. Nucleic Acid Res. 1991, 19, 4780.
FISH—in Birds 279
36. Yang, F.; Carter, N.P.; Shi, L.; Ferguson-Smith, M.A. A comparative study of karyotypes
of muntjacs by chromosome painting. Chromosoma 1995, 103, 642–652.
37. Yang, F.; O’Brien, P.C.M.; Milne, B.S.; Graphodatsky, A.S.; Solanky, N.; Trifonov, V.;
Rens, W.; Sargan, D.; Ferguson-Smith, M.A. A complete comparative chromosome map
for the dog, red fox and human and its integration with canine genetic maps. Genomic.
1999, 62, 189–202.
38. Telenius, H.; Ponder, B.A.J.; Tunnacliffe, A.; Carter, N.P.; Behmel, A.; Ferguson-Smith,
M.A.; Nordenskjöld, M.; Pfragner, R.; Ponder, B.A. Cytogenetic analysis by chromosome
painting using DOP-PCR amplifed fow-sorted chromosomes. Genes Chromosomes
Cancer 1992, 4, 257–263.
24 FISH—in Fish
Chromosomes
Francisco de M. C. Sassi, Gustavo A. Toma
and Marcelo de Bello Cioff
CONTENTS
Introduction............................................................................................................ 282
Material .................................................................................................................. 282
FISH Probe Labeling and Precipitation........................................................ 283
Via Polymerase Chain Reaction (PCR) ............................................ 283
Via Nick-Translation Kit................................................................... 283
Precipitation...................................................................................... 283
Slide Pre-FISH Treatment............................................................................. 283
FISH Workfow ............................................................................................. 284
If an Indirectly Labeled Probe Is Used ............................................. 284
Methods.................................................................................................................. 285
Samples......................................................................................................... 285
Cell Suspension and Slides Preparation ....................................................... 285
Probes ........................................................................................................... 285
Ribosomal DNA ........................................................................................... 286
Genomics ...................................................................................................... 286
Transposable Elements (TEs) ....................................................................... 287
Small Nuclear RNAs (snRNAs) ................................................................... 287
Microdissection............................................................................................. 287
Comparative Genomic Hybridization (CGH)............................................... 287
Probe Labeling.............................................................................................. 288
Multicolor FISH................................................................................ 288
Precipitation and Hybridization Mixture ...................................................... 288
FISH Probes...................................................................................... 288
CGH and WCP Probes...................................................................... 288
Fish-FISH Protocol....................................................................................... 289
Variations of FISH Protocol for WCP and CGH .............................. 292
Sequential FISH................................................................................ 293
Image Capture and Treatment....................................................................... 293
Acknowledgments.................................................................................................. 293
References.............................................................................................................. 293
INTRODUCTION
Representing more than half of the vertebrate species richness, fishes are a
very diverse group of organisms, with approximately 36,000 valid species.[1, 2, 3]
They include distinct groups such as bony (ray-finned fish, lungfish, coel-
acanth), cartilaginous (sharks, rays), and jawless fishes (hagfishes, lampreys).[3]
Their worldwide distribution, basal phylogenetic position among vertebrates,
and niche diversity make them valuable experimental models in many scien-
tific fields, such as ecological, evolutionary, and genetics.[3] Furthermore, with
the increased number of newly described sex chromosome systems (with the
coexistence of both ZZ/ZW- and XX/XY-derived systems), characterization of
inter- and intra-specific chromosomal rearrangements, and the implementation
of novel genomic research tools, fish cytogenetics has steadily grown over the
years. Many investigations on chromosomes have helped to solve taxonomic
issues, elucidate meiotic behaviors, and even provided information regarding
the evolution of repetitive DNA sequences, such as satellite DNAs and transpo-
sons.[4–6] In all these cases, chromosomal mapping of DNA by fluorescence in
situ hybridization (FISH) has been an indispensable tool, allowing a more refined
look into the genomic content of chromosomes, and providing a way to search
for DNA sequences that may have a relevant role in chromosomal differentia-
tion (e.g., the origin of B and sex chromosomes, identification of chromosomal
rearrangements, etc.).
Despite that frst applications of FISH occurred in end 1980s,[7] FISH in fshes
was frst applied only around 1993 using primarily rRNA genes and other tan-
dem repeats as probes.[8–11] These protocols were mostly similar to those used
in human molecular cytogenetics,[7, 12–14] with some differences in post-hybrid-
ization washes (especially in concentration of solutions and time of washes) and
other minor adaptations. However, with the improvement of hybridization probes
and rising numbers of FISH-derived techniques, such as genome in situ hybrid-
ization (GISH), comparative genomic hybridization (CGH), and whole chromo-
some painting (WCP), a new optimized FISH in fsh protocol was designed.[15]
This, instead, greatly differ from those previously used, as many changes were
proposed to have a more meticulous hybridization procedure, which could be
used in any fsh group.
Here we present an updated fsh-FISH protocol, in which we show a fgure-guided
protocol for the whole procedure and discuss some minor adaptations made over
the years, such as adjustments for a sequential FISH and problems that have usually
emerged using an overabundance of labeled probes.
MATERIAL
As usual, the standard cytogenetics equipment and reagents (centrifuge, pipettes,
colchicine, methanol, etc.) are obligatory, and specialized items are listed next,
including their working solutions.
FISH—in Fish Chromosomes 283
FISH WORKFLOW
• 20×SSC (Saline Sodium Citrate) stock solution, store at 4oC. For 1×SSC
and 2×SSC dilutions, store at RT (Cat. No.: 15557–036; Invitrogen, USA)
or prepare: 1l solution using 175.3g of sodium chloride (NaCl) and 88.23g
of trisodium citrate dihydrate (Na3C6H507.2H2O). Dissolve in 1l of distilled
water.
• Deionized formamide (Cat. No.: 1 09684 2500, Merck, Germany; aliquot
and store at 4oC).
• Denaturation buffer: 70% (v/v) deionized formamide, 20% (v/v) fltered
double distilled water, 10% (v/v) 20×SSC; make fresh as required.
• Dextran sodium sulfate (Cat. No.: D8906–10G, Sigma, USA).
• Phosphate buffer: prepare 0.5 M Na2HPO4 and 0.5 M NaH2PO4, mix these
two solutions (1:1) to get pH 7.0, and then aliquot and store at −20°C.
• Hybridization buffer: Dissolve 2 g dextran sodium sulfate in 10 ml 50%
deionized formamide/2×SSC/50 mM phosphate buffer for 3 h at 70°C; pH
adjusted to 7 with phosphate buffer; hydrochloric acid destabilizes buffer
solution. Aliquot and store at −20°C.
• Tween 20 = polyoxyethylene-sorbitan monolaurate (Cat. No.: 10670–1000,
Sigma, Germany, store at RT).
• Washing buffer (4×SCCT): 4×SSC, 0.05% Tween 20; make fresh as
required.
• Vectashield Mounting Medium with DAPI FR/10ml (Cat. No.: H-1200,
Vector, USA).
METHODS
Since fsh-FISH methods were recently revisited, here we provide a review and an
update on the technique. For the complete PCR cycles, conditions of amplifcation,
and probe labeling.[15]
SAMPLES
Mitotic chromosomes can be obtained by the classical air-drying method,[16, revisited in 17]
which consists of the application of colchicine to stop chromosomes in metaphase,
followed by hypotonization of cells and fxation in Carnoy 2 (methanol 3:1 glacial
acetic acid). The kidney is the preferred organ to obtain metaphase chromosomes,
due to its hematopoietic function in adult teleosts,[18] but good preparations were
already obtained from other organs such as the spleen.[19] Cell culture could also be
an alternative for mitotic chromosome obtainment, using a tissue fragment that usu-
ally is muscle or fn.[20] To obtain meiotic chromosomes of fshes, the most common
protocol follows the squash of the testes to liberate cells and proceed to hypotoniza-
tion in KCl 0.075 M, followed by fxation in Carnoy 2.[21]
PROBES
The most important step for the success of a FISH procedure is the establishment of
a probe of good quality. Such probe can be obtained by different methods, such as
target DNA amplifcation via PCR, microdissection, and whole-genome extraction.
Besides, commercially available probes (as microsatellite and telomeric sequences)
286 Cytogenetics and Molecular Cytogenetics
FIGURE 24.1 Examples of a good (a) and a bad (b) metaphase plate to analyze with FISH.
a) Mitotic chromosomes of Chalceus erythrurus (Teleostei, Characiformes) hybridized with
ribosomal DNA probes (a) 5S rDNA in green, and 18S rDNA in red.
b) Lebiasina bimaculata (Teleostei, Characiformes) chromosomes hybridized with GC
microsatellite.
Observe that in a) chromosomes are separated and well defned, while in b) they present some
overlapped regions and nuclei around (arrowheads), which may interfere with the hybridiza-
tion and the detection of hybridized regions.
can also be produced. We list next the methods for obtaining the most common
probes used in fsh cytogenetics.
RIBOSOMAL DNA
The chromosomal localization of ribosomal genes is an important tool for the com-
prehension of the evolutionary process on fshes.[22] These multigene families are
highly conserved and give origin to the ribosomal RNA, that actively participate in
protein synthesis. They are expressed by two multigene families: the 45S rDNA and
5S rDNA. In the 45S rDNA are codifed the 5.8S, the 18S, and the 28S, separated by
two internal transcribed spacers (ITS1 and ITS2), two external transcribed spacers
(ETS1 and ETS2), and a non-transcribed spacer (NTS). On other hand, 5S rDNA
is organized into a series of repetitions, separated by an NTS, also being found as
pseudogenes dispersed on chromosomes.[23] In fsh cytogenetics, the most common
probes of rDNA are 5S and 18S. For the obtainment of such probes, we use previ-
ously designed primers (18S from [24] and 5S from[25]) to isolate the sequences from
a targeted genome, which usually is from Hoplias malabaricus (Characiformes,
Erythrinidae), due to the previous existence of well-isolated libraries.
GENOMICS
More recently, genomics has become an important mechanism for probes obtain-
ment in fsh cytogenetics. Briefy, sequences from next-generation sequencing
FISH—in Fish Chromosomes 287
(NGS) are obtained, and bioinformatics pipelines are applied to isolate specifc
libraries, where primers can be designed for the targeted sequences. Then, these
primers are used in a PCR reaction to amplify the target sequence and thus be
used as a FISH probe after labeling. Satellite DNAs and TE probes are the most
common class of DNA used,[e.g. 26] but studies are still scarce when compared to the
high diversity of fshes, and consequently of the possibilities of this technique for
more DNA classes.
MICRODISSECTION
For the whole-chromosome painting (WCP) procedure, single chromosomes (or spe-
cifc regions of them) can be directly isolated by microdissection, which involves the
use of a glass needle to scuff up out the chromosome from the slide, followed by its
amplifcation via PCR and its label with a fuorescent dUTP. The complete procedure
is reviewed in [38].
PROBE LABELING
The probe needs to be hapten- or fuorescent-labeled for FISH. For this purpose, the
isolated DNA can be directly labeled with a fuorescent molecule (e.g., Cy5, Atto
550, Atto 488) or be linked with haptens (e.g., biotin and digoxigenin), thus constitut-
ing indirect labeling. As previously described, two main techniques can be used for
this: a standard PCR reaction (using designed or degenerated primers) or via Nick
Translation (NT). While for the frst the labeled nucleotides are incorporated during
the amplicon syntheses, the second method is based on the incorporation of fuores-
cent/haptens-dNTPs in small aleatory gaps generated by a DNAse. When labeling
by NT, it’s very important to control the fnal size of fragments, which should vary
between 200 and 600 base pairs (bp). For this purpose, several commercially NT
reactions kits can be widely found.
Multicolor FISH
To hybridize different probes in a single experiment, these should be labeled with
distinct fuorophores. Usually, the combination of red (Atto550, Rhodamine, Cy3,
Spectrum Orange, etc.) and green (Atto488, FITC, Spectrum green, among others)
probes produces good and distinguishable patterns, since DAPI (blue) is the most
common counterstain used.
FISH-FISH PROTOCOL
Day 1 (Figure 24.2):
15. Heat a Coplin jar with 1×SSC to 65°C, remove the coverslip and wash the
slides for 5 min on a shaker;
16. Transfer the slides to the washing buffer (4×SSC/Tween solution) and
wash for 5 min in on shaker at RT;
17. Wash the slides in 1×PBS for 1 min at RT using a shaker;
18. Quickly submerge the glass slide in distilled water;
19. Dehydrate the slides in ethanol 70%, 85%, and 100% for 2 min each, at
RT. Completely air-dry the slides;
20. Add 20 µl of DAPI + antifade onto slides, cover with a 24×50 mm cover-
slip and stabilize the fuorophores by leaving the slides at 4°C for 30 min
before the analysis in a fuorescence microscope.
290 Cytogenetics and Molecular Cytogenetics
FIGURE 24.2 Illustrated fsh-FISH protocol of the frst day of the procedure. The steps
correspond to the ones described in the fsh-FISH protocol (Day 1).
FISH—in Fish Chromosomes 291
FIGURE 24.3 The second day of the illustrated fsh-FISH protocol, with variations accord-
ing to the label method used (a: directly labeled; b: indirectly labeled). The steps correspond
to the ones described in the fsh-FISH protocol (Day 2).
292 Cytogenetics and Molecular Cytogenetics
15. Heat a Coplin jar with 2×SSC at 42°C, remove the coverslip and wash the
slides for 5 min in a shaker;
16. Wash the slides in 1×SSC at RT for 5 min on a shaker;
17. Incubate the slides in 3% NFDM/4×SSC for 5 min at RT;
18. Apply 50 µl of the detection solution (will vary according to the combina-
tion of fuorophores used), cover with 24×50mm coverslips, and incubate
at 37 °C in a humid dark chamber for 1h;
19. Remove the coverslips, transfer the slides to the washing buffer (4×SSC/
Tween solution), and wash for 5 min on a shaker at RT;
20. Repeat the wash as done in step 19 twice;
21. Wash the slides in 1×PBS for 1 min at RT with a shaker;
22. Quickly submerge the glass slide in distilled water;
23. Dehydrate the slides in ethanol 70%, 85%, and 100% for 2 min each, at
RT. Completely air-dry the slides;
24. Add 20 µl of DAPI + antifade onto slides, cover with a 24×50mm cover-
slip and stabilize the fuorophores by leaving the slides at 4°C for 30 min
before the analysis in a fuorescence microscope.
13. Denature the probes in a thermal cycler at 85°C for 10 min, followed by a
pre-annealing at 37°C for 30 min, followed by cooling at 4°C.
14. Add 20 µl of the hybridization mixture (probes + hybridization buffer) to
the slides. Cover with a 24 × 50mm coverslip, seal the borders of the cov-
erslip with rubber cement and incubate at 37°C overnight in a dark humid
chamber.
15. Heat two Coplin jars with 1×SSC at 65°C, remove the coverslip and wash
the slides for 5 min on a shaker;
16. Repeat step 15 to washing the slides again;
17. Transfer the slides to the washing buffer (4×SSC/Tween solution) and
wash for 5 min on a shaker at RT;
18. Wash the slides in 1×PBS for 1 min at RT with a shaker;
19. Finish as described before
FISH—in Fish Chromosomes 293
Sequential FISH
Depending on the degree of chromosomal damage during the FISH technique, a
second round of hybridization can be realized. For this, the previous probe-chromo-
some assembly must be disassembled as described next:
1. Remove the coverslip and wash the slides in 1×SSC (65°C) for 5 min on a shaker;
2. Repeat step 1;
3. Transfer the slides to the washing buffer (4×SSC/Tween solution) and wash
for 5 min an a shaker at RT;
4. Repeat step 3;
5. Wash the slides in 1×PBS for 1 min at RT with a shaker;
6. Quickly submerge the glass slide in distilled water;
7. Dehydrate the slides in ethanol 70%, 85%, and 100% for 2 min each, at RT.
Completely air-dry the slides.
This should be enough to disassembly the previously applied FISH probes from
chromosomes and the FISH procedure should proceed from step 11 (denaturation of
slides). Be careful with the time of denaturation in corresponding buffer, since chro-
mosomes can be very fragile. This sequential FISH can be very helpful to identify
shared regions of distinct probes.
ACKNOWLEDGMENTS
The authors are thankful for the funding of Fundação de Amparo à Pesquisa do Estado
de São Paulo (Process numbers 2020/02681–9; 2018/14677–6 and 2020/11772–8).
Additionally, we would like to thank CNPq and the Humboldt Foundation for fnan-
cial support.
REFERENCES
1. Nelson, J.S.; Grande, T.C.; Wilson, M.V. Fishes of the World. John Wiley & Sons, New
York, 2016.
2. Salzburger, W. Understanding explosive diversifcation through cichlid fsh genomics.
Nat. Rev. Genet. 2018, 19, 705–717.
3. Fricke, R.; Eschmeyer, W.N.; Van der Laan, R. (Eds.). Eschmeyer’s catalog of fshes:
Genera, species, references. http://researcharchive.calacademy.org/research/ichthyol-
ogy/catalog/fshcatmain.asp [accessed on 01/06/2022].
294 Cytogenetics and Molecular Cytogenetics
4. Lehmann, R.; Kovařík, A.; Ocalewicz, K.; Kirtiklis, L.; Zuccolo, A.; Tegner, J.N.;
Wanzenböck, J.; Bernatchez, L.; Lamatsch, D.K.; R. Symonová. DNA transposon expan-
sion is associated with genome size increase in Mudminnows. Genome Biol. Evol. 2021,
13, evab228.
5. Santos, R.Z.; Calegari, R.M.; Silva, D.M.Z.A.; Ruiz-Ruano, F.J.; Melo, S.; Oliveira, C.;
Foresti, F.; Uliano-Silva, M.; Porto-Foresti, F.; Utsunomia, R. A long-term conserved
satellite DNA that remais unexpanded in several genomes of Characiformes fsh is
actively transcribed. Genome Biol. Evol. 2021, 13, evab002.
6. Yano, C.F.; Sember, A.; Kretschmer, R.; Bertollo, L.A.C.; Ezaz, T.; Hatanaka, T.;
Liehr, T.; Ráb, P.; Al-Rikabi, A.; Viana, P.F.; Feldberg, E.; Oliveira, E.A.; Toma, G.A.;
Cioff, M.B. Against the mainstream: Exceptional evolutionary stability of ZW sex
chromosomes across the fsh families Triportheidae and Gasteropelecidae (Teleostei:
Characiformes). Chromosome Res. 2021, 29, 1–26.
7. Pinkel, D.; Straume, T.; Gray, J.W. Cytogenetic analysis using quantitative, high sensitiv-
ity, fuorescence hybridization. Proc. Nat. Acad. Sci. 1986, 83, 2934–2938.
8. Pendás, A.M.; Morán, P.; Garcia-Vázquez, E. Ribosomal RNA genes are interspersed
throughout a heterochromatic chromosome arm in Atlantic salmon. Cytogenet. Genome
Res. 1993, 63, 128–130.
9. Pendás, A.M.; Morán, P.; García-Vázquez, E. Multi-chromosomal location of ribosomal
RNA genes and heterochromatin association in brown trout. Chromosome Res. 1993, 1,
63–67.
10. Pendás, A.M.; Moran, P.; Freije, J.P.; Garcia-Vazquez, E. Chromosomal mapping and
nucleotide sequence of two tandem repeats of Atlantic salmon 5S rDNA. Cytogenet.
Genome Res. 1994, 67, 31–36.
11. Phillips, R.B. Application of fuorescence in situ hybridization (FISH) to fsh genetics
and genome mapping. Marine Biotechnol. 2001, 3, S145–S152.
12. Wiegant, J.; Ried, T.; Nederlof, P.M.; Ploeg, M.V.D.; Tanke, H.J.; Raap, A.K. In situ
hybridisation with fuoresceinated DNA. Nucl. Acids Res. 1991, 19, 3237–3241.
13. Wiegant, J.; Galjart, N.J.; Raap, A.K.; d’Azzo, A. The gene encoding human protective
protein (PPGB) is on chromosome 20. Genomics 1991, 10(2), 345–349.
14. Freije, J.P.; Pendas, A.M.; Velasco, G.; Roca, A.; Abrahamson, M.; Lopez-Otin, C.
Localization of the human cystatin D gene (CST5) to chromosome 20p11. 21 by in situ
hybridization. Cytogenet. Genome Res. 1993, 62, 29–31.
15. Yano, C.F.; Bertollo, L.A.C.; Cioff M.B. Fish‑FISH: Molecular Cytogenetics in
Fish Species: In Fluorescence In Situ Hybridization (FISH). Springer, Berlin, 2017,
pp. 429–443.
16. Bertollo, L.A.C. Cytotaxonomic considerations on Hoplias lacerdae (Pisces,
Erythrinidae). Brazil. J. Genet. 1978, 1, 103–120.
17. Bertollo, L.A.C.; Cioff, M.B.; Moreira-Filho, O. Direct chromosome preparation
from freshwater teleost fshes. In: Fish Cytogenetic Techniques (Chondrichthyans and
Teleosts); Ozouf-Costaz, C.; Pisano, E.; Foresti, F.; Almeida Toledo, L.F., Eds., CRC
Press/Taylor & Francis Group, Oxon, 2015, pp. 21–26.
18. Zapata, A. Ultrastructural study of the teleost fsh kidney. Development. Comp. Immunol.
1979, 3, 55–65.
19. Oliveira, E.A.; Bertollo, L.A.C.; Rab, P.; Ezaz, T.; Yano, C.F.; Hatanaka, T.; Jegede, O.I.;
Tanomtong, A.; Liehr, T.; Sember, A.; Maruyama, S.R.; Feldberg, E.; Viana, P.F.; Cioff,
M.B. Cytogenetics, genomics and biodiversity of the South American and African
Arapaimidae fsh family (Teleostei, Osteoglossiformes). PloS One 2019, 14, e0214225.
20. Paim, F.G.; Maia, L.; Landim-Alvarenga, F.D.C.; Foresti, F.; Oliveira, C. New protocol
for cell culture to obtain mitotic chromosomes in fshes. Meth. Protoc. 2018, 1, 47.
FISH—in Fish Chromosomes 295
21. Traut, W.; Winking, H. Meiotic chromosomes and stages of sex chromosome evolution
in fsh: Zebrafsh, platyfsh and guppy. Chromosome Res. 2001, 9, 659–672.
22. Rebordinos, L.; Cross, I.; Merlo, A. High evolutionary dynamism in 5S rDNA of fsh:
State of the art. Cytogenet. Genome Res. 2013, 141, 103–113.
23. Lafontaine, D.L.; Tollervey, D. The function and synthesis of ribosomes. Nat. Rev. Mol.
Cell Biol. 2001, 2, 514–520.
24. Cioff, M.B.; Martins, C.; Centofante, L.; Jacobina, U.; Bertollo, L.A.C. Chromosomal vari-
ability among allopatric populations of Erythrinidae fsh Hoplias malabaricus: Mapping
of three classes of repetitive DNAs. Cytogenet. Genome Res. 2009, 125, 132–141.
25. Martins, C.; Galetti, P.M. Chromosomal localization of 5S rDNA genes in Leporinus fsh
(Anostomidae, Characiformes). Chromosome Res. 1999, 7, 363–367.
26. Crepaldi, C.; Martí, E.; Gonçalves, É.M.; Martí, D.A.; Parise-Maltempi, P.P. Genomic
differences between the sexes in a fsh species seen through satellite DNAs. Front.
Genet. 2021, 12, 728670.
27. Volff, J.N.; Schartl, M. Variability of genetic sex determination in poeciliid fshes.
Genetica. 2001, 111, 101–110.
28. Ferreira, D.C.; Porto-Foresti, F.; Oliveira, C.; Foresti, F. Transposable elements as a
potential source for understanding the fsh genome. Mobile Genet. Elements. 2011, 1,
112–117.
29. Chalopin, D.; Naville, M.; Plard, F.; Galiana, D.; Volff, J.N. Comparative analysis of
transposable elements highlights mobilome diversity and evolution in vertebrates.
Genome Biol. Evol. 2015, 7, 567–580.
30. Carducci, F.; Barucca, M.; Canapa, A.; Carotti, E.; Biscotti, M.A. Mobile elements in
ray-fnned fsh genomes. Life. 2020, 10, 221.
31. Ferreira, A.M.V.; Marajó, L.; Matoso, D.A.; Ribeiro, L.B.; Feldberg, E. Chromosomal
mapping of Rex retrotransposons in Tambaqui (Colossoma macropomum Cuvier, 1818)
exposed to three climate change scenarios. Cytogenet. Genome Res. 2019, 159, 39–47.
32. da Silva, F.A.; Guimarães, E.M.C.; Carvalho, N.D.; Ferreira, A.M.; Schneider, C.H.;
Carvalho-Zilse, G.A.; Feldberg, E.; Gross, M.C. Transposable DNA elements in
Amazonian fsh: From genome enlargement to genetic adaptation to stressful environ-
ments. Cytogenet. Genome Res. 2020, 160, 148–155.
33. Volff, J.N.; Körting, C.; Sweeney, K.; Schartl, M. The non-LTR retrotransposon Rex3
from the fsh Xiphophorus is widespread among teleosts. Mol. Biol. Evol. 1999, 16,
1427–1438.
34. Marz, M.; Kirsten, T.; Stadler, P.F. Evolution of spliceosomal snRNA genes in metazoan
animals. J. Mol. Evol. 2008, 67, 594–607.
35. Cabral-de-Mello, D.C.; Valente, G.T.; Nakajima, R.T.; Martins, C. Genomic organization
and comparative chromosome mapping of the U1 snRNA gene in cichlid fsh, with an
emphasis in Oreochromis niloticus. Chromosome Res. 2012, 20, 279–292.
36. Silva, D.M.; Utsunomia, R.; Pansonato-Alves, J.C.; Oliveira, C.; Foresti, F. Chromosomal
mapping of repetitive DNA sequences in fve species of Astyanax (Characiformes,
Characidae) reveals independent location of U1 and U2 snRNA sites and association of
U1 snRNA and 5S rDNA. Cytogenet. Genome Res. 2015, 146, 144–152.
37. Utsunomia, R.; Silva, D.M.Z.A.; Ruiz-Ruano, F.J.; Araya-Jaime, C.; Pansonato-Alves,
J.C.; Scacchetti, P.C.; Hashimoto, D.T.; Oliveira, C.; Trifonov, V.A.; Porto-Foresti, F.;
Camacho, J.P.M.; Foresti, F. Uncovering the ancestry of B chromosomes in Moenkhausia
sanctaeflomenae (Teleostei, Characidae). PLoS One. 2016, 11, e0150573.
38. Yang, F.; Trifonov, V.; Ng, B.L.; Kosyakova, N.; Carter, N.P. Generation of Paint
Probes from Flow‑Sorted and Microdissected Chromosomes: In Fluorescence In Situ
Hybridization (FISH). Springer, Berlin; 2017, pp. 63–79.
296 Cytogenetics and Molecular Cytogenetics
39. Sassi, F.M.C.; Deon, G.A.; Moreira-Filho, O.; Vicari, M.R.; Bertollo, L.A.C.; Liehr, T.;
de Oliveira, E.A.; Cioff, M.B. Multiple sex chromosomes and evolutionary relation-
ships in Amazonian catfshes: The outstanding model of the genus Harttia (Siluriformes:
Loricariidae). Genes. 2020, 11, 1179.
40. Sassi, F.M.C.; Moreira-Filho, O.; Deon, G.A.; Sember, A.; Bertollo, L.A.C.; Liehr, T.;
Oliveira, V.C.S.; Viana, P.F.; Feldberg, E.; Vicari, M.R.; Cioff, M.B. Adding new pieces
to the puzzle of karyotype evolution in Harttia (Siluriformes, Loricariidae): Investigation
of Amazonian species. Biology. 2021, 10, 922.
41. Sassi, F.M.C.; Perez, M.F.; Oliveira, V.C.S.; Deon, G.A.; de Souza, F.H.S.; Ferreira,
P.H.N.; de Oliveira, E.A.; Hatanaka, T.; Liehr, T.; Bertollo, L.A.C.; Cioff, M.B. High
genetic diversity despite conserved karyotype organization in the giant Trahiras from
genus Hoplias (Characiformes, Erythrinidae). Genes. 2021, 12, 252.
25 FISH—and the
Characterization of
Synaptonemal Complex
Victor Spangenberg
CONTENTS
Introduction ............................................................................................................ 297
Immuno-FISH Approach on Total Preparations of Synaptonemal Complexes ..... 298
The Logical Scheme of Immuno-FISH Is as Follows............................................ 299
DNA FISH Probes and Spread Preparation of Synaptonemal Complexes ............ 300
Spreading Technique .............................................................................................. 302
Materials ....................................................................................................... 302
Procedure ...................................................................................................... 302
Conclusions ............................................................................................................ 303
References .............................................................................................................. 304
INTRODUCTION
Meiotic prophase I is the key stage of meiosis and includes several sequential sub-
stages connected with synaptonemal complex (SC), which is the skeletal structure
of meiotic bivalents. Therefore, studies of SC-preparations are not focused on one
specifc stage. On the contrary, it is a complex analysis of dynamic changes in the
nuclei of germ cells of all prophase I stages of meiosis: leptotene, zygotene, pachy-
tene, diplotene and diakinesis. In biological objects without strict synchronization
in meiosis, it is usually possible to obtain all of the listed stages on one cytological
preparation. The stages of assembly and subsequent disassembly of SC are associ-
ated with numerous protein markers of DNA double-strand breaks (DSB) processing
(their formation and subsequent repair), meiotic recombination, chromatin remod-
eling and many others. Thus, the SC is a highly dynamic nucleoprotein structure.
The study of protein markers of meiosis using immunocytochemical (ICC) methods
makes SC-karyotyping an important extension to the analysis of metaphase mitotic
chromosomes.
There is a wide variety of methods for obtaining preparations of meiotic chro-
mosomes, i.e. total preparations of SCs. Most applicable methods for the analysis of
meiotic nuclei are spread techniques.[1, 2] The main feature of spreading approaches
is obtaining a fat (spread) preparation from a native three-dimensional nucleus.
Notably, spread preparation of SCs should not contain too many overlapped biva-
lents. Although such confgurations (overlapping) are unavoidable when studying
TABLE 25.1
Basic Morphological Criteria of Meiotic Prophase I Stages and Most-Used
Protein Markers—Synaptonemal Complex Associated Proteins
Stages of Meiotic Synaptonemal Complex Basic Protein Markers Used for
Prophase I Identifcation of Stages
Leptotene formation of axial elements of SPO11, RAD51, γH2AFX
chromosomes, intense DSB processing
and repair.
Early zygotene clustering of chromosome’s telomeres— SYCP3, SYCP1, RAD51,
formation of “chromosomal bouquet” γH2AFX
structure. Initiation of peritelomeric/
interstitial synapsis.
Late zygotene prolongation of chromosomal synapsis, SYCP3, SYCP1, RAD51,
declustering of telomere ends. γH2AFX
Pachytene complete assembly of synaptonemal SYCP3, SYCP1, MLH1,
complexes (meiotic bivalents). γH2AFX(on the XY-bivalent only)
Early diplotene initiation of homologous chromosomes SYCP3, SYCP1, MLH1
desynapsis—desynaptic “forks”, or
interstitial regions of desynapsis.
Late diplotene progression of desynapsis, elongation and SYCP3
fragmentation of axial elements,
synaptonemal complex disassemly.
Diakinesis complete removal of SYCP3 from SYCP3
chromosome axes, but remnants near
centromeres.
species with long chromosomes, following the spread protocols one can achieve
analyzable total preparations of SCs.
Methods for obtaining total preparations of SCs were developed for mammals,[1, 2]
widely used for reptiles,[3, 4] and also adapted for other groups, such as fshes,[5]
insects,[6] plants,[7] and fungi,[8] with some modifcations. Particular care should be
taken when going for preparations from meiocytes of organisms that have strong
enzymatic systems, for example fungi.[8] Ice-cold conditions or use of protease inhib-
itors are highly recommended, since during hypotonic shock and cell destruction,
enzymes can destroy proteins of SC. Interestingly, some authors recommend to use
protease inhibitors for mammalian cells as well.[9] The most useful protein markers
of SC (i.e. morphologi-cal criteria for determining the stages of meiotic prophase I)
are presented in Table 25.1.
After FISH, it is recommended to make images not only the optical channel
under study, but all those available on the microscope assembly. This is important
because chromatin becomes less contrasting after denaturation. The more opti-
cal channels are captured, the easier it is to colocalize the ICC and FISH images
300 Cytogenetics and Molecular Cytogenetics
using morphological structures as reference. The most convenient for this are
the SC axial structures (immunostaining of major SC protein SYCP3 is usually
detectable after FISH if stable antibody conjugates are used), or secondly, chro-
matin contrasted with 4′,6-diamidino-2-phenylindole (= DAPI) before and after
FISH.
Thus, immuno-FISH on SC-karyotypes allows to study chromatin changes (DNA
and proteins) in dynamics—during successive stages of meiotic prophase I. It is
important to know that meiotic bivalents are on average 10 times longer than meta-
phase mitotic chromosomes. Therefore, many tasks related to precise positioning of
DNA-loci can be more easily solved on SC spreads. On the other hand, important
discoveries on the elimination of part of the genomes also have been made using
immuno-FISH on SC-preparations: germline-restricted chromosome (GRC), elimi-
nated from somatic cells and spermatids,[16] or elimination of extra X-chromosome
from 47,XXY karyotype in human.[9]
FIGURE 25.2 Immuno-FISH study of 160 Kb (yellow) and 90 Kb (violet) DNA loci on
chromosome 17 in human spermatocyte I, early diplotene.
a) 160 Kb probe (yellow) on the bivalent 17.
b) 160 Kb probe (yellow) visualizes chromatin loops on the bivalent 17.
c) Immunostaining of crossing over marker, MLH1 (red) on the bivalent 17 near 160 Kb
probe (yellow) Scale bar—5μm (from[18] with modifcations).
302 Cytogenetics and Molecular Cytogenetics
chromosomes.[26, 27] This approach also showed applicability when used on prepara-
tions of SCs to detect single-copy genes.[28]
In general, FISH techniques on preparations of SCs are similar to the main proto-
cols used on mitotic metaphase chromosomes. Particularly to those protocols where
signal amplifcation is used (biotin-avidin, tyramide detection systems). Minor dif-
ferences relate to less compaction, and hence greater accessibility of chromatin on
meiotic prophase I preparation. Therefore, for example, RNAse treatment can be
done in half the time compared to somatic metaphase plates.
SPREADING TECHNIQUE
MATERIALS
• Microscope slides: All preparations of primary spermatocyte’s of oocytes
spread nuclei (i.e. total preparations of SCs) should be done on slides with
an adhesive coating, such as poly-L-lysine. This is important for attachment
and preservation of chromatin during several rounds: ICC (immunostaining
and washing) and FISH (denaturation, annealing, washing).
• Fixative for spreads: 1% paraformaldehyde, pH 8.4 containing 0.15% Triton
X-100. Bi-distilled water with the addition of paraformaldehyde powder
must be heated to 60–65°C. Add 1–2 small drops (10–15 µl) of 1N NaOH
until a transparent solution is obtained (avoid boiling). Let the fxative cool,
flter using flter paper, add Triton X-100, and adjust to pH8.4 with 0.1M
sodium tetraborate.
• Hypotonic solution: 0.1 M sucrose (1.7g sucrose in 50ml bi-distilled water).
• Wash solution: 0.4% Photofo (Kodak) adjusted to pH 8.2–8.4 with borate
buffer. Can be stored as a 4% stock solution and diluted before use.
PROCEDURE
1. Isolate gonadal tissues and transfer to cold Eagle’s medium (without
L-Glutamine) or 1×PBS (pH 7.4) in a small Petri dish. Samples should be
isolated shortly before preparations. Alternatively, storage is allowed in
cold (+4°C) conditions in the Eagle medium (or similar) or in 1×PBS for
several hours. Avoid drying of tissues. Using two forceps, disaggregate
the tissue of the seminiferous tubules. In the case of ovarian tissue, cell
disaggregation can be carried out mechanically between glass slide and
coverslip (better to use concave microscope slide). Using a stereomicro-
scope, gently press the dissecting needle on the coverslip over the pressed
piece of tissue and collect the suspension with a pipette. Avoid drying out
and keep the temperature near 4°C using cold plate thermostat.
2. Transfer 50 μl of cell suspension from step 1 in a 1.5 ml reaction tube and
continue to disrupt aggregates using pipette with a 200 μl tip with one mm
shortened tip. Avoid formation of bubbles. Check a small amount of the
suspension in a light microscope for large spermatocytes and in case of
FISH—and the Characterization of Synaptonemal Complex 303
CONCLUSIONS
In general, the immuno-FISH method on preparations of meiotic chromosomes is
promising for working with a variety of biological objects due to the availability of
antibodies with a wide range of immune homology, as well as due to the accumula-
tion and analysis of genomic data. Thus, it is possible to study the key stage of the
life cycle of an organism using both, (i) a variety of meiosis associated protein mark-
ers, and (ii) DNA markers (whole chromosome painting, unique DNA loci, satellite
DNA, telomeric repeats, rDNA). In the near future, it is obvious that a large num-
ber of new protein markers associated with the SC in particular, and with meiosis
prophase I in general, will become available.[29] Therefore, when using immuno-
FISH-protocols, frst of all highest quality preparation of spread meiotic bivalents
is required, with preservation of the native form of protein markers. Second, FISH
protocols should take into account the lower chromatin density in meiosis compared
to the metaphase of mitosis. Overall, this means that methods of amplifying detected
signals are important.
304 Cytogenetics and Molecular Cytogenetics
REFERENCES
1. Navarro, J.; Vidal, F.; Guitart, M.; Egozcue, J. A method for the sequential study of syn-
aptonemal complexes by light and electron microscopy. Hum. Genet. 1981, 59, 419–421.
2. Peters, A.H.F.M.; Plug, A.W.; Van Vugt, M J.; De Boer, P. A drying-down technique
for the spreading of mammalian meiocytes from the male and female germline.
Chromosome Res. 1997, 5, 66–68.
3. Lisachov, A.; Poyarkov, N.; Pawangkhanant, P.; Borodin, P.; Srikulnath, K. New karyo-
type of Lygosoma bowringii (Günther, 1864) suggests cryptic diversity. Herpet. Notes.
2018, 11, 1083–1088.
4. Spangenberg, V.; Arakelyan, M.; Galoyan, E.; Martirosyan, I.; Bogomazova, A.;
Martynova, E.; de Bello Cioff, M.; Liehr, T.; Al-Rikabi, A.; Osipov, F.; Petrosyan, V.;
Kolomiets, O. Meiotic synapsis of homeologous chromosomes and mismatch repair
protein detection in the parthenogenetic rock lizard Darevskia unisexualis. Molecular
Reproduction and Development. 2021, 88, 119–127.
5. Araya-Jaime, C.; Serrano, É.A.; de Andrade Silva, D.M.Z.; Yamashita, M.; Iwai, T.;
Oliveira, C.; Foresti, F. Surface-spreading technique of meiotic cells and immunodetec-
tion of synaptonemal complex proteins in teleostean fshes. Mol. Cytogenet. 2015, 8,
1–6.
6. Viera, A.; Santos, J.L.; Parra, M.T.; Calvente, A.; Gómez, R.; de la Fuente, R.; Suja, J.Á.;
Page, J.; Rufas, J.S. Cohesin axis maturation and presence of RAD51 during frst meiotic
prophase in a true bug. Chromosoma. 2009, 118(5), 575–589.
7. Loidl, J.; Jones, G.H. Synaptonemal complex spreading in Allium. Chromosoma. 1986,
93, 420–428.
8. Mazheika, I.S.; Kolomiets, O.L. A method for preparation of synaptonemal complexes of
meiotic chromosomes from basidial protoplasts of the white button mushroom Agaricus
bisporus (Lange) Imbach. Russ J Genet. 2003, 39, 283–287.
9. Sciurano, R.B.; Luna Hisano, C.V.; Rahn, M.I.; Brugo Olmedo, S.; Rey Valzacchi, G.;
Coco, R.; Solari, A.J. Focal spermatogenesis originates in euploid germ cells in classical
Klinefelter patients. Hum. Reprod. 2009, 24, 2353–2360.
10. Sepsi, A.; Fábián, A.; Jäger, K.; Heslop-Harrison, J.S.; Schwarzacher, T. ImmunoFISH:
Simultaneous visualisation of proteins and DNA sequences gives insight into meiotic
processes in nuclei of grasses. Front Plant Sci. 2018, 9, 1193.
11. Kolomiets, O.L.; Lelekova, M.A.; Kashintsova, A.A.; Kurilo, L.F.; Bragina, E.E.;
Chernykh, V.B.; Gabliya, M.Y.; Vinogradov, I.V.; Vityazeva, I.I.; Bogolyubov, S.V.;
Spangenberg, V.E. Detection of human meiotic and spermatogenetic anomalies using
light, electron and fuorescence microscopy. Androl Genit Surgery. 2018, 19, 24–35.
12. Pigozzi, M.I. Localization of single-copy sequences on chicken synaptonemal complex
spreads using fuorescence in situ hybridization (FISH). Cytogenet Genome Res. 2007,
119, 105–112.
13. Spangenberg, V.; Losev, M.; Volkhin, I.; Smirnova, S.; Nikitin, P.; Kolomiets, O. DNA
Environment of centromeres and non-homologous chromosomes interactions in mouse.
Cells. 2021, 10, 3375.
14. Sciurano, R.B.; Solari, A.J. Ultrastructural and immunofuorescent methods for the
study of the XY body as a biomarker. In: Functional Analysis of DNA and Chromatin;
Humana Press, Totowa, NJ, 2014, pp. 137–149.
15. Spangenberg, V.; Arakelyan, M.; Galoyan, E.; Pankin, M.; Petrosyan, R.; Stepanyan,
I.; Grishaeva, T.; Danielyan, F.; Kolomiets, O. Extraordinary centromeres: Differences
FISH—and the Characterization of Synaptonemal Complex 305
in the meiotic chromosomes of two rock lizards species Darevskia portschinskii and
Darevskia raddei. Peer J. 2019, 7, e6360.
16. Torgasheva, A.A.; Malinovskaya, L.P.; Zadesenets, K.S.; Karamysheva, T.V.; Kizilova,
E.A.; Akberdina, E.A.; Pristyazhnyuk, I.E.; Shnaider, E.P.; Volodkina, V.A.; Saiftdinova,
A.F.; Galkina, S.A.; Larkin, D.M.; Rubtsov, N.B.; Borodin, P.M. Germline-restricted
chromosome (GRC) is widespread among songbirds. Proc. Natl. Acad. Sci. 2019, 116,
11845–11850.
17. Zadesenets, K.S.; Katokhin, A.V.; Mordvinov, V.A.; Rubtsov, N.B. Telomeric DNA in
chromosomes of fve opisthorchid species. Parasitol Internat. 2012, 61, 81–83.
18. Spangenberg, V. Dynamics of the structural organization of the chromosomes during
the meiotic prophase I in human and mouse. PhD thesis. NI Vavilov Institute of General
Genetics RAS (Moscow, 2013).
19. Lisachov, A.P.; Borodin, P.M. Microchromosome polymorphism in the sand lizard,
Lacerta agilis Linnaeus, 1758 (Reptilia, Squamata). Comparat. Cytogenet. 2016, 10, 387.
20. Beliveau, B.J.; Joyce, E.F.; Apostolopoulos, N.; Yilmaz, F.; Fonseka, C.Y.; McCole,
R.B.; Wu, C.T. Versatile design and synthesis platform for visualizing genomes with
Oligopaint FISH probes. Proc. Natl. Acad. Sci. 2012, 109, 21301–21306.
21. Froenicke, L.; Anderson, L.K.; Wienberg, J.; Ashley, T. Male mouse recombination maps
for each autosome identifed by chromosome painting. Am. J. Hum. Genet. 2002, 71,
1353–1368.
22. Bogdanov, Y.F.; Spangenberg, V.E.; Dadashev, S.Y.; Vityazeva, I.I.; Bogoliubov, S.V.;
Kolomiets, O.L. Morphological manifestation of unique DNA segments in human mei-
otic prophase I. Cell Tiss. Biol. 2012, 6, 407–411.
23. Grishaeva, T.M.; Spangenberg, V.E.; Kolomiets, O.L.; Dadashev, S.Y.; Bogdanov, Y.F.
Peculiarities of chromosome organization in meiosis. Cell Tiss. Biol. 2013, 7, 343–346.
24. Liehr, T. Commercial FISH probes. In: Fluorescence In Situ Hybridization (FISH);
Springer, Berlin, 2017, pp. 49–61.
25. Swennenhuis, J.F.; Foulk, B.; Coumans, F.A.; Terstappen, L.W. Construction of repeat-
free fuorescence in situ hybridization probes. Nucl. Acids Res. 2012, 40, e20.
26. Khrustaleva, L.I.; Kik, C. Localization of single-copy T-DNA insertion in transgenic
shallots (Allium cepa) by using ultra-sensitive FISH with tyramide signal amplifcation.
Plant J. 2001, 25, 699–707.
27. Kudryavtseva, N.; Ermolaev, A.; Karlov, G.; Kirov, I.; Shigyo, M.; Sato, S.; Khrustaleva,
L. A dual-color tyr-FISH method for visualizing genes/markers on plant chromosomes
to create integrated genetic and cytogenetic maps. Int J Mol Sci. 2021, 22, 5860.
28. Kirov, I.V.; Van Laere, K.; Khrustaleva, L.I. High resolution physical mapping of single
gene fragments on pachytene chromosome 4 and 7 of Rosa. BMC Genet. 2015, 16, 1–10.
29. da Cruz, I.; Rodríguez-Casuriaga, R.; Santiñaque, F.F.; Farías, J.; Curti, G.; Capoano,
C.A.; Geisinger, A. Transcriptome analysis of highly purifed mouse spermatogenic cell
populations: Gene expression signatures switch from meiotic-to postmeiotic-related pro-
cesses at pachytene stage. BMC Genomics. 2016, 17, 1–19.
26 RNA-FISH—
on Lampbrush
Chromosomes:
Visualization of Individual
Transcription Units
Tatiana Kulikova and Alla Krasikova
CONTENTS
Introduction............................................................................................................ 308
Description of Methods.......................................................................................... 309
Chromosome Isolation and Pretreatment...................................................... 309
RNA Preservation ......................................................................................... 309
Chromosome Fixation .................................................................................. 309
Probe Labeling.............................................................................................. 310
RNA-FISH Procedure................................................................................... 310
RNA FISH Procedure for Double Stranded DNA-Probes
(PCR- or Nick-Translation Products) ............................................... 311
Hybridization Mixture and In Situ Hybridization ............................ 311
Post-Hybridization Washings............................................................ 311
Detection of Hapten Labeled Probes ................................................ 311
RNA FISH Procedure for Oligonucleotide Probes....................................... 312
Hybridization Mixture and In Situ Hybridization ............................ 312
Post-Hybridization Washings............................................................ 312
Detection of Hapten Labeled Oligonucleotide Probes ..................... 312
Microscopy and Image Analysis................................................................... 313
Control Experiments .............................................................................................. 313
Interpretation of Results......................................................................................... 315
Conclusions............................................................................................................ 315
Acknowledgements................................................................................................ 315
References.............................................................................................................. 315
INTRODUCTION
RNA directed fuorescence in situ hybridization (FISH) is an effective approach
to localize RNA sequences in situ, visualize gene transcripts, and estimate gene
expression in time and space.[1–3] In a typical interphase nucleus, transcribed genes,
together with nascent transcripts at the site of their synthesis, are visible as dot-
like foci.[4, 5] The only exception are several highly expressed genes with very
long introns that form loop-like structures with nascent transcripts in Drosophila
spermatocytes,[6] and in human somatic cells.[7] However, in fshes, amphibians,
reptiles, birds, and certain insects, hypertranscription during oogenesis leads to
decompactization of hundreds of transcribed genomic regions and transforma-
tion of the meiotic chromosomes into highly elongated lampbrush chromosomes
(LBCs).[8–11] Lateral loops of LBCs representing sites of ongoing transcription
serve as a model for exploration into nascent RNA synthesis and processing at
the cytological level.[9, 12] Single transcription units on the lateral loops of amphib-
ian and avian LBCs with a gradient of RNP matrix can be seen even by light
microscopy.[13–15]
First in situ hybridization experiments on LBCs of newt Notophthalmus viride‑
scens were performed with radioactively labeled cRNA probes detected by autora-
diography.[16] In further studies, radioactive 5S rDNA probes were hybridized with
non-denatured N. viridescens LBCs to reveal nascent RNA.[17] In these early DNA/
RNA in situ hybridization experiments nascent 5S RNA transcripts were revealed
on a set of lateral loops. In control preparations, RNase pretreatment eliminated
5S RNA transcripts from the ribonucleoprotein (RNP) matrix of these loops. By
hybridization of the radioactively labelled DNA-probe to nascent RNA transcripts
on LBCs of crested newt, Triturus cristatus carnifex, Varley and co-authors evi-
denced transcription of highly repetitive DNA during oogenesis.[18] Similarly, radio-
active probes to satellite I DNA hybridized to non-denatured LBCs of N. viridescens
without RNase digestion demonstrated intense labeling of lateral loops.[19] Next
FISH method was developed and employed to detect nascent RNA on the lateral
loops of amphibian and avian LBCs.[13, 20–23] Nowadays, RNA-FISH can be success-
fully applied to reveal newly synthesized pre-mRNA and non-coding RNA on LBC
lateral loops. For example, probes made by in vitro transcription and bacterial artif-
cial chromosome (BAC)–derived probes were effectively hybridized to transcription
units of intensively expressed protein-coding genes on the lateral loops of axolotl and
chicken LBCs.[15, 24]
Visualization of nascent gene transcripts on individual transcription units could
be accompanied by immunofuorescent staining as described elsewhere.[25] For
example, by RNA-FISH after immunofuorescent detection of the elongating form
of RNA polymerase II we demonstrated that short 41 bp repeats are transcribed on
the lateral loops of chicken LBCs by RNA polymerase II with hyperphosphorylated
C-terminal domain.[13] For other examples of RNA-FISH after LBC immunostain-
ing, see in references [22] and [26]. RNA-FISH can also be combined with the target-
ing of labeled protein components to the RNP matrix of LBC lateral loops. Besides,
FISH according to nascent RNA hybridization protocol (see later) can be performed
on LBC preparations after incorporation of RNA precursors such as bromouridine
RNA-FISH—on Lampbrush Chromosomes 309
DESCRIPTION OF METHODS
CHROMOSOME ISOLATION AND PRETREATMENT
Methods for obtaining preparations of LBCs and nuclear bodies from amphibian and
avian diplotene oocytes are described in detail elsewhere (for example in [27] and
[28], or on https://projects.exeter.ac.uk/lampbrush/protocols.htm). In these protocols,
oocyte nucleus content is spread within an isolation chamber mounted on a standard
microscope slide. After microdissection from the oocyte nucleus LBC spreads are
fxed in paraformaldehyde (PFA) in phosphate buffered saline (PBS) followed by
alcohol-based fxation. Depending on the antigen detected, other fxation protocols
may be used: without ethanol or with reduced time of fxation in PFA.
Permeabilization can be omitted, since preparations of native chromosomes iso-
lated from the oocyte nucleus by microdissection are used. The absence of permea-
bilization results in the preservation of native chromosome morphology.
Nascent RNA on LBC preparations exists in a complex with a number of RNA
binding proteins and other RNPs; therefore, pretreatment with proteinases is not
performed, as this may reduce the specifc hybridization signal. For the RNA FISH
protocol no ribonuclease pretreatment should be done to detect nascent RNA targets
on LBC spreads. However, certain control slides imply RNase digestion step (see
later in the section “Control Experiments”).
RNA PRESERVATION
To achieve optimal results, RNA preservation measures must be followed, from chro-
mosome isolation procedure to post-hybridization washings. Bench surfaces should
be treated with 5% H2O2 or commercial RNase inhibitors. Slides and chambers for
chromosome isolation, lab beakers and other glassware, as well as metal instruments
should be washed in 2% 7X-detergent, thoroughly rinsed in distilled water and dried
in a drying oven at 200°C or autoclaved. Deionized water used for buffers should
be autoclaved. Whole chromosome isolation procedure and slide handling should be
performed with sterile laboratory gloves.
CHROMOSOME FIXATION
1. Fix freshly isolated LBC preparations in 2% PFA in a PBS for 30 minutes
at room temperature.
2. Dehydrate in 50% ethanol and 70% ethanol for 5 minutes each.
310 Cytogenetics and Molecular Cytogenetics
PROBE LABELING
A variety of probes can be used for RNA-FISH on LBC preparations including
PCR-derived DNA probes, probes obtained via nick-translation, cDNA probes bio-
tinylated by random-primed oligonucleotide labeling, RNA probes made by in vitro
transcription, and synthetic oligonucleotide or locked nucleic acid probes. Protocol
for labeling DNA probes for FISH on LBC preparations by (degenerated oligo-
nucleotide primed = DOP-) polymerase chain reaction (PCR) was given earlier in
[23]. Labeling of longer probes (for example isolated BAC clone DNA) by Nick-
translation can be performed as described in [29]. Oligonucleotide probes are usually
labeled by terminal deoxynucleotidyl transferase.[30, 31] Probes can be labeled directly
by fuorochrome-modifed nucleotides or indirectly by hapten-modifed nucleotides
further detected by a fuorochrome. The quality of all labeled probes can be verifed
by DNA-FISH on metaphase chromosome preparations.
To visualize multiple gene transcripts in the same experiment, a mixture of ~10
BAC clone-based probes can be applied on a single LBC set. If gene targets localize
in a relatively small genomic region (2–2.5 Mbp), BAC probes should be labeled with
different haptens and fuorochromes.
RNA-FISH PROCEDURE
RNA-FISH on LBC preparations is performed according to conventional protocol[32]
with modifcations.[20] Stringency of the hybridization can be adjusted by varying
the concentration of formamide, the incubation temperature during hybridization,
concentration of Na+ in the washing buffer, and the temperature of the washing buf-
fer. The duration of hybridization should not be greatly reduced since it may lead to
underrepresentation of specifc nascent RNA targets; however, in case of oligonucle-
otide probes, hybridization time can be reduced up to 4 hours. To detect nascent
transcripts on LBC lateral loops, amplifcation of the FISH signal is usually not
required due to the presence of multiple closely spaced RNA targets on a single
transcription unit.
Re-FISH according to RNA hybridization protocol can be performed on LBC
spreads after image acquisition. In this case, after coverslip removal, slides with
LBCs are washed in 4×SSC (saline-sodium citrate buffer) with 0.01% Tween 20 at
37°C. Then RNA FISH procedure is applied with another probe set. However, it
should be noted that RNA on the lateral loops could be partially degraded during
RNA-FISH—on Lampbrush Chromosomes 311
this procedure. Thus, Re-FISH protocol can be best applied to co-localize probes
which, as shown earlier, hybridize with nascent RNA.
Post-Hybridization Washings
12. Gently remove rubber cement with the forceps.
13. Place slides into a Coplin jar or 50 ml beaker with 2×SSC for 5–10 min-
utes to rinse coverslips.
14. Wash slides through three changes of 0.2×SSC at 60°C in a water bass
with rocking.
15. Wash slides through two changes of 2×SSC at 45°C in a water bass with
rocking.
17. Remove paraflm and excess of the blocking solution and apply 50–100 μl
of primary antibodies against a hapten or avidin/streptavidin conjugated
with a fuorochrome dissolved in blocking solution. Immediately cover the
drops with squares of paraflm. Incubate in a moist chamber at 37°C for
40 minutes.
18. Wash slides through three changes of 4×SSC with 0.01% Tween 20 at
42°C in a water bass with rocking.
19. Put slides into a moist chamber and apply 50–100 μl of secondary
antibodies or anti-avidin/streptavidin antibodies in blocking solution.
Immediately cover the drops with squares of paraflm. Incubate at 37°C
for 40 minutes.
20. Wash slides through three changes of 4×SSC with 0.01% Tween 20 at
42°C in a water bass with rocking.
21. If biotin is used, repeat steps 17 and 18.
22. Rinse slides with deionized water.
23. Dehydrate in increasing concentration of ethanol: 70% and 96%, for
5 minutes each.
24. Dry on air.
25. Apply 7 μl of antifade solution (65% glycerol, 0.01M TrisHCl, 2%
DABCO, pH 7.5) containing 1 μg/μl DAPI and mount 24×24 coverslips.
Post-Hybridization Washings
5. Gently remove rubber cement with the forceps.
6. Place slides into a Coplin jar or 50 ml beaker with 2×SSC for 5–10 minutes
to rinse coverslips.
7. Wash slides through three changes of 2×SSC at 37°C in a water bass with
rocking.
CONTROL EXPERIMENTS
A combination of control experiments is necessary to accurately estimate genuine
gene transcription on LBC spreads by RNA-FISH (Figure 26.1):
INTERPRETATION OF RESULTS
When we interpret the results of an RNA-FISH experiment on LBC preparations,
we should take into consideration the quality of spreads, RNA preservation, results
of control experiments and genomic characteristics of the targeted region. Control
experiments are essential to reliably establish the transcriptional activity of genes
on LBCs by RNA-FISH, for example [20, 21]. It should be noted that different RNA
species could be partially and nonuniformly degraded leading to variability across
RNA FISH results.[33] Moreover, analysis of gene expression on LBC spreads can
depend on the transcriptional activity of a certain gene, i.e. the number of nascent
RNA transcripts associated with individual transcription units. In addition, nascent
transcripts of tandem repeats could be still detectable on those LBC preparations
on which single-gene transcripts are already undetectable due to RNA degradation.
CONCLUSIONS
Here we describe an optimized RNA-FISH protocol directed to effective visual-
ization of transcribed genes on LBC preparations. In contrast to the basic DNA/
(DNA+RNA-transcript) FISH protocol on LBC spreads,[23] critical steps necessary
to detect nascent gene transcripts on lampbrush lateral loops are emphasized. The
delineated protocol was successfully implemented to establish transcription of pro-
tein-coding and non-coding RNA genes on LBCs of birds,[24] and can be applied to
LBC preparations of other diverse model organisms.
ACKNOWLEDGEMENTS
The research was supported by the RSF (grant #19–74–20075). Microscopy was
performed in the research resource center “Molecular and cell technologies” of
St. Petersburg State University.
REFERENCES
1. Chartrand, P.; Bertrand, E.; Singer, R.H.; Long, R.M. Sensitive and high-resolution
detection of RNA in situ. Methods Enzymol. 2000, 318, 493–506.
2. Brown, J.M.; Buckle, V.J. Detection of nascent RNA transcripts by fuorescence in situ
hybridization. Methods Mol. Biol. 2010, 659, 33–50.
3. Long, X.; Colonell, J.; Wong, A.M.; Singer, R.H.; Lionnet, T. Quantitative mRNA imag-
ing throughout the entire Drosophila brain. Nat. Methods. 2017, 14, 703–706.
4. Femino, A.M.; Fay, F.S.; Fogarty, K.; Singer, R.H. Visualization of single RNA tran-
scripts in situ. Science. 1998, 280, 585–590.
5. Ferraro, T.; Lucas, T.; Clémot, M.; De Las Heras Chanes, J.; Desponds, J.; Coppey, M.;
Walczak, A.M.; Dostatni, N. New methods to image transcription in living fy embryos:
The insights so far, and the prospects. WIREs Dev. Biol. 2016, 5, 296–310.
6. Fingerhut, J.M.; Moran, J.V.; Yamashita, Y.M. Satellite DNA-containing gigantic introns
in a unique gene expression program during Drosophila spermatogenesis. PLoS Genet.
2019, 15, e1008028.
7. Leidescher, S.; Ribisel, J.; Ullrich, S.; Feodorova, Y.; Hildebrand, E.; Galitsyna, A.;
Bultmann, S.; Link, S.; Thanisch, K.; Mulholland, C.; Dekker, J.; Leonhardt, H.; Mirny,
316 Cytogenetics and Molecular Cytogenetics
L.; Solovei, I. Spatial organization of transcribed eukaryotic genes. Nat. Cell Biol. 2022,
24, 327–339.
8. Callan, H.G. Lampbrush chromosomes. Springer, Berlin; 1986.
9. Morgan, G.T. Lampbrush chromosomes and associated bodies: New insights into prin-
ciples of nuclear structure and function. Chromosome Res. 2002, 10, 177–200.
10. Gaginskaya, E.; Kulikova, T.; Krasikova, A. Avian lampbrush chromosomes: A pow-
erful tool for exploration of genome expression. Cytogenet. Genome Res. 2009, 124,
251–267.
11. Krasikova, A.; Kulikova, T. Lampbrush Chromosomes: Current Concepts and Research
Perspectives. St Petersburg University Publishing House, St Petersburg, 2019.
12. Morgan, G.T. Imaging the dynamics of transcription loops in living chromosomes.
Chromosoma. 2018, 127, 361–374.
13. Deryusheva, S.; Krasikova, A.; Kulikova, T.; Gaginskaya, E. Tandem 41-bp repeats in
chicken and Japanese quail genomes: FISH mapping and transcription analysis on lamp-
brush chromosomes. Chromosoma. 2007, 116, 519–530.
14. Kaufmann, R.; Cremer, C.; Gall, J. G. Superresolution imaging of transcription units on
newt lampbrush chromosomes. Chromosome Res. 2012, 20, 1009–1015.
15. Keinath, M.C.; Davidian, A.; Timoshevskiy, V.; Timoshevskaya, N.; Gall, J.G.
Characterization of axolotl lampbrush chromosomes by fuorescence in situ hybridiza-
tion and immunostaining. Exp. Cell Res. 2021, 401, 112523.
16. Barsacchi, G.; Gall, J.G. Chromosomal localization of repetitive DNA in newt, Triturus.
J Cell Biol. 1972, 54, 580–591.
17. Pukkila, P.J. Identifcation of the lampbrush chromosome loops which transcribe 5S
ribosomal RNA in Notophthalmus (Triturus) viridescens. Chromosoma. 1975, 53,
71–89.
18. Varley, J.M.; Macgregor, H.C.; Erba, H.P. Satellite DNA is transcribed on lampbrush
chromosomes. Nature. 1980, 283, 686–688.
19. Diaz, M.O.; Barsacchi-Pilone, G.; Mahon, K.A.; Gall, J.G. Transcripts from both strands
of a satellite DNA occur on lampbrush chromosome loops of the newt Notophthalmus.
Cell. 1981, 24, 649–659.
20. Weber, T.; Schmidt, E.; Scheer, U. Mapping of transcription units on Xenopus laevis
lampbrush chromosomes by in situ hybridization with biotin-labeled cDNA probes. Eur.
J. Cell Biol. 1989, 50, 144–153.
21. Solovei, I.; Gaginskaya, E.R.; Macgregor, H.C. The arrangement and transcription of
telomere DNA sequences at the ends of lampbrush chromosomes of birds. Chromosome
Res. 1994, 2, 460–470.
22. Sallacz, N.B.; Jantsch, M.F. Chromosomal storage of the RNA-editing enzyme ADAR1
in Xenopus oocytes. Mol. Biol. Cell. 2005, 16, 3377–3386.
23. Zlotina, A.; Krasikova, A. FISH in lampbrush chromosomes. In Fluorescence In Situ
Hybridization (FISH); Springer Protocols Handbooks; Liehr, T., Ed. Springer Berlin
Heidelberg, Berlin; 2017, pp. 445–457.
24. Kulikova, T.; Maslova, A.; Starshova, P.; Rodriguez, S.J.; Krasikova, A. Comparison of
the somatic TADs and lampbrush chromomere-loop complexes in transcriptionally active
prophase I oocytes. Chromosoma. 2022. https://doi.org/10.1007/s00412-022-00780-5.
25. Krasikova, A.V.; Kulikova, T.V. Distribution of heterochromatin markers in lampbrush
chromosomes in birds. Russ. J. Genet. 2017, 53, 1022–1029.
26. Kulikova, T.; Chervyakova, D.; Zlotina, A.; Krasikova, A.; Gaginskaya, E. Giant
poly(A)-rich RNP aggregates form at terminal regions of avian lampbrush chromo-
somes. Chromosoma. 2016, 125, 709–724.
RNA-FISH—on Lampbrush Chromosomes 317
27. Morgan, G.T. Working with oocyte nuclei: Cytological preparations of active chromatin
and nuclear bodies from amphibian germinal vesicles. Methods Mol. Biol. 2008, 463,
55–66.
28. Gall, J.G.; Nizami, Z.F. Isolation of giant lampbrush chromosomes from living oocytes
of frogs and salamanders. J. Vis. Exp. 2016, 118, 54103.
29. Green, M.R.; Sambrook, J. Labeling of DNA probes by nick translation. Cold Spring
Harb Protoc. 2020, 2020, prot100602.
30. Green, M.R.; Sambrook, J. Labeling the 3′ termini of oligonucleotides using terminal
deoxynucleotidyl transferase. Cold Spring Harb Protoc. 2021, 2021, prot100685.
31. Green, M.R.; Sambrook, J. Preparation of labeled DNA, RNA, and oligonucleotide
probes. Cold Spring Harb Protoc. 2022, 2022, pdb.top100578.
32. Young, A.P.; Jackson, D.J.; Wyeth, R.C. A technical review and guide to RNA fuores-
cence in situ hybridization. Peer J. 2020, 8, e8806.
33. Baudrimont, A.; Jaquet, V.; Wallerich, S.; Voegeli, S.; Becskei, A. Contribution of RNA
degradation to intrinsic and extrinsic noise in gene expression. Cell Rep. 2019, 26, 3752–
3761.e5.
27 FISH—in Insect
Chromosomes
Vladimir E. Gokhman and Valentina G. Kuznetsova
CONTENTS
Introduction............................................................................................................ 319
Samples/Tissues ..................................................................................................... 320
Description of Methods.......................................................................................... 320
General Notes ............................................................................................... 320
FISH Protocol for Insect Chromosomes....................................................... 321
Chemicals (in Alphabetic Order) ...................................................... 322
General Procedure (Based on [24]) .................................................. 323
Alternative Procedure (Based on [17]) ............................................. 324
Use of C0t-1 DNA (Based on the Preceding Protocol,
Instead of Step 3)............................................................... 325
Yields ..................................................................................................................... 325
Physical Mapping of DNA Sequences.......................................................... 325
Repetitive Sequences ........................................................................ 325
Unique Sequences............................................................................. 326
Chromosome Painting (= Multicolor FISH)................................................. 327
Study of Various Properties of Particular Chromosomes ................. 327
Comparative Genomic Hybridization (CGH) and Genomic In Situ
Hybridization (GISH) ....................................................................... 328
Conclusions............................................................................................................ 328
Acknowledgement ................................................................................................. 329
References.............................................................................................................. 329
INTRODUCTION
Insects, i.e., the class Insecta within the subphylum Hexapoda, are an extremely
diverse and species-rich group of predominantly terrestrial animals. Recent esti-
mates suggest that there are about 5 to 15 million species of insects in the world
fauna, with more than a million already described.[1, 2] In fact, these creatures play an
essential role in many, if not all, terrestrial and freshwater ecosystems.[3] Given this
information, one can conclude that both the morphological variation and ecological
importance of insects are enormous.[1, 3] Moreover, this vast group is also substan-
tially diverse in terms of cytology and genetics, including chromosome morphol-
ogy and behavior.[4–12] For example, different types of the chromosome structure,
SAMPLES/TISSUES
To obtain reliable results of FISH, this technique must be performed on a suff-
cient number of high-quality mitotic and/or meiotic divisions. In turn, appropriate
developmental stages and tissues of insects must be chosen to achieve this goal.
Specifcally, male testes often harbor numerous spermatocytes undergoing either
mitotic or meiotic divisions.[23, 24] Nevertheless, certain numbers of cell divisions
can be found in the midgut as well as in the developing ovaries.[23, 25–28] Both
adults and immature stages of insects can generally become a valuable source of
mitoses and meioses, depending on the taxon under study.[25–29] However, devel-
oping tissues of insect larvae (and, to a lesser extent, pupae) often contain cells of
varying ploidy, thus making the precise identifcation of the chromosome num-
ber substantially diffcult, if at all possible. Exceptions from this rule include
salivary glands of dipteran larvae, which contain giant polytene chromosomes,
and imaginal discs of many Holometabola,[26, 30] for example, cerebral ganglia of
hymenopteran and coleopteran prepupae.[7, 23] Embryos extracted from develop-
ing eggs or viviparous females also provide high-quality mitotic divisions.[25, 31]
In some special cases, e.g., in many members of the suborder Symphyta (= lower
Hymenoptera), unfertilized eggs can also be taken out of adult females and incu-
bated in a moist chamber to stimulate their development.[32] It must also be noted,
however, that a precise morphological identifcation of feld-collected immature
stages is often diffcult, especially in holometabolous insects, but use of addi-
tional characters (for example, molecular ones) could offer a possible solution to
this problem.
DESCRIPTION OF METHODS
GENERAL NOTES
At present, two main groups of methods are used to produce high-quality chromo-
some preparations of insects, i.e., squash and air-drying techniques.[13, 31] All these
methods are aimed to ensure preservation of the chromosome structure, good spread-
ing of chromosomes as well as their reliable attachment to the slide. Nevertheless,
squash technique achieves these goals by applying physical pressure to a piece of
FISH—in Insect Chromosomes 321
preliminarily fxed tissue, which is turned into a monolayer of cells on the slide. This
preparation can be subsequently converted into a permanent one using dry ice or liq-
uid nitrogen.[30, 33] On the other hand, air-drying technique implies initial hypotonic
treatment of the tissue; after that, it is macerated and turned into cell suspension,
which is then applied to a slide, fxed and air-dried.[34] Both methods have their own
advantages and drawbacks. Specifcally, squash technique can readily utilize mate-
rial preserved with e.g. Carnoy’s fxative, whereas air-drying one predominantly
deals with living tissues. On the other hand, the air-drying method usually ensures
better chromosome morphology, but it apparently needs additional post-fxation to
keep the chromosomes attached to the slide.
In addition to the conventional FISH, a few studies of insect chromosomes were
conducted using the so-called fber-FISH.[25, 35] With this technique, FISH is performed
on the stretched DNA fbers (= mechanically stretched chromosomes) obtained from
interphase nuclei destroyed by either heat or chemical shock.[36] Fiber-FISH can detect
DNA sequences with a substantially increased resolution of 1–400 Kb, as opposed to
1–3 Mb provided by regular FISH on metaphase chromosomes.[25, 37]
As noted earlier, probes of different types are used to perform FISH. Among
these probes, both repetitive and unique DNA sequences, together with the so-called
chromosome paints (see later) can be listed.[38] On the other hand, these sequences
are either directly synthesized or amplifed from the existing genomic material. In
turn, amplifcation of the probes is substantially facilitated using bacterial artif-
cial chromosomes (BACs).[38–41] Regardless of the technique used for the prepara-
tion of chromosomes and probes, the process of FISH includes certain essential
stages, which can be subdivided into pre-hybridization, hybridization per se, and
post-hybridization[18, 19]:
Rinse in Milli-Q water briefy, pass through the ethanol series (70, 95 and
100 %) for 3 min each at RT, and air-dry. Apply 20 μl of Fluoroshield™
with DAPI and cover with coverslip. Examine under the microscope.
YIELDS
PHYSICAL MAPPING OF DNA SEQUENCES
Repetitive Sequences
FISH in insect chromosomes is most often used for mapping repetitive DNA
sequences, in which it is especially effcient.[19] These sequences can be provisionally
subdivided into the so-called satellite DNA, which usually occurs in clusters,[53] and
transposable elements.[54] Among the satellite sequences, ribosomal DNAs (rDNAs)
are the most frequently used as probes.[20, 43, 51, 55–57] Eukaryotic rDNA is composed
of tandem repeats divided into large (45S) and small (5S) transcriptional units. In
turn, these units form characteristic clusters, which usually contain hundreds or even
thousands of copies.[58] The large unit contains genes encoding 18S, 5.8S and 28S
rRNAs, which are separated by internal transcribed spacers.[59, 60] The small unit
encodes multiple 5S rRNA genes separated by non-transcribed spacers. 5S rDNA
is mostly distributed independently of 45S rDNA throughout the genome, although
it often forms clusters of its own.[25, 35, 61–67] However, 45S and 5S rRNA genes are
interspersed in the coleopteran Lagria villosa (Fabricius).[68] Nevertheless, the main
rDNA clusters of insects remain the most studied, with 18S rDNA being the most
frequently used probe.[24, 46, 48, 52, 61, 63, 66, 67, 69–74, etc.] The number and localization of
the rDNA clusters vary among closely related forms,[33, 75–81] and, occasionally, even
among particular populations and individuals of the same species. In the latter case,
different sections of the 45S rDNA cistron can evolve more or less independently.[60]
Moreover, certain rDNA-like sequences sometimes spread over the whole genome
and can therefore be detected on every chromosome.[82]
In addition to rDNA, other repetitive sequences can also be studied using
FISH. For example, histones are basic proteins, which, together with DNA, form
the fundamental units of chromatin structure.[83] Histone-coding genes are usually
arranged in clusters, and these clusters also can be physically mapped with this
technique.[35, 61, 63, 64, 67, 74, 78, 84–86] In insects, clusters of these genes are often (but not
always) co-localized with those of 5S rDNA.[61–63, 87, 88] Histone-coding genes can be
arranged either in clusters of tandem repeats formed by sequences by all types (H1,
H2A, H2B, H3 and H4), spaced by non-coding DNA, as individual genes or their
326 Cytogenetics and Molecular Cytogenetics
random combinations. For example, these genes are grouped into quartets and quin-
tets in several Drosophila species.[89]
Microsatellites represent short tandemly repeated DNA sequences (usually about
two to six base pairs),[58, 90] which can be effciently mapped using FISH.[35, 66, 71, 77, 91, 92]
Although these sequences are predominantly associated with heterochromatic seg-
ments[53] (but see[93]), microsatellites can have diverse distribution patterns within
the given chromosome set. Specifcally, they can be either scattered along the whole
genome or restricted to certain chromosomes and/or their segments, being either
dispersed or united into clusters.[77, 93, 94]
Small nuclear DNAs (snDNAs) encode corresponding RNAs (snRNAs), which
play important roles in processing pre-messenger RNA (= splicing), telomere main-
tenance, and a few other processes.[95] Various types of these DNAs (e.g. U1 and U2
snDNA), which are organized in clusters, can also be mapped using FISH.[64–67, 71, 96–98]
Tandemly repeated centromeric sequences of satellite DNA[53] are located in the
pericentromeric regions of chromosomes.[99] FISH visualized specifc sequences of
that kind in certain grasshoppers, beetles and ants,[100–102] but microdissection exper-
iments also showed that centromeric sequences can vary not only between different
species, but between different chromosomes of the same species as well (see e.g.[103]).
Telomeric sequences are located in the terminal regions of chromosomes.[9] In
most insects, as well as in other arthropods, TTAGG is the most widespread and
apparently ancestral telomeric repeat.[48, 52, 77, 104–107] However, telomeric motifs of
many insect taxa are highly variable, being often substituted by other sequences with
either known or, more frequently, unknown structure.[9, 108, 109] In addition, telomeric
repeats are substituted by transposons in higher Diptera, and these transposons can
be detected using FISH as well (see later).[19]
Although mobile repetitive DNA sequences, i.e., transposable elements,[54] are
often not arranged in clusters, they also can be revealed in insect chromosomes using
FISH.[19, 25, 110 –113] Sometimes, whole fractions of repetitive DNA, e.g., C0t-1 DNA,
can be physically mapped to insect chromosomes as well.[35, 66, 88, 114] This analysis
can provide an insight on spreading of repetitive sequences among different chromo-
somes, especially within their heterochromatic regions.[65, 115]
Unique Sequences
In addition to repetitive DNA sequences, certain unique, single-copy genes also can
be physically mapped on insect chromosomes using FISH. Previously, regular FISH
could only reveal longer sequences.[32] Later on, sensitivity of FISH was substantially
enhanced using e.g. tyramide signal amplifcation (TSA-FISH).[41, 74, 116]
During the last decades, an important role of chromosomal inversions in main-
taining genetic diversity among and within different populations became obvi-
ous.[117] In particular, these inversions preserve certain gene complexes within the
same chromosome, i.e., the so-called supergenes,[118] by preventing crossing over.
A few cases of supergenes are found in insects, including ants, in which their pres-
ence is demonstrated using FISH.[119] In addition, recent research revealed that
genomes of many parasitoid Hymenoptera harbor symbiotic polydnaviruses (dou-
ble-stranded DNA viruses) that are integrated into insect chromosomes, and then
FISH—in Insect Chromosomes 327
excised from the genome and further injected into the host during oviposition.[120]
Although DNA sequences of these viruses were detected in chromosomes of a
particular parasitic wasp using non-fuorescence in situ hybridization,[121] similar
studies could be conducted using FISH. Moreover, FISH also demonstrated that
a number of physiologically important genes are sex-linked in most clades of the
moth family Tortricidae.[40]
CONCLUSIONS
The previously mentioned results demonstrate that FISH represents a powerful tool
used in the cytogenetic study of insects (Figure 27.1). In addition to physical mapping
of various DNA sequences, it can highlight different genomic and chromosomal
rearrangements, and therefore can outline certain pathways of the karyotype and
genome evolution in this enormous group of animals.[11, 23, 25, 26, 31, 43, 44, 53, 73, 76, 82, 86, 138, 139]
We expect that, either by itself or in combination with other methods, this tech-
nique will reveal many new details of the genome change at various scales of space
and time.
FISH—in Insect Chromosomes 329
ACKNOWLEDGEMENT
The authors are grateful to Boris A. Anokhin (Zoological Institute, Russian Academy
of Sciences) for his useful advice on practical aspects of performing FISH on insect
chromosomes.
REFERENCES
1. Grimaldi, D.; Engel, M.S. Evolution of the Insects. Cambridge University Press,
Cambridge; 2005, 755 p.
2. Stork, N.E. How many species of insects and other terrestrial arthropods are there on
Earth? Ann. Rev. Entomol. 2018, 63, 31–45.
3. Scudder, G.G.E. The importance of insects. In: Insect Biodiversity: Science and
Society; Foottit, R.G., Adler, P.H., Eds.; 2nd Edition, 1; Wiley Blackwell, Oxford, 2017,
pp. 9–43.
4. White, M.J.D. Animal Cytology and Evolution. 3rd Edition. Cambridge University
Press, Cambridge; 1973, 961 p.
5. Lukhtanov, V.A. Sex chromatin and sex chromosome systems in nonditrysian
Lepidoptera “Insecta”. J. Zool. Syst. Evol. Res. 2000, 38, 73–79.
6. Nokkala, S.; Kuznetsova, V.G.; Maryańska-Nadachowska, A.; Nokkala, C. Holocentric
chromosomes in meiosis. I. Restriction of the number of chiasmata in bivalents.
Chromosome Res. 2004, 12, 733–739.
330 Cytogenetics and Molecular Cytogenetics
26. Sharakhova, M.V.; George, P.; Timoshevskiy, V.; Sharma, A.; Peery, A.; Sharakhov, I.V.
Mosquitoes (Diptera). In: Protocols for Cytogenetic Mapping of Arthropod Genomes;
Sharakhov, I.V., Ed. CRC Press, Boca Raton, FL; 2015, pp. 93–170.
27. Angus, R.B.; Jeangirard, C.; Stoianova, D.; Grozeva, S.; Kuznetsova, V.G. A chromo-
somal analysis of Nepa cinerea Linnaeus, 1758 and Ranatra linearis (Linnaeus, 1758)
(Heteroptera, Nepidae). Comp. Cytogenet. 2017, 11, 641–657.
28. Karagyan, G.; Golub, N.; Sota, T. Cytogenetic characterization of periodical cicadas
(Hemiptera: Cicadidae: Magicicada). Eur. J. Entomol. 2020, 117, 474–480.
29. Yoshido, A.; Sahara, K.; Yasukochi, Y. Silk moths (Lepidoptera). In: Protocols for
Cytogenetic Mapping of Arthropod Genomes; Sharakhov, I.V., Ed. CRC Press, Boca
Raton, FL; 2015, pp. 219–256.
30. Sharakhova, M.V.; Timoshevskiy, V.A.; Yang, F.; Demin, S.I.; Severson, D.W.;
Sharakhov, I.V. Imaginal discs—a new source of chromosomes for genome mapping of
the yellow fever mosquito Aedes aegypti. PLoS Negl. Trop. Dis. 2011, 5, e1335.
31. Mandrioli, M.; Manicardi, G.C. Aphids (Hemiptera). In: Protocols for Cytogenetic
Mapping of Arthropod Genomes; Sharakhov, I.V., Ed. CRC Press, Boca Raton, FL;
2015, pp. 327–350.
32. Matsumoto, K.; Yamamoto, D.S.; Sumitani, M.; Lee, J.M.; Hatakeyama, M.; Oishi, K.
Detection of a single copy gene on a mitotic metaphase chromosome by fuorescence
in situ hybridization (FISH) in the sawfy, Athalia rosae (Hymenoptera). Arch. Ins.
Biochem. Physiol. 2002, 49, 34–40.
33. Grozeva, S.; Anokhin, B.A.; Kuznetsova, V.G. Bedbugs (Hemiptera). In: Protocols for
Cytogenetic Mapping of Arthropod Genomes; Sharakhov, I.V., Ed. CRC Press, Boca
Raton, FL; 2015, pp. 285–326.
34. König, C.; Paschke, S.; Pollmann, M.; Reinisch, R.; Gantert, C.; Weber, J.; Krogmann,
L.; Steidle, J.L.M.; Gokhman, V.E. Molecular and cytogenetic differentiation within
the Lariophagus distinguendus (Förster, 1841) species complex (Hymenoptera,
Pteromalidae). Comp. Cytogenet. 2019, 13, 133–145.
35. Palacios-Gimenez, O.M.; Carvalho, C.R.; Ferrari Soares, F.A.; Cabral-de-Mello, D.C.
Contrasting the chromosomal organization of repetitive DNAs in two Gryllidae crick-
ets with highly divergent karyotypes. PLoS One. 2015, 10, e0143540.
36. Heiskanen, M.; Kallioniemi, O.; Palotie, A. Fiber-FISH: Experiences and a refned pro-
tocol. Genet. Anal.: Biomol. Eng. 1996, 12, 179–184.
37. Raap, A.K.; Florijn, R.J.; Blonden, L.A.J.; Wiegant, J.; Vaandrager, J.; Vrolijk, H.;
Dunnen, J.; Tanke, H.J.; van Ommen, G.-J. Fiber FISH as a DNA mapping tool.
Methods. 1996, 9, 67–73.
38. Liehr, T. Homemade locus-specifc FISH probes: Bacterial artifcial chromosomes. In:
Fluorescence In Situ Hybridization (FISH): Application Guide; Springer Protocols
Handbooks; Liehr, T., Ed. 2nd Edition. Springer Verlag, Berlin; 2017, 101–106.
39. Yoshido, A.; Yasukochi, Y.; Marec, F.; Abe, H.; Sahara, K. FISH analysis of the W
chromosome in Bombyx mandarina and several other species of Lepidoptera by means
of B. mori W-BAC probes. J. Ins. Biotechnol. Sericol. 2007, 76, 1–7.
40. Nguyen, P.; Sýkorová, M.; Šíchová, J.; Kůta, V.; Dalíková, M.; Čapková Frydrychová,
R.; Neven, L.G.; Sahara, K.; Marec, F. Neo-sex chromosomes and adaptive potential in
tortricid pests. Proc. Natl. Acad. Sci. USA. 2013, 110, 6931–6936.
41. Carabajal Paladino, L.Z.; Nguyen, P.; Šíchová, J.; Marec, F. Mapping of single-copy
genes by TSA-FISH in the codling moth, Cydia pomonella. BMC Genet. 2014,
15, S15.
42. Liehr, T.; Kreskowski, K.; Ziegler, M.; Piaszinski, K.; Rittscher, K. The standard
FISH procedure. In: Fluorescence In Situ Hybridization (FISH): Application Guide;
332 Cytogenetics and Molecular Cytogenetics
Springer Protocols Handbooks; Liehr, T., Ed. 2nd Edition; Springer Verlag, Berlin;
2017, 109–118.
43. Gokhman, V.E.; Anokhin, B.A.; Kuznetsova, V.G. Distribution of 18S rDNA sites and
absence of the canonical TTAGG insect telomeric repeat in parasitoid Hymenoptera.
Genetica. 2014, 142, 317–322.
44. Kuznetsova, V.G.; Grozeva, S.M.; Hartung, V.; Anokhin, B.A. First evidence for
(TTAGG)n telomeric sequence and sex chromosome post-reduction in Coleorrhyncha
(Insecta, Hemiptera). Comp. Cytogenet. 2015, 9, 523–532.
45. Kuznetsova, V.G.; Khabiev, G.N.; Anokhin, B.A. Cytogenetic study on antlions
(Neuroptera, Myrmeleontidae): First data on telomere structure and rDNA location.
Comp. Cytogenet. 2016, 10, 647–656.
46. Kuznetsova, V.G.; Maryańska-Nadachowska, A.; Shapoval, N.A.; Anokhin, B.A.;
Shapoval, A.P.; Cytogenetic characterization of eight Odonata species originating from
the Curonian spit (the Baltic Sea, Russia) using C-banding and FISH with 18S rDNA
and telomeric (TTAGG)n probes. Cytogenet. Genome Res. 2018, 153, 147–157.
47. Lachowska-Cierlik, D.; Maryańska-Nadachowska, A.; Kuznetsova, V.; Picker, M. First
chromosomal study of Mantophasmatodea: Karyotype of Karoophasma biedouwense
(Austrophasmatidae). Eur. J. Entomol. 2015, 112, 599–605.
48. Vershinina, A.O.; Anokhin, B.A.; Lukhtanov, V.A. Ribosomal DNA clusters and telo-
meric (TTAGG)n repeats in blue butterfies (Lepidoptera, Lycaenidae) with low and
high chromosome numbers. Comp. Cytogenet. 2015, 9, 161–171.
49. Gokhman, V.E.; Kuznetsova, V.G. Presence of the canonical TTAGG insect telomeric
repeat in the Tenthredinidae (Symphyta) suggests its ancestral nature in the order
Hymenoptera. Genetica. 2018, 146, 341–344.
50. Maryańska-Nadachowska, A.; Kuznetsova, V.G.; Golub, N.V.; Anokhin, B.A. Detection
of telomeric sequences and ribosomal RNA genes in holokinetic chromosomes of
fve jumping plant-lice species: First data on the superfamily Psylloidea (Hemiptera:
Sternorrhyncha). Eur. J. Entomol. 2018, 115, 632–640.
51. Golub, N.; Anokhin, B.; Kuznetsova, V. Comparative FISH mapping of ribosomal
DNA clusters and TTAGG telomeric sequences to holokinetic chromosomes of eight
species of the insect order Psocoptera. Comp. Cytogenet. 2019, 13, 403–410.
52. Gapon, D.A.; Kuznetsova, V.G.; Maryańska-Nadachowska, A. A new species of the
genus Rhaphidosoma Amyot et Serville, 1843 (Heteroptera, Reduviidae), with data on
its chromosome complement. Comp. Cytogenet. 2021, 15, 467–505.
53. Palomeque, T.; Lorite, P. Satellite DNA in insects: A review. Heredity. 2008, 100,
564–573.
54. Wells, J.N.; Feschotte, C. A feld guide to eukaryotic transposable elements. Ann. Rev.
Genet. 2020, 54, 539–561.
55. Cabrero, J.; Camacho, J.P.M. Location and expression of ribosomal RNA genes in
grasshoppers: Abundance of silent and cryptic loci. Chromosome Res. 2008, 16,
595–607.
56. Warchałowska-Śliwa, E.; Grzywacz, B.; Maryańska-Nadachowska, A.; Karamysheva,
T.V.; Heller, K.-G.; Lehmann, A.W.; Lehmann, G.U.C.; Chobanov, D.P. Molecular and
classical chromosomal techniques reveal diversity in bushcricket genera of Barbitistini
(Orthoptera). Genome. 2013, 56, 667–676.
57. Sochorová, J.; Garcia, S.; Gálvez, F.; Symonová, R.; Kovařík, A. Evolutionary trends
in animal ribosomal DNA loci: Introduction to a new online database. Chromosoma.
2018, 127, 141–150.
58. Richard, G.F.; Kerrest, A.; Dujon, B. Comparative genomics and molecular dynamics
of DNA repeats in eukaryotes. Microbiol. Mol. Biol. Rev. 2008, 72, 686–727.
FISH—in Insect Chromosomes 333
59. Hillis, D.M.; Dixon, M.T. Ribosomal DNA: Molecular evolution and phylogenetic
inference. Quart. Rev. Biol. 1991, 66, 411–453.
60. Ferretti, A.B.S.M.; Ruiz-Ruano, F.J.; Milani, D.; Loreto, V.; Martí, D.A.; Ramos, E.;
Martins, C.; Cabral-de-Mello, D.C. How dynamic could be the 45S rDNA cistron? An
intriguing variability in a grasshopper species revealed by integration of chromosomal
and genomic data. Chromosoma. 2019, 128, 165–175.
61. Cabral-de-Mello, D.C.; Martins, C.; Souza, M.J.; Moura, R.C. Cytogenetic mapping of
5S and 18S rRNAs and H3 histone genes in 4 ancient Proscopiidae grasshopper spe-
cies: Contribution to understanding the evolutionary dynamics of multigene families.
Cytogenet. Genome Res. 2011, 132, 89–93.
62. Cabral-de-Mello, D.C.; Oliveira, S.G.; Moura, R.C.; Martins, C. Chromosomal orga-
nization of the 18S and 5S rRNAs and histone H3 genes in Scarabaeinae coleopterans:
Insights into the evolutionary dynamics of multigene families and heterochromatin.
BMC Genet. 2011, 12, 88.
63. Oliveira, N.L.; Cabral-de-Mello, D.C.; Rocha, M.F.; Loreto, V.; Martins, C.; Moura,
R.C. Chromosomal mapping of rDNAs and H3 histone sequences in the grasshopper
Rhammatocerus brasiliensis (Acrididae, Gomphocerinae): Extensive chromosomal
dispersion and co-localization of 5S rDNA/H3 histone clusters in the A complement
and B chromosome. Mol. Cytogenet. 2011, 4, 24.
64. Bueno, D.; Palacios-Gimenez, O.M.; Cabral-de-Mello, D.C. Chromosomal mapping of
repetitive DNAs in the grasshopper Abracris favolineata reveal possible ancestry of
the B chromosome and H3 histone spreading. PLoS One. 2013, 8, e66532
65. Palacios-Gimenez, O.M.; Castillo, E.R.; Martí, D.A.; Cabral-de-Mello, D. C. Tracking
the evolution of sex chromosome systems in Melanoplinae grasshoppers through chro-
mosomal mapping of repetitive DNA sequences. BMC Evol. Biol. 2013, 13, 167.
66. Palacios-Gimenez, O.M.; Cabral-de-Mello, D.C. Repetitive DNA chromosomal orga-
nization in the cricket Cycloptiloides americanus: A case of the unusual X1X20 sex
chromosome system in Orthoptera. Mol. Genet. Genomics. 2015, 290, 623–631.
67. Bardella, V.B.; Fernandes, J.A.M.; Cabral de Mello, D.C. Chromosomal evolutionary
dynamics of four multigene families in Coreidae and Pentatomidae (Heteroptera) true
bugs. Mol. Genet. Genomics. 2016, 291, 1919–1925.
68. Gusso Goll, L.; Matiello, R.R.; Artoni, R.F.; Vicari, M.R.; Nogaroto, V.; de Barros,
A.V.; Almeida, M.C. High-resolution physical chromosome mapping of multigene fam-
ilies in Lagria villosa (Tenebrionidae): Occurrence of interspersed ribosomal genes in
Coleoptera. Cytogenet. Genome Res. 2015, 146, 64–70.
69. Maryańska-Nadachowska, A.; Anokhin, B.A.; Gnezdilov, V.M.; Kuznetsova, V.G.
Karyotype stability in the family Issidae (Hemiptera, Auchenorrhyncha) revealed by
chromosome techniques and FISH with telomeric (TTAGG)n and 18S rDNA probes.
Comp. Cytogenet. 2016, 10, 347–369.
70. Maryańska-Nadachowska, A.; Kuznetsova, V.G.; Karamysheva, T.V. Chromosomal
location of rDNA clusters and TTAGG telomeric repeats in eight species of the spittle-
bug genus Philaenus (Hemiptera: Auchenorrhyncha: Aphrophoridae). Eur. J. Entomol.
2013, 110, 411–418.
71. Piccoli, M.C.A.; Bardella, V.B.; Cabral-de-Mello, D.C. Repetitive DNAs in Melipona
scutellaris (Hymenoptera: Apidae: Meliponidae): Chromosomal distribution and
test of multiple heterochromatin amplifcation in the genus. Apidologie. 2018, 49,
497–504.
72. Micolino, R.; Cristiano, M.P.; Travenzoli, N.P.; Lopes, D.M.; Cardoso, D.C.
Chromosomal dynamics in space and time: Evolutionary history of Mycetophylax ants
across past climatic changes in the Brazilian Atlantic coast. Sci. Rep. 2019, 9, 18800.
334 Cytogenetics and Molecular Cytogenetics
73. Menezes, R.S.T.; Cabral-de-Mello, D.C.; Milani, D.; Bardella, V.B.; Almeida, E.A.B.
The relevance of chromosome fssions for major ribosomal DNA dispersion in
hymenopteran insects. J. Evol. Biol. 2021, 34, 1466–1476.
74. Provazníková, I.; Hejníčková, M.; Visser, S.; Dalíková, M.; Carabajal Paladino, L.Z.;
Zrzavá, M.; Voleníková, A.; Marec, F.; Nguyen, P. Large-scale comparative analysis of
cytogenetic markers across Lepidoptera. Sci. Rep. 2021, 11, 12214.
75. Cabrero, J.; Bugrov, A.; Warchałowska-Śliwa, E.; López-León, M.D.; Perfectti, F.;
Camacho, J.P.M. Comparative FISH analysis in fve species of Eyprepocnemidine
grasshoppers. Heredity. 2003, 90, 377–381.
76. Nguyen, P.; Sahara, K.; Yoshido, A.; Marec, F. Evolutionary dynamics of rDNA clus-
ters on chromosomes of moths and butterfies (Lepidoptera). Genetica. 2010, 138,
343–354.
77. Travenzoli, N.M.; Lima, B.A.; Cardoso, D.C.; Dergam, J.A.; Fernandes-Salomão, T.M;
Lopes, D.M. Cytogenetic analysis and chromosomal mapping of repetitive DNA in
Melipona species (Hymenoptera, Meliponini). Cytogenet. Genome Res. 2019,158, 213–224.
78. Martí, E.; Milani, D.; Bardella, V.B.; Albuquerque, L.; Song, H.; Palacios-Gimenez,
O.M.; Cabral-de-Mello, D.C. Cytogenomic analysis unveils mixed molecular evolu-
tion and recurrent chromosomal rearrangements shaping the multigene families on
Schistocerca grasshopper genomes. Evolution. 2021, 75, 2027–2041.
79. Panzera, F.; Pita, S.; Lorite, P. Chromosome structure and evolution of Triatominae:
A review. In: Triatominae—The Biology of Chagas Disease Vectors; Guarneri, A.;
Lorenzo, M., Eds. Springer Nature Switzerland AG, Cham, 2021, pp. 65–99.
80. Pereira, J.A.; Travenzoli, N.M.; de Oliveira, M.P.; Werneck, H.A.; Fernandes Salomão,
T.M.; Lopes, D.M. Molecular cytogenetics in the study of repetitive sequences help-
ing to understand the evolution of heterochromatin in Melipona (Hymenoptera,
Meliponini). Genetica. 2021, 149, 55–62.
81. Teixeira, G.A.; de Aguiar, H.J.A.C.; Petitclerc, F.; Orivel, J.; Lopes, D.M.; Barros,
L.A.C. Evolutionary insights into the genomic organization of major ribosomal DNA
in ant chromosomes. Ins. Mol. Biol. 2021, 30, 340–354.
82. Sproul, J.S.; Barton, L.M.; Maddison, D.R. Repetitive DNA profles reveal evidence
of rapid genome evolution and refect species boundaries in ground beetles. Syst. Biol.
2020, 69, 1137–1148.
83. Maxson, R.; Cohn, R.; Kedes, L. Expression and organization of histone genes. Annu.
Rev. Genet. 1983, 17, 239–277.
84. Cabrero, J.; Lopez-Leon, M.D.; Teruel, M.; Camacho, J.P. Chromosome mapping of H3
and H4 histone gene clusters in 35 species of acridid grasshoppers. Chromosome Res.
2009, 17, 397–404.
85. Mandrioli, M.; Manicardi, G.C. Chromosomal mapping reveals a dynamic organiza-
tion of the histone genes in aphids (Hemiptera: Aphididae). Entomologia. 2013, 1, e2.
86. Toscani, M.A.; Pigozzi, M.I.; Papeschi, A.G.; Bressa, M.J. Histone H3 methylation and
autosomal vs. sex chromosome segregation during male meiosis in Heteroptera. Front.
Ecol. Evol. 2022, 10, 836786.
87. Cabral-de-Mello, D.C.; Moura, R.C.; Martins, C. Chromosomal mapping of repetitive
DNAs in the beetle Dichotomius geminatus provides the frst evidence for an asso-
ciation of 5S rRNA and histone H3 genes in insects, and repetitive DNA similarity
between the B chromosome and A complement. Heredity. 2010, 104, 393–400.
88. Cabral-de-Mello, D.C.; Moura, R.C.; Melo, A.S.; Martins, C. Evolutionary dynamics of
heterochromatin in the genome of Dichotomius beetles based on chromosomal analy-
sis. Genetica. 2011, 139, 315–325.
FISH—in Insect Chromosomes 335
89. Nagoda, N.; Fukuda, A.; Nakashima, Y.; Mats, Y. Molecular characterization and evo-
lution of the repeating units of histone genes in Drosophila americana: Coexistence of
quartet and quintet units in a genome. Insect Mol. Biol. 2005, 14, 713–717.
90. Jonika, M.; Lo, J.; Blackmon, H. Mode and tempo of microsatellite evolution across
300 million years of insect evolution. Genes. 2020, 11, 945.
91. Ruiz-Ruano, F.J.; Cuadrado, A.; Montiel, E.E.; Camacho, J.P.; López-Leon, M.D. Next
generation sequencing and FISH reveal uneven and nonrandom microsatellite distribu-
tion in two grasshopper genomes. Chromosoma. 2015, 124, 221–234.
92. dos Santos, J.M.; Diniz, D.; Rodrigues, T.A.S.; Cioff, M.B.; Waldschmidt, A.M.
Heterochromatin distribution and chromosomal mapping of microsatellite repeats
in the genome of Frieseomelitta stingless bees (Hymenoptera: Apidae: Meliponini).
Florida Entomol. 2018, 101, 33–39.
93. Milani, D.; Cabral-de-Mello, D.C. Microsatellite organization in the grasshopper
Abracris favolineata (Orthoptera: Acrididae) revealed by FISH mapping: Remarkable
spreading in the A and B chromosomes. PLoS One. 2014, 9, e97956.
94. Elizeu, A.M.; Travenzoli, N.M.; Ferreira, R.P.; Lopes, D.M.; Tavares, M.G. Comparative
study on the physical mapping of ribosomal genes and repetitive sequences in Friesella
schrottkyi (Friese 1900) (Hymenoptera: Apidae, Meliponini). Zool. Anz. 2021, 292,
225–230.
95. Matera, A.G.; Terns, R.M.; Terns, M.P. Non-coding RNAs: Lessons from the small
nuclear and small nucleolar RNAs. Nat. Rev. Mol. Cell Biol. 2007, 8, 209–220.
96. Anjos, A.; Ruiz-Ruano, F.J.; Camacho, J.P.; Loreto, V.; Cabrero, J.; de Souza, M.J.;
Cabral-de-Mello, D.C. U1 snDNA clusters in grasshoppers: Chromosomal dynamics
and genomic organization. Heredity. 2015, 114, 207–219.
97. Milani, D.; Palacios-Gimenez, O.M.; Cabral-de-Mello, D.C. The U2 snDNA is a use-
ful marker for B chromosome detection and frequency estimation in the grasshopper
Abracris favolineata. Cytogenet. Genome Res. 2017, 151, 36–40.
98. Milani, D.; Ruiz-Ruano, F.J.; Camacho, J.P.M.; Cabral-de-Mello, D.C. Out of pat-
terns, the euchromatic B chromosome of the grasshopper Abracris favolineata is not
enriched in high-copy repeats. Heredity. 2021, 127, 475–483.
99. Plohl, M.; Luchetti, A.; Meštrović, N.; Mantovani, B. Satellite DNAs between selfsh-
ness and functionality: Structure, genomics and evolution of tandem repeats in centro-
meric (hetero)chromatin. Gene. 2008, 409, 72–82.
100. Huang, Y.C.; Lee, C.C.; Kao, C.Y.; Chang, N.C.; Lin, C.C.; Shoemaker, D.; Wang, J.
Evolution of long centromeres in fre ants. BMC Evol. Biol. 2016, 16, 189.
101. Mora, P.; Vela, J.; Ruiz-Mena, A.; Palomeque, T.; Lorite, P. Isolation of a pericen-
tromeric satellite DNA family in Chnootriba argus (Henosepilachna argus) with an
unusual short repeat unit (TTAAAA) for beetles. Insects. 2019, 10, 306.
102. Ruiz-Torres, L.; Mora, P.; Ruiz-Mena, A.; Vela, J.; Mancebo, F.J.; Montiel, E.E.;
Palomeque, T.; Lorite, P. Cytogenetic analysis, heterochromatin characterization and
location of the rDNA genes of Hycleus scutellatus (Coleoptera, Meloidae): A species
with an unexpected high number of rDNA clusters. Insects. 2021, 12, 385.
103. Fernandes, A.; Scudeler, P.A.S.; Diniz, D.; Foresti, F.; Campos, L.A.O.; Lopes, D.M.
Microdissection: A tool for bee chromosome studies. Apidologie. 2011, 42, 743–748.
104. Frydrychová, F.; Grossmann, P.; Trubač, P.; Vítková, M.; Marec, F. Phylogenetic distri-
bution of TTAGG telomeric repeats in insects. Genome. 2004, 47, 163–178.
105. Kuznetsova, V.; Grozeva, S.; Anokhin, B.A. The frst fnding of (TTAGG)n telomeric
repeat in chromosomes of true bugs (Heteroptera, Belostomatidae). Comp. Cytogenet.
2012, 6, 341–346.
336 Cytogenetics and Molecular Cytogenetics
106. Pita, S.; Panzera, F.; Mora, P.; Vela, J.; Palomeque, T.; Lorite, P. The presence of the
ancestral insect telomeric motif in kissing bugs (Triatominae) rules out the hypothesis
of its loss in evolutionarily advanced Heteroptera (Cimicomorpha). Comp. Cytogenet.
2016, 10, 427–437.
107. Chirino, M.G.; Dalíková, M.; Marec, F.R.; Bressa, M.J. Chromosomal distribution of
interstitial telomeric sequences as signs of evolution through chromosome fusion in six
species of the giant water bugs (Hemiptera, Belostoma). Ecol. Evol. 2017, 7, 5227–5235.
108. Mravinac, B.; Meštrović, N.; Čavrak, V.V.; Plohl, M. TCAGG, an alternative telomeric
sequence in insects. Chromosoma. 2011, 120, 367–376.
109. Menezes, R.S.T.; Bardella, V.B.; Cabral-de-Mello, D.C.; Lucena, D.A.A.; Almeida,
E.A.B. Are the TTAGG and TTAGGG telomeric repeats phylogenetically conserved in
aculeate Hymenoptera? Sci. Nat. 2017, 104, 85.
110. Lorite, P.; Maside, X.; Sanllorente, O.; Torres, M.I.; Periquet, G.; Palomeque, T. The
ant genomes have been invaded by several types of mariner transposable elements.
Naturwissenschaften. 2012, 99, 1007–1020.
111. Palacios-Gimenez, O.M.; Bueno, D.; Cabral-de-Mello, D.C. Chromosomal mapping
of two Mariner-like elements in the grasshopper Abracris favolineata {Orthoptera:
Acrididae} reveals enrichment in euchromatin. Eur. J. Entomol. 2014, 111,
329–334.
112. Li, Y.; Jing, X.A.; Aldrich, J.C.; Clifford, C.; Chen, J.; Akbari, O.A.; Ferree, P.M.
Unique sequence organization and small RNA expression of a “selfsh” B chromosome.
Chromosoma. 2017, 126, 753–768.
113. Amorim, I.C.; Costa, R.G.C.; Xavier, C.; Moura, R.C. Characterization and chro-
mosomal mapping of the DgmarMITE transposon in populations of Dichotomius
(Luederwaldtinia) sericeus species complex (Coleoptera: Scarabaeidae). Genet. Mol.
Biol. 2018, 41, 419–425.
114. Xavier, C.; Cabral-de-Mello, D.C.; Moura, R.C. Heterochromatin and molecular char-
acterization of DsmarMITE transposable element in the beetle Dichotomius schifferi
(Coleoptera: Scarabaeidae). Genetica. 2014, 142, 575–581.
115. Cunha, M.S.; Campos, L.A.O.; Lopes, D.M. Insights into the heterochromatin evolu-
tion in the genus Melipona (Apidae: Meliponini). Insectes Soc. 2020, 67, 391–398.
116. Fominaya, A.; Loarce, Y.; González, J.M.; Ferrer, E. Tyramide signal amplifcation:
Fluorescence in situ hybridization for identifying homoeologous chromosomes. In:
Plant Cytogenetics: Methods and Protocols; Kianian, S.F., Kianian, P.M.A., Eds.
Springer Science+Business Media, New York, NY; 2016, pp. 35–48.
117. Kirkpatrick, M. How and why chromosome inversions evolve. PLoS Biol. 2010, 8,
e1000501.
118. Thompson, M.J.; Jiggins, C.D. Supergenes and their role in evolution. Heredity. 2014,
113, 1–8.
119. Wang, J.; Wurm, Y.; Nipitwattanaphon, M.; Riba-Grognuz, O.; Huang, Y.-C.;
Shoemaker, D.; Keller, L. A Y-like social chromosome causes alternative colony orga-
nization in fre ants. Nature. 2013, 493, 664–668.
120. Fleming, J.G.W.; Summers, M.D. Polydnavirus DNA is integrated in the DNA of its
parasitoid wasp host. Proc. Natl. Acad. Sci. USA. 1991, 88, 9770–9774.
121. Belle, E.; Beckage, N.E.; Rousselet, J.; Poirié, M.; Lemeunier, F.; Drezen, J.-M.
Visualization of polydnavirus sequences in a parasitoid wasp chromosome. J. Virol.
2002, 76, 5793–5796.
122. Rütten, K.B.; Pietsch, C.; Olek, K.; Neusser, M.; Beukeboom, L.W.; Gadau, J.
Chromosomal anchoring of linkage groups and identifcation of wing size QTL using
FISH—in Insect Chromosomes 337
137. Hejníčková, M.; Dalíková, M.; Potocký, P.; Tammaru, T.; Trehubenko, M.; Kubíčková,
S.; Marec, F.; Zrzavá, M. Degenerated, undifferentiated, rearranged, lost: High vari-
ability of sex chromosomes in Geometridae (Lepidoptera) identifed by sex chromatin.
Cells. 2021, 10, 2230.
138. Lukhtanov, V.A.; Shapoval, N.A.; Anokhin, B.A.; Saiftdinova, A.F.; Kuznetsova, V.G.
Homoploid hybrid speciation and genome evolution via chromosome sorting. Proc. R.
Soc. B. 2015, 282, 20150157.
139. Pita, S.; Panzera, F.; Sánchez, A.; Panzera, Y.; Palomeque, T.; Lorite, P. Distribution and
evolution of repeated sequences in genomes of Triatominae (Hemiptera-Reduviidae)
inferred from genomic in situ hybridization. PLoS One. 2014, 9, e114298.
28 FISH—in Plant
Chromosomes
Susan Liedtke, Sarah Breitenbach
and Tony Heitkam
CONTENTS
Introduction............................................................................................................ 340
Samples/Tissues ..................................................................................................... 340
Description of Methods: Fixation .......................................................................... 341
Chemicals ..................................................................................................... 341
Fixation of Leaf Tissue ................................................................................. 341
Fixation of Primary Root Tissue................................................................... 341
Description of Methods: Preparation of Mitotic Plant Chromosomes................... 342
Chemicals ..................................................................................................... 343
Enzyme Treatment ........................................................................................ 343
Washing and Nuclei Purifcation .................................................................. 344
Preparation of the Dropping Preparation...................................................... 344
Description of Methods: Fluorescence In Situ Hybridization ............................... 344
Equipment..................................................................................................... 344
Chemicals ..................................................................................................... 345
FISH with Indirect Labeling......................................................................... 345
RNase Treatment .............................................................................. 345
Pepsin Treatment .............................................................................. 346
Fixation of Chromosomes................................................................. 346
Dehydration of the Preparations ....................................................... 346
Hybridization .................................................................................... 346
Washing (All Steps in Coplin Jars)................................................... 347
Antibody Reaction ............................................................................ 347
DAPI Staining................................................................................... 347
Fluorescence Microscopy ................................................................. 347
Typical Applications .............................................................................................. 348
Conclusions............................................................................................................ 350
Acknowledgements................................................................................................ 350
References.............................................................................................................. 350
INTRODUCTION
With the advent of fuorescence in situ hybridization (FISH) methods since the
1980s–1990s, we have rapidly gained insights into the structure, organization and evo-
lution of plant genomes.[1, 2] The potential of molecular cytogenetics was also recog-
nized in the plant breeding sector, often with a focus on hybridization, introgressions
and chromosomal additions. Now, with the advances in long-read sequencing and with
the new genome-editing techniques in place, the plant cytogenetics feld is reposition-
ing itself at the interface between microscopy and genomics. FISH methods are more
relevant than ever, for example to better resolve the inaccessible, repetitive genome
fraction,[3] the genomes’ three-dimensional organization,[4, 5] but also to identify
chromosomes,[6] assess haplotypes, predict recombination,[7] and determine genomic
variation among individuals.[8] On top, FISH may also assist the synthetic genomics
feld seeking to steer chromosome mutations and to build artifcial chromosomes.[9, 10]
Nevertheless, the application of FISH to plant chromosomes remains challenging.
First, plant genomes can contain over 80% of repetitive DNA,[11] and up to 149
gigabasepairs.[12] Although these numbers reside at the extreme ends of the corre-
sponding scales, they serve well to illustrate that plant genomes are often large and
repetitive. Therefore, they remain diffcult targets for sequence-based methods.
Second, only limited genome and haplotype information has been available for
many plants, leading to less fexibility in experimental design. This gap is now clos-
ing at high speed, with many new crop and wild plant genome builds being released
every year (see www.plabipd.de/timeline_view.ep). Similarly, many karyotyping
tools that were originally designed for human chromosomes are now fexible in
organism choice and can be used for plant chromosomes as well.
Finally, the preparation of plant chromosomes—a prerequisite for FISH—is not
straight-forward. Plants contain a high amount of fbers and their cells are enclosed
by a cell wall with a variable chemical composition, depending on species, tissue and
developmental stage. Finding the balance between cell wall degradation and removal
of cytoplasm on the one hand, and preserving chromosome integrity on the other
hand, is one of the key challenges of plant cytogenetics, a process which needs to be
adapted or even established for each new plant species and tissue coming to the lab.
SAMPLES/TISSUES
As most FISH experiments require spreads of mitotic chromosomes, typically,
dividing plant cells are harvested. In plants, mitotic cells occur in the meristems,
out of which the root apical meristems are best accessible. Hence, the wide majority
of plant cytogenetics labs uses root tip tissues to prepare mitotic chromosomes, with
primary roots being especially serviceable. However, for some plants this is not an
option: root crops for example accumulate carbohydrates in thickened roots, and
grown trees may have buried their root tips inaccessibly below the soil. For these
plants, very young leaves in leaf buds may provide a suitable alternative. Examples
for this are beet crops for which a barely visible young leaf can be picked from the
middle of the rosette (as performed for example by[13, 14]), and trees or shrubs such as
beeches or camellias, for which young shoot tips can be taken in spring season.[15, 16]
FISH—in Plant Chromosomes 341
To study meiosis, tissue of the developing fower has to be harvested; i.e., the anthers
for male meiosis and the ovules for female meiosis (please see for the collection of
protocols for the analysis of meiotic chromosomes by[17]).
CHEMICALS
• Fixative: methanol glacial acetic acid (3:1).
• Enrichment of metaphases: 8-hydroxyquinoline.
CHEMICALS
• Enzyme solution: to remove the cell wall components, the plant tissue is
usually incubated in an enzyme solution. Please note that a wide range of
enzyme solutions with plenty variations exist. Depending on the species,
developmental stage and tissue, this solution needs to be adapted to the
tissue at hands. Here, we give one example which shows typical enzyme
components and concentrations, as applied to sugar beet,[13] camellia,[16] and
saffron crocus[8]:
4% (w/v) cellulase ‘Onozuka’ R-10 from Trichoderma viride (Serva 16419)
2% (w/v) cellulase from Aspergillus niger (Sigma C1184)
2% (w/v) cytohelicase from Helix pomatia (Sigma C8274)
OR
2% (w/v) hemicellulase from Aspergillus niger (Sigma H2125)
0.5% (w/v) pectolyase from Aspergillus japonicus (Sigma P3026)
5–20% (w/v or v/v) pectinase from Aspergillus niger (Sigma 17389 or Sigma
P4716)
• Enzyme buffer: 4mM citric acid, 6mM sodium citrate, prepared with dis-
tilled water, pH4.5 adjusted with HCl; if autoclaved, storable for up to
12 months at 4°C.
• Fixative: methanol glacial acetic acid (3:1).
ENZYME TREATMENT
1. Wash fxed leaves with distilled water on a platform shaker at 50 rpm for
5 min.
2. Transfer the leaf onto a petri dish with enzyme buffer and continue shaking
at 50 rpm for 5 min. Replace the enzyme buffer once and repeat the wash-
ing step once.
3. Incubate the leaf in a reaction tube with 100 µl enzyme solution for 3.5–4 h
at 37°C. In many cases, enzyme solution, timeframe and temperature have
to be modifed to meet the optimal balance between digesting the cell wall
and cytoplasm, but keeping the nuclei intact.
4. Macerate tissue with forceps and remove undigested components directly
with the forceps or a micropistil.
5. Pre-pipette macerated tissue up and down with 200 µl pipette. Be careful to
not create any air bubbles.
6. Incubate for another 15–20 min at 37°C and homogenize every 5 min with
a 200 µl pipette.
344 Cytogenetics and Molecular Cytogenetics
• Two shaking water baths; one heated to 37°C and the other to 42°C.
• Thermoblock/thermoshaker.
• Pasteur pipettes: disposable, plastic.
• Incubator heated to 37°C.
• Touchdown in situ system (for the controlled heating and cooling of slides).
• Phase-contrast light microscope.
• Fluorescence microscope.
CHEMICALS
• Enzyme buffer: 4mM citric acid, 6mM sodium citrate, prepared with dis-
tilled water, pH4.5 adjusted with HCl; if autoclaved, storable for up to 12
months at 4°C.
• Fixative: methanol glacial acetic acid (3:1).
• 20×SSC: 3M NaCl, 0.3M sodium citrate, fll up to 1 l with distilled
water
• RNaseA: 0.1 µg/µl in 2×SSC (freshly prepared from a 10 µg/µl stock
solution)
• 0.01M HCl: dilute 0.25M HCl to 0.01M HCl with distilled water
• Pepsin solution: 10 µg/ml in 0.01M HCl (freshly prepare from a 500 µg/ml
stock solution).
• 4% paraformaldehyde (freshly prepared): heat 2.8g paraformaldehyde in
70 ml distilled water to 70°C in a water bath; add 50 µl 1M NaOH until the
solution becomes clear; cool to RT.
• 4×SSC/0.2% Tween 20: use stock solutions of 20×SSC and Tween 20
(100%); fll up with distilled water to 1 l.
• Formamide wash solution (freshly prepared): 20% formamide in
0.2×SSC.
• Blocking solution: use 10×Block from Roche, dilute to 1×Block in
4×SSC/0.2% Tween 20
• Antibody solution (prepare fresh):
• Dilute the anti-Dig solution 1:75.
• Dilute the Streptavidin solution 1:200 in 1× Blocking solution.
• 4′,6-diamidino-2-phenylindole (DAPI) solution: use a DAPI stock solution
(100 µg/ml in ultrapure water); dilute 1:50 in Citifuor (fnal concentration
2 µg/ml).
Pepsin Treatment
3. Pour off 2×SSC and replace with pre-warmed 0.01M HCl, followed by wash-
ing for 1min at 37°C. This is to equilibrate the preparations to an acidic pH.
4. Pipette 200 µl pepsin onto each slide, cover slide with plastic strips, and
place slides into a pre-warmed humid chamber for 15 min at 37°C. This
will serve to further digest the cytoplasm.
5. Carefully transfer the slides to a coplin jar, already flled with 2×SSC at
37°C. Only now, while the slides are submerged, remove plastic strips and
wash for 3×5min at 37°C on a shaking plate.
Fixation of Chromosomes
6. In a coplin jar, incubate the slides in 4% paraformaldehyde solution for 15
min at 37°C without shaking.
7. In a coplin jar, wash preparations in 2×SSC 3×10min at 37°C on a shaking plate.
Hybridization
The hybridization and washing stringencies are infuenced by a range of parameters,
e.g., the base composition of the probe, the formamide and salt concentration as well
as the temperature. Stringency guide values are laid out in Table 28.1. The following
protocol uses our standard procedure, which entails hybridization at approximately
76% and washing at 79% stringency (see Table 28.1).
Antibody Reaction
21. Wash with 4×SSC/0.2% Tween 20 in a shaking water bath for 1×5 min at
37°C (to equilibrate the salt concentration).
22. Pipette 200 µl blocking solution onto each preparation, cover with plastic
strip to block non-specifc binding.
23. Place preparations in a humid chamber for 20 min at 37°C.
24. Carefully remove plastic strips, pipette 50 µl of antibody solution onto
each slide.
25. Cover again and place in a humid chamber for 1 h at 37°C.
26. Remove plastic strips, wash in 4×SSC/0.2% Tween 20 in a shaking
water bath for 3×10 min at 37°C. This serves to wash off the unbound
antibody.
DAPI Staining
27. Pipette 15 µl DAPI solution (in Citifuor) onto the slide and carefully cover
it with a large coverslip (50×24 mm). Try to avoid any air bubbles.
28. Squeeze out any excess liquid with flter paper; the cover slip should lose
the ability to move freely on the slide.
Fluorescence Microscopy
29. Observe the slides under a fuorescence microscope.
348 Cytogenetics and Molecular Cytogenetics
TABLE 28.1
Guide Values for Hybridization and Washing Stringency in Relation to
Temperature, Salt and Formamide Concentration
Formamide Concentration [%]
60 55 50 45 40 35 30 25 20 15 10 5 0
2×SSC 82 79 76 73
b) 70 67 64 61 57 54 51 48 45
Tm at 37 °Ca)
1×SSC 87 84 81 78 75 72 69 66 62 59 56 53 50
Stringency
0.5×SSC 92 89 86 83 80 77 74 71 67 64 61 58 55
0.2×SSC 98 95 92 89 86 83 80 77 74 71 68 65 62
0.1×SSC 103 100 97 94 91 86 85 82 79 74 73 70 67
2×SSC 87 84 81 78 75 72 69 66 62 59 56 53 50
Tm at 42 °C a)
1×SSC 92 89 86 83 80 77 74 71 67 64 61 58 55
Stringency
0.5×SSC 97 94 91 88 85 82 79 76 72 69 66 63 60
0.2×SSC 103 100 97 94 91 88 85 82 79 76 73 70 67
0.1×SSC 108 105 102 99 96 93 90 87 84 81 78 75 72
The exemplary hybridization stringency of Tm = 76% used in this protocol is marked.
a) based on the following equation: Tm = 81.5°C + 16.6logM + 0.41(%G+C)—0.61(% formamide)—
(600/n); where M = sodium concentration [mol/l], n = number of base pairs in smallest duplex
b) standard hybridization parameters in our laboratory
TYPICAL APPLICATIONS
FISH to plant chromosomes enables a range of different applications. Depending
on the question, the respective probes, plant tissues and cell division stages need to
be chosen. In the following, we showcase several examples for FISH applications in
plant chromosomes (Figure 28.2):
(1) One of the earliest FISH applications has been the unequivocal assign-
ment of chromosomes and karyotyping. For this, typically, mitotic chro-
mosomes are prepared and hybridized with a probe cocktail producing
chromosome-specifc signals. These probes can be repeat-derived,[8]
derived from long-insert clones,[14] or from bulked oligonucleotides.[6] As
example, we follow a research question that asks for the genetic origin
of a polyploid plant, namely the triploid saffron crocus. This crop’s trip-
loidization has occurred only once in history, likely in the time of the
Aegean Bronze Age, and has spread since then across the globe as a clonal
line.[23] Nevertheless, the parental species contribution of saffron has been
debated for nearly a century. In this case, repeat-based probes have been
used for comparative FISH between saffron and potential parental wild
crocuses (Figure 28.2A). This FISH-based strategy clearly revealed the
triploid saffron crocus emerged from cytotypes of wild Crocus cart‑
wrightianus[8]; a fnding that was also mirrored through genotyping-by-
sequencing strategies.[24] With increasing sequence information available,
FISH—in Plant Chromosomes 349
FIGURE 28.2 Application examples of FISH onto mitotic and meiotic chromosomes of plants. (A) The
DAPI-stained mitotic chromosomes of triploid saffron are shown in grey. In total, 72 chromosomal
landmarks are produced by hybridization of the four satellite DNAs, CroSat1 (blue), CroSat2 (green),
CroSat3 (light blue), CroSat4 (red), the 18S rRNA gene (yellow) and the 5S rRNA gene (white). This
six-color hybridization of repetitive DNA probes allows the unequivocal identifcation of saffron’s
chromosomes.[8] The scale bar is 10 µm. (B) The DAPI-stained mitotic chromosomes of banana (Musa
acuminata ssp. malaccensis) are shown in blue. This example of chromosome painting targets the chro-
mosomes 6 (red) and 7 (green). For this, bulked oligonucleotide probes were derived from the genome
assembly and hybridized to allow the clear identifcation of chromosome 6– and chromosome 7–derived
regions.[25] The scale bar is 5 µm. (C) The DAPI-stained mitotic chromosomes of poplar (Populus tricho‑
carpa) are shown in blue. To assess the suitability of repeat-derived multilocus markers, it is important
to verify their spread across all chromosomes and chromosome regions. In this example, a probe derived
from the abundant transposable element SaliS-I (red) shows hybridization signals along all poplar chro-
mosomes.[26] The scale bar is 5 µm. (D–H) In this example, the closure of the sugar beet genome assem-
bly has been evaluated by hybridizing a terminal marker (red), the telomere (blue), and a subtelomeric
satellite DNA (yellow) to meiotic chromosome spreads of sugar beet. Based on the distance between
the terminal marker and the distal repeats, the closeness of the terminal marker to the telomere can be
evaluated. (D) Schematic representation of the hybridized probes (red, yellow, blue) onto the ends of a
pachytene chromosome (grey). (E, F) Terminal markers of the chromosome arms 1N (E, arrowhead)
and 9N (G, arrowhead) have been hybridized to pachytene spreads.[14] The close-up of 1N (F) shows the
overlapping of signals, indicating the closeness of the 1N terminal marker to the end of the chromosome.
In contrast, the close-up of 9N shows a large distance between the marker and the telomere, indicating
missing sequence information in the assembly of chromosome 9.
Source: (A) Saffron crocus FISH image reused from [8], Figure 2Q with permission from John Wiley
and Sons; (B) Banana FISH image reused from[25] under CC-BY; (C) Poplar FISH image reused from[26]
under CC-BY; (E–H) Figure 5 reused from[14] with permission from John Wiley and Sons.
CONCLUSIONS
Taken together, for the analysis of plant genome structure and evolution as well as for
plant breeding, FISH is still a powerful technique.
ACKNOWLEDGEMENTS
The protocols laid out in the chapter have been established in the Dresden cytogenet-
ics lab, initially by the late Thomas Schmidt and the late Ines Walter. We remember
their efforts in setting up plant cytogenetics in Dresden and still admire their eye to
detail.
REFERENCES
1. Jiang, J. Fluorescence in situ hybridization in plants: Recent developments and future
applications. Chromosome Res. 2019, 27, 153–165.
2. Heslop-Harrison, J.S.; Schwarzacher, T. Organization of the plant genome in chromo-
somes. Plant J. 2011, 66, 18–33.
3. Naish, M.; Alonge, M.; Wlodzimierz, P.; Tock, A.J.; Abramson, B.W.; Schmücker, A.;
Mandáková, T.; Jamge, B.; Lambing, C.; Kuo, P.; Yelina, N.; Hartwick, N.; Colt, K.;
Smith, L.M.; Ton, J.; Kakutani, T.; Martienssen, R.A.; Schneeberger, K.; Lysak, M.A.;
Berger, F.; Bousios, A.; Michael, T.P.; Schatz, M.C.; Henderson, I.R. The genetic and
epigenetic landscape of the Arabidopsis centromeres. Science. 2021, 374, eabi7489.
4. Perničková, K.; Koláčková, V.; Lukaszewski, A.J.; Fan, C.; Vrána, J.; Duchoslav, M.;
Jenkins, G.; Phillips, D.; Šamajová, O.; Sedlářová, M.; Šamaj, J.; Doležel, J.; Kopecký,
D. Instability of alien chromosome introgressions in wheat associated with improper
positioning in the nucleus. Int. J. Mol. Sci. 2019, 20, 1448.
5. Lysak, M.A. Celebrating Mendel, McClintock, and Darlington: On end-to-end chromo-
some fusions and nested chromosome fusions. Plant Cell. 2022, 34, 2475–2491.
6. Braz, G.T.; He, L.; Zhao, H.; Zhang, T.; Semrau, K.; Rouillard, J.-M.; Torres, G.A.; Jiang,
J. Comparative oligo-FISH mapping: An effcient and powerful methodology to reveal
karyotypic and chromosomal evolution. Genetics. 2018, 208, 513.
FISH—in Plant Chromosomes 351
7. do Vale Martins, L.; Yu, F.; Zhao, H.; Dennison, T.; Lauter, N.; Wang, H.; Deng, Z.;
Thompson, A.; Semrau, K.; Rouillard, J.M.; Birchler, J.A.; Jiang, J. Meiotic crossovers
characterized by haplotype-specifc chromosome painting in maize. Nat. Commun.
2019, 10, 4604.
8. Schmidt, T.; Heitkam, T.; Liedtke, S.; Schubert, V.; Menzel, G. Adding color to a cen-
tury-old enigma: Multi-color chromosome identifcation unravels the autotriploid nature
of saffron (Crocus sativus) as a hybrid of wild Crocus cartwrightianus cytotypes. New
Phytol. 2019, 222, 1965–1980.
9. Beying, N.; Schmidt, C.; Pacher, M.; Houben, A.; Puchta, H. CRISPR-Cas9-mediated
induction of heritable chromosomal translocations in Arabidopsis. Nat. Plants. 2020, 6,
638–645.
10. Yu, W.; Yau, Y.-Y.; Birchler, J.A. Plant artifcial chromosome technology and its poten-
tial application in genetic engineering. Plant Biotechnol. J. 2016, 14, 1175–1182.
11. Schnable, P.S.; Ware, D.; Fulton, R.S.; Stein, J.C.; Wei, F.; Pasternak, S.; Liang, C.;
Zhang, J.; Fulton, L.; Graves, T.A.; Minx, P.; Reily, A.D.; Courtney, L.; Kruchowski,
S.S.; Tomlinson, C.; Strong, C.; Delehaunty, K.; Fronick, C.; Courtney, B.; Rock, S.M.;
Belter, E.; Du, F.; Kim, K.; Abbott, R.M.; Cotton, M.; Levy, A.; Marchetto, P.; Ochoa, K.;
Jackson, S.M.; Gillam, B.; Chen, W.; Yan, L.; Higginbotham, J.; Cardenas, M.; Waligorski,
J.; Applebaum, E.; Phelps, L.; Falcone, J.; Kanchi, K.; Thane, T.; Scimone, A.; Thane, N.;
Henke, J.; Wang, T.; Ruppert, J.; Shah, N.; Rotter, K.; Hodges, J.; Ingenthron, E.; Cordes,
M.; Kohlberg, S.; Sgro, J.; Delgado, B.; Mead, K.; Chinwalla, A.; Leonard, S.; Crouse,
K.; Collura, K.; Kudrna, D.; Currie, J.; He, R.; Angelova, A.; Rajasekar, S.; Mueller,
T.; Lomeli, R.; Scara, G.; Ko, A.; Delaney, K.; Wissotski, M.; Lopez, G.; Campos, D.;
Braidotti, M.; Ashley, E.; Golser, W.; Kim, H.; Lee, S.; Lin, J.; Dujmic, Z.; Kim, W.;
Talag, J.; Zuccolo, A.; Fan, C.; Sebastian, A.; Kramer, M.; Spiegel, L.; Nascimento, L.;
Zutavern, T.; Miller, B.; Ambroise, C.; Muller, S.; Spooner, W.; Narechania, A.; Ren, L.;
Wei, S.; Kumari, S.; Faga, B.; Levy, M.J.; McMahan, L.; Van Buren, P.; Vaughn, M.W.;
Ying, K.; Yeh, C.T.; Emrich, S.J.; Jia, Y.; Kalyanaraman, A.; Hsia, A.P.; Barbazuk, W.B.;
Baucom, R.S.; Brutnell, T.P.; Carpita, N.C.; Chaparro, C.; Chia, J.M.; Deragon, J.M.;
Estill, J.C.; Fu, Y.; Jeddeloh, J.A.; Han, Y.; Lee, H.; Li, P.; Lisch, D.R.; Liu, S.; Liu, Z.;
Nagel, D.H.; McCann, M.C.; SanMiguel, P.; Myers, A.M.; Nettleton, D.; Nguyen, J.;
Penning, B.W.; Ponnala, L.; Schneider, K.L.; Schwartz, D.C.; Sharma, A.; Soderlund,
C.; Springer, N.M.; Sun, Q.; Wang, H.; Waterman, M.; Westerman, R.; Wolfgruber, T.K.;
Yang, L.; Yu, Y.; Zhang, L.; Zhou, S.; Zhu, Q.; Bennetzen, J.L.; Dawe, R.K.; Jiang, J.;
Jiang, N.; Presting, G.G.; Wessler, S.R.; Aluru, S.; Martienssen, R.A.; Clifton, S.W.;
McCombie, W.R.; Wing, R.A.; Wilson, R.K. The B73 maize genome: Complexity, diver-
sity, and dynamics. Science. 2009, 326, 1112–1115.
12. Pellicer, J.; Fay, M.F.; Leitch, I.J. The largest eukaryotic genome of them all? Bot. J.
Linn. Soc. 2010, 164, 10–15.
13. Schmidt, N.; Seibt, K.M.; Weber, B.; Schwarzacher, T.; Schmidt, T.; Heitkam, T. Broken,
silent, and in hiding: Tamed endogenous pararetroviruses escape elimination from the
genome of sugar beet (Beta vulgaris). Ann. Bot. 2021, 128, 281–299.
14. Paesold, S.; Borchardt, D.; Schmidt, T.; Dechyeva, D. A sugar beet (Beta vulgaris L.)
reference FISH karyotype for chromosome and chromosome-arm identifcation, integra-
tion of genetic linkage groups and analysis of major repeat family distribution. Plant J.
2012, 72, 600–611.
15. Anamthawat-Jónsson, K. Preparation of chromosomes from plant leaf meristems for
karyotype analysis and in situ hybridization. Methods Cell Sci. 2004, 25, 91–95.
16. Heitkam, T.; Petrasch, S.; Zakrzewski, F.; Kögler, A.; Wenke, T.; Wanke, S.; Schmidt, T.
Next-generation sequencing reveals differentially amplifed tandem repeats as a major
352 Cytogenetics and Molecular Cytogenetics
CONTENTS
Introduction ............................................................................................................ 353
Samples/Tissues ..................................................................................................... 354
Description of Method ........................................................................................... 354
Yields ..................................................................................................................... 355
Conclusions ............................................................................................................ 355
References .............................................................................................................. 355
INTRODUCTION
Fluorescence in situ hybridization (FISH) is an approach that enables the mapping of
DNA-probes in nuclei and/or on chromosomes. This is an option being not available
until ~50 years ago; still research and diagnostic possibilities of FISH-technique are
still far from being fully utilized.[1] A major shortcut of FISH is that it normally can
only be done on fxed, dead and in most cases heat denatured tissues. In other words,
native chromatin structure is impaired and insights into in vivo situation of a living
and metabolic active cell, is not possible.[2]
However, results similar to those obtained by normal FISH approach can be
achieved on fxed and on living cells by taking advantage of the so-called “gene scis-
sor system” based on CRISPR/Cas9, being originally a defense system of bacterial
cells against foreign viral DNA. CRISPR stands for “Clustered Regularly Interspaced
Short Palindromic Repeats” and Cas9 is a CRISPR-associated gene #9.[3]
The CRISPR/Cas9 system is used for targeted genomic editing, and high suc-
cess rates are reported for such experiments.[3–4] However, far from all high expecta-
tions it was clear from the beginning that even rates of ~99.9% correct targeting of
a gene defect in a patient is not specifc enough to justify a responsible use in gene
therapy.[5] Furthermore, recent research showed that CRISPR/Cas9 is able to induce
chromothripsis, an effect which is defnitely unwanted, deleterious and/or a starting
point of malignifcation of cells.[6]
Still, for the topic of this book, FISH and the new possibilities in connection with
the “gene scissor system” have to be mentioned. First there is (a) FISH-like labeling
of living cells,[7–8] requiring a sophisticated cell-biological equipment and experience,
being not related to what a “standard” molecular cytogenetic laboratory normally
can do. Besides there are yet three other CRISPR/Cas9 system based approaches on
fxed cells. The so-called (b) “Genome Oligopaint via Local Denaturation” (GOLD)
FISH assay can detect single copy sequences using Cas9-RNA and local (relatively
low temperature) denaturation.[9] However, CAS-FISH-,[10] and RNA-guided endo-
nuclease-in situ labeling (RGEN-ISL) (synonym CRISPR-FISH) protocols,[11] allow
detection of repetitive DNA in fxed cells, without denaturation.
Here exclusively the protocol for CRISPR-FISH is treated.[11]
SAMPLES/TISSUES
CRISPR-FISH is done in fxed cells.[11] Here a ribonucleoprotein (RNP) consisting
of a target-specifc CRISPR RNA (crRNA), a fuorescent-labelled transactivating
crRNA (tracrRNA), and Cas9 endonuclease are used. Several fuorochromes can be
applied also in parallel in this approach.[11]
DESCRIPTION OF METHOD
The method of CRISPR-FISH is described acc. to[11].
10. Tip over the slide(s) to remove Buffer 2, add ~30 µl of fuid from step 6,
and cover with coverslip.
11. Incubate at 26°C, acc. to species and DNA-target-sequence, for 2–4 hours
in a humid chamber.
12. Wash slide(s) in 5 min in 1×PBS on ice.
13. Fix slide(s) 5 min in 4% formaldehyde/1×PBS on ice.
14. Wash with 1×PBS (5 min on ice)
15. Dehydrated in an ethanol series (70%, 90%, 96%; 2 min each) at room
temperature.
16. Counterstain with 4′,6-diamino-2-phenylindole (DAPI) in mounting
medium (Vectashield, Vector Laboratories, Burlingame, CA, USA).
17. Evaluate in a suited fuorescence microscope—laser scanning microscopy
is recommended.
YIELDS
Similar results as by standard FISH using repetitive probes (as telomeric or centro-
meric sequences) are possible using CRISPR-FISH. The advantage of CRISPR-FISH
is that tissue is not denatured. Thus, e.g. an in parallel immunostaining can be done
targeting non-degraded proteins.[12]
CONCLUSIONS
Results obtainable by taking advantage of CRISPR/Cas9 “gene scissor system” sys-
tem as discussed in this chapter remember pictures known from standard FISH-
approaches. Still the way how they are obtained is a completely different one. Thus,
the use of the designation CRISPR-FISH is traceable, but may not be the best choice
in terms of potentially misleading molecular cytogenetic labs to make them think
CRISPR-FISH is an easily adaptable alternative for them. The latter seems yet not to
be the case, and CRISPR-FISH is only to be introduced in conditions well suited for
standard FISH with a medium to high investment of time and resources.
REFERENCES
1. Liehr, T. Molecular cytogenetics in the era of chromosomics and cytogenomic
approaches. Front. Genet. 2021, 12, 720507.
2. Potlapalli, B.P.; Schubert, V.; Metje-Sprink, J.; Liehr, T.; Houben, A. Application of Tris-
HCl allows the specifc labeling of regularly prepared chromosomes by CRISPR-FISH.
Cytogenet. Genome Res. 2020, 160, 156–165.
3. Adli, M. The CRISPR tool kit for genome editing and beyond. Nat. Commun. 2018, 9,
911.
4. Li, H.; Yang, Y.; Hong, W.; Huang, M.; Wu, M.; Zhao, X. Applications of genome edit-
ing technology in the targeted therapy of human diseases: Mechanisms, advances and
prospects. Signal Transduct. Target. Ther. 2020, 5, 1.
5. Uddin, F.; Rudin, C.M.; Sen, T. CRISPR gene therapy: Applications, limitations, and
implications for the future. Front. Oncol. 2020, 10, 1387.
356 Cytogenetics and Molecular Cytogenetics
6. Leibowitz, M.L.; Papathanasiou, S.; Doerfer, P.A.; Blaine, L.J.; Sun, L.; Yao. Y.; Zhang,
C.Z.; Weiss, M.J.; Pellman, D. Chromothripsis as an on-target consequence of CRISPR-
Cas9 genome editing. Nat. Genet. 2021, 53, 895–905.
7. Wang, H.; Xu, X.; Nguyen, C.M.; Liu, Y.; Gao, Y.; Lin, X.; Daley, T.; Kipniss, N.H.;
La Russa, M.; Qi, L.S. CRISPR-mediated programmable 3D genome positioning and
nuclear organization. Cell. 2018, 175, 1405–1417.
8. Wang, H.; Nakamura, M.; Abbott, T.R.; Zhao, D.; Luo, K.; Yu, C.; Nguyen, C.M.; Lo, A.;
Daley, T.P.; La Russa, M.; Liu, Y.; Qi, L.S. CRISPR-mediated live imaging of genome
editing and transcription. Science. 2019, 365, 1301–1305.
9. Wang, Y.; Cottle, W.T.; Wang, H.; Feng, X.A.; Mallon, J.; Gavrilov, M.; Bailey, S.; Ha, T.
Genome oligopaint via local denaturation fuorescence in situ hybridization. Mol. Cell.
2021, 81, 1566–1577.
10. Deng, W.; Shi, X.; Tjian, R.; Lionnet, T.; Singer, R.H. CASFISH: CRISPR/Cas9-
mediated in situ labeling of genomic loci in fxed cells. Proc. Natl. Acad. Sci. U. S. A.
2015, 112, 11870–11875.
11. Ishii, T.; Schubert, V.; Khosravi, S.; Dreissig, S.; Metje-Sprink, J.; Sprink, T.; Fuchs, J.;
Meister, A.; Houben, A. RNA-guided endonuclease—in situ labelling (RGEN-ISL): A
fast CRISPR/Cas9-based method to label genomic sequences in various species. New
Phytol. 2019, 222, 1652–1661.
12. Ishii, T.; Kiyotaka, N.; Houben, A. Application of CRISPR/Cas9 to visualize defned
genomic sequences in fxed chromosomes and nuclei. In: Cytogenomics; Liehr, T., Eds.
Academic Press, New York, 2021, pp. 147–153.
Index
Page locators in bold indicate a table. Page locators in italics indicate a fgure.
357
358 Index
H M
hematological neoplasms, 19, 95–96 malignant cells, 73, 77, 213
hematology, 60, 73 maternal
hematopoietic stem cells (HSCs), 75–76 chromosome separation, 122
heparin (anticoagulant), 83, 216 testing, cytogenetic, 17, 53
homologous recombination (HR), 54, 122, 130, mesenchymal stem cells (MSCs), 253
172, 190, 203 MGMT (O6-methylguanine-DNA
Human Genome Project, 49, 74, 157 methyltransferase), 212–213
human papilloma virus (HPV), 243 microdeletion, detection of, 53, 54, 64, 131
microduplication syndromes, 53
I Miller-Dieker lissencephaly syndrome (MDLS),
53
imaging/image analysis, 45–46, 212, 313 mitochondria, 251–254
immuno-FISH, 184, 298–300, 301, 303 molecular combing, 2, 4, 37, 207, 208, 209
immunocytochemistry (ICC), 297–299, 302 momiome (mobile functions of mitochondria
infertility, human sperm and, 14, 55, 111–112, and the mitochondrial genome), 253
124, 128, 135, 148 monoclonal gammopathy of undetermined
insect signifcance (MGUS), 59–60
chromosome painting, 327 mosaicism
chromosome preparation, 323–324 chromosomal, 45
chromosome, FISH protocol, 321 gonadal, 18
sequence mapping, 325–326 somatic, 16, 55
interphase cells, 35, 56, 105, 125, 152, 202 multicolor banding (MCB), 153–154
interphase FISH, 60, 105, 112–114, 181–182 interphase chromosome-specifc, 184
inversion myelodysplastic syndrome (MDS), 18, 20, 58, 61
paracentric, 135, 136, 240, 241 myxoid round cell liposarcoma, 64
pericentric, 14, 136–137, 238, 240
isochromosomes, 124, 128–129, 133–134 N
L O
lampbrush chromosomes (LBC), 314 Okazaki fragments, 190
lateral loops, 308, 313, 314, 315 Online Mendelian Inheritance in Man (OMIM),
methods for obtaining, 309 163
360 Index