Brain Development 2021
Brain Development 2021
Brain
Development
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY™
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Simon G. Sprecher
Department of Biology, University of Fribourg, Fribourg, Switzerland
Editor
Simon G. Sprecher
Department of Biology
University of Fribourg
Fribourg, Switzerland
The brain is without any doubt the most complex organ. Particularly, cellular diversity, con-
nectivity among neurons, formation of neuronal networks, the use of distinct neurotrans-
mitter system, and the underlying function for behavior raise the question of how this
highly interconnected organ develops. It is therefore not surprising that the intersection
between developmental biology and neuroscience provides an exceptional field to address
and investigate impacting biological questions. Complementing findings of an array of dis-
tinct animal model systems provide the basis of brain development research. Our current
understanding is based on widely used genetic model systems including the fruit fly, zebra
fish, chicken, and mouse. These animal models are impacting in particular since they allow
elaborate genetic manipulations including transgenic expression systems and conditional
knockout or knockdown of developmental genes. Genetic developmental studies are fur-
ther complemented by several non-genetic animal models, which further substantiate gen-
eral principles and mechanisms in brain development. Questions that can be investigated
often depend on the methodological accessibility. Therefore, progress and developments in
the constantly improving laboratory technologies provide an essential foundation for the
advancement in the field.
This book aims to provide a broad overview and introduction of widely used leading-
edge techniques in genetic model systems as well as some of the complementing animal
models. The main focus lies on two key technical aspects of developmental neurobiology:
Detection of gene expression and functional characterization of developmental control
genes. The basic principle of expression and function studies are shared between different
model systems. This includes in situ hybridization, reporter gene expression, and immuno-
histochemical staining methods, as well as RNA interference, Morpholino, or transgenic
techniques. However the experimental procedure, such as, for instance, tissue treatment,
fixation, dissection, genetic manipulation, and imaging, often differs substantially between
animal models or even distinct stages in the same species. The collection of protocols aims
to provide precise technical protocols but also allows for comparing a wide range of proto-
cols in different tissues and species.
v
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315
Contributors
IRWIN ANDERMATT • Institute of Molecular Life Sciences and Neuroscience Center Zurich,
University of Zurich, Zurich, Switzerland
JENNIFER E. BESTMAN • The Dorris Neuroscience Center, The Scripps Research Institute,
La Jolla, CA, USA
GEORGE BOYAN • Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-
Universität, Martinsried, Germany
THOMAS G. BRACEWELL • Department of Cell and Developmental Biology, University College
London, London, UK
HOLLIS T. CLINE • The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla,
CA, USA
MICHAEL J. CRAWFORD • Biological Sciences, University of Windsor, Windsor, ON, Canada
DANIELLE C. DIAPER • Department of Neuroscience, MRC Centre for Neurodegeneration
Research, Institute of Psychiatry, King’s College London, London, UK
BORIS EGGER • Zoology Unit, Department of Biology, University of Fribourg, Fribourg,
Switzerland
ADÈLE FAUCHERRE • Département de Physiologie, Institut de Génomique Fonctionnelle,
CNRS UMR 5203, INSERM U661, Universtités Montpellier 1 & 2, Montpellier, France
QIZHI GONG • Department of Cell Biology and Human Anatomy, University of California,
Davis, CA, USA
FRANÇOIS GUILLEMOT • Division of Molecular Neurobiology, MRC National Institute for
Medical Research, London, UK
DAFNI HADJIECONOMOU • Division of Molecular Neurobiology, MRC National Institute for
Medical Research, London, UK
GISELBERT HAUPTMANN • Department of Biosciences and Nutrition, NOVUM, Karolinska
Institutet, Huddinge, Sweden
THOMAS A. HAWKINS • Department of Cell and Developmental Biology, University College
London, London, UK
RONG-QIAO HE • State Key Laboratory of Brain and Cognitive Science, Institute of
Biophysics, Chinese Academy of Sciences, Beijing, China
FRANK HIRTH • Department of Neuroscience, MRC Centre for Neurodegeneration
Research, Institute of Psychiatry, King’s College London, London, UK
LARA HOOKER • Biological Sciences, University of Windsor, Windsor, ON, Canada
DAVID JUSSEN • Institute of Genetics, University of Mainz, Mainz, Germany
KAROLINE F. KRAFT • Institute of Genetics, University of Mainz, Mainz, Germany
CLAUDIUS F. KRATOCHWIL • Friedrich Miescher Institute for Biomedical Research,
Basel, Switzerland
GILBERT LAUTER • Department of Biosciences and Nutrition, NOVUM, Karolinska
Institutet, Huddinge, Sweden
ZI-LONG LI • State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics,
Chinese Academy of Sciences, Beijing, China
ix
x Contributors
AIMIN LIU • Department of Biology, Eberly College of Science, Center for Cellular Dynamics,
Huck Institute of Life Sciences, The Penn State University, University Park, PA, USA
JINLING LIU • Department of Biology, Eberly College of Science, Center for Cellular Dynamics,
Huck Institute of Life Sciences, The Penn State University, University Park, PA, USA
KAI-LI LIU • State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese
Academy of Sciences, Beijing, China
YING LIU • State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese
Academy of Sciences, Beijing, China
YU LIU • Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-Universität,
Martinsried, Germany
HERNÁN LÓPEZ-SCHIER • Unit of Sensory Biology & Organogenesis, Helmholtz Zentrum
München, Munich, Germany
EMILIE PACARY • Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale,
U862, Institut National de la Santé et de la Recherche Médicale, Université de Bordeaux,
Bordeaux, France
BENJAMIN PERRUCHOUD • Zoology Unit, Department of Biology, University of Fribourg,
Fribourg, Switzerland
FILIPPO M. RIJLI • Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
SAQIB S. SACHANI • Biological Sciences, University of Windsor, Windsor, ON, Canada
IRIS SALECKER • Division of Molecular Neurobiology, MRC National Institute for Medical
Research, London, UK
NANA SHIMOSAKO • Division of Molecular Neurobiology, MRC National Institute for Medical
Research, London, UK
CRISTINE SMOCZER • Biological Sciences, University of Windsor, Windsor, ON, Canada
IRIS SÖLL • Department of Biosciences and Nutrition, NOVUM, Karolinska Institutet,
Huddinge, Sweden
CLAUDIO D. STERN • Department of Cell & Developmental Biology, University College London,
London, UK
ESTHER T. STOECKLI • Institute of Molecular Life Sciences and Neuroscience Center Zurich,
University of Zurich, Zurich, Switzerland
ANDREA STREIT • Department of Craniofacial Development & Stem Cell Biology, King’s College
London, London, UK
RICHARD P. TUCKER • Department of Cell Biology and Human Anatomy, University of
California, Davis, CA, USA
KATHERINE J. TURNER • Department of Cell and Developmental Biology, University College
London, London, UK
ROLF URBACH • Institute of Genetics, University of Mainz, Mainz, Germany
GUDRUN VIKTORIN • Biozentrum, University of Basel, Basel, Switzerland
XIU-MEI WANG • State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics,
Chinese Academy of Sciences, Beijing, China
ANDREAS WANNINGER • Faculty of Life Sciences, Department of Integrative Zoology, University
of Vienna, Vienna, Austria
TIM WOLLESEN • Department of Integrative Zoology, Faculty of Life Sciences, University of
Vienna, Vienna, Austria
Part I
Drosophila Protocols
Chapter 1
Abstract
Immunostaining is used to visualize the spatiotemporal expression pattern of developmental control genes
that regulate the genesis and specification of the embryonic and larval brain of Drosophila. Immunostaining
uses specific antibodies to mark expressed proteins and allows their localization to be traced throughout
development. This method reveals insights into gene regulation, cell-type specification, neuron and glial
differentiation, and posttranslational protein modifications underlying the patterning and specification of
the maturing brain. Depending on the targeted protein, it is possible to visualize a multitude of regions of
the Drosophila brain, such as small groups of neurons or glia, defined subcomponents of the brain’s axon
scaffold, or pre- and postsynaptic structures of neurons. Thus, antibody probes that recognize defined
tissues, cells, or subcellular structures like axons or synaptic terminals can be used as markers to identify
and analyze phenotypes in mutant embryos and larvae. Several antibodies, combined with different labels,
can be used concurrently to examine protein co-localization. This protocol spans over 3–4 days.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_1, © Springer Science+Business Media, LLC 2014
3
4 Danielle C. Diaper and Frank Hirth
Fig. 1 The embryonic brain of Drosophila. (a–d) Laser confocal microscopy, superimposition of optical sections
of stage 14 embryo immunolabeled with anti-HRP-FITC. (a) Frontal view of brain; (b) ventral view of ventral
nerve cord; (c) lateral view of brain; (d) optical section along the midline of (c); (e) cartoon summarizing major
neural segments and axon scaffolding which together constitute the embryonic Drosophila brain. Abbreviations:
sec supraesophageal commissure, fc frontal connective, cnn circumesophageal connective, sco subesophageal
commissure, an antennal nerve, ac anterior commissure, pc posterior commissure, pcn protocerebral con-
nective, lc longitudinal connective, PC protocerebrum, DC deutocerebrum, TC tritocerebrum, MD mandibular
neuromere, MX maxillary neuromere, LB labral neuromere, es esophagus. Scale bar: 25 μm
gene of interest has been cloned and antibodies raised against its
encoded protein product(s), immunostaining can be used to
visualize the spatiotemporal pattern of protein expression during
embryonic and larval brain development. Second, antibody probes
that recognize defined tissues, cells, or subcellular structures like
axons or synaptic terminals can be used as markers to identify and
analyze phenotypes in mutant embryos and larvae. Thus, following
protein localization throughout Drosophila development reveals
insights into gene regulation, cell-type specification, neuron and
glial differentiation, and posttranslational protein modifications
underlying the patterning, specification, and neuronal connectivity
of the maturing brain.
In this respect, whole-mount immunohistochemistry (IHC),
which is the process of using antibody probes to detect antigens
(i.e., proteins), is among the most valuable of tools for analysis.
It is a relatively cheap and reliable way to visualize discrete struc-
tures or cell types within the developing embryonic and larval brain
of Drosophila. With the use of multiple antibodies, or when used in
conjunction with genetic labeling, such as mosaic analysis with a
repressible cell marker (MARCM) [5], it is possible to follow
several different proteins, examining co-localization, or identify
specific cells and follow their neuronal projections from the cell
body to its terminal dendritic arborizations [6–8]. Immunostaining
of Drosophila brain tissue is often used to help deconstruct the
complexities of neural circuit development or neurodegeneration
[9, 10], in many cases with the aim of understanding human disease
pathogenesis [11, 12]. The availability of Drosophila antibodies to
human homologues can pave the way for follow-up studies in
mammals or human tissue [13, 14].
There is a growing supply of Drosophila-specific antibodies
available that bind to all manner of cellular proteins, from organ-
elle components to synaptic vesicle markers [15]. Immunostaining
can be done in basically two ways, which are defined by way of
visualization method. One option visualizes proteins/antigens
with secondary antibodies that carry labels, which, upon enzymatic
reactions, lead to precipitates that are visible under the light micro-
scope. Typical examples are alkaline phosphatase or horseradish
peroxidase reactions that lead to brown, black, or purple precipi-
tates (for details, see ref. 16). These histochemical staining methods
have obvious limitations: enzymatic reactions do not penetrate
well into tissue, and resulting precipitates do not allow 3D recon-
structions unless the tissue is embedded in plastic, microdissected,
and subsequently scanned, which is laborious and time-consuming.
The advent of laser confocal and super-resolution microscopy
together with computer algorithms nowadays allows rapid and
reproducible 3D reconstructions of optical sections derived from
fluorescence immunostaining, which has thus become the method
of choice. It is based on secondary antibodies that are conjugated
6 Danielle C. Diaper and Frank Hirth
1.1.1 Alter the Fixation Formaldehyde is suitable for deep penetration of the tissue; it
Technique forms strong cross-links between proteins and is suitable for long-
term storage [19] and preserving chromosome morphology [20].
You may want to consider replacing the paraformaldehyde with
freshly prepared formaldehyde solution. Glutaraldehyde may also
be used [21, 22]; however, as it is a larger molecule, it does not
diffuse as well through deep tissue. Its cross-links span a larger
distance, so it can stably fix proteins that are further apart.
Glutaraldehyde fixation is not ideal for immunohistochemistry as
further treatment of the tissue is necessary to avoid aldehyde
Immunostaining of Embryonic and Larval Drosophila Brain 7
1.1.2 Antibody Too high or low concentrations of antibody may cause either
Optimization antibody aggregation on the surface of the tissue or poor penetra-
tion of the tissue [26]; therefore, carrying out a dilution series will
help to determine the most effective titer. To improve the signal-
to-noise ratio, try using affinity-purified antibodies or altering the
temperature, e.g., room temperature instead of 4 °C, or length of
antibody incubation, e.g., 3 h instead of overnight [22].
2 Materials
3 Methods
3.1 Embryo Fixation 1. Recover plates from egg collection and remove any remaining
yeast or dead flies with a spatula or brush being careful not to
damage the agar (see Note 5).
2. Dechorionate the embryos by adding 50 % hypochlorite and
agitate for 2–5 min until dechorionated embryos float to the
surface (see Note 6).
3. Pour the embryos and hypochlorite through the nylon mesh
and rinse thoroughly with dH2O (see Note 7) (see Fig. 2).
4. Transfer embryos to a 2 ml tube containing 1 ml heptane and
1 ml PEM-FA (see Note 8).
5. Agitate on a rotator at high speed for 10–30 min (no longer
than 30 min).
6. Allow embryos to settle—they should be at the interface
between the two phases.
7. Remove the lower phase, then the upper phase (see Note 9).
Fig. 2 Embryo collection apparatus. An open-bottom container (a) has a hole cut
in the lid (b); the hole is closed by a fine mesh (c), which is included into the lid
when screwed on top of the container. A 50 ml plastic container can also be
adapted in this way
10 Danielle C. Diaper and Frank Hirth
3.2 Embryo 1. Transfer embryos into a 1.5 ml tube (see Note 12).
Immunostaining 2. Rehydrate the embryos by removing the methanol and washing
2 × 5 min, then 1 × 30 min with PBT 0.1 %.
3. Incubate embryos for 30 min in 5 % PBT-NGS.
4. Remove PBT-NGS and add appropriate amount of primary
antibody diluted in 5 % PBT-NGS to a reaction volume of
100 μl (see Note 13). Incubate overnight at 4 °C.
5. Remove antibody solution and wash 1 × 1 min, 3 × 5 min, and
4 × 30 min with PBT 0.1 % (see Note 11).
6. Incubate embryos for 30 min in 5 % PBT-NGS.
7. Remove PBT-NGS and add appropriate amount of secondary
antibody diluted in 5 % PBT-NGS to a reaction volume of
100 μl. Incubate at 4 °C over night in the dark (see Note 14).
8. Remove antibody solution and wash 3 × 5 min and 4 × 30 min
with PBT 0.1 % (see Note 11).
9. Add 1 drop of VECTASHIELD mounting medium and incu-
bate at 4 °C over night in the dark.
10. Mount on glass slide with 22 × 50 mm cover slip (see Note 15).
11. Your samples are now ready for image acquisition (see Note 16).
3.3 Larval 1. Collect larvae and place into a watch glass containing cold PBS.
Immunostaining 2. Fill a 0.5 ml tube with cold PBS and keep on ice with the
lid open.
3. Roughly remove the larval CNS (see Note 17), placing dissected
brains into the PBS-containing 0.5 ml tube.
4. After 30 min of dissection, remove PBS and add 500 μl of PLP.
Agitate on a rotator for 1 h at room temperature.
5. Remove PLP and wash 3 × 10 min in PBT (see Note 18).
6. Block by incubating the brains in 5 % PBT-NGS (for L1 and L2)
or 10 % PBT-NGS (for L3) on the rotator for 15 min.
7. Remove PBT-NGS and add appropriate amount of primary
antibody diluted in 5 or 10 % PBT-NGS to a reaction volume
Immunostaining of Embryonic and Larval Drosophila Brain 11
Fig. 3 Immunostaining of whole mount embryonic CNS and 3rd instar larval CNS. Top row: The embryonic
nervous system is staining with horseradish peroxidase Cy3 (HRP-cy3; red) and visualized under a standard
fluorescence microscope. The anterior brain is visible (arrow) as well as the ladderlike axon tracts of the
ventral nerve cord (arrowhead); compare to Fig. 1. The nuclei of all embryonic cells are visualized with DAPI.
Merge shows co-labeling of HRP-cy3 and DAPI. Bottom row: 3rd instar larval CNS immunostained with anti-
Bruchpilot counterlabeled with goat anti-mouse 568, and nuclei are highlighted with DAPI; merge shows co-
labeling of nc82 and DAPI. The nc82 antibody recognizes the Bruchpilot protein which is specifically enriched
in the active zone of synapses
4 Notes
11. For each wash, remove the old solution and add the same
amount of fresh solution. Washes should be agitated on a rotator
at full speed.
12. Pipette as fast as you can; as soon as the liquid stops being
turbulent, the embryos tend to stick to the sides of the pipette
and are very difficult to remove. This means you should have
your two tubes ready, with lids open and suck up the liquid and
embryos and transfer as quickly as possible.
13. Depending on what you are attempting to visualize, you can
add several primary antibodies at once. Take care to avoid using
similar species that may cross-react, for example, if you use a
mouse and rat primary antibody, the secondary anti-mouse will
bind to the rat antibody and the secondary anti-rat will bind to
the mouse antibody. If you are visualizing an endogenous GFP
signal (e.g., using the GAL4>UAS system), you may get a better
signal by also using an antibody against GFP and a secondary
with a 488 (green) fluorophore. Some primary antibodies are
already conjugated to a fluorophore and do not need a secondary
antibody, such as horseradish peroxidase Cy3 (HRP-cy3), which
specifically labels the Drosophila nervous system.
14. You will need a secondary antibody that targets the animal
species in which the primary antibody was raised. You can use
up to four separate channnels by picking fluorophores that are
unlikely to interfere with each other, e.g., DAPI, UV; 488,
green; 568, red; and 647, far red. Of course, the fluorophore
you use will depend on what filters, lamps, or lasers are avail-
able for image capture.
15. Suck up VECTASHIELD and embryos and pipette out in a
wide zigzag line on the labeled microscope slide (see Fig. 7a).
Roll 4 small balls of modeling clay and place in the corners
where the cover slip will go. Lower one short end of the cover
slip in place and gently lower the cover slip, avoiding air bub-
bles. Gently press cover slip at the 4 corners.
16. To avoid bleaching the brains, first use a fluorescent microscope
to select your best brains or embryos. When imaging using a
laser scanning microscope, scan each channel sequentially to
avoid interference between fluorophores. If you have issues with
the noise-to-signal ratio, check that you are using the antibody
at the appropriate dilution, ensure you are completing all wash
steps, and check the pH of the buffer solutions.
17. This bit takes practice! There are several ways to pull the CNS
from the larvae, but the thing that will help the most is being
able to differentiate the CNS from the imaginal discs and fat
bodies (see Figs. 4 and 5). I find it easier to gently clamp the
body of the larvae about two thirds the way towards the mouth
hooks; then when the larvae extends its mouth hooks, grab
14 Danielle C. Diaper and Frank Hirth
Fig. 4 Cartoon illustrating a rough larval CNS dissection. (1) First grab the
extended mouth hooks and make a gentle tear in the cuticle. (2) Rip the larvae in
half. (3) Holding the mouth hooks again, slowly pull off the remaining cuticle until
(4) you are left with the larval brain still attached to the head, imaginal discs, and
fat bodies
Fig. 5 Identifying the 3rd instar larval brain and CNS. (a) Several imaginal discs
(filled arrow), fat bodies (arrowhead), and digestive components (empty arrow)
will be attached to the 3rd instar larval brain and CNS. Identifying the brain
among these other tissues will aid successful dissections. The larval brain and
CNS are outlined with a black line. (b) DAPI labeled 3rd instar larval brain and
CNS visualized under the fluorescence microscope after successful dissection;
note that wing and leg imaginal discs are still attached
them with the forceps (see Fig. 4). Gently pull the mouth hooks
until the cuticle rips and the internal matter is exposed.
The round optic lobes of the CNS may be visible. Place both
forceps on the body and rip the lower half of the larvae off.
Grab the mouth hooks and body again and gently pull until
the mouth hooks and CNS come away from the rest of the
larvae. Holding onto the mouth hooks, transfer to the 0.5 ml
tube that is sat on ice. If you do not have much luck this way,
or find that the ventral nerve cord is lost using this method,
you can also pinch both forceps just below the mouth hooks
(by the scruff of its neck!) and rip the cuticle open and then bit by
bit remove the lower part of the larvae, the digestive tract, fat,
and some imaginal discs.
Immunostaining of Embryonic and Larval Drosophila Brain 15
Fig. 6 Cartoon illustrating fine dissection of the larval brain and CNS. (1) Using
the dissecting pins like a knife and fork, remove the mouth hooks from the brain
and then (2) gently remove all other tissues taking care (3) not to damage or
puncture the brain
18. When washing it is best to leave brains to settle for 15 s and then
pipette off the solution. Eject the pipetted solution into a clean
watch glass, check under the microscope, and rescue any brains
by pipetting them back into the tube. If carrying out several
dissections, you can leave samples washing for an extended
period of time until all preparations have reached the same stage.
19. As the secondary antibodies are photosensitive, you should
keep your preparation in the dark as much as possible to avoid
bleaching the fluorophores. Cover the tubes with tinfoil or
repurpose a small container to hold your tubes and attach to
the rotator using Velcro strips.
20. Cut around 2 cm off the end of a P200 pipette tip and transfer
brains and VECTASHIELD to a microscope slide. Label the
microscope slide with a dissection code (for cross-reference
with your records, e.g., Initials_01), genotype, the primary
antibody and secondary fluorophore used, and the date.
Remove the majority of the VECTASHIELD, so that the
brains are no longer floating, and pipette back into your dissec-
tion tube. Use the pin holder tools to remove unwanted imagi-
nal discs, fat bodies, and mouth hooks. You can use them like a
knife and fork to slice away tissue, or pin an imaginal disc into
place with one needle and cut the connecting tissue with the
other (see Fig. 6). Take care not to pin or damage the CNS itself.
21. Arrange brains in concentric circles with the ventral nerve cord
pointing outwards (see Fig. 7b). Orientate the brains in a mix-
ture of dorsal or ventral side up. Add a small amount of
VECTASHIELD in a circle around the brains—not too much
or they will float out of position. Once the cover slip is in place,
gently press on the four corners to flatten the brains slightly, and
be careful as they are easily ruptured by squashing. Add more
VECTASHIELD if necessary by pipetting small amounts at the
edge of the cover slip. Remove excess VECTASHIELD with
cotton buds or tissues dipped in ethanol—again, be very careful
not to dislodge the cover slip as this will destroy your brains!
16 Danielle C. Diaper and Frank Hirth
Acknowledgements
References
1. Skeath JB, Thor S (2003) Genetic control of development in Drosophila. Bioessays 26:
Drosophila nerve cord development. Curr Opin 739–751
Neurobiol 13:8–15 4. Hirth F, Reichert H (1999) Conserved genetic
2. Kim DW, Hirth F (2009) Genetic mechanisms programs in insect and mammalian brain devel-
regulating stem cell self-renewal and differen- opment. Bioessays 21:677–684
tiation in the central nervous system of 5. Lee T, Luo L (2001) Mosaic analysis with a
Drosophila. Cell Adh Migr 3:402–411 repressible cell marker (MARCM) for Drosophila
3. Urbach R, Technau GM (2004) Neuroblast neural development. Trends Neurosci 24:
formation and patterning during early brain 251–254
Immunostaining of Embryonic and Larval Drosophila Brain 17
6. Grueber WB, Ye B, Yang CH, Younger S, New York, NY, For an amended and updated
Borden K, Jan LY, Jan YN (2007) Projections version, follow the link: http://patelweb.
of Drosophila multidendritic neurons in the berkeley.edu/Images/Protocols/pdf%20files/
central nervous system: links with peripheral Antibody%20Methods%202006.pdf
dendrite morphology. Development 134: 17. Ashburner M (1989) Drosophila: a laboratory
55–64 manual. Cold Spring Harbor Laboratory Press,
7. Selcho M, Pauls D, Han KA, Stocker RF, New York, NY
Thum AS (2009) The role of dopamine in 18. Hoffman, G. (2008) Seeing is believing: Use
Drosophila larval classical olfactory condition- of antibodies in immunohistochemistry and in
ing. PLoS One 126:e5897 situ hybridization. In: Short course II of SfN’s
8. White KE, Humphrey DM, Hirth F (2010) 38 annual meeting: 15–19 November 2008;
The dopaminergic system in the aging brain of Washington, DC. Society for Neuroscience
Drosophila. Front Neurosci 4:205 19. Rothwell WF, Sullivan W (2000) Fluorescent
9. Muqit MK, Feany MB (2002) Modelling neu- analysis of Drosophila embryos. In: Sullivan W,
rodegenerative diseases in Drosophila: a fruitful Ashburner M, Hawley RS (eds) Drosophila
approach? Nat Revs Neurosci 3:237–243 protocols. Cold Spring Harbor Laboratory
10. Koizumi K, Higashida H, Yoo S et al (2007) Press, New York, NY, p 141
RNA interference screen to identify genes 20. Bonaccorsi S, Giansanti MG, Cenci G, Gatti M
required for Drosophila embryonic nervous (2012) Formaldehyde fixation of Drosophila
system development. Proc Natl Acad Sci USA testes. Cold Spring Harb Protoc. doi:10.1101
104:5626–5631 21. Heimbeck G, Bugnon V, Gendre N, Häberlin
11. Stochmanski SJ, Therrien M, Laganière J et al C, Stocker RF (1999) Smell and taste percep-
(2012) Expanded ATXN3 frameshifting events tion in Drosophila melanogaster larva: toxin
are toxic in Drosophila and mammalian neuron expression studies in chemosensory neurons.
models. Hum Mol Genet 21:2211–2218 J Neurosci 19:6599–6609
12. Dunlop J, Morin X, Corominas M et al (2004) 22. Stocker RF, Heimbeck G, Gendre N, de Belle
glaikit is essential for the formation of epithelial JS (1997) Neuroblast ablation in Drosophila
polarity and neuronal development. Curr Biol P[GAL4] lines reveals origins of olfactory
14:2039–2045 interneurons. J Neurobiol 32:443–456
13. Tsuji T, Higashida C, Yoshida Y et al (2011) 23. Hassell J, Hand AR (1974) Tissue fixation with
Ect2, an ortholog of Drosophila’s pebble, nega- diimidoesters as an alternative to aldehydes. I.
tively regulates neurite outgrowth in neuroblas- Comparison of cross-linking and ultrastructure
toma × glioma hybrid NG108-15 cells. Cell Mol obtained with dimethylsuberimidate and glu-
Neurobiol 31:663–668 taraldehyde. J Histochem Cytochem 22:
14. Pandey UB, Nichols CD (2011) Human disease 223–229
models in Drosophila melanogaster and the 24. Wieschaus E, Nüsslein-Volhard C (1998)
role of the fly in therapeutic drug discovery. Looking at embryos. In: Roberts DB (ed)
Pharmacol Rev 63:411–436 Drosophila, a practical approach. Oxford
15. Developmental Studies Hybridoma Bank University Press Inc, New York, NY, p 205
developed under the auspices of the NICHD 25. Ripper D, Schwarz H, Stierhof YD (2008)
and maintained by The University of Iowa, Cryo-section immunolabelling of difficult to
Department of Biology, Iowa City, IA 52242 preserve specimens: advantages of cryofixation,
http://dshb.biology.uiowa.edu/ freeze-substitution and rehydration. Biol Cell
16. Patel N (1994) Imaging neuronal subsets and 100:109–123
other cell types in whole mount Drosophila 26. Rebay I, Fehon R (2000) Generating antibod-
embryos and larvae using antibody probes. In: ies against Drosophila proteins. In: Sullivan W,
Goldstein LSB, Fryberg E (eds) Methods in Ashburner M, Hawley RS (eds) Drosophila
Cell Biology, Vol 44. Drosophila melanogaster: protocols. Cold Spring Harbor Laboratory
Practical Uses in Cell Biology. Academic, Press, New York, NY, p 400
Chapter 2
Abstract
In Drosophila, the brain arises from about 100 neural stem cells (called neuroblasts) per hemisphere which
originate from the neuroectoderm. Products of developmental control genes are expressed in spatially
restricted domains in the neuroectoderm and provide positional cues that determine the formation and
identity of neuroblasts. Here, we present a protocol for non-fluorescent double in situ hybridization com-
bined with antibody staining which allows the simultaneous representation of gene expression patterns in
Drosophila embryos in up to three different colors. Such visible multiple stainings are especially useful to
analyze the expression and regulatory interactions of developmental control genes during early embryonic
brain development. We also provide protocols for whole mount and flat preparations of Drosophila embryos,
which allow a more detailed analysis of gene expression patterns in relation to the cellular context of the early
brain (and facilitate the identification of individual brain neuroblasts) using conventional light microscopy.
Key words Drosophila, Embryonic brain, Neuroectoderm, Neuroblast identification, In situ hybrid-
ization, Antibody staining
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_2, © Springer Science+Business Media, LLC 2014
19
20 David Jussen and Rolf Urbach
2 Materials
2.6 Equipment 1. Cold light source (halogen or LED) equipped with fiber light
for Microscopy guides (e.g., KL1500 by Schott).
2. Digital microscope camera (e.g., ProgRes® series, Jenoptik).
3. Upright light microscope (equipped with differential interfer-
ence contrast and 40×–100× objectives, e.g., Axioscope Zeiss).
4. Stereo microscope (e.g., MZ series by Leica Microsystems).
3 Methods
3.1 Embryo 1. Place flies on apple juice agar for egg laying (see Note 1).
Collection and 2. Dechorionize embryos after egg laying by covering the apple
Dechorionization juice agar with chlorine bleach (6 %) for 3 min. Slightly rotate
once in a while. Dechorionized embryos will float up.
3. Collect embryos by transferring the chlorine bleach into an
egg basket (see Note 2).
4. Wash embryos with water.
24 David Jussen and Rolf Urbach
3.2 Fixation 1. Open the egg basket. Take embryos from the nylon mesh with
a small scalpel and transfer them into an Eppendorf tube with
fixative.
2. Fix embryos by vigorously shaking for 20 min (900 rpm).
3. Let the two phases of the fixative separate.
4. Remove the lower phase of the fixative without removing too
many embryos.
5. Add 500 μl methanol.
6. Remove the vitelline membrane by vortexing at maximum
speed for 2 min.
7. Let the devitellinized embryos sink down and remove lower
phase of the solution.
8. Add 500 μl methanol.
9. Vortex for 1 min.
10. Let the embryos sink down and remove as much of the solu-
tion as possible without removing too many embryos.
11. Rinse 4× with methanol.
● Embryos may be stored in methanol at −20 °C (see Note 3).
3.3 (Double) In Situ Use gloves, filtered tips, and DEPC-treated PBTween for the
Hybridization following in situ hybridization procedure:
1. Rinse 5× with PBTween.
2. Incubate 5 min in PBTween/hybridization buffer (1:1).
3. Incubate 5 min in hybridization buffer.
4. Perform prehybridization by incubating 1 h in hybridization
buffer + ssDNA (1:100) at 55 °C upon shaking (300 rpm).
5. Perform hybridization by incubating in hybridization buffer +
ssDNA (1:100) containing your probe (both of your probes
for double in situ hybridization) at the appropriate working
dilution(s) (see Note 4).
6. Incubate overnight at 55 °C upon shaking (300 rpm).
7. Incubate 30 min in hybridization buffer at 65 °C upon shaking
(350 rpm).
8. Incubate 30 min in PBTween/hybridization buffer (1:1)
upon shaking (350 rpm).
9. Wash 4 × 20 min with PBTween at 65 °C upon shaking
(350 rpm).
Standard tips and untreated PBTween may be used from
now on.
10. Wash 10 min with PBTween at RT.
Non-fluorescent Multiple Stainings in the Embryonic Brain of Drosophila 25
3.11 Manufacturing Two different kinds of needles are needed for the preparation of
of Preparation Needles embryos. One will be used for handling the embryos (i.e., moving in
glycerol, transferring onto microscope slide, holding during prepara-
tion; termed “handling needle”), while the other is used for cutting
the tissue during preparation (termed “cutting needle”) (Fig. 1a).
For that, the pinpoint of both needles will be processed differently.
Non-fluorescent Multiple Stainings in the Embryonic Brain of Drosophila 27
Fig. 1 Filet preparation of Drosophila embryos. (a) The cutting needle has a flattened tip with sharp edges,
while the tip of the handling needle is rounded. (b) Main steps performing filet preparation of embryos at dif-
ferent stages. The posterior part of the abdomen is removed by cutting the embryo transversally. Then, the
remaining ectoderm is cut along the dorsal midline towards the anterior tip of the embryo to open the head
capsule. Finally, the ectoderm has to be flattened. (c–e) Instructions for flat preparations. Black and white dots
indicate corresponding positions of the head capsule. Arrowheads indicate how to move the upper (small)
cover slip. Arrows indicate the flow of glycerol caused by moving the cover slip. (c) An embryo with opened
head capsule is positioned in glycerol between a large (24 × 60 mm) and a small (18 × 18 mm) cover slip
(separated by aluminum spacers) with anterior facing forwards and ventral facing down. (d) The small cover
slip is carefully moved in posterior direction (regarding the body axis of the embryo), which causes the hemi-
spheres to erect. (e) The small cover slip is carefully moved back until the hemispheres are unfurled flatly on
the cover slip. a anterior, p posterior, d dorsal, vML ventral midline, pNE procephalic neuroectoderm
28 David Jussen and Rolf Urbach
3.13 Whole Mount 1. Fix two cover slips (22 × 22 mm) side by side on a microscope
Preparation of slide with a drop of nail polish. The distance between both
Embryos should be approx. 15 mm.
2. Transfer selected embryos between the cover slips with a
pipette.
3. Place another cover slip (22 × 22 mm) on the embryos. Make
sure that it stays on top of both spacing cover slips.
4. Examine under a microscope (see Note 16; Fig. 2a, d)
5. Seal for documentation and storage by applying nail polish to
the edges of the cover slip
3.14 Filet The following filet preparation is recommended for embryos until
Preparation of Early to developmental stage 12 and is performed under a stereo micro-
Mid-Stage Embryos scope (see Note 17).
1. Fill the pit of a concavity slide with 70 % glycerol.
2. Transfer the selected embryo into the pit with the handling
needle.
3. Place the embryo on the lateral side and hold it carefully with
the handling needle.
4. Remove the posterior part of the abdomen and cut along the
dorsal midline from posterior to anterior with a cutting needle
to open the head capsule (Fig. 1b).
5. Spread the hemispheres slightly with both needles or a Dumont
forceps.
Non-fluorescent Multiple Stainings in the Embryonic Brain of Drosophila 29
Fig. 2 Non-fluorescent multiple stainings of the head neuroectoderm and brain neuroblasts in whole mounts
and flat preparations. (a) Ventral view on the head region of a stage 8 whole mount embryo (anterior is up)
stained for Nkx6-mRNA (via NBT/BCIP, blue) and single-minded protein (via DAB, brown). (b) Schematic repre-
sentation of the details presented in C/D′. (c) View on the head region (left hemisphere) of a filet prepared stage
6 embryo with almost all neuroectodermal cells in one plane: blue is ind-mRNA (via NBT/BCIP), magenta is
msh-mRNA (via Vector Red) and brown is Vnd-Protein (via DAB). (D/D′) Early brain neuroblast pattern of a late
stage 9 embryo as whole mount (d) and after filet preparation of the same embryo (D′), stained against Engrailed
(via NBT/BCIP, blue) and svp-LacZ (via DAB, brown). Note that filet preparation allows a more clear view, and
thus the identification of individual neuroblasts by marker expression and relative position (as illustrated in D″)
according to [7, 8]. a anterior, p posterior, v ventral, d dorsal, CF cephalic furrow, vML ventral midline, hs
engrailed head spot, as engrailed antennal stripe, is engrailed intercalary stripe. D-D″ Adapted from [7]
11. Carefully pull the small cover slip (e.g., with a forceps) back-
wards (i.e., towards yourself). This will cause the hemispheres
to erect (Fig. 1d).
12. Push the small cover slip forwards (i.e., away from yourself).
By that, glycerol will float towards the erected hemispheres
causing them to unfurl flatly on the cover slip (Fig. 1e; see
Note 18).
13. When the embryo is appropriately flattened, fix the small cover
slip by applying a drop of nail polish to every corner. Let the
nail polish dry, remove excess glycerol (e.g., with small stripes
of tissue paper), and seal the cover slip by applying nail polish
to the edges (two times).
14. Clean the cover slip from glycerol and fix it on a microscope
slide with small stickers.
15. Under a microscope, the filet preparation can be examined
from both sides (with a view from ventral or dorsal) by turning
the cover slides including the filet preparation (Fig. 2c, D′;
see Note 19).
3.16 Analysis of the The development of brain neuroblasts occurs in a stereotypic spa-
Developing Pattern of tiotemporal pattern, with every neuroblast expressing a unique
Brain Neuroblasts combination of marker genes. Both have been described in detail
for Drosophila [7, 8], which allows the identification of individual
brain neuroblasts. The brain neuroblast pattern is best examined in
filet preparation of embryos from late stage 8 to late stage 11 using
100× magnification and differential interference contrast. We rec-
ommend (combinatorial) antibody stainings for distinct molecular
markers (particularly against Deadpan, svp-LacZ, and Engrailed
protein) to analyze the patterns of brain neuroblasts in more detail.
Deadpan (Dpn) is a general neuroblast marker. In early develop-
mental stages (i.e., late stage 8–stage 9), Dpn antibody may even
be sufficient to identify single neuroblasts according to their posi-
tion within the entire brain neuroblast pattern [7]. From stage 9
onwards, identification of single brain neuroblasts becomes increas-
ingly difficult with general markers. We recommend the use of svp-
LacZ enhancer trap line (available at Bloomington Stock Center,
Bloomington, Indiana, USA). The expression pattern of svp-LacZ
lines is stable, well described, and persists throughout embryonic
brain development. svp-LacZ marks a subset of (about 40) brain
neuroblasts in a characteristic pattern, which covers all three main
subdivisions of the brain (i.e., proto-, deuto-, and tritocerebrum).
By that, it can be used to identify the svp-LacZ-labeled neuroblasts
within the entire pattern of brain neuroblasts but also the adjacent
(unlabeled) ones (Fig. 2D-D″) according to ref. 7. In any case, it is
helpful to combine stainings with Engrailed, which is segmentally
expressed in the posterior part of each subdivision of the brain
neuroectoderm (engrailed head spot—protocerebrum, engrailed
antennal stripe—deutocerebrum, engrailed intercalary stripe—
tritocerebrum) and in distinct subsets of brain neuroblasts that
emerge from the corresponding Engrailed neuroectodermal
domains (Fig. 2D-D″). These Engrailed-positive brain neuroblasts
additionally serve as reference points for orientation within the
Dpn- or svp-LacZ-labeled neuroblast patterns [7, 8]. The (combi-
natorial) use of the markers mentioned above may in most cases be
sufficient to identify single brain neuroblasts. However, further
markers are listed in [8].
4 Notes
Acknowledgments
The authors thank Janina Seibert and Dagmar Volland for sharing
protocols and considerable expertise, Janina Seibert for Fig. 2c,
and Karoline F. Kraft for critically reading the manuscript. This
work was supported by grants from the Deutsche
Forschungsgemeinschaft (UR163/2-1 and UR163/3-1).
References
1. Kosman D, Mizutani CM, Lemons D, Cox brain and ventral nerve cord. Dev Biol 346:
WG, McGinnis W, Bier E (2004) Multiplex 332–345
Detection of RNA Expression in Drosophila 7. Urbach R, Schnabel R, Technau GM (2003)
Embryos. Science 305:846 The pattern of neuroblast formation, mitotic
2. Skeath JB, Thor S (2003) Genetic control of domains and proneural gene expression during
Drosophila nerve cord development. Curr early brain development in Drosophila.
Opin Neurobiol 13:8–15 Development 103:3589–3606
3. Urbach R, Technau GM (2004) Neuroblast 8. Urbach R, Technau GM (2003) Molecular
formation and patterning during early brain markers for identified neuroblasts in the devel-
development in Drosophila. Bioessays 26: oping brain of Drosophila. Development 103:
739–751 3621–3637
4. Urbach R, Volland D, Seibert J, Technau GM 9. Campos-Ortega J, Hartenstein V (1997) The
(2006) Segment-specific requirements for dor- embryonic development of Drosophila mela-
soventral patterning genes during early brain nogaster. Springer, Berlin Heidelberg
development in Drosophila. Development 10. Rothwell WF, Sullivan W (2000) Fluorescent
133:4315–4330 analysis of Drosophila embryos. In: Sullivan
5. Seibert J, Volland D, Urbach R (2009) Ems W, Ashburner M, Hawley RS (eds) Drosophila
and Nkx6 are central regulators in dorsoven- protocols, 1st edn. CSHL Press, Cold Spring
tral patterning of the Drosophila brain. Harbor, New York, pp 143–145
Development 136:3937–3947 11. Hauptmann G (2001) One-, two-, and
6. Seibert J, Urbach R (2010) Role of en and three-color whole-mount in situ hybridiza-
novel interactions between msh, ind, and vnd tion to Drosophila embryos. Methods
in dorsoventral patterning of the Drosophila 23:359–372
Chapter 3
Abstract
Proper functioning of the brain relies on an enormous diversity of neural cells generated by neural stem
cell-like neuroblasts (NBs). Each of the about 100 NBs in each side of brain generates a nearly invariant
and unique cell lineage, consisting of specific neural cell types that develop in defined time periods. In this
chapter we describe a method that labels entire NB lineages in the embryonic brain. Clonal DiI labeling
allows us to follow the development of a NB lineage starting from the neuroectodermal precursor cell up
to the fully developed cell clone in the first larval instar brain. We also show how to ablate individual cells
within a NB clone, which reveals information about the temporal succession in which daughter cells are
generated. Finally, we describe how to combine clonal DiI labeling with fluorescent antibody staining that
permits relating protein expression to individual cells within a labeled NB lineage. These protocols make
it feasible to uncover precise lineage relationships between a brain NB and its daughter cells, and to assign
gene expression to individual clonal cells. Such lineage-based information is a critical key for understanding
the cellular and molecular mechanisms that underlie specification of cell fates in spatial and temporal
dimension in the embryonic brain.
Key words Drosophila, Embryonic brain, Neural stem cell, Neuroblast lineage, DiI labeling, Antibody
staining
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_3, © Springer Science+Business Media, LLC 2014
37
38 Karoline F. Kraft and Rolf Urbach
2 Materials
10. Fixatives: For antibody staining after DiI labeling, mix 200 μl
37 % formaldehyde in 900 μl 1× PBS. For photoconversion,
mix 260 μl 37 % formaldehyde in 900 μl 1× PBS.
11. Glycerol 70 %: Dissolve 70 ml 100 % glycerol in 30 ml 1× PBS.
12. Fluorocarbon oil Voltalef 10S.
13. Fetal calf serum.
2.2 Equipment 1. Inverse microscope (e.g., Leitz Fluovert FU) equipped with
different fluorescent filters (Fluorescein FITC, Rhodamin, etc.).
2. 100 W halogen lamp.
3. 100 W mercury lamp.
4. Stereomicroscope.
5. Cold light source.
6. Capillary grinder (e.g., Bachofer Type 462).
7. Capillary puller (e.g., Sutter P97).
8. Micromanipulator (e.g., Leitz M).
9. Bunsen burner.
10. Cover slips (22 × 22 mm, 24 × 60 mm).
11. Glass slide (76 × 26 × 1 mm).
12. Plasticine.
13. Borosilicate glass microcapillaries (Hilgenberg normal
“Meterware”).
14. Glass cutter.
15. Wet chamber (e.g., Petri dish containing wet pieces of filter
paper).
16. Petri dish (60 × 20 mm).
17. Scalpel.
18. Preparation needle.
19. Fine forceps (e.g., Dumont 5SF).
20. Black block dish.
21. Single-use syringe (5 or 10 ml).
22. Polyethylene tube (6 × 4 × 1 mm).
23. Transparent nail polish.
24. Tape (single sided, double sided).
25. Dissecting knife.
26. Cage (60 × 70 mm).
27. Weighing tray (41 × 41 × 8 mm).
DiI-labeling in the Drosophila Embryonic Brain 41
3 Methods
3.3 Preparation 1. Take a piece of polyethylene tube (ca. 8 cm) and heat it briefly
of Polyethylene Tube over a small flame of a Bunsen burner (until the polyethylene
starts “bubbling”).
2. Pull apart slowly from both ends until the tube reaches a
length of about 50 cm.
3. Cut one end of the tube at appropriate position so that the
inner diameter fits the capillary (Fig. 1c).
3.4 Manufacturing 1. Capillaries most suitable for DiI labeling have the following
Labeling Capillaries characteristics: (a) a relatively long shank and (b) a relatively
small tip diameter. The outer diameter at the distal tip of the
capillary should be around 3–5 μm (Fig. 1d) (see Note 1).
2. Grind the capillary on a commercial capillary grinder in an
angle of ca. 30°; the resulting capillary will be sharp enough to
easily penetrate the vitelline membrane. For best results, the
capillary should be wet-ground. In the following, this capillary
type will be called “labeling capillary,” whereas a pulled but
unground capillary, needed for transfer and preparation of
embryos, will be called “flat capillary.”
42 Karoline F. Kraft and Rolf Urbach
Fig. 1 Set up for DiI labeling. (a) Gastrula-stage embryo (at stage 7) glued to the
cover slip for DiI labeling in the brain neuroectoderm. cf cephalic furrow; dML
dorsal midline. (b) Schematic cross section through the head region boxed in
(a); the embryo is fixed on a cover slip in appropriate orientation to label cells in
the ventral or intermediate neuroectoderm (NE) under an inverse microscope.
vML ventral midline. Modified from [29]. (c) Micromanipulator equipped with a
labeling capillary coupled with a polyethylene tube which is connected to a
syringe. (d) The angle of the bevel of the labeling capillary should be around 30°,
and the outer diameter at the distalmost tip between 3 and 5 μm. (e) Labeling of
two neighboring neuroectodermal cells with DiI (magenta) and DiD (green).
Dashed line indicates the adhesive border (color figure online)
3.6 Egg Collection 1. At least 2 days before you start collecting embryos for DiI
and Staging labelings: Place the flies (between 4 and 14 days old) in a fly
cage on an apple juice plate with yeast. Change the plate every
24 h (see Note 4).
2. For DiI labeling, precisely staged embryos within a range of
1 h can be obtained when changing the plate every hour at
25 °C.
3.7 Dechorionization 1. Fix a piece of double-sided tape to a cover slip (22 × 22 mm)
and Mounting (Fig. 2b).
of Embryos for 2. Take cooled, fresh apple juice agar plates, and cut the agar into
DiI Labeling blocks of 2 × 2 cm. Place two blocks of agar in a distance of
about 1.5 cm on a glass slide (76 × 26 mm), and the cover slip
on top, leaving a part of each agar block exposed (Fig. 2b1).
3. Make 3 lines of 5–7 holes each, by pressing a preparation nee-
dle into the agar blocks at an angle of about 20° (Fig. 2b2).
4. Transfer and scatter a few embryos onto the tape with a dis-
secting knife (Fig. 2b3).
5. Wait about 3 min to let the chorion dry.
6. Collect eggs at the blastoderm stage and dechorionate by
rotating the embryo slightly along the dorsoventral axis with
the tip of a preparation needle. The chorion breaks and releases
the embryo (see Note 5).
7. Carefully pick up each dechorionated embryo and transfer it
to one of the agar blocks to prevent further drying (Fig. 2b4).
44 Karoline F. Kraft and Rolf Urbach
Fig. 2 Preparation of embryos for DiI labeling. (a) Spread a drop of heptane glue on
a cover slip. Let the heptane glue dry for at least 10 min. Stick a single-sided tape
frame on the glue-coated cover slip. (b) (1) Fix a piece of double-sided tape on a
cover slip. Place the cover slip on top of two blocks of agar. (2) Make holes into the
agar. (3) Transfer embryos onto the tape. Dechorionate embryos. (4) Transfer each
embryo on an agar block and orientate them. (5) Cut up the agar blocks so that
every embryo is on an individual block. (6) Check for optimal orientation. (7) Fix the
embryo on the prepared cover slip. (8) When embryos are properly dried, cover
them with Voltalef 10S oil. a anterior, p posterior, d dorsal, v ventral
3.8 Filling the 1. Put the unbeveled end of the capillary (as indicated by the pen
Capillary with Dye mark, see Subheading 3.4) into the polyethylene tube, and a
Solution single-use syringe on the other end of the tube.
2. Fix the capillary with the topside up (as indicated by the pen
mark) in the holder of a micromanipulator (Fig. 1c).
3. Place a drop of 1 μl dye/oil on a cover slip (24 × 60 mm) and
bring both the dye drop and the capillary tip under the micro-
scope in focus using a 10× objective.
4. Now change to a 50× objective, and slowly draw up the dye
into the capillary.
5. As soon as the tip is filled with dye, the syringe has to be
removed from the polyethylene tube, the tip of the capillary to
be lifted up, and the syringe, with the plunger pulled out, to
be inserted again into the tube.
6. To reuse the drop of dye later, you can store the cover slip in
a dark, dry, and dust-free box.
3.9 Labeling 1. Place the cover slip with the embryos under the microscope.
of Individual 2. If the embryos are properly desiccated, there should be a rela-
Neuroectodermal Cells tively sharp adhesive border around the area where each
and Determination embryo is stacked to the glue and several rows of neuroecto-
of Their Position dermal cells should lie in one focal plane (Fig. 1b).
46 Karoline F. Kraft and Rolf Urbach
3. To identify the optimal time point for labeling and the posi-
tion of the cell to be labeled, it is useful to have at least a 600×
or higher magnification (e.g., using a 50× immersion oil objec-
tive and a 12.5× ocular magnification). Optionally, the label-
ing can be controlled on a monitor using a standard digital
camera (CCD/CMOS).
4. Check under the microscope the developmental stage of the
embryos. By stage 7 (staging according to [30]) you can
detect, for example, the dorsal and ventral midline, the cephalic
furrow (see Note 9). The latest time point for successful label-
ing seems to be around stage 9.
5. Determine the neuroectodermal cell to be labeled by making
use of the above mentioned, morphological landmarks that
help to recognize the position of individual cells (see Note 9).
6. Via the micromanipulator, lower the tip of the capillary until it
is in focus, but still in a certain distance to the embryo. From
now on, do not work with the micromanipulator anymore, as
all movements should be effected via the microscope table.
7. Penetrate the vitelline membrane slowly and carefully with the
tip of the capillary in the position where the neuroectodermal
cell of interest is located, and approach the tip as closely as
possible towards its cell membrane (see Note 10).
8. By slowly pushing the plunger of the syringe, deposit a small
drop of dye next to this cell. The diameter of the applied drop
is critical for successful labeling: about 1/2–1/5 of the diam-
eter of a neuroectodermal cell seems to be most promising
(Fig. 1e). Immediately after depositing the drop, the dye dif-
fuses into the cell membrane leaving behind the nonfluores-
cent drop of oil solvent.
9. Slowly, pull the capillary out of the embryo.
10. For double labeling in the same embryo, place two capillaries
filled with different dyes (each on a different micromanipula-
tor) as close as possible to the embryo before labeling. If no
additional micromanipulator is available, you can also quickly
exchange the two capillaries between the labeling.
11. After labeling, briefly inspect the quality of the labeling under
fluorescent light using a halogen lamp (see Note 11).
12. Finally, control the position of the labeled cell via DIC optics.
13. Take notes of each labeling concerning position and behavior
of the labeled cell (see Subheading 3.10).
14. Store the labeled embryos on the cover slip in a wet chamber
at 18–20 °C.
DiI-labeling in the Drosophila Embryonic Brain 47
3.10 Control of Cell 1. About 1 h after labeling (around late stage 8/early stage 9),
Behavior After judge the behavior of the labeled cell using the same fluores-
Labeling cence and magnification settings (see Note 12).
2. If the neuroectodermal cell has delaminated, the NB was suc-
cessfully labeled. To follow the development of the NB lineage
in more detail, it is possible to inspect the labeling in shorter
time intervals (e.g., every 30 min) without disturbing the
labeled cells.
3. The identification of a labeled NB clone is facilitated when
labeling is done in embryos of appropriate Gal4 strains (most
can be ordered in the Bloomington or Kyoto Stock Center) in
which the Gal4 pattern is visualized via a GFP reporter.
Alternatively, you can combine your labeling with an antibody
staining (see Subheading 3.15).
4. For further development, the embryos are kept under fluoro-
carbon oil Voltalef 10S at 18 °C until they reach the desired
stage. To obtain fully developed stage 17 NB clones, embryos
are kept at 18 °C overnight.
5. Before further preparation, monitor the embryos and eliminate
all those which do not show properly labeled cell clones.
3.11 Preparation 1. For better handling, we recommend removing the parts of the
of Embryos at cover slip outside of the single-sided tape frame using a glass
Developmental Stage cutter.
17 (See Fig. 3) 2. Tip the cover slip at an ~80° angle for several minutes so that
the fluorocarbon oil can drain off.
3. Then, place the cover slip in a weighing tray (41 × 41 × 8 mm)
and spread about 1 ml heptane on the leftover fluorocarbon
oil in order to remove it.
4. Rock the cover slip slowly for about 10–20 s. Do not rock too
long since the heptane will also solve the glue and the embryos
will detach.
5. Quickly place the cover slip on a glass slide for better
handling.
6. Pour away the remaining heptane carefully. Wait a few seconds
until the heptane is almost completely evaporated and cover
the embryos with PBS immediately to prevent drying.
7. Get the embryos, one by one, out of the vitelline membrane
using a flat capillary.
8. Dissect the CNS out of the embryo by placing one fine forceps
at the anterior end of the embryo and another one in its
middle/posterior part. Carefully lacerate the embryo to make
the CNS visible. Remove all remaining tissue from the CNS.
48 Karoline F. Kraft and Rolf Urbach
Fig. 3 Different applications of clonal DiI labeling. (a) Combined confocal images
of a DiI labeled mushroom body NB clone (a) in a Ok107 (ey)-Gal4; UAS-CD8::GFP
brain (a′) at mid/late stage 17. Note that some cells of the DiI labeled clone
express GFP, indicating that this is a mushroom body NB clone. (b) DiI labeled
mushroom body NB clone at late stage 17 after ablation of the NB at stage 11.
Accordingly, only the early-born part of its lineage has developed. (c) 3D recon-
struction of a DiI labeled mushroom body NB clone (magenta) in the early first
larval instar in comparison to the structures of the entire mushroom body (green)
as revealed by Ok107 (ey)-Gal4; UAS-CD8::GFP expression. (d) A mushroom
body NB clone (magenta) and another protocerebral NB clone (green) at late
stage 17 obtained after labeling of two neighboring cells with DiI (magenta) and
DiD (green) (see inset). A–D Adapted from [28]. (e) Photoconverted protocerebral
NB clone at early stage 17. (f) DiI labeled protocerebral NB clone (at early first
larval instar) before (f) and after (f′) fluorescent antibody staining against the
gene products Eyes absent (f″a) and Retinal homeobox (f″b). Note that antibody
staining leads to partial loss of DiI signal. (f″) Merge
3.12 Preparation After about 30 h at 18 °C, embryos are checked for eclosion under
of First Instar Larvae a stereomicroscope in intervals of about 15 min. Take notes on
which larva hatches at which position on the cover slip, as this
information is necessary to make correlations to the notes you took
during the labeling (see Note 13).
1. Immediately after eclosion, take the larvae out of the oil by
using a flat capillary. Transfer each larva separately to a glue-
coated cover slip prepared with a tape frame (as explained in
Subheading 3.5) filled in with PBS.
2. To prepare the CNS, tear the larva in half with a pair of fine
forceps. Then turn the cuticle “inside out” and the CNS will
appear attached to the cuticle. Remove the CNS entirely from
the tissue, transfer it with a capillary to another glue-coated
cover slip prepared with a tape frame (as explained in
Subheading 3.5) filled with PBS, and stick the ventral nerve
cord with its ventral side on the glue.
3. The preparation can be stored in a wet chamber at 4 °C.
If you want to photoconvert the clone continue as described
in Subheading 3.17, otherwise go on with fixation
(see Subheading 3.13).
3.13 Fixation 1. Remove the PBS under visual control using a stereomicro-
scope, and replace it immediately with fixation solution
(see Subheading 2.1). Note, as fixatives are toxic, take precautions
(e.g., place the stereomicroscope on a portable fume hood).
2. Fix 20 min for subsequent antibody staining (as described in
Subheading 3.15), otherwise 10 min.
3. Thereafter, carefully replace the fixative with PBS.
4. Change the PBS 4–5 times.
3.14 Documentation 1. Fix the cover slip (24 × 60 mm) carrying the preparation on a
glass slide with tape for better handling.
2. Remove excessive PBS so that the border of the liquid slightly
overtops the single-sided tape frame (see Note 14).
3. Now carefully place a cover slip (22 × 22 mm) onto the single-
sided tape frame.
4. By slightly moving the upper cover slip, the brain hemispheres
that get in contact with this cover slip (as explained in
Subheadings 3.11 and 3.12) can be brought into the best pos-
sible position for exposure.
5. Fix the cover slip to the underlying cover slip by applying nail
polish at the corners.
50 Karoline F. Kraft and Rolf Urbach
3.15 Optional: As far it was only possible to combine photoconverted DiI labelings
DiI Labeling Combined with nonfluorescent histochemical antibody stainings (visualized,
with Fluorescent e.g., via the alkaline phosphatase reaction) with the disadvantage
Antibody Stainings that the photoconverted cells themselves, which have accumulated
strong levels of diaminobenzidine (DAB), could not be antibody
stained. Now this limitation can be avoided by combining labeling,
performed with the improved fixable DiI derivatives (e.g., CM-DiI),
with fluorescent antibody stainings. To carry out fluorescent anti-
body stainings with fluorescent DiI labeled clones, another critical
issue is the proper usage of detergents, needed for antibodies to
penetrate the membranes of cell tissues. As the carbocyanine dyes
are lipophilic, conventional detergents in widely used concentra-
tions (such as 0.1–0.3 % Triton X-100 or 0.05 % saponin) result in
extensive diffusion of the DiI label out of the tissue (own observa-
tions; see also ref. [31]). Presented below is a protocol in which we
apply a low concentration of 0.1 % Tween20, and in addition,
mechanically perforate the neurilemma that envelops the brain.
1. After fixation, to facilitate the penetration of the antibodies
into the brain tissue, carefully perforate the neurilemma with a
flat capillary. Perforate generously but do not damage the area
containing the labeled clones.
2. Thereafter, cover the specimen for 48 h with primary anti-
body/antibodies dissolved in 0.1 % PBTween (~200 μl). Put
the cover slip in a wet closed chamber at 4 °C.
3. Then take off the primary antibody/antibodies and replace
with 0.1 % PBTween twice.
4. Cover three times for 20 min with 0.1 % PBTween.
5. Dissolve the secondary antibody/antibodies (take in consider-
ation that you use appropriate fluorescent conjugates which
do not interfere with applicated carbocyanine dyes) in 0.1 %
PBTween and incubate for 18–20 h at 4 °C. In case of weak
antibody stainings, a Tyramide Signal Amplification System
(TSA) is recommended (according to the manufacturers’ pro-
tocol of PerkinElmer, Waltham, USA).
6. After taking off the secondary antibody/antibodies, replace
with 0.1 % PBTween twice and then cover for 15 min with
0.1 % PBTween twice.
7. Subsequently, wash twice with PBS.
8. Finally, put a cover slip on the tape frame, fix with nail polish,
and follow up with documentation immediately as the DiI sig-
nal fades relatively fast (see Subheading 3.14).
DiI-labeling in the Drosophila Embryonic Brain 51
3.16 Optional: 1. For cell ablations, use embryos that have been labeled as
Ablation of DiI Labeled described in Subheading 3.9 and have developed into the
NBs and/or Daughter desired stage. Use a microscope equipped with a 100 W halo-
Cells gen lamp and appropriate filter settings. Bring the specimen in
which cell ablations will be done under a sufficient magnifica-
tion (600× or higher) in focus. Connect the ablation capillary
(see Subheading 3.4) with the tube, then clamp the capillary
into the micromanipulator and lower it into the Voltalef oil.
Finally, connect the syringe to the other end of the tube to
prevent Voltalef oil entering the tip.
2. Identify the labeled cell(s) to be ablated under the halogen
lamp.
3. Using the micromanipulator, move the capillary towards the
embryo under transmitted light.
4. Slowly and carefully move the cross table of the microscope
towards the capillary tip until the capillary penetrates the vitel-
line membrane, and then carefully direct it to the cell(s) desig-
nated for ablation.
5. Once the tip of the capillary reaches the cell to be ablated, cau-
tiously pull the syringe plunger in order to suck off the cell(s)
(see Note 16).
6. Remove the capillary from the embryo by moving the cross
table.
7. Store the treated embryos in a wet chamber at 18 °C until
they have reached the desired developmental stage.
8. Then prepare, fix, and document the specimen as described in
Subheadings 3.11–3.14.
3.17 Optional: Photoconversion transforms the DiI label into a permanent reac-
Photoconversion tion product via the Maranto reaction in which illumination
of DiI Labeled releases a singlet oxygen that oxidizes DAB [32] (see also ref. [33]).
Preparations DiI labeled photoconverted clones can be investigated using con-
ventional light microscopy and preserved for many years.
1. Incubate for 1 h in calf serum.
2. Exchange the drop of PBS with a drop of DAB solution (that
has been briefly centrifuged before). Note, DAB is a hazard-
ous chemical—handle with appropriate precautions!
3. Photoconvert the labeled NB clone under the microscope
with a 100 W mercury lamp and rhodamine filter using a water
or oil immersion objective with about 50× magnification.
Photoconversion is completed when all fluorescent signal is
gone, this takes usually 10–20 min. Check regularly to make
sure that the preparation is not over dyed.
4. After photoconversion, remove the DAB.
5. Rinse the preparation with PBS 3–4 times.
52 Karoline F. Kraft and Rolf Urbach
4 Notes
15. As DiI signal may become weaker over time, it is often useful
to document a labeled NB clone before and after the antibody
staining. Because of the relatively long procedure, you risk to
lose the signal.
16. For cell ablations, halogen lamps with rhodamine filters and
transmitted light at once are recommended. In order to recog-
nize the DiI fluorescent signal of the cell(s) to be ablated, and
simultaneously, the surrounding tissue, reduce the intensity of
the transmitted light to an appropriate low level.
Acknowledgments
References
1. Technau GM, Berger C, Urbach R (2006) 9. Jefferis GS, Marin EC, Stocker RF et al (2001)
Generation of cell diversity and segmental pat- Target neuron prespecification in the olfactory
tern in the embryonic central nervous system map of Drosophila. Nature 414:204–208
of Drosophila. Dev Dyn 235:861–869 10. Lai S-L, Awasaki T, Ito K et al (2008)
2. Skeath JB (1999) At the nexus between pat- Clonal analysis of Drosophila antennal lobe
tern formation and cell-type specification: the neurons: diverse neuronal architectures in the
generation of individual neuroblast fates in the lateral neuroblast lineage. Development 135:
Drosophila embryonic central nervous system. 2883–2893
Bioessays 21:922–931 11. Pearson BJ, Doe CQ (2004) Specification of
3. Urbach R, Technau GM (2004) Neuroblast for- temporal identity in the developing nervous
mation and patterning during early brain devel- system. Annu Rev Cell Dev Biol 20:619–647
opment in Drosophila. Bioessays 26:739–751 12. Yu H-H, Lee T (2007) Neuronal temporal
4. Jacob J, Maurange C, Gould AP (2008) identity in post-embryonic Drosophila brain.
Temporal control of neuronal diversity: com- Trends Neurosci 30:520–526
mon regulatory principles in insects and verte- 13. Yu H-H, Kao C-F, He Y et al (2010) A com-
brates? Development 135:3481–3489 plete developmental sequence of a Drosophila
5. Kao C-F, Lee T (2010) Birth time/order- neuronal lineage as revealed by twin-spot
dependent neuron type specification. Curr MARCM. PLoS Biol 8
Opin Neurobiol 20:14–21 14. Golic KG, Lindquist S (1989) The FLP recom-
6. Lin S, Lee T (2012) Generating neuronal binase of yeast catalyzes site-specific recombi-
diversity in the Drosophila central nervous nation in the Drosophila genome. Cell 59:
system. Dev Dyn 241:57–68 499–509
7. Skeath JB, Thor S (2003) Genetic control of 15. Xu T, Rubin GM (1993) Analysis of genetic
Drosophila nerve cord development. Curr mosaics in developing and adult Drosophila
Opin Neurobiol 13:8–15 tissues. Development 117:1223–1237
8. Baumgardt M, Karlsson D, Terriente J et al 16. Evans CJ, Olson JM, Ngo KT et al (2009)
(2009) Neuronal subtype specification within G-TRACE: rapid Gal4-based cell lineage
a lineage by opposing temporal feed-forward analysis in Drosophila. Nat Methods 6:
loops. Cell 139:969–982 603–605
56 Karoline F. Kraft and Rolf Urbach
17. Lai S-L, Lee T (2006) Genetic mosaic with of Drosophila melanogaster. II. Neuroblast
dual binary transcriptional systems in lineages derived from the dorsal part of the
Drosophila. Nat Neurosci 9:703–709 neuroectoderm. Dev Biol 189:186–204
18. Lee T, Luo L (1999) Mosaic analysis with a 27. Schmid A, Chiba A, Doe CQ (1999) Clonal
repressible cell marker for studies of gene func- analysis of Drosophila embryonic neuroblasts:
tion in neuronal morphogenesis. Neuron neural cell types, axon projections and muscle
22:451–461 targets. Development 126:4653–4689
19. Lee T (2009) New genetic tools for cell lin- 28. Kunz T, Kraft KF, Technau GM, Urbach R
eage analysis in Drosophila. Nat Methods 6: (2012) Origin of Drosophila mushroom
566–568 body neuroblasts and generation of divergent
20. Yu HH, Chen CH, Shi L et al (2009) Twin- embryonic lineages. Development 139:
spot MARCM to reveal the developmental ori- 2510–2522
gin and identity of neurons. Nat Neurosci 29. Bossing T, Technau GM (1994) The fate of
12:947–953 the CNS midline progenitors in Drosophila as
21. Larsen C, Shy D, Spindler SR et al (2009) revealed by a new method for single cell label-
Patterns of growth, axonal extension and axo- ling. Development 120:1895–1906
nal arborization of neuronal lineages in the 30. Campos-Ortega JA, Hartenstein V (1997)
developing Drosophila brain. Dev Biol The embryonic development of Drosophila
335:289–304 melanogaster. Springer, Berlin
22. Luo L (2005) A practical guide: single-neuron 31. Elberger AJ, Honig MG (1990) Double-
labeling using genetic methods. In: Yuste R, labeling of tissue containing the carbocyanine
Konnerth A (eds) Imaging in neuroscience dye DiI for immunocytochemistry. J Histochem
and development. A laboratory manual. Cold Cytochem 38:735–739
Spring Harbor Laboratory, New York, NY, 32. Maranto AR (1982) Neuronal mapping: a
pp 99–110 photooxidation reaction makes Lucifer yellow
23. Honig MG, Hume RI (1986) Fluorescent car- useful for electron microscopy. Science
bocyanine dyes allow living neurons of identi- 217:953–955
fied origin to be studied in long-term cultures. 33. von Bartheld CS, Cunningham DE, Rubel EW
J Cell Biol 103:171–187 (1990) Neuronal tracing with DiI: decalcifica-
24. Honig MG, Hume RI (1989) Dil and DiO: ver- tion, cryosectioning, and photoconversion
satile fluorescent dyes for neuronal labelling and for light and electron microscopic analysis.
pathway tracing. Trends Neurosci 12:333–340 J Histochem Cytochem 38:725–733
25. Bossing T, Udolph G, Doe CQ et al (1996) 34. Foe VE (1989) Mitotic domains reveal early
The embryonic central nervous system lin- commitment of cells in Drosophila embryos.
eages of Drosophila melanogaster. I. Development 107:1–22
Neuroblast lineages derived from the ventral 35. Urbach R, Schnabel R, Technau GM (2003)
half of the neuroectoderm. Dev Biol 179: The pattern of neuroblast formation, mitotic
41–64 domains and proneural gene expression during
26. Schmidt H, Rickert C, Bossing T et al (1997) early brain development in Drosophila.
The embryonic central nervous system lineages Development 130:3589–3606
Chapter 4
Abstract
Visualization of single neurons within their complex environment is a pivotal step towards uncovering the
mechanisms that control neural circuit development and function. This chapter provides detailed technical
information on how to use Drosophila variants of the mouse Brainbow-2 system, called Flybow, for sto-
chastic labeling of cells with different fluorescent proteins in one sample. We first describe the genetic
strategies and the heat shock regime required for induction of recombination events. This is followed by a
detailed protocol as to how to prepare samples for imaging. Finally, we provide specifications to facilitate
multichannel image acquisition using confocal microscopy.
Key words Drosophila, Brainbow, Multicolor cell labeling, Genetics, Immunostaining, Confocal laser
scanning microscopy
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_4, © Springer Science+Business Media, LLC 2014
57
58 Nana Shimosako et al.
Fig. 1 Schematic representation of Flybow transgenes and color outcomes. Expression of the modified FLP
recombinase mFLP5 induces inversions and excisions of cassettes each flanked by inward-facing mFRT71
recognition sites (black triangles). Cassettes consist of two FP cDNAs with opposite orientations. In FB2.0, an
additional FLP-out cassette flanked by canonical FRT sites facing in the same orientation (grey triangles) pre-
cedes the FP-containing cassettes. (a) A detailed description of FB1.0, FB1.1, and FB2.0 transgenes is provided
in [3]. (b) In FB1.0B, FB1.1B, and FB2.0B transgenes, Cerulean-V5 has been replaced by mTurquoise (mTq)
Fig. 2 Expression of B set of FB transgenes in the adult Drosophila optic lobe. Examples for original transgenes
can be found in [3]. Photoreceptor cells (R1–R8) visualized with mAb24B10 (blue) extend axons into the optic
lobe: R1–R6 axons innervate the lamina; R8 and R7 axons terminate in the medulla [15]. NP4151-Gal4, an
enhancer trap insertion into the Netrin B locus [16, 17], drives expression of FB transgenes in lamina neurons
L3 and medulla neuron subtypes, which extend axons into the lobula and lobula plate. (a, a′) Heat shock appli-
cation induces expression of mFLP5 to trigger inversion events in FB1.0B260b (n = 14), labeling individual neu-
rons with mTurquoise instead of mCherry. (b, b′) In FB1.1B260b (n = 19), excision and inversion events lead to
expression of mCitrine, mCherry, or mTurquoise instead of the default marker EGFP. (c, c′) FB2.0B260b (n = 29)
facilitates sparse labeling of single neurons in a population because it relies on heat shock-induced expression
of FLP for excision of the stop cassette flanked by FRT sites, and of mFLP5 for stochastic expression of the four
fluorescent proteins. Confocal images represent single optic sections. Images in b, b′, c, and c′ were pro-
cessed using channel separation software. Heat shock exposure: 3 × 45 min (a–b′) and 3 × 30 min (c, c′) at
48, 72, and 96 h after puparium formation. Scale bars: 20 μm
Table 1
Basic Flybow transgene toolkit
in the nervous system and other tissues [3]. While Flybow also can
be applied to studies in embryos, in this chapter, we focus on its
use in the larval, pupal, and adult nervous system.
2 Materials
3 Methods
3.1 Genetic Crosses 1. Build and expand the driver stock, which contains both an hs-
and Clone Induction mFLP5 and a Gal4 transgene active in a cell type, brain area,
Protocol or other tissue of interest (see Note 6).
Flybow Approach 63
Fig. 3 Example of experimental strategy to induce recombination events. After setting up or transferring the
cross into a fresh vial (t = 0), flies are left to lay eggs for 1 day (t = 24 h after egg laying (AEL)). Subsequently,
parents are transferred into a fresh vial or removed. Progeny in vials are heat shocked up to three times at 48,
72, and 96 h AEL in a 37 °C water bath. Brains can be dissected from larvae, staged pupae, or adults. APF, after
puparium formation
3.2 Immunolabeling 1. Dissect 3rd instar larval, pupal, or adult brains (see Note 9) in
drops of cold PBS using forceps on a dissecting pad. To maxi-
mize the number of dissected samples with recombination
events, some Gal4 drivers make it possible to preselect flies with
clones under a fluorescence-dissecting microscope. Transfer
the brains into a glass embryo dish filled with PBS using forceps
or a glass pipette. Keep the dish on ice until all samples of a
given genotype are dissected (see Notes 10 and 11).
2. Transfer the brains into a mesh basket (see Note 12) posi-
tioned in one well of a 24-well multidish filled with 1 mL PLP.
Fix for 1 h at room temperature (see Note 13).
64 Nana Shimosako et al.
Table 2
Example of a confocal microscopy scanning method for imaging FB transgenes
4 Notes
Acknowledgements
References
1. Livet J, Weissman TA, Kang H, Draft RW, Lu J, Improved monomeric red, orange and yellow
Bennis RA et al (2007) Transgenic strategies for fluorescent proteins derived from Discosoma
combinatorial expression of fluorescent pro- sp. red fluorescent protein. Nat Biotechnol
teins in the nervous system. Nature 450:56–62 22:1567–1572
2. Hampel S, Chung P, McKellar CE, Hall D, 10. Rizzo MA, Springer GH, Granada B, Piston
Looger LL, Simpson JH (2011) Drosophila DW (2004) An improved cyan fluorescent
Brainbow: a recombinase-based fluorescence protein variant useful for FRET. Nat
labeling technique to subdivide neural expres- Biotechnol 22:445–449
sion patterns. Nat Methods 8:253–259 11. Shaner NC, Steinbach PA, Tsien RY (2005) A
3. Hadjieconomou D, Rotkopf S, Alexandre C, guide to choosing fluorescent proteins. Nat
Bell DM, Dickson BJ, Salecker I (2011) Methods 2:905–909
Flybow: genetic multicolor cell labeling for 12. Griesbeck O, Baird GS, Campbell RE,
neural circuit analysis in Drosophila melano- Zacharias DA, Tsien RY (2001) Reducing the
gaster. Nat Methods 8:260–266 environmental sensitivity of yellow fluorescent
4. Brand AH, Perrimon N (1993) Targeted gene protein. Mechanism and applications. J Biol
expression as a means of altering cell fates and Chem 276:29188–29194
generating dominant phenotypes. Development 13. Goedhart J, van Weeren L, Hink MA, Vischer
118:401–415 NO, Jalink K, Gadella TW Jr (2010) Bright
5. Voziyanov Y, Konieczka JH, Stewart AF, cyan fluorescent protein variants identified by
Jayaram M (2003) Stepwise manipulation of fluorescence lifetime screening. Nat Methods
DNA specificity in Flp recombinase: progres- 7:137–139
sively adapting Flp to individual and combina- 14. Lee T, Luo L (1999) Mosaic analysis with a
torial mutations in its target site. J Mol Biol repressible cell marker for studies of gene func-
326:65–76 tion in neuronal morphogenesis. Neuron
6. Liaw CW, Zamoyska R, Parnes JR (1986) 22:451–461
Structure, sequence, and polymorphism of the 15. Hadjieconomou D, Timofeev K, Salecker I
Lyt-2 T cell differentiation antigen gene. J (2011) A step-by-step guide to visual circuit
Immunol 137:1037–1043 assembly in Drosophila. Curr Opin Neurobiol
7. Zacharias DA, Violin JD, Newton AC, Tsien 21:76–84
RY (2002) Partitioning of lipid-modified 16. Hayashi S, Ito K, Sado Y, Taniguchi M,
monomeric GFPs into membrane microdo- Akimoto A, Takeuchi H et al (2002) GETDB,
mains of live cells. Science 296:913–916 a database compiling expression patterns and
8. Dietzl G, Chen D, Schnorrer F, Su KC, molecular locations of a collection of Gal4
Barinova Y, Fellner M et al (2007) A genome- enhancer traps. Genesis 34:58–61
wide transgenic RNAi library for conditional 17. Timofeev K, Joly W, Hadjieconomou D,
gene inactivation in Drosophila. Nature 448: Salecker I (2012) Localized netrins act as posi-
151–156 tional cues to control layer-specific targeting of
9. Shaner NC, Campbell RE, Steinbach PA, photoreceptor axons in Drosophila. Neuron
Giepmans BN, Palmer AE, Tsien RY (2004) 75:80–93
Chapter 5
Abstract
The Drosophila visual system is an excellent model system to study the switch from proliferating to differ-
entiating neural stem cells. In the developing larval optic lobe, symmetrically dividing neuroepithelial cells
transform to asymmetrically dividing neuroblasts in a highly ordered and sequential manner. This chapter
presents a protocol to visualize neural stem cell types in the Drosophila optic lobe by fluorescence confocal
microscopy. A main focus is given on how to dissect, fix, immunolabel, and mount brains to reveal cellular
morphology during early larval brain development.
Key words Neural stem cell, Drosophila, Brain, Optic lobe, Immunofluorescent labeling
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_5, © Springer Science+Business Media, LLC 2014
71
72 Benjamin Perruchoud and Boris Egger
2 Materials
3 Methods
3.2 Staging 1. Collect embryos on apple juice plates for 4–6 h and let embryos
of Larvae develop at 25 °C (see Note 2).
2. 24 h after midpoint of egg collection, pick about 80–100
freshly hatched larvae and place them in a food plate contain-
ing a drop of wet yeast.
3. Let larvae develop at 25 °C to the appropriate larval stage
(e.g., 24 h ALH, after larval hatching or 48 h ALH).
3.3 Dissection 1. Pick the larvae from food plate and place them on a paper tis-
of Larval Brains sue soaked with PBS (see Note 3).
2. Place several drops of 1× PBS in a circle on the inside of a
plastic petri dish lid. Add one larva to each drop (see Fig. 1a).
3. To dissect small larvae (0–48 h ALH), use two syringe needles
on cotton buds (see Fig. 1a). Carefully hold the larva in place
with one needle pressing down the middle of the larval body.
Stick the tip of the other needle firmly into the very anterior
part of the larva where the mouth hooks are located. Draw the body
slowly away from the sticked down mouthpart. As the anterior
head part breaks away from the rest of the body, the brain spills
out. The brain might be still attached to the mouth hooks.
Dissect off the mouth hooks and imaginal discs as necessary
(see Note 4). To dissect larger larvae (72–96 h ALH), use fine
forceps and the so-called “inverted sock” technique. Pull the
larva in the middle apart and discard posterior part. Grab the
larval mouth hooks at the anterior half with one forceps and
invert the larval cuticle over the tip of the forceps in a manner
like to invert a sock. The interior organs of the larva are now
on the outside and the brain can be further dissected.
74 Benjamin Perruchoud and Boris Egger
a b
c d
1 2 3
f
g
h
Fig. 1 (a) Larvae are placed in a drop of 1× PBS in a petri dish lid and dissected by using needles on cotton
buds or fine forceps. (b) Dissected brains are transferred by using a pipette (small brains) or forceps (larger
brains) and oriented in desired position on Poly-L-Lysine-coated cover slips. (c) Columbia jar holds maximal
four cover slips. (d) Cover slips with attached brains are transferred to Columbia jars for fixation and washing
steps. (e) A humid chamber is used for primary and secondary antibody incubation. (f) After the final washing
step, a strip of Vaseline is added to each cover slip corner as a spacer. (g) Cover slip is carefully placed on a
slide containing a drop of Vectashield and corners are gently pushed down. (h) Samples can be sealed with
transparent nail varnish and stored at 4 °C
Immunofluorescent Labelling on Drosophila Optic Lobes 75
4. Place a drop of PBS onto the coated cover slip. For small
brains (0–48 h ALH), cut off the very tip of a 200 μl pipette
tip and transfer the brains to the cover slip (see Note 5 and
Fig. 1b). For larger brains (72–96 h ALH) use the forceps to
transfer the brains.
5. Place brains to the Poly-L-Lysine-coated area in the desired
orientation by using cotton bud needles or forceps (see Note 6).
3.4 Fixation 1. Rinse cover slips with attached brains in a Columbia jar
and Staining (see Fig. 1c) containing 1× PBS.
2. Fix brains on cover slips in a Columbia jar containing 4 ml
fixative by rotating on a platform for 18 min (see Fig. 1d).
3. Replace fixative by PBST ~6 ml and wash the brains 1 × 1 min,
2 × 5 min, and 1 × 15 min on a rotating platform.
4. Place the cover slips into a humid chamber and add primary
antibodies (see Note 7) diluted in PBST (100 μl total volume)
on top of slide (see Fig. 1e). Close lid of humid chamber and
incubate overnight at 4 °C (see Note 8).
5. Wash the cover slips in a Columbia jar with PBST 1 × 1 min,
3 × 5 min, and 2 × 15 min.
6. Place the cover slips in a humid chamber and add secondary
antibodies diluted in PBST (100 μl total volume) on top of the
slide. Close the lid of humid chamber and incubate overnight
at 4 °C (see Note 9).
7. Wash cover slips in a Columbia jar with PBST 1 × 1 min,
3 × 5 min, and 3 × 15 min.
3.5 Mounting 1. Transfer cover slips using a forceps from the Columbia jar and
rinse off PBST with ddH2O from top of cover slips (see Note 10).
2. Remove excess liquid on a paper tissue and add a strip of
Vaseline to each corner as a space holder by using the syringe
(see Fig. 1f).
3. Add a drop of Vectashield on a clean microscopy slide.
4. Place cover slips with attached brains onto microscopy slides
and tap or gently press down each corner using cotton buds
(see Note 11 and Fig. 1g).
5. Seal cover slips with transparent nail varnish (see Fig. 1h).
6. Samples can be stored in slide folder at 4 °C.
76 Benjamin Perruchoud and Boris Egger
4 Notes
Acknowledgments
We thank Mike Bate for his advice on how to dissect brains of 1st
and 2nd instar larvae. B.P. and B.E. are funded by the Swiss
University Conference (SUK/CUS) Project P-01 Bio-BEFRI.
References
1. Hofbauer A, Campos-Ortega JA (1990) negatively regulated by JAK/STAT.
Proliferation pattern and early differentiation Development 135:1471–1480
of the optic lobes in Drosophila melanogaster. 7. Reddy BV, Rauskolb C, Irvine KD (2010)
Roux’s Arch Dev Biol 198:264–274 Influence of fat-hippo and notch signaling on
2. Egger B, Boone JQ, Stevens NR et al (2007) the proliferation and differentiation of
Regulation of spindle orientation and neural Drosophila optic neuroepithelia. Development
stem cell fate in the Drosophila optic lobe. 137:2397–2408
Neural Develop 2:1 8. Ngo KT, Wang J, Junker M et al (2010)
3. Egger B, Gold KS, Brand AH (2010) Notch Concomitant requirement for Notch and Jak/
regulates the switch from symmetric to asym- Stat signaling during neuro-epithelial differen-
metric neural stem cell division in the tiation in the Drosophila optic lobe. Dev Biol
Drosophila optic lobe. Development 137: 346:284–295
2981–2987 9. Orihara-Ono M, Toriya M, Nakao K et al
4. Egger B, Gold KS, Brand AH (2011) (2011) Downregulation of Notch mediates
Regulating the balance between symmetric the seamless transition of individual Drosophila
and asymmetric stem cell division in the devel- neuroepithelial progenitors into optic medul-
oping brain. Fly 5(3):237–241 lar neuroblasts during prolonged G1. Dev Biol
5. Yasugi T, Sugie A, Umetsu D et al (2010) 351:163–175
Coordinated sequential action of EGFR and 10. Wang W, Liu W, Wang Y et al (2010) Notch sig-
Notch signaling pathways regulates proneural naling regulates neuroepithelial stem cell mainte-
wave progression in the Drosophila optic lobe. nance and neuroblast formation in Drosophila
Development 137:3193–3203 optic lobe development. Dev Biol 350:414–428
6. Yasugi T, Umetsu D, Murakami S et al (2008) 11. Gotz M, Huttner WB (2005) The cell biology
Drosophila optic lobe neuroblasts triggered by of neurogenesis. Nat Rev Mol Cell Biol
a wave of proneural gene expression that is 6:777–788
Chapter 6
Abstract
Mosaic analysis with a repressible cell marker (MARCM) generates positively labeled, wild-type or mutant
mitotic clones by unequally distributing a repressor of a cell lineage marker, originally tubP-driven GAL80
repressing the GAL4/UAS system. Variations of the technique include labeling of both sister clones (twin
spot MARCM), the simultaneous use of two different drivers within the same clone (dual MARCM), as
well as the use of different repressible transcription systems (Q-MARCM). MARCM can be combined
with any UAS-based construct, such as localized GFP fusions to visualize subcellular compartments, genes
for rescue and ectopic expression, and modifiers of neural activity. A related technique, the twin spot gen-
erator, generates positively labeled clones without the use of a repressor, thus minimizing the lag time
between clone induction and appearance of label. The present protocol provides a detailed description of a
standard MARCM analysis of brain development that includes generation of MARCM stocks and crosses,
induction of clones, brain dissection at various stages of development, immunohistochemistry, and confo-
cal microscopy, and can be modified for similar experiments involving mitotic clones.
Key words Drosophila, MARCM, Twin spot generator, Flp-out, Clones, Somatic recombination,
Protocol, Brain development, Cell labeling, Neuroblast lineage
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_6, © Springer Science+Business Media, LLC 2014
79
80 Gudrun Viktorin
a b
FRT (m)
(m) (m) 80 80 (m) 80 (m) 80 80
(m)
FLP (m) 80 tubP
GAL80
FRT
tubP
GAL4
UAS
gfp
UAS
gene
hs
FLP
Fig. 1 (a) Schematic representation of unequal repressor (orange) distribution in MARCM by induced somatic
recombination between homologous FRT sites, leading to positively labeled clones (green) and unlabeled sister
clones. Only the recombining chromosome pair is shown, containing Flp recombinase target recognition (FRT)
sites (black triangles) and repressor transgene (orange). Inset is a maximum projection of a confocal stack of
a type II neuroblast clone that was induced around hatching, with wide secondary arborizations (Leica SP5,
63×/1.30 glycerol immersion objective). (b) Complete set of elements in cells of heterozygous offspring of a
MARCM cross prior to recombination, and its interactions. Flp/FRT system, black; GAL4/UAS system, white;
GAL80 repressor driven by ubiquitous promoter, orange; GFP marker, green. Some optional elements are
included in parentheses: a mutation on the GAL80-free FRT chromosome (m), and an additional UAS-driven,
GAL80-repressible transgene for rescue or ectopic expression of genes within the clone
2 Materials
2.1 Fly Stocks A pair of MARCM stocks contains the following genetic elements
(Fig. 1b), available through Bloomington [44]:
1. FRT chromosome with distally located tubP-GAL80 trans-
gene (see Note 1).
2. Homologous FRT chromosome, with or without distally
located mutation.
3. GAL4 driver (see Note 2).
4. UAS marker (see Note 3).
5. hs-flp or promoter-driven flp transgene (see Note 4).
6. Balancer chromosomes with markers visible at the time of
dissection (see Note 5).
2.4 Solutions for 1. PBS: Tablets (Sigma) or diluted from 10× PBS: 2.56 g/l
Dissection and NaH2PO4, 11.94 g/l Na2HPO4, 102.2 g/l NaCl, pH at
Immunohisto- 7.4 [46].
chemistry 2. PBT: 0.1–0.5 % Triton X-100 in PBS.
3. PBL: [47] (see Note 9) (0.1 M lysine, 0.05 M phosphate buffer,
pH 7.4—dissolve lysine in half the volume of water, adjust pH
to 7.4 with 0.1 M Na2HPO4, adjust volume to final concen-
trations with 0.1 M NaH2PO4/Na2HPO4 buffer pH 7.4).
4. 8 % paraformaldehyde stock solution: 0.8 g paraformaldehyde
dissolved in 10 ml water with 70 μl 1 M NaOH, incubate in
37 °C water bath with occasional shaking until just dissolved,
prepare fresh or freeze aliquots at −20 °C.
5. Methanol.
6. BT-NGS: PBT with 10 % normal goat serum that had been
heat inactivated for 30 min at 56 °C and frozen in aliquots.
7. Antibody stocks (see Note 9).
3 Methods
3.2 Egg Collection, 1. Transfer MARCM crosses into egg collectors, cover with
Heat-Shock Induction yeasted grape juice plates or fly bottles, and collect at 25 °C in
of Clones, Brain the dark for the desired time period.
Dissection 2. Keep offspring at 25 °C until the desired stage for heat shock
(or dissection, if flp is not induced by heat shock).
3. Immerse sealed grape juice plates or fly bottles in a 37 °C water
bath for the desired duration (see Note 15).
4. Raise larvae at 25 °C on fly food until dissection (see Note 16).
Grape juice plates can be cut into pieces and transferred into fly
food bottles.
5. Harvest animals as larvae, pupae, or adults: For pre-wandering
larvae, fill bottle with 30 % glycerol, skim larvae off the surface
with spoon, wash with tap water, and immobilize on ice for
genotyping; take wandering third-instar larvae or pupae off the
bottle wall with forceps or a wet brush; Anesthetize adults with
CO2 and keep immobilized on ice.
6. Select offspring of the correct genotype, and discard any off-
spring with balancer chromosomes.
7. Dissect brains of larvae, pupae, or adults (see Note 17) in PBS.
For immunostaining, keep dissected tissue on ice and fix after
20–30 min of dissection. For subsequent live imaging, dissect
at room temperature in live imaging media [14, 51].
3.3 Fixation, 1. Fix freshly dissected tissue for 30–60 min in 2 or 4 % parafor-
Immunostaining, and maldehyde in PBL or PBS (see Note 18).
Microscopy 2. Wash with PBT four times within 45 min.
3. Preincubate in PBT–NGS solution for 15 min.
4. Incubate in primary antibody in PBT–NGS overnight at 4 °C.
5. Wash with PBT four times within 1 h.
6. Incubate in secondary antibody for 2–3 h at room temperature
or, preferably, overnight at 4 °C in the dark.
7. Wash with PBT ≥ 4 times within 1 h.
8. Take off liquid with a pipette, and place a drop of Vectashield
onto the brains. Without stirring incubate for several hours to
overnight, to slowly diffuse in glycerol without osmotically
shocking the tissue. Brains should be imaged soon but can be
kept in Vectashield at −20 °C for later processing.
9. Place brains onto the slide, remove Vectashield as much as
possible so that surface tension holds the brains in place,
and arrange them in rows and columns for microscopy, using
an eyelash tool or forceps. Larval brains that are still attached
MARCM in Drosophila Brain Development 85
4 Notes
Table 1
Sample MARCM driver stocks that have been used routinely and remained stable over years
(constructed by B. Bello)
9. Some labs routinely fix in PBL [47], not older than 3 months,
and others in PBS or other buffers [46]. PBL is derived from a
periodate–lysine solution used to fix glycoproteins [68]; lysine
alone may help to preserve certain antigens. Commonly used
antibodies for postembryonic brains label either synaptic neu-
ropil or axon tracts, described in [69]: BP106/mouse anti-
neurotactin (1:20, DSHB [70]) labels larval and pupal
secondary lineages and axon tracts. BP106 requires a 5–10-
min methanol incubation after fixation (Kathy Ngo and Volker
Hartenstein, pers. comm.) that also makes larval and pupal
brains less fragile and does not interfere with many other anti-
body stainings; BP104/mouse anti-neuroglian (1:20, DSHB;
[71]) labels secondary axon tracts in the adult brain; nc82/
mouse anti-bruchpilot (1:10; DSHB; [72]) is used for the
Drosophila standard brain [73] and labels synaptic neuropil. nc82
staining in adult brains greatly improves when incubated over two
nights and in 0.5 % Triton X-100 [40, 74]. Chicken anti-GFP
(1:1,000, Abcam, Cambridge, UK), rabbit anti-RFP (1:200,
Abcam), and rabbit anti-beta-Galactosidase (1:1,000, Cappel/
MP Biomedicals) can be used for transgenic markers, and Alexa
488/568/647 (1:300, Molecular Probes/Life Technologies)
secondary antibodies work well for triple labeling.
10. For third-instar larvae, pupae, and adult brains, a spacer slightly
thinner than a coverslip will hold brains in place without
squashing them, e.g., two layers of scotch tape. For smaller
brains, use one layer of tape, or apply a flexible spacer such as
Vaseline to the edges of the coverslip, and gently push down.
Minor-quality nail polish will not seal well and may interfere with
fluorescence. Hardening mounting media are also in use [9].
11. At 20× magnification, the whole brain can be fully scanned; for
single-cell resolution (a neuronal cell body from a postembry-
onic lineage has a diameter around 4–5 μm), 40× or higher
magnification is necessary. At 63× magnification, a fixed brain
hemisphere from a wandering third-instar larva just fits into the
field of view. For whole-hemisphere scans of brains mounted in
glycerol-based Vectashield, a glycerol immersion objective min-
imizes spherical aberrations [75]. In addition, a long working
distance is useful, since working distance of higher magnifying
oil objectives can be shorter than the thickness of the brain plus
coverslip. Valuable aspects of confocal microscopy and image
processing artifacts are discussed in [53].
12. If a transgenic chromosome for a planned stock is difficult to bal-
ance for introducing the other elements, consider redistributing
the MARCM elements in case the desired stock is not viable. It is
not infrequent to obtain around 15 flies or less of the correct
genotype from a bottle during the final crossing generations.
13. A bottle with 100–120 virgins and 10–30 males, left to lay eggs
within 1–2 h at 25 °C, can give rise to 100–200 live larvae depend-
MARCM in Drosophila Brain Development 89
ing on the fecundity of the stock used, and the time of egg collec-
tion, highest during the flies’ afternoon/evening [50]. Flies that
had been raised under crowded conditions are small and lay few
eggs; split up bottles with too many larvae into 2–3 bottles prior
to virgin collection. If very large amounts of virgins need to be
collected, it can save time to cross a virginator such as hs-hid (Y)
(M. Van Doren in [76]) into the MARCM driver that can kill off
most males in a 1-h-long heat shock at the third instar prior to
virgin collection (see descriptions of Bloomington stocks 8846,
24638). Each element that is homozygous in the parent stock
doubles the amount of larvae of the correct genotype in the off-
spring; thus, if homozygous viable and fertile, avoid males con-
taining a balancer, and allow a day longer for mating to occur with
fewer males.
14. Virgin collections and crosses can be lost instantly when shaken
into a bottle with sticky food, and females need to remain well
fed with yeast paste. Between egg collections, shake MARCM
crosses into fresh bottles with dry surface as soon as the drop
of yeast is eaten. If bottles tend to be humid, keep flies less
densely, and place a rolled-up paper tissue in the food to absorb
moisture when not collecting eggs. Crosses can be kept at
18 °C when not in use to prolong the fertile period. Females
lay best during the week following mating, but can be used for
3–4 weeks until the amount of eggs declines.
15. Larvae will crawl rapidly to escape the heat; submerge sealed
plates completely, or push down the stopper of the fly food
bottle below the surface of the water bath. Larvae crawl onto
mite-proof stoppers but not cheap foam ones. The optimal
time and duration of heat shock need to be determined experi-
mentally to reach the desired clone density. As an estimate,
using hs-flp122, a heat shock of 6 min at 37 °C around hatching
most frequently induces 0–2 neuroblast clones per brain, while
a heat shock of 8 min most frequently induces 1–5 neuroblast
clones per brain (own observations). Background clone induc-
tion is usually negligible, but single experiments can show con-
siderable clone frequencies without heat shock (own
observations). Certain neuroblast clones are only recovered
upon heat shock during certain instars, and in smaller develop-
mental time windows than may be expected from other experi-
ments (B. Bello, personal communication).
16. The density at which larvae are reared greatly affects their
developmental rate [38, 50]. In my hands, a maximum of 120
larvae in a bottle of 6 cm diameter will develop rather synchro-
nously, but tends to dry out; add water as needed. If larvae
are too dense, split them up into several bottles. Larval and
pupal development at 18 °C takes quite exactly half as long as
development at 25 °C [50], which can be used to avoid nighttime
dissections, unless staging needs to be exact.
90 Gudrun Viktorin
Fig. 2 Dissection of larvae (a, b), prepupae (c, d), and pupae (e, f) using scissors and eyelash tool. Scissors in
(a) are not positioned at the proposed cut site
Acknowledgments
References
1. Blair SS (2003) Genetic mosaic techniques for 3. Xu T, Rubin GM (1993) Analysis of genetic
studying Drosophila development. Development mosaics in developing and adult Drosophila tis-
130(21):5065–5072. doi:10.1242/dev.00774 sues. Development 117(4):1223–1237
130/21/5065 [pii] 4. Lee T, Luo L (1999) Mosaic analysis with a
2. Golic KG (1991) Site-specific recombination repressible cell marker for studies of gene function
between homologous chromosomes in in neuronal morphogenesis. Neuron 22(3):451–
Drosophila. Science 252(5008):958–961 461. doi:doi:S0896-6273(00)80701-1 [pii]
MARCM in Drosophila Brain Development 93
5. Lee T, Luo L (2001) Mosaic analysis with a 16. Viktorin G, Riebli N, Popkova A, Giangrande
repressible cell marker (MARCM) for A, Reichert H (2011) Multipotent neural
Drosophila neural development. Trends stem cells generate glial cells of the central
Neurosci 24(5):251–254. doi:S0166- complex through transit amplifying interme-
2236(00)01791-4 [pii] diate progenitors in Drosophila brain devel-
6. Jefferis GS, Marin EC, Stocker RF, Luo L opment. Dev Biol 356(2):553–565.
(2001) Target neuron prespecification in the doi:S0012-1606(11)01035-9 [pii] 10.1016/
olfactory map of Drosophila. Nature j.ydbio.2011.06.013
414(6860):204–208. doi:10.1038/35102574 17. Lin S, Lai SL, Yu HH, Chihara T, Luo L, Lee
7. Lee T, Lee A, Luo L (1999) Development of T (2010) Lineage-specific effects of Notch/
the Drosophila mushroom bodies: sequential Numb signaling in post-embryonic develop-
generation of three distinct types of neurons ment of the Drosophila brain. Development
from a neuroblast. Development 126(18): 137(1):43–51. doi:137/1/43 [pii] 10.1242/
4065–4076 dev.041699
8. Pereanu W, Hartenstein V (2006) Neural lin- 18. Truman JW, Moats W, Altman J, Marin EC,
eages of the Drosophila brain: a three- Williams DW (2010) Role of Notch signaling
dimensional digital atlas of the pattern of lineage in establishing the hemilineages of secondary
location and projection at the late larval stage. neurons in Drosophila melanogaster.
J Neurosci 26(20):5534–5553. doi:26/20/5534 Development 137(1):53–61. doi:137/1/53
[pii] 10.1523/JNEUROSCI.4708-05.2006 [pii] 10.1242/dev.041749
9. Truman JW, Schuppe H, Shepherd D, Williams 19. Bello BC, Hirth F, Gould AP (2003) A pulse
DW (2004) Developmental architecture of of the Drosophila Hox protein Abdominal-A
adult-specific lineages in the ventral CNS of schedules the end of neural proliferation via
Drosophila. Development 131(20):5167–5184 neuroblast apoptosis. Neuron 37(2):209–219.
10. Pereanu W, Shy D, Hartenstein V (2005) doi:S0896627302011819 [pii]
Morphogenesis and proliferation of the larval 20. Komiyama T, Johnson WA, Luo L, Jefferis
brain glia in Drosophila. Dev Biol 283(1):191– GS (2003) From lineage to wiring specificity
203. doi:S0012-1606(05)00228-9 [pii] POU domain transcription factors control
10.1016/j.ydbio.2005.04.024 precise connections of Drosophila olfactory
11. Awasaki T, Lai SL, Ito K, Lee T (2008) projection neurons. Cell 112(2):157–167.
Organization and postembryonic development doi:S0092867403000308 [pii]
of glial cells in the adult central brain of 21. Liu Z, Steward R, Luo L (2000) Drosophila Lis1
Drosophila. J Neurosci 28(51):13742–13753. is required for neuroblast proliferation, dendritic
doi:28/51/13742 [pii] 10.1523/ elaboration and axonal transport. Nat Cell Biol
JNEUROSCI.4844-08.2008 2(11):776–783. doi:10.1038/35041011
12. Boone JQ, Doe CQ (2008) Identification of 22. Kohatsu S, Koganezawa M, Yamamoto D
Drosophila type II neuroblast lineages contain- (2011) Female contact activates male-specific
ing transit amplifying ganglion mother cells. interneurons that trigger stereotypic courtship
Dev Neurobiol 68(9):1185–1195. behavior in Drosophila. Neuron 69(3):498–
doi:10.1002/dneu.20648 508. doi:S0896-6273(10)01040-8 [pii]
13. Bowman SK, Rolland V, Betschinger J, Kinsey 10.1016/j.neuron.2010.12.017
KA, Emery G, Knoblich JA (2008) The tumor 23. Amanai K, Jiang J (2001) Distinct roles of cen-
suppressors Brat and Numb regulate transit- tral missing and dispatched in sending the
amplifying neuroblast lineages in Drosophila. Dev Hedgehog signal. Development 128(24):
Cell 14(4):535–546. doi:10.1016/j.dev- 5119–5127
cel.2008.03.004; S1534-5807(08)00112-3 [pii] 24. Wang J, Zugates CT, Liang IH, Lee CH, Lee T
14. Bello BC, Izergina N, Caussinus E, Reichert (2002) Drosophila Dscam is required for diver-
H (2008) Amplification of neural stem cell gent segregation of sister branches and sup-
proliferation by intermediate progenitor cells presses ectopic bifurcation of axons. Neuron
in Drosophila brain development. Neural Dev 33(4):559–571. doi:S0896627302005706
3:5. doi:1749-8104-3-5 [pii] 10.1186/1749 [pii]
-8104-3-5 25. Zheng X, Wang J, Haerry TE, Wu AY, Martin
15. Izergina N, Balmer J, Bello B, Reichert H J, O’Connor MB, Lee CH, Lee T (2003)
(2009) Postembryonic development of transit TGF-beta signaling activates steroid hormone
amplifying neuroblast lineages in the receptor expression during neuronal remodeling
Drosophila brain. Neural Dev 4:44. doi:1749- in the Drosophila brain. Cell 112(3):303–315.
8104-4-44 [pii] 10.1186/1749-8104-4-44 doi:S0092867403000722 [pii]
94 Gudrun Viktorin
26. Zhu S, Lin S, Kao CF, Awasaki T, Chiang AS, 72(4):527–540. doi:0092-8674(93)90072-X
Lee T (2006) Gradients of the Drosophila [pii]
Chinmo BTB-zinc finger protein govern neu- 38. Rooke JE, Theodosiou NA, Xu T (2000)
ronal temporal identity. Cell 127(2):409–422. Clonal analysis in the examination of gene
doi:S0092-8674(06)01230-X [pii] 10.1016/j. function in Drosophila. Methods Mol Biol
cell.2006.08.045 137:15–22. doi:1-59259-066-7-15 [pii]
27. Lai SL, Lee T (2006) Genetic mosaic with dual 10.1385/1-59259-066-7:15
binary transcriptional systems in Drosophila. 39. Theodosiou NA, Xu T (1998) Use of FLP/
Nat Neurosci 9(5):703–709. doi:nn1681 [pii] FRT system to study Drosophila development.
10.1038/nn1681 Methods 14(4):355–365. doi:S1046-2023
28. Potter CJ, Luo L (2011) Using the Q system (98)90591-6 [pii] 10.1006/meth.1998.0591
in Drosophila melanogaster. Nat Protoc 40. Wu JS, Luo L (2006) A protocol for dissecting
6(8):1105–1120. doi:nprot.2011.347 [pii] Drosophila melanogaster brains for live imag-
10.1038/nprot.2011.347 ing or immunostaining. Nat Protoc 1(4):2110–
29. Yu HH, Kao CF, He Y, Ding P, Kao JC, Lee T 2115. doi:nprot.2006.336 [pii] 10.1038/
(2010) A complete developmental sequence of nprot.2006.336
a Drosophila neuronal lineage as revealed by 41. Wu JS, Luo L (2006) A protocol for mosaic
twin-spot MARCM. PLoS Biol 8 (8) analysis with a repressible cell marker
doi:10.1371/journal.pbio.1000461 (MARCM) in Drosophila. Nat Protoc
30. Kunz T, Kraft KF, Technau, GM, Urbach R 1(6):2583–2589. doi:nprot.2006.320 [pii]
(2012) Origin of Drosophila mushroom body 10.1038/nprot.2006.320
neuroblasts and generation of divergent embry- 42. Kao CF, Lee T (2010) Genetic mosaic analysis
onic lineages. Development 139:2510–2522. of Drosophila brain by MARCM. In: Zhang B,
doi:10.1242/dev.077883 Freeman MR, Waddell S (eds) Drosphila neu-
31. Griffin R, Sustar A, Bonvin M, Binari R, del robiology. Cold Spring Harbor Laboratory
Valle RA, Hohl AM, Bateman JR, Villalta C, Press, Cold Spring Harbor, NY, pp 125–140
Heffern E, Grunwald D, Bakal C, Desplan C, 43. Stork T, Bernardos R, Freeman MR (2012)
Schubiger G, Wu CT, Perrimon N (2009) The Analysis of glial cell development and function
twin spot generator for differential Drosophila in Drosophila. Cold Spring Harb Protoc
lineage analysis. Nat Methods 6(8):600–602. 2012(1):1–17. doi:2012/1/pdb.top067587
doi:nmeth.1349 [pii] 10.1038/nmeth.1349
[pii] 10.1101/pdb.top067587
32. Wong AM, Wang JW, Axel R (2002) Spatial
44. BDSC http://flystocks.bio.indiana.edu/
representation of the glomerular map in the
Drosophila protocerebrum. Cell 109(2):229– 45. Ashburner M, Roote J (2000) Laboratory cul-
241. doi:S0092867402007079 [pii] ture of Drosophila. In: Sullivan W, Ashburner
33. Larsen C, Shy D, Spindler SR, Fung S, Pereanu M, Hawley S (eds) Drosophila protocols. Cold
W, Younossi-Hartenstein A, Hartenstein V Spring Harbor Laboratory Press, Cold Spring
(2009) Patterns of growth, axonal extension and Harbor, pp 585–599
axonal arborization of neuronal lineages in the 46. Patel NH (2006) http://www.patellab.org/ >
developing Drosophila brain. Dev Biol Protocols
335(2):289–304. doi:S0012-1606(09)00926-9 47. Hadjieconomou D, Rotkopf S, Alexandre C,
[pii] 10.1016/j.ydbio.2009.06.015 Bell DM, Dickson BJ, Salecker I (2011)
34. Pignoni F, Zipursky SL (1997) Induction of Flybow: genetic multicolor cell labeling for
Drosophila eye development by decapentaple- neural circuit analysis in Drosophila melano-
gic. Development 124(2):271–278 gaster. Nat Methods 8(3):260–266.
35. Ito K, Awano W, Suzuki K, Hiromi Y, doi:nmeth.1567 [pii] 10.1038/nmeth.1567
Yamamoto D (1997) The Drosophila mush- 48. Eliceiri KW, Berthold MR, Goldberg IG,
room body is a quadruple structure of clonal Ibanez L, Manjunath BS, Martone ME,
units each of which contains a virtually identi- Murphy RF, Peng H, Plant AL, Roysam B,
cal set of neurones and glial cells. Development Stuurmann N, Swedlow JR, Tomancak P,
124(4):761–771 Carpenter AE (2012) Biological imaging soft-
36. de Celis JF, Bray S (1997) Feed-back mecha- ware tools. Nat Methods 9(7):697–710.
nisms affecting Notch activation at the dorso- doi:nmeth.2084 [pii] 10.1038/nmeth.2084
ventral boundary in the Drosophila wing. 49. Greenspan RJ (1997) Fly pushing: the theory
Development 124(17):3241–3251 and practice of drosophila genetics. Cold
37. Struhl G, Basler K (1993) Organizing activ- Spring Harbor Laboratory Press, Cold Spring
ity of wingless protein in Drosophila. Cell Harbor, NY
MARCM in Drosophila Brain Development 95
50. Ashburner M, Golic KG, Hawley RS (eds) 60. Chou TB, Perrimon N (1992) Use of a yeast
(2005) Drosophila: a laboratory manual, 2nd site-specific recombinase to produce female
edn. Cold Spring Harbor Laboratory Press, germline chimeras in Drosophila. Genetics
Cold Spring Harbor, NY 131(3):643–653
51. Cabernard C, Doe CQ (2011) Live imaging of 61. Golic KG, Lindquist S (1989) The FLP recom-
neuroblast lineages within intact larval brains binase of yeast catalyzes site-specific recombi-
in Drosophila. In: Sharpe J, Wong R, Yuste R nation in the Drosophila genome. Cell
(eds) Imaging in developmental biology: a 59(3):499–509. doi:0092-8674(89)90033-0
laboratory manual. Cold Spring Harbor [pii]
Laboratory Press, Cold Sprint Harbor, NY, pp 62. Newsome TP, Asling B, Dickson BJ (2000)
217–227 Analysis of Drosophila photoreceptor axon
52. Lin DM, Fetter RD, Kopczynski C, guidance in eye-specific mosaics. Development
Grenningloh G, Goodman CS (1994) 127(4):851–860
Genetic analysis of Fasciclin II in Drosophila: 63. Silies M, Yuva Y, Engelen D, Aho A, Stork T,
defasciculation, refasciculation, and altered Klambt C (2007) Glial cell migration in the eye
fasciculation. Neuron 13(5):1055–1069. disc. J Neurosci 27(48):13130–13139.
doi:doi:0896-6273(94)90045-0 [pii] doi:27/48/13130 [pii] 10.1523/
53. Ito K, Okada R, Tanaka NK, Awasaki T (2003) JNEUROSCI.3583-07.2007
Cautionary observations on preparing and 64. Lattao R, Bonaccorsi S, Guan X, Wasserman
interpreting brain images using molecular biol- SS, Gatti M (2011) Tubby-tagged balancers
ogy-based staining techniques. Microsc Res for the Drosophila X and second chromo-
Tech 62(2):170–186. doi:10.1002/ somes. Fly (Austin) 5(4). doi:17283 [pii]
jemt.10369 65. Pina C, Pignoni F (2012) Tubby-RFP balanc-
54. Han C, Jan LY, Jan YN (2011) Enhancer- ers for developmental analysis: FM7c 2xTb-
driven membrane markers for analysis of non- RFP, CyO 2xTb-RFP, and TM3 2xTb-RFP.
autonomous mechanisms reveal neuron-glia Genesis 50(2):119–123. doi:10.1002/
interactions in Drosophila. Proc Natl Acad Sci dvg.20801
U S A 108(23):9673–9678. doi:1106386108 66. Blair SS (2000) Imaginal discs. In: Sullivan W,
[pii] 10.1073/pnas.1106386108 Ashburner M, Hawley S (eds) Drosophila pro-
55. Zhang YQ, Rodesch CK, Broadie K (2002) tocols. Cold Spring Harbor Laboratory Press,
Living synaptic vesicle marker: synaptotagmin- Cold Spring Harbor, NY, pp 159–173
GFP. Genesis 34(1–2):142–145. doi:10.1002/ 67. Ito K (1999) How to dissect Drosophila brains.
gene.10144 http://jfly.iam.u-tokyo.ac.jp/html/movie/
56. Nicolaï LJ, Ramaekers A, Raemaekers T, index.html
Drozdzecki A, Mauss AS, Yan J, Landgraf 68. Van Vactor DL Jr, Cagan RL, Kramer H,
M, Annaert W, Hassan BA (2011) Zipursky SL (1991) Induction in the develop-
Genetically encoded dendritic marker sheds ing compound eye of Drosophila: multiple
light on neuronal connectivity in Drosophila. mechanisms restrict R7 induction to a single
Proc Natl Acad Sci U S A 107(47):20553– retinal precursor cell. Cell 67(6):1145–1155.
20558. doi: 1010198107 [pii] 10.1073/ doi:0092-8674(91)90291-6 [pii]
pnas.1010198107
69. Pereanu W, Kumar A, Jennett A, Reichert H,
57. Jefferis GS, Potter CJ, Chan AM, Marin EC, Hartenstein V (2010) Development-based
Rohlfing T, Maurer CR Jr, Luo L (2007) compartmentalization of the Drosophila cen-
Comprehensive maps of Drosophila higher tral brain. J Comp Neurol 518(15):2996–
olfactory centers: spatially segregated fruit and 3023. doi:10.1002/cne.22376
pheromone representation. Cell 128(6):1187–
1203. doi:S0092-8674(07)00204-8 [pii] 70. Hortsch M, Patel NH, Bieber AJ, Traquina
10.1016/j.cell.2007.01.040 ZR, Goodman CS (1990) Drosophila neuro-
tactin, a surface glycoprotein with homology
58. Simpson JH (2009) Mapping and manipulat- to serine esterases, is dynamically expressed
ing neural circuits in the fly brain. Adv Genet during embryogenesis. Development 110(4):
65:79–143. doi:S0065-2660(09)65003-3 1327–1340
[pii] 10.1016/S0065-2660(09)65003-3
71. Bieber AJ, Snow PM, Hortsch M, Patel NH,
59. Hampel S, Chung P, McKellar CE, Hall D, Jacobs JR, Traquina ZR, Schilling J, Goodman
Looger LL, Simpson JH (2011) Drosophila CS (1989) Drosophila neuroglian: a member
brainbow: a recombinase-based fluorescence of the immunoglobulin superfamily with
labeling technique to subdivide neural expres- extensive homology to the vertebrate neural
sion patterns. Nat Methods 8(3):253–259. adhesion molecule L1. Cell 59(3):447–460.
doi:nmeth.1566 [pii] 10.1038/nmeth.1566 doi:0092-8674(89)90029-9 [pii]
96 Gudrun Viktorin
Other Arthropods
Chapter 7
Abstract
Injection of fluorochromes such as Alexa Fluor® 568 into single cells in brain slices reveals a network of
dye-coupled cells to be associated with the central complex. Subsequent immunolabeling shows these cells
to be repo positive/glutamine synthetase positive/horseradish peroxidase negative, thus identifying them
as astrocyte-like glia. Dye coupling fails in the presence of n-heptanol indicating that dye spreads from cell
to cell via gap junctions. A cellular network of dye-coupled, astrocyte-like, glia surrounds and infiltrates
developing central complex neuropils. Intracellular dye injection techniques complement current molecular
approaches in analyzing the functional properties of such networks.
Key words Insect, Brain slices, Dye injection, Immunolabeling, Glia, Gap junctions
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_7, © Springer Science+Business Media, LLC 2014
99
100 George Boyan and Yu Liu
2 Materials
2.2 Dissection and 1. Fine forceps, sizes #3, #5 (both Dumont, Switzerland).
Associated Supplies 2. Iridectomy scissors (Martin, Germany).
3. Micropipette (Eppendorf) with exchangeable tips.
4. Packages of exchangeable 1 μL tips for antibody delivery,
and 50 μL tips for medium exchange (we use Ultratip Greiner
Bio-one).
Dye Coupling Between Glia in Brain Slices 101
2.3 Solutions, Media 1. 0.1 M PBS: 2.6 g/L NaH2PO4, 22.5 g/L Na2HPO4,
102.2 g/L NaCl, adjust to pH 7.4 with NaOH. Store at 4 °C.
Take 100 mL PBS and store over ice on the day of making
frozen sections.
2. Prepare an ice-cold 1 % methylene blue solution in 0.1 M PBS
and place an aliquot in each of several ice-cold Eppendorf
(100 μL) containers. Embryonic grasshopper heads are small
(up to 1 mm) so several heads can be placed in one Eppendorf.
3. Physiological saline: 8.8 g/L NaCl, 0.2 g/L KCl, 0.3 g/L
CaCl2, 2.5 g/L MgSO4, 0.9 g/L TES buffer. Adjust to pH 7.0
with the buffer. Store at 4 °C. Decant 10 mL for each experi-
mental sitting and allow to reach room temperature prior to
the experiment.
4. Fetal Calf Serum (FCS), store frozen (−18 °C). Maintain an
aliquot of 10 mL in an Eppendorf at 4 °C. This fluid will be
used for dipping the electrode tip into and for internally coating
the glass pipettes with which heads/brains are transferred.
5. Aqueous bouin: picric acid 71 %, formaldehyde 24 %, glacial
acetic acid 5 %. Fixative for immunolabeling with both anti-
glutamine synthetase (for glia) and horseradish peroxidase (for
neurons). Store under a fume hood at room temperature.
6. 3.7 % paraformaldehyde (PFA) as the fixative for double gluta-
mine synthetase/repo immunolabeling. Store in a fume hood
at room temperature.
7. PBT containing NHS: 1 % normal horse serum (NHS), 0.1 %
bovine serum albumin (BSA), PBT (1 % Triton X-100 in 0.1 M
PBS). Adjust to pH 7.4 with buffer. Store at 4 °C in the dark.
8. After dye injection and immunolabeling, we use Vectashield®
(Vector laboratories) to cover brain slices and minimize bleach-
ing during fluorescence microscopy.
9. The fluorochrome we use here is Alexa Fluor® 568 (Invitrogen,
emission maximum 600 nm), but we also used Alexa Fluor®
488 (Invitrogen, emission maximum 517 nm) and Lucifer
Yellow CH (Aldrich, emission maximum 528 nm) (see ref. 30
and Note 1). Alexa Fluor® 568 as a hydrazine has a low molec-
ular weight (790 Da) and so flows readily throughout cells on
injection. Store the stock at −18 °C. Make up a 0.1 mL aliquot
of 10 mM Alexa Fluor® 568 in 200 mM KCl and store in a
glass ampule (3-1801, neoLab, Heidelberg) at 4 °C in the
dark. Maintain 10 mL of 200 mM KCl at room temperature
for backfilling each micropipette.
102 George Boyan and Yu Liu
2.7 Electronics 1. DC amplifier (we use a Getting 5) equipped with a head stage
that is positioned as near to the preparation as possible to
reduce electrical interference, and with current passing facility
via a virtual ground circuit.
2. A simple 2-channel oscilloscope for monitoring current injection
and electrode resistance. Use the electrode resistance-testing
capability of the amplifier to check that the electrode has a
104 George Boyan and Yu Liu
2.8 Fixed-Stage 1. In a fixed-stage microscope, the optics not the stage are raised
Compound Microscope and lowered, allowing focusing of tissues with intracellular
and Camera electrode in position. You will need a fixed-stage compound
microscope (e.g., Zeiss Axioskop 2) equipped with filters for
both epifluorescence illumination (tetramethyl rhodamine iso-
thiocyanate, TRITC, emission 580 nm; fluorescein isothiocya-
nate, FITC, emission 528 nm) and differential interference
contrast (DIC)/transmitted light optics. The microscope
should stand inside an earthed Faraday cage to minimize elec-
trical interference from lamps and circuits in the building, and
on a vibration-free table (preferably supported by compressed
air) for stability so as to optimize microelectrode positioning.
2. The microscope should be equipped with a low-power (×10)
objective (e.g., Zeiss Plan-Neofluar, 440330, n.A. 0.30) for
centering the brain slice and electrode tip and a high-power
(×63) water-immersion objective with long working distance
(2.1 mm) for monitoring the actual intracellular penetration
(e.g., Zeiss Achroplan ceramic-coated ×63 objective, 440067-
9901, n.A. 0.95 W). The ceramic renders the objective elec-
trically inert when placed in the saline solution covering the
brain slice.
3. A color CCD camera (1.3 MP is adequate) mounted on the
microscope, driver software for a computer, a flatscreen monitor
(at least 19 in. diagonal and 1,640 × 1,050 pixel resolution is
recommended). A micrometer scale etched into a glass slide
(Zeiss 473390-9901) is used for checking that the microscope/
camera adapter provides a 1:1 image size on the monitor.
2.9 Imaging 1. To reveal fine structures such as gliopodia in 3D, you need
to acquire optical sections (a so-called Z stack) of prepara-
tions using a confocal laser scanning microscope. We used a
Leica TCS SP5 equipped with Leica Hc Pl Apo CS ×20 and
Leica Hcx Pl Apo lambda blue ×63 oil immersion objectives
(see Note 4).
2. Fluorochromes were visualized using an argon laser with exci-
tation wavelengths of 488 nm for Alexa Fluor® 488, 578 nm
for both Alexa Fluor® 568 and Cy3, and 633 nm for Cy5.
3. Z-stacks of confocal images were processed using public
domain software (ImageJ).
Dye Coupling Between Glia in Brain Slices 105
3 Methods
3.1 Preparing Frozen 1. Using the #5 forceps and iridectomy scissors, which have
Brain Slices previously been dipped in FCS to prevent tissue adhesion, dis-
sect embryos out of the egg into ice-cold 0.1 M PBS and free
embryos from surrounding membranes. For grasshoppers,
embryos are staged according to percentage of development
(see ref. 40).
2. For younger embryos (<60 %), decapitate embryos with a single
snip of the iridectomy scissors. For older embryos (>70 %), the
cuticle becomes an obstacle during cryosection. Therefore
open the head capsule by cutting away the front with the iri-
dectomy scissors, cut through the left and right optic stalks,
and extract the brain carefully from the head capsule.
3. Take a modified glass pipette and suck up FCS repeatedly to
coat the inside. This prevents tissue adhering to the glass.
Use the modified pipette to now suck up single heads/brains
and transfer to an ice-cold 1 % methylene blue solution in
0.1 M PBS for 5 min. to stain the tissue surface and thus facili-
tate visual orientation of tissue within the frozen blocks for
sectioning.
4. Transfer the heads/brains, unfixed, into the wells containing
Tissue Freezing Medium®. Shock freeze the wells by placing
them in a cuvette containing 2-methylbutane maintained at
−20 °C. Do not add sucrose as this adversely affects the quality
of later dye coupling.
5. Orient the frozen block containing the head/brain in the
Cryostat according to the desired plane of sectioning. We gen-
erally sectioned horizontally with respect to the body axis and
at 16 μm thickness using a Leica low profile 819 microtome
blade. Replace this blade as required. This slice thickness
allowed individual cells surrounding the central body of the
brain to be easily targeted for intracellular dye injection under
DIC optics and without damaging primary glial processes.
6. Each brain slice (still frozen and attached to the blade) is trans-
ferred by direct application onto a separate frozen Superfrost®
Plus (Menzel-Gläser) microscope slide. For older embryos the
depth of the brain could mean that 20 or so slides are neces-
sary. This 1:1 procedure is important because it means that
only one brain slice is being investigated (at room tempera-
ture) at any given time. Any additional brain slices on the slide
would dry out during the experiment.
7. Draw a complete ring around the section with the Pap pen
for frozen slides. This ring will limit the spread of the saline
solution covering the tissue slice and allow maximum fluid
contact between the slide and the water-immersion objective.
106 George Boyan and Yu Liu
3.2 Intracellular 1. In preparation for dye injection, transfer the frozen slide with
Dye Injection its single brain slice to the fixed-stage microscope. Immediately
cover with physiological saline at room temperature to a depth
of at least 5 mm to allow good contact with the immersion
objective. Too little fluid and the surface tension forces gener-
ated by the objective might tear the slice from the slide. Allow
1–2 min for temperature acclimatization. Confirm using DIC
optics at ×10 magnification that the preparation is centered
within the optical field of the microscope. This is critical because
once the ×63 objective is in use, repositioning of the slice is
limited by the restricted visual field offered by the objective.
2. Take the Eppendorf containing FCS from the refrigerator and
allow to come to room temperature.
3. Remove an electrode carefully from the array within the petri
dish (too sudden a movement may cause the tip to snap off!).
Fill the electrode with the fluorochrome solution by placing its
blunt end vertically into the solution and allowing it to travel
via capillary action along the internal filament into the tip.
This will take about 1–2 min. The entire electrode should then
be filled with 200 mM KCl from its blunt end via the fine
syringe needle to enable a good electrical contact with the Ag/
AgCl electrode within the electrode holder. Dip the electrode
tip several times into the FCS to act as a lubricant facilitating
penetration and subsequent removal of the electrode from the
target cell.
4. Slide the electrode into the perspex microelectrode holder
(1.0 mm diameter, WPI Inc., FL, USA). Ensure there are no
salt residues on the electrode holder by wiping down with a
tissue wetted with aqua dest.
5. Position the electrode holder into an appropriate microma-
nipulator equipped with adjustment (manual or motorized) of
X, Y, Z axes. The shielded cable from the microelectrode
holder is connected to the positive pole on the head stage of a
DC amplifier (in our case a Getting 5) equipped with current
passing facility via a virtual ground circuit.
6. Place the tip of the chlorided silver wire acting as a reference
electrode into the physiological solution covering the prepa-
ration and connect to the negative pole of the amplifier head
stage.
Dye Coupling Between Glia in Brain Slices 107
Fig. 1 Intracellular injection of Alexa Fluor® 568 reveals dye-coupled glia cells associated with the central
complex of the embryonic grasshopper brain. Photomicrographs of horizontal embryonic brain slices: (a–c)
normal, (d–f) after addition of bathing medium containing 1 × 10−5 M n-heptanol (a gap junctional blocker).
(a, d) DIC images show dye-filled (purple) electrode (white arrowheads) on penetration of a single target cell.
Central complex neuropils (CB central body, EB ellipsoid body, N nodulus) are outlined dashed white. Arrow
indicates anterior (a). Combined transmitted light/fluorescence images show the brain slices on commence-
ment (b, e: 0 min) and after 10 min. (c, f) Dye injection. Note the large collection of dye-coupled cells normally
(c) and their complete absence when n-heptanol is present (f). Scale bar represents 70 μm. Panels modified
from ref. 26
3.3 Gap Junctional 1. Alexa Fluor® 568 has a small molecular weight [41] so that its
Blocker passage between glia cells might be mediated by gap junctions.
We tested this possibility by applying TES buffer containing
1 × 10−5 M n-heptanol—a proven gap junctional blocker
(see refs. 28, 29)—and then repeating the dye injection experi-
ment as described above (Fig. 1d).
2. In each experiment involving n-heptanol we found that only
the penetrated cell was stained after 10 min of injection with
Alexa Fluor® 568 (Fig. 1e). This shows that the injection of the
dye itself was not affected by the presence of n-heptanol.
Complete absence of dye coupling (Fig. 1f) supports the
presence of gap junctions between the labeled cells in our
brain slices.
Dye Coupling Between Glia in Brain Slices 109
Fig. 2 Immunolabeling confirms glial identity of cells. (a) Confocal image of a brain
slice following agarose embedding and double immunolabeling shows cells
associated with the central body (CB) to be glutamine synthetase (GS) positive
(green)/HRP negative (red). Two cells (white stars) direct glia podia around the CB
(open white arrowhead) as well as (white arrowhead) into neighboring neuropil
of the protocerebrum (PC). Arrow indicates anterior (a). (b, c) All GS-positive cells
(green) associated with the ellipsoid body (EB) (b), and central body (CB) (c),
co-express the glial-specific nuclear antigen Repo (red, white stars) confirming
their identity as astrocyte-like glia. Note the extensive GS-positive gliopodia
(white arrowheads) surrounding these neuropils. (d–f) Confocal images confirm
glial identity of a pair of dye-coupled cells (black, white stars) associated with the
central body (CB) following Alexa Fluor® 568 injection in a brain slice. (d) Red
channel shows dye-injected (black star) and dye-coupled (white star) cells
following dye injection. (e) Green channel shows both cells to be glutamine syn-
thetase positive. (f) Combined channels confirm co-labeling (yellow) of anti-GS
and Alexa and therefore glial identity of both cells. Scale bar in (a) represents
12 μm, 25 μm in (b), 20 μm in (c–f). Panel (a) modified from ref. 26; panels (b, c)
modified from ref. 9
4 Notes
Acknowledgements
References
1. Jones BW, Fetter RD, Tear G, Goodman CS barrier of an insect: recordings from identified
(1995) Glial cells missing: a genetic switch that neuroglia. J Exp Biol 109:307–318
controls glial versus neuronal fate. Cell 82: 11. Swales LS, Lane NJ (1985) Embyronic develop-
1013–1023 ment of glial cells and their junctions in the locust
2. Jones BW (2001) Glial cell development in the central nervous system. J Neurosci 5:117–127
Drosophila embryo. Bioessays 23:877–887 12. Schmidt J, Deitmar JW (1996) Photoin-
3. Hidalgo A (2003) Neuron-glia interactions activation of the giant neuropil glial cells in the
during axon guidance in Drosophila. Biochem leech Hirudo medicinalis: effects on neuronal
Soc Trans 31:50–55 activity and synaptic transmission. J Neuro-
4. Klämbt C (2009) Modes and regulation of physiol 76:2861–2871
glial migration in vertebrates and invertebrates. 13. Alexopoulos H, Böttger A, Fischer S et al
Nat Rev Neurosci 10:769–779 (2004) Evolution of gap junctions: the missing
5. Vanhems E (1995) Insect glial cells and their link? Curr Biol 14:879–880
relationships with neurons. In: Vernadakis A, 14. Phelan P, Goulding LA, Tam JLY et al (2008)
Roots B (eds) Neuron-Glia interrelations dur- Molecular mechanism of rectification at an
ing phylogeny: II. Plasticity and regeneration. identified electrical synapse in the Drosophila
Humana, Totowa, NJ, pp 49–77 giant fibre system. Curr Biol 18:1955–1960
6. Awasaki T, Lai S-L, Ito K, Lee T (2008) 15. Koussa MA, Tolbert LP, Oland LA (2011)
Organization and postembryonic development Development of a glial network in the olfactory
of glial cells in the adult central brain of nerve: role of calcium and neuronal activity.
Drosophila. J Neurosci 28:13742–13753 Neuron Glia Biol 6(4):245–261
7. Edwards TN, Meinertzhagen IA (2010) The 16. Zahs KR, Newman E (1997) Asymmetric gap
functional organisation of glia in the adult junctional coupling between glial cells in the
brain of Drosophila and other insects. Prog rat retina. Glia 20:10–22
Neurobiol 90:471–497 17. Newman EA (2001) Propagation of intercel-
8. Awasaki T, Lee T (2011) New tools for the lular calcium waves in retinal astrocytes and
analysis of glial cell biology in Drosophila. Glia Müller cells. J Neurosci 21:2215–2223
59:1377–1386 18. Theiss C, Meller K (2002) Aluminum impairs
9. Boyan G, Loser M, Williams L, Liu Y (2011) gap junctional intercellular communication
Astrocyte-like glia associated with the embry- between astroglial cells in vitro. Cell Tissue Res
onic development of the central complex in the 310:143–154
grasshopper Schistocerca gregaria. Dev Genes 19. Houades V, Koulakoff A, Ezan P et al (2008)
Evol 221:141–155 Gap junction-mediated astrocytic networks
10. Schofield PK, Swales LS, Trehern JE (1984) in the mouse barrel cortex. J Neurosci 28:
Potentials associated with the blood-brain 5207–5217
Dye Coupling Between Glia in Brain Slices 113
Molluscs
Chapter 8
Abstract
Representatives of the phylum Mollusca have long been important models in neurobiological research.
Recently, the routine application of immunocytochemistry in combination with confocal laser scanning
microscopy has allowed fast generation of highly detailed reconstructions of neural structures of even the
smallest multicellular animals, including early developmental stages. As a consequence, large-scale com-
parative analyses of neurogenesis—an important prerequisite for inferences concerning the evolution of
animal nervous systems—are now possible in a reasonable amount of time. Herein, we describe immuno-
cytochemical staining protocols for both whole-mount preparations of developmental stages—usually
70–300 μm in size—as well as for vibratome sections of complex brains. Although our procedures have
been optimized for marine molluscs, they may easily be adapted for other (marine) organisms by the
creative neurobiologist.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_8, © Springer Science+Business Media, LLC 2014
117
118 Andreas Wanninger and Tim Wollesen
2 Materials
2.1 Whole-Mount 1. Make solution for relaxation of live embryos and larvae. 7.14 %
Staining of Neural stock solution: 38.1 g MgCl2 × 6H2O, 100 mL water. Use
Tissue parts of it to make a 3.5–3.6 % MgCl2 solution.
2. 4 % Paraformaldehyde (PFA) in 0.1 M phosphate buffer (PB)
for fixation: 33.48 g of Na2HPO4 × 2H2O, 7.93 g
NaH2PO4 × H2O, and 100 mL H2O. Alternatively: 26.7 g
Na2HPO4, 6.9 g NaH2PO4, and 100 mL H2O. Adjust pH to
7.3. Dissolve 4 g of PFA powder in 100 mL PB or make a
1:10 dilution of custom-made 4 % PFA solution in PB (see
Notes 1 and 2).
3. Washing solution: 0.1 % NaN3 to PB is recommended to avoid
bacterial or fungal growth (not necessary for the fixative).
4. Many molluscan larvae bear shells, which are best decalcified
after fixation in 0.05 M EGTA (dissolve 1.9 g EGTA powder
in 100 mL H2O) (see Note 3).
5. PBT: 0.2–10 % Triton X-100 (concentration depending largely
on specimen size and tissue), 0.1 M PB + 0.1 % NaN3.
6. Block-PBT solution: 6 % normal goat serum (NGS) in PBT.
7. Antibody solution: dilute all primary and secondary anti-
bodies at the respective working concentration in Block-PBT
(see Note 4).
8. Fluorescent counterstains: e.g., DAPI (for nuclei), phalloidin
or phallacidin (for F-actin including musculature).
120 Andreas Wanninger and Tim Wollesen
3 Methods
11. Trim block with razor blade to pyramidal shape and cut off a
single corner. This will facilitate orientation of the sections
(see Fig. 3).
12. Fill tray with chilled 0.1 M PB.
13. Glue trimmed sample with instant glue on stage and let dry
for 10 s.
14. Submerse sample in 0.1 M PB and adjust thickness of sections
(see Note 12).
15. Adjust begin and end, frequency, and speed of sectioning
process.
16. Section trimmed block with a vibratome and collect all sections
of interest with forceps. Ensure that problematic hard parts
of the specimen are located at the distal end of each section.
By doing so, only a little portion of the specimen will rupture
in case that these parts are impossible to section.
17. Apply immunocytochemistry protocol to the sections of interest
as described above (see Subheading 3.2, steps 2–6).
18. Mount vibratome sections on coated objective slides.
19. Orient and dry sections with forceps and paper towels (do not
overdry).
20. Apply approximately 100 μl of mounting medium alongside
the objective slide. Gently cover sections with a cover slip and
avoid air bubbles.
21. Let sample harden in the dark at 4 °C for a few hours to sev-
eral days.
4 Notes
Acknowledgments
References
1. Haszprunar G, Wanninger A (2012) Molluscs. 10. Braubach OR, Dickinson AJG, Evans CCE,
Curr Biol 13:R510–R514 Croll RP (2006) Neural control of the velum
2. Bullock TH, Horridge GA (1965) Structure and in larvae of the gastropod, Ilyanassa obsoleta.
function in the nervous systems of invertebrates. J Exp Biol 209:4676–4689
Freeman and Company, San Francisco, p 1611 11. Voronezhskaya EE, Nezlin LP, Odintsova NA,
3. Lin MF, Leise EM (1996) Gangliogenesis in Plummer JT, Croll RP (2008) Neuronal devel-
the prosobranch gastropod Ilyanassa obsoleta. opment in larval mussel Mytilus trossulus
J Comp Neurol 374:180–193 (Mollusca: Bivalvia). Zoomorphology 127:
97–110
4. Shigeno S, Tsuchiya K, Segawa S (2001)
Embryonic and paralarval development of the 12. Todt C, Wanninger A (2010) Of tests, trochs,
central nervous system of the loliginid squid shells, and spicules: development of the basal
Sepioteuthis lessoniana. J Comp Neurol 437: mollusk Wirenia argentea (Solenogastres) and
449–475 its bearing on the evolution of trochozoan lar-
val key features. Front Zool 7:6
5. Chase R (2002) Behavior & its neural control
13. Wollesen T, Loesel R, Wanninger A (2009)
in gastropod molluscs. Oxford University
Pygmy squids and giant brains: mapping the
Press, Oxford, p 314
complex cephalopod CNS by phalloidin stain-
6. Nixon M, Young JZ (2003) The brains and ing of vibratome sections and whole-mount
lives of cephalopods. Oxford University Press, preparations. J Neurosci Methods 179:63–67
Oxford, p 392 14. Wollesen T, Cummins SF, Degnan BM,
7. Marois R, Carew TJ (1997) Projection pat- Wanninger A (2010) FMRFamide gene and
terns and target tissues of the serotonergic cells peptide expression during central nervous sys-
in larval Aplysia californica. J Comp Neurol tem development of the cephalopod mollusk,
386:491–506 Idiosepius notoides. Evol Dev 12:113–130
8. Friedrich S, Wanninger A, Brückner M, 15. Wollesen T, Nishiguchi MK, Seixas P, Degnan
Haszprunar G (2002) Neurogenesis in the BM, Wanninger A (2012) The VD1/RPD2
mossy chiton, Mopalia muscosa (Gould) α1-neuropeptide is highly expressed in the
(Polyplacophora): evidence against molluscan brain of cephalopod mollusks. Cell Tissue Res
metamerism. J Morphol 253:109–117 348:439–452
9. Wanninger A, Haszprunar G (2003) The devel- 16. Rodriguez J, Deinhardt F (1960) Preparation
opment of the serotonergic and FMRF-amidergic of a semipermanent mounting medium for
nervous system in Antalis entalis (Mollusca, fluorescent antibody studies. Virology 12:
Scaphopoda). Zoomorphology 122:77–85 316–317
Part IV
Xenopus Protocols
Chapter 9
Abstract
The dynamic expression pattern analysis provides the primary information of gene function. Differences of
the RNA and/or protein location will provide valuable information for gene expression regulation.
Generally, in situ hybridization (ISH) and immunohistochemistry (IHC) are two main techniques to
visualize the locations of gene transcripts and protein products in situ, respectively. Here we describe the
protocol for the whole brain dissection, the in situ hybridization and immunostaining of the developing
Xenopus brain sections. Additionally, we point out the modification of in situ hybridization for microRNA
expression detection.
Key words Expression pattern, Brain dissection, Section, Immunostaining, In situ hybridization,
Xenopus, microRNA
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_9, © Springer Science+Business Media, LLC 2014
129
130 Kai-li Liu et al.
2 Materials
2.2 Chemical Representative chemical reagents for ISH and IHC are listed as
Reagents follows:
1. HEPES, tricaine methanesulfonate (Sigma).
2. Formaldehyde.
3. Paraformaldehyde (PFA).
4. Tween-20.
5. Methanol.
6. Ethanol.
7. Sucrose.
8. OCT.
9. Paraffin.
10. Xylene.
11. Proteinase K.
12. BSA (BSA V; Sigma).
In Situ and Immunostaining in Xenopus 131
2.3 Solutions Solutions for in situ hybridization should be prepared using ultrapure
for In Situ water (18.2 MΩ at 25 °C). For IHC, double distilled water
Hybridization and (ddH2O) is feasible. Chemical reagents are the analysis pure:
Immunostaining 1. MMR: 0.1 M NaCl, 2.0 mM KCl, 1 mM MgSO4, 2 mM
CaCl2, 5 mM HEPES (pH 7.8), 0.1 mM EDTA. For 1 L 10×
MMR, add NaCl 58.44 g, KCl 1.49 g, MgCl2·6H2O 2.03 g,
CaCl2(anhyd) 2.22 g, and HEPES 11.92 g into 800 mL water,
mix dissolve and adjust pH to 7.5 with NaOH, and bring to
1 L with water. Autoclave and store at room temperature (RT).
2. MS222 anesthetic solution: MS222 0.2 mg/mL in 0.1×
MMR, pH 7.5–7.8.
3. 1× PBS: For 1 L 10× PBS, dissolve NaH2PO4·2H2O 2.89 g,
Na2HPO4·12H2O 26.73 g, and NaCl 102.2 g in 800 mL
ultrapure water, and then adjust pH to 7.4 with NaOH. Bring
volume to 1 L and autoclave. Store this solution at RT. Dilute
the stock solution with water before use and adjust pH if
necessary.
4. MEMFA: 0.1 M MOPS (pH 7.4), 2 mM EGTA, 1 mM
MgSO4, 3.7 % formaldehyde (see Note 1).
5. 4 % PFA/1× PBS: For 50 mL, dilute 10 mL 20% PFA with
5 mL 10× PBS with water prior to use (see Note 2).
6. 20 % Sucrose/1× PBS: For 50 mL, dissolve 10 g RNase-free
sucrose with 1× PBS, sterilize the solution through 0.45 μM
filter, and store at 4 °C.
132 Kai-li Liu et al.
3 Methods
2. When the embryos stop moving, place them into agarose plate
with precooling 1× PBS. Use the forceps with blunt tips to
hold the embryo at the trunk; use another forceps with sharp
tips to make a cut through the spinal cord at the dorsal truck
just behind the hindbrain. Tip up the skin of the front cut with
forceps and carefully peel off the skin of the dorsal head to
expose the brain. Then insert the tips of the forceps from the
cut to the bottom of the brain, and lift up the brain by a careful
forward movement of the forceps and shear the nerve bundles
with forceps tips or scissors. Transfer the dissected brain imme-
diately into fixation solution with a pipette or forceps.
3. For the brain dissection of developing frogs to adult, make the
frog anesthetized in the MS222 for around 20 min and then
place it on the ice. Cut off the spine and tear the skin from
foramen magnum toward the head with a sharp scissors and
then use bone clamp to open the skull and expose the brain
(see Note 8). The procedure of dissecting brain follows as above
(step 2). If this step takes long time, to prevent the decay of
brain tissue, add several drops of fixation solution onto the brain
to fix it in situ for 1 h before taking it out (see Note 9).
4. Fix the dissected brain in MEMFA or 4 % PFA at 4 °C over-
night (see Note 10).
5. For whole-mount in situ hybridization or paraffin section prepa-
ration. Then the brain could be dehydrated gradually by replacing
the fixation solution with 25 % ethanol/PBS, 50 % etha-
nol/50% PBS, 75% ethanol, 100% ethanol sequentially, 5–10 min
each; and stored in 100% ethanol at −20 °C (see Note 11).
For whole-mount immunohistochemistry, ethanol should be
replaced with methanol. For cryosection section preparation,
go on Subheading 3.2 directly without dehydration.
3.2 Cryosection 1. Pipette off the fixation solution and add precooling 1× PBS to
Preparation and wash for 3 times.
Pretreatment Prior to 2. Pipette off the PBS, and add 20 % sucrose/1× PBS to cryoprotect
Hybridization the brains at 4 °C for 4 h to overnight until the brains sink to
the bottom.
3. Label molds the name and the direction of specimens. Transfer
the brain(s) into the mold and remove the sucrose solution as
much as possible. Add appropriate volume (enough to immerse
the whole specimen) of OCT in the molds. Orientate the brains
in right directions with a long syringe needle, and then carefully
move the mold on the iron plate upon dry ice or into the 70 %
ethanol precooling at −80 °C. After the OCT has been com-
pletely frozen, store the block at −80 °C before sectioning.
4. Slightly tickle or press the mold to take out the frozen block,
mount the block on the specimen stand of cryostat at right
direction, and cut the edges to modify it into right shape before
134 Kai-li Liu et al.
3.3 Paraffin Section 1. Dissolve paraffin at 60 °C before the day of embedding, and
Preparation and keep it at 60 °C.
Pretreatment Prior to 2. Wash the stored dehydrated brains with 100 % ethanol, and
Hybridization then transfer specimens to xylene by gradually replacing the
solution with 75 % ethanol/25 % xylene, 50 % ethanol/50 %
xylene, 25 % ethanol/75 % xylene, and 100 % xylene, and shake
at RT for 5–10 min each time.
3. Wash the brains with new 100 % xylene, pour the specimens
with xylene into a small beaker (specimens should be immersed
in xylene), and incubate at 60 °C for 20 min (see Note 12).
4. Embedding: Add the same volume of prewarmed paraffin,
incubate at 60 °C for 45 min, and then replace with 100 %
paraffin, incubated at 60 °C for 20 min. Replace with 100 %
new paraffin, incubated at 60 °C for 3 h; wash specimens with
100 % new paraffin, incubate at 60 °C for 20 min; replace with
new paraffin, pour specimens with paraffin into prewarmed
mold or plastic disc, and set the specimens at the right orienta-
tion and positions with prewarmed needle. Turn off the oven,
and leave the paraffin block with specimens to be solidified
slowly in the oven overnight (see Note 13).
5. Modify the embedded block and cut 5–10 μm sections in a
microtome. Transfer sections on the surface of 0.2 % ethanol
on slides prewarmed at 37 °C (see Note 14). Discard the solu-
tion when sections have been completely extended, dry the
sections at 37 °C overnight, and then store the slides at 4 °C.
6. For hybridization or immunostaining. Dewax the section in
100 % xylene by three washes, 10 min each time, 100 % etha-
nol wash twice, and then rehydrate the section by washes with
graded alcohol sequentially reverse to the dehydration, 2 min
each wash.
3.4.2 Washing Steps 1. Set a glass trough with washing solution stand in water bath.
and Antibody Visualization Warm up the solution and the glass trough to the hybridization
temperature. Add 100 mL washing solution to a glass trough
for ten slides.
2. Transfer slides to the glass trough with prewarmed washing
solution (see Note 17), and take away the coverslips after they
detach off the slides (usually in 1–2 min). Then, for ordinary
probe, make two washes at 65 °C with stirring or shaking,
30 min each; for LNA probe, make two washes in 2× SSC at
RT, 10 min each, followed with another wash in 0.2× SSC at
RT for 10 min.
3. Wash the slides 2 times in MABT at RT, 30 min each for ordinary
probe, or five times in PBT, 3 min each, for LNA probe.
4. Take the slide out one by one with forceps, wipe away the
excessive solution with filter paper, and then lay the slides on
the holder of the humid chamber with filter paper soaked in 1×
PBS or water. Add around 1mL blocking buffer per slide to
cover all the sections (without coverslip), and block for a half
to 1 h at RT.
5. Discard the blocking buffer, add 150 μL antibody mix (anti-
body diluted 1:2,000 to 1:8,000 in blocking buffer) per slide,
cover with a piece of parafilm cut as the same size of coverslip,
and incubate the slide in a humid chamber at RT overnight.
6. Discard the antibody solution, transfer the slides in a glass
trough with MABT, and make five washes on a shaker, 30 min
each wash at RT (see Note 18), followed with another five
washes in PBT, 5 min each at RT.
7. Staining: Wash the slides three times in alkaline-phosphatase
buffer for 5 min each at RT. Take out the slides and set them
in a humid chamber, add 150 μL NBT/BCIP or Fast Red
staining buffer per slide, cover with parafilm, and then develop
136 Kai-li Liu et al.
Fig. 1 In situ hybridization (ISH) of st.46 Xenopus brain sections for detecting tran-
scripts of zic2 (a, b), ath3/neurod4 (c), microRNA-181 (d). ISH for Zic2 was carried
out in cryosection, stained by using AP substrate Fast Red (red), which shows
expressed in the dorsal region of the developing brain (b), the nuclei of which were
counterstained with Hoechst (a). ISH for ath3/neurod4 and microRNA-181 were
conducted in paraffin sections and stained by applying AP substrate BM purple
(c, d). Ath3/neurod4 was expressed higher in ventricular zone (proximal) while lower
in marginal zone (distal) of developing brain/neural tube (c). microRNA-181 is rarely
detectable in developing brain while relatively highly expressed in the retina inner
nuclear layer (d). Br brain, Re retina, Vz ventricular zone, Mz marginal zone
4 Notes
Fig. 2 Immunostaining of Xenopus brain sections for detecting protein locations of N-tubulin (a, b) and
phosphohistone-H3 (pH3) (c–e). IHC for n-tubulin was carried out in cryosection in (a, b), stained by TRITC-
conjugated secondary antibody showing red fluorescence and expressed in the whole developing brain/neural
tube and otic vesicle (b), the nuclei of which were counterstained with Hoechst (a). IHC for pH3 was conducted
in paraffin sections and stained with TRITC-conjugated secondary antibody (c, d) and HRP substrate AEC (e).
pH3 is specifically expressed in the proliferating cells in the midline of the developing brain as shown by the
arrows. Br brain, Ov otic vesicle
Acknowledgments
References
1. Harland RM (1991) In situ hybridization: an 6. Decembrini S, Bressan D, Vignali R, Pitto L,
improved whole-mount method for Xenopus Mariotti S, Rainaldi G, Wang X, Evangelista M,
embryos. Methods Cell Biol 36:685–695 Barsacchi G, Cremisi F (2009) MicroRNAs cou-
2. Lupo G, Liu Y, Qiu R, Chandraratna RA, ple cell fate and developmental timing in retina.
Barsacchi G, He RQ, Harris WA (2005) Proc Natl Acad Sci USA 106(50):21179–21184
Dorsoventral patterning of the Xenopus eye: 7. Liu K, Liu Y, Mo W, Qiu R, Wang X, Wu JY,
a collaboration of Retinoid, Hedgehog and He R (2011) MiR-124 regulates early neuro-
FGF receptor signaling. Development 132: genesis in the optic vesicle and forebrain,
1737–1748 targeting NeuroD1. Nucleic Acids Res 39:
3. Liu Y, Lupo G, Marchitiello A, Gestri G, He 2869–2879
RQ, Banfi S, Barsacchi G (2001) Expression of 8. Qiu R, Liu Y, Wu JY, Liu K, Mo W, He R
the Xvax2 gene demarcates presumptive ventral (2009) Misexpression of miR-196a induces eye
telencephalon and specific visual structures in anomaly in Xenopus laevis. Brain Res Bull
Xenopus laevis. Mech Dev 100:115–118 79:26–31
4. Lan L, Liu M, Liu Y, He R (2007) Expression 9. Qiu R, Liu K, Liu Y, Mo W, Flynt AS, Patton
and antibody preparation of POU transcription JG, Kar A, Wu JY, He R (2009) The role of
factor qBrn-1. Protein Pept Lett 14:7–11 miR-124a in early development of the Xenopus
5. Lan L, Liu M, Liu Y, Zhang W, Xue J, Xue Z, eye. Mech Dev 126:804–816
He R (2006) Expression of qBrn-1, a new 10. Sive HL, Grainger RM, Richard RM (2002)
member of the POU gene family, in the early Early development of Xenopus laevis: a labora-
developing nervous system and embryonic tory manual. Cold Spring Harbor Laboratory
kidney. Dev Dyn 235:1107–1114 Press, Cold Spring Harbor, NY
In Situ and Immunostaining in Xenopus 141
11. Nieuwkoop PD, Faber J (1994) Normal table 12. Kloosterman WP, Wienholds E, de Bruijn E,
of Xenopus laevis (Daudin): a systematical and Kauppinen S, Plasterk RH (2006) In situ detec-
chronological survey of the development from tion of miRNAs in animal embryos using LNA-
the fertilized egg till the end of metamorphosis. modified oligonucleotide probes. Nat Methods
Garland, New York 3:27–29
Chapter 10
Abstract
Microinjection has a long and distinguished history in Xenopus and has been used to introduce a surprisingly
diverse array of agents into embryos by both intra- and intercellular means. In addition to nuclei, investiga-
tors have variously injected peptides, antibodies, biologically active chemicals, lineage markers, mRNA,
DNA, morpholinos, and enzymes. While enumerating many of the different microinjection approaches
that can be taken, we will focus upon the mechanical operations and options available to introduce mRNA,
DNA, and morpholinos intracellularly into early stage embryos for the study of neurogenesis.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_10, © Springer Science+Business Media, LLC 2014
143
144 Cristine Smoczer et al.
1.1 Intercellular The object of intercellular injection has been to introduce agents
Injection either between germinal layers or into the blastocoel.
1.1.2 Blastocoel Peptides have been injected into the blastocoel either to activate
[6–8] or to competitively block cell surface or matrix-mediated
pathways [8].
A similar approach that used antibodies as a blocker has also
been successfully used [9]. Other, more complex mixtures contain-
ing biological agents have been tested in this manner, for example,
the contents of germinal vesicles [10].
1.2.1 Exogenous DNA Several decades ago, linearized DNA was microinjected into fertil-
ized Xenopus eggs to study tissue-specific expression of the intro-
duced gene [14]. Although a majority of the transgenic embryos
were highly mosaic, a very small percentage survived to adulthood
and transmitted the transgene to the next generations [15, 16].
1.2.2 REMI (Restriction Many years passed following pioneering nuclear transfer studies
Enzyme-Mediated [1, 17] before nuclear transplantation coincident with the introduction
Integration) of plasmid evolved to facilitate the generation of transgenic frogs [18].
Microinjection Manipulations in the Elucidation… 145
1.2.5 Lineage Markers Several strategies are available to deploy fluoresceinated lineage
markers. Intracellular injection of fluorescently labelled hydrophilic
dextrans has a well-tried history [28, 29]. An advantage that dex-
trans confer is that they tend to diffuse relatively well throughout
the injected cell, and yet they remain bounded by their large
molecular mass to that cell and its progeny. Tissues derived from
injected embryos can be combined with wild type or differently
injected embryos via heterotypic grafting [12, 30]. Alternatively,
the mRNA for GFP [31] or fluorescent fusion markers [32] can be
deployed either by co-injection or as bi-cistronic transcripts derived
from IRES vectors.
1.2.6 Biologically Transcripts are routinely injected into Xenopus embryos to perturb
Active In Vitro neurological development. Ectopic gain-of-function experiments
Synthesized mRNAs are straightforward [33]. Alternatively, chimeric constructs that
deploy fused activator (VP16) or repressor (engrailed) domains are
used to deliver phenotypes that can reveal whether a candidate
gene exerts an activating or repressive effect [34]. Mutations can
be introduced to alter the behavior or to define the role of encoded
sub-domains. For example, a point mutation introduced to Mix.1
impaired the DNA binding functions of its encoded homeodomain
[34]. Finally, mRNA “rescue” experiments can be mounted to
assess hierarchical relationships between genes. If morpholino-
mediated knockdowns or chimeric activator/repressor transcripts are
introduced, then the capacity of potential downstream signalling
partners can be assessed by co-injecting them in complementation
studies. All of these approaches suffer some disadvantages: transcripts
will be nonspecifically distributed throughout all the progeny of an
injected blastomere; ectopic mRNAs have a limited lifespan—their
sensitivity to degradation limits their utility to early developmental
stages; and different species of transcript might vary with regard to
longevity and distribution pattern in a context-specific manner.
2 Materials
2.1 Frogs 1. Xenopus laevis males and females (Xenopus I, Michigan, USA).
and Embryos Wild caught specimens preferred.
2. Human chorionic gonadotropin (HCG) (AkzoNobel or Sigma).
Prepare a 1,000 U/ml solution in the manufacturer provided buffer.
Unused remainder can be stored at −20 °C for several weeks.
Microinjection Manipulations in the Elucidation… 147
2.2 Solutions Stock Part A (20×): 1.16 M NaCl, 13.4 mM KCl, 16.6 mM
MgSO4, 6.7 mM Ca(NO3)2, pH to 7.4.
2.2.1 Steinberg’s
Solution Stock Part B (20×): 92.5 mM Tris base, 0.08 N HCl.
Prepare both parts using distilled water, autoclave, and store at
room temperature.
Dilutions of stock solution for typically used Steinberg’s solutions:
200 % 100 % 80 % 20 %
Part A per liter 100 ml 50 ml 40 ml 10 ml
Part B per liter 100 ml 50 ml 40 ml 10 ml
2.2.2 MBS (Modified Part A. 10× MBS: 880 mM NaCl, 25 mM NaHCO3, 10 mM KCl,
Barth’s Saline, Stored in 50 mM HEPES of pH 7.8, 10 mM MgSO4. Adjust pH to 7.8 with
Two Parts) NaOH, top up with distilled water, and then autoclave. Store
at room temperature and use within the month.
Part B. 0.1 M CaCl2—into 7 ml aliquots. Store frozen.
On the day of experiments, prepare 1× MBS by adding 100 ml of
10× MBS and 7 ml 0.1 M CaCl2 to distilled water—volume
adjusted to 1 l.
2.2.4 Ficoll Ficoll 400 is dissolved in 0.3× MBS in a ratio 2–5 % weight to
volume, depending upon the needle size. Typically, RNA and
morpholino injections deploy very fine needles and embryos only
require 2–3 % Ficoll for structural support during injection and
recovery. Larger needles required for nuclear transfer will require
5 % Ficoll. Prepare the morning of use and refrigerate to 12–13 °C.
148 Cristine Smoczer et al.
2.3 Glass Needles 1. Microinjection needles. Drawn from borosilicate glass capillaries
(1 mm outside diameter and 0.58 mm inside diameter—
Harvard Apparatus Ltd., Edenbridge, UK, or Drummond
3-000-203-G/X) using a microneedle puller (such as model
PC-10, Narishige, Tokyo, Japan) (see Note 1).
2. 1 cc inoculation (tuberculin) syringe with a 26-gauge needle.
3. Mineral oil (Eppendorf).
4. Micromanipulator.
5. Positive displacement injector (e.g., Drummond Nanoject
II—see Note 2).
6. Stereomicroscope.
7. Paraplast film or glass slide.
3 Methods
3.1 Preparation of 1. Select two females exhibiting prominent red cloacae. Inject
Frogs and Fertilization each with 0.75 cc HCG and incubate overnight (if frogs are to
of Eggs [38] be kept at 18 °C, inject at 6:00 pm in the afternoon the day
before needed or at 11:00 if females are to be held at room
temperature 22–23 °C).
2. Next morning, check to see if the females are shedding eggs.
If they are, then anesthetize the male in tricaine methanesulfo-
nate (MS-222) and sacrifice by cervical section or decapitation.
Remove the testes to 200 % Steinberg’s solution and keep cool
either on ice or in a refrigerator.
3. Macerate one quarter testes in 2 ml of 80 % Steinberg’s solution
and draw up and down in 1 cc syringe (no needle), to break the
tissue up further and to evenly distribute the sperm.
4. Strip eggs from the female into a Petri dish and add 0.5 cc
sperm suspension. Leave at room temperature for 10 min.
5. After 10 min has elapsed, flood the fertilized embryos with
0.1× MBS.
6. In the meantime, prepare fresh 2 % L-cysteine solution.
7. Successfully fertilized embryos rotate so that their dark animal
pole faces up, and the cortex visibly contracts.
8. Once rotation has occurred, remove the embryos to a vessel
for de-jellying (finger bowl or 80 ml beaker). Remove as much
of the 0.1× MBS as easily possible, and flood with L-cysteine
solution. Swirl them very gently periodically. After 2–4 min, the
jelly should have dissolved, and the eggs will pack close together
in the bottom of the beaker when it is tilted.
9. Immediately decant the bulk of the solution being careful
never to fully expose the embryos to air, and gently replace
Microinjection Manipulations in the Elucidation… 149
10. Repeat this 9 more times taking great care not to violently
agitate the embryos.
11. Remove to a Petri dish of 0.1× MBS, and cool the embryos
to 14 °C.
12. For subsequent manipulation/transfers of embryos, we use a
Pasteur pipette that has been cut and flame polished at the end
leaving an opening approximately 1.5–2 mm wide (wider that
an embryo by a generous margin).
Good quality and undamaged embryos will look round and
firm and will not adhere to the plastic (see Note 3).
3.3 Preparation Morpholinos can be obtained through Gene Tools LLC, and their
of Injectables administration and qualities are described elsewhere in this
Morpholinos volume.
plate and then decanting the solution until the Ficoll has been
largely replaced. Embryos may float, but can be immersed
again by dripping fluid gently overtop.
11. At this point survey the embryos under a stereomicroscope
and remove all dead or nondividing embryos. Remove the
surviving embryos to a fresh dish of 0.1× MBS. Continue to
incubate at 12–13 °C.
12. Each day, be sure to change the 0.1× MBS and to remove dead
embryos before they render the media toxic.
13. If a fluorescent marker has been employed, use a fluorescence
stereomicroscope to remove unsuccessfully injected embryos
from the plates. Incubate the remainder to the developmental
stage desired.
4 Notes
1. Needles can also be pulled manually over a small flame, but this
takes practice and skill. Affix a modified large gauge needle to
a stand, and use it as a miniature gas burner to heat a small
region of a glass capillary. Be careful to keep flammables away
from the metal that can become very hot. Roll the capillary
between your fingers above the flame until the glass become
soft and ductile, and then pull the two halves apart while lifting
the glass away from the flame. This takes caution, care, and
practice.
2. We have previously employed a homemade air pressure
displacement rig to inject embryos. This is accomplished by
attaching a “y” of tubing to a source of pressurized air. One
arm of the “y” is partially clamped, and the other goes to the
glass needle held in a micromanipulator. A glass needle is back-
filled with injectable fluid and then plugged into one end of
the air delivery tube. The clamp on the other arm is adjusted
to allow the bulk of air to escape while permitting the fluid to
remain in the needle. When delivery of injection fluid is desired,
the partially clamped tube is pinched by folding to prevent any
air from bleeding out, and this in turn raises pressure sufficient
to expel injection fluid. Injection volumes are calibrated by
time and diameter, and recalibration has to be repeated peri-
odically. The process is slow and cumbersome, but surprisingly
accurate and reproducible.
3. Flattened or floppy eggs/embryos or sticking to the bottom of
the Petri dish reflect damaged materials. Possible causes include
poor fertilization (check sperm motility and Steinberg’s solu-
tion pH), poor egg quality from the female, too much time in
L-cysteine, incomplete washing in 0.1× MBS after de-jellying,
Microinjection Manipulations in the Elucidation… 153
Acknowledgment
References
1. Briggs R, King TJ (1952) Transplantation of embryos. Proc Natl Acad Sci USA 91(8):
living nuclei from blastula cells into enucleated 3049–3053
frogs’ eggs. Proc Natl Acad Sci USA 38(5): 9. Purcell L, Gruia-Gray J, Scanga S, Ringuette
455–463 M (1993) Developmental anomalies of
2. Gurdon JB (2006) Nuclear transplantation in Xenopus embryos following microinjection
Xenopus. Methods Mol Biol 325:1–9 of SPARC antibodies. J Exp Zool 265(2):
3. Drysdale TA, Crawford MJ (1994) Effects of 153–164
localized application of retinoic acid on 10. Kao KR, Elinson RP (1985) Alteration of the
Xenopus laevis development. Dev Biol 162(2): anterior-posterior embryonic axis: the pattern
394–401 of gastrulation in macrocephalic frog embryos.
4. Dolbeare F (1995) Bromodeoxyuridine: a Dev Biol 107(1):239–251
diagnostic tool in biology and medicine, part II: 11. Shi J, Severson C, Yang J, Wedlich D,
oncology, chemotherapy and carcinogenesis. Klymkowsky MW (2011) Snail2 controls
Histochem J 27(12):923–964 mesodermal BMP/Wnt induction of neural
5. Roy S, Gardiner DM, Bryant SV (2000) crest. Development 138(15):3135–3145
Vaccinia as a tool for functional analysis in 12. Khosrowshahian F, Wolanski M, Chang WY,
regenerating limbs: ectopic expression of Shh. Fujiki K, Jacobs L, Crawford MJ (2005) Lens
Dev Biol 218(2):199–205 and retina formation require expression of
6. Otto C, Schutz G, Niehrs C, Glinka A (2000) Pitx3 in Xenopus pre-lens ectoderm. Dev Dyn
Dissecting GHRH- and pituitary adenylate 234(3):577–589
cyclase activating polypeptide-mediated sig- 13. Zuber ME, Gestri G, Viczian AS, Barsacchi G,
nalling in Xenopus. Mech Dev 94(1–2): Harris WA (2003) Specification of the verte-
111–116 brate eye by a network of eye field transcription
7. Cooke J, Smith JC (1989) Gastrulation and factors. Development 130(21):5155–5167
larval pattern in Xenopus after blastocoelic 14. Rusconi S, Schaffner W (1981) Transformation
injection of a Xenopus-derived inducing factor: of frog embryos with a rabbit beta-globin gene.
experiments testing models for the normal Proc Natl Acad Sci USA 78(8):5051–5055
organization of mesoderm. Dev Biol 131(2): 15. Etkin LD, Pearman B, Ansah-Yiadom R (1987)
383–400 Replication of injected DNA templates in
8. Saint-Jeannet JP, Dawid IB (1994) Vertical Xenopus embryos. Exp Cell Res 169(2):
versus planar neural induction in Rana pipiens 468–477
154 Cristine Smoczer et al.
16. Etkin LD, Pearman B (1987) Distribution, 28. Wetts R, Fraser SE (1991) Microinjection of
expression and germ line transmission of exog- fluorescent tracers to study neural cell lineages.
enous DNA sequences following microinjec- Development Suppl 2:1–8
tion into Xenopus laevis eggs. Development 29. Gimlich RL, Braun J (1985) Improved fluo-
99(1):15–23 rescent compounds for tracing cell lineage.
17. Gurdon JB (1962) The transplantation of Dev Biol 109(2):509–514
nuclei between two species of Xenopus. Dev 30. Keller R, Tibbetts P (1989) Mediolateral cell
Biol 5:68–83 intercalation in the dorsal, axial mesoderm of
18. Kroll KL, Amaya E (1996) Transgenic Xenopus laevis. Dev Biol 131(2):539–549
Xenopus embryos from sperm nuclear trans- 31. Zernicka-Goetz M, Pines J, Ryan K, Siemering
plantations reveal FGF signaling requirements KR, Haseloff J, Evans MJ, Gurdon JB (1996)
during gastrulation. Development 122(10): An indelible lineage marker for Xenopus using
3173–3183 a mutated green fluorescent protein.
19. Amaya E, Kroll KL (1999) A method for gen- Development 122(12):3719–3724
erating transgenic frog embryos. Methods Mol 32. Itoh K, Sokol SY (2011) Polarized transloca-
Biol 97:393–414 tion of fluorescent proteins in Xenopus ecto-
20. Bronchain OJ, Hartley KO, Amaya E (1999) A derm in response to Wnt signaling. J Vis Exp
gene trap approach in Xenopus. Curr Biol (51). doi:2700 [pii] 10.3791/2700
9(20):1195–1198 33. Wolanski M, Khosrowshahian F, Kelly LE,
21. Sparrow DB, Latinkic B, Mohun TJ (2000) El-Hodiri HM, Crawford MJ (2009) xArx2: an
A simplified method of generating trans- aristaless homolog that regulates brain regional-
genic Xenopus. Nucleic Acids Res 28(4): ization during development in Xenopus laevis.
E12 Genesis 47(1):19–31
22. Loeber J, Pan FC, Pieler T (2009) Generation 34. Lemaire P, Darras S, Caillol D, Kodjabachian L
of transgenic frogs. Methods Mol Biol 561: (1998) A role for the vegetally expressed
65–72 Xenopus gene Mix.1 in endoderm formation
23. Ogino H, McConnell WB, Grainger RM (2006) and in the restriction of mesoderm to the mar-
High-throughput transgenesis in Xenopus ginal zone. Development 125(13):2371–2380
using I-SceI meganuclease. Nat Protoc 1(4): 35. Bayramov AV, Martynova NY, Eroshkin FM,
1703–1710 Ermakova GV, Zaraisky AG (2004) The home-
24. Pan FC, Chen Y, Loeber J, Henningfeld K, odomain-containing transcription factor X-nkx-
Pieler T (2006) I-SceI meganuclease-mediated 5.1 inhibits expression of the homeobox gene
transgenesis in Xenopus. Dev Dyn 235(1): Xanf-1 during the Xenopus laevis forebrain
247–252 development. Mech Dev 121(12):1425–1441
25. Allen BG, Weeks DL (2005) Transgenic 36. Pei W, Noushmehr H, Costa J, Ouspenskaia
Xenopus laevis embryos can be generated using MV, Elkahloun AG, Feldman B (2007) An
phiC31 integrase. Nat Methods 2(12): early requirement for maternal FoxH1 during
975–979 zebrafish gastrulation. Dev Biol 310(1):10–22
26. Allen BG, Weeks DL (2006) Using phiC31 37. Sive H, Grainger RM, Harland RM (2000)
integrase to make transgenic Xenopus laevis Early development of Xenopus laevis. A labora-
embryos. Nat Protoc 1(3):1248–1257 tory outline, 1st edn. Cold Spring Harbor
27. Sekkali B, Tran HT, Crabbe E, De Beule C, Laboratory Press, Cold Spring Harbor
Van Roy F, Vleminckx K (2008) Chicken beta- 38. Drysdale TA, Elinson RP (1991) Development
globin insulator overcomes variegation of trans- of the Xenopus laevis hatching gland and its
genes in Xenopus embryos. FASEB J 22(7): relationship to surface ectoderm patterning.
2534–2540 Development 111:469–478
Chapter 11
Abstract
Antisense morpholino oligonucleotides (MOs) have become a valuable method to knock down protein
levels, to block mRNA splicing, and to interfere with miRNA function. MOs are widely used to alter gene
expression during development of Xenopus and zebra fish, where they are typically injected into the fertil-
ized egg or blastomeres. Here, we present methods to use electroporation to target delivery of MOs to the
central nervous system of Xenopus laevis or Xenopus tropicalis tadpoles. Briefly, MO electroporation is
accomplished by injecting MO solution into the brain ventricle and driving the MOs into cells in the brain
with current passing between two platinum plate electrodes, positioned on either side of the target brain
area. The method is straightforward and uses standard equipment found in many neuroscience labs. A
major advantage of electroporation is that it allows spatial and temporal control of MO delivery and there-
fore knockdown. Co-electroporation of MOs with cell-type specific fluorescent protein expression plas-
mids allows morphological analysis of cellular phenotypes. Furthermore, co-electroporation of MOs
with rescuing plasmids allows assessment of specificity of the knockdown and phenotypic outcome. By
combining MO-mediated manipulations with sophisticated assays of neuronal function, such as electro-
physiological recording, behavioral assays, or in vivo time-lapse imaging of neuronal development,
the functions of specific proteins and miRNAs within the developing nervous system can be elucidated.
These methods can be adapted to apply antisense morpholinos to study protein and RNA function in a
variety of complex tissues.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_11, © Springer Science+Business Media, LLC 2014
155
156 Jennifer E. Bestman and Hollis T. Cline
2 Materials
Grass Stimulator
Picospritzer
micropipette electrode
holder
Fig. 1 Configuration of electroporation equipment. (a) A typical setup of the apparatus needed to electroporate
includes a Grass Stimulator square pulse generator, a Picospritzer pressure injector, dissecting microscope,
and micromanipulators to hold the electrode and micropipette for injection. (b) Simple wiring diagram and
settings for the Grass Stimulator and Picospritzer and diagram to indicate the position of the injection micro-
pipette and the plates of the electrode
Morpholino Studies in Xenopus Brain Development 159
Fig. 2 Custom platinum electrode. (a) The plates of the electrode are constructed out of platinum foil that is
soldered to insulated wire leads. The plates are isolated from one another with shrink tubing. A larger piece of
shrink tubing holds the aligned plates in place, and a still larger piece of shrink tubing holds the electrode onto
the rod that fits in the micromanipulator. Scale bar = 5 mm. (b) A magnified view of the tip of the electrode
showing the folded platinum foil that is folded and cut into ~1 mm wide plates. The bracket indicates the
shrink tubing that insulates electrodes from one another. Scale bar = 2 mm
3 Methods
3.1 Animal Tadpoles are reared as groups of ~100 in large bowls at 23 °C with
Husbandry a 12 h light/12 h dark diurnal cycle. For our experiments, we typi-
cally use animals that are between Nieuwkoop and Faber stage 46
(5 days postfertilization, dpf) and stage 49 (~12 dpf) [26], though
electroporation is effective on younger and old animals. Once
experiments begin, single tadpoles are kept in 6-well tissue culture
plates so that time-lapse images from identified individual tadpoles
can be acquired. All procedures are conducted with Xenopus tad-
poles anesthetized with 0.02 % MS-222 solution. Before electro-
poration or imaging protocols, the tadpoles are transferred into
the MS-222 solution, and within minutes, they are immobile,
162 Jennifer E. Bestman and Hollis T. Cline
3.4 In Vivo Imaging 1. We wait about 24 h after electroporation before the first image is
of Xenopus Tadpoles acquired to allow sufficient fluorescent protein to be expressed.
to Screen for CNS Another factor to consider in deciding when to collect images
Phenotypes is the rate of protein turnover of the gene targeted by the MO.
Images should be acquired over an interval when the MO is
capable of knocking down protein levels.
2. Individual tadpoles should be transferred into MS-222 and
anesthetized.
3. Anesthetized tadpoles in the MS-222 solution are transferred
to the Sylgard imaging chamber, which is mounted on a micro-
scope slide (Fig. 3). Under a dissecting microscope, tadpoles
are positioned within the carved indentation of the Sylgard so
that the dorsal side of the tadpole is up. Place a coverslip over
the tadpole/Sylgard and press it down so that it attaches to the
wet surface of the Sylgard and presses gently on the surface of
the tadpole’s head. A bit of liquid from the tadpole chamber
on the Sylgard will ensure that the coverslip sticks to the
Sylgard. If tadpoles are selected for confocal/multiphoton
image acquisition, care must be taken so that the tadpole does
not shift midway through the acquisition of the z-stack. See
Note 5 on positioning tadpoles.
4. (Optional) Prescreen tadpoles for imaging experiments.
Tadpoles may be viewed quickly under the screening microscope
166 Jennifer E. Bestman and Hollis T. Cline
3.5 MO Uptake MOs are more efficiently electroporated into tissue than are
and Fluorescent DNA plasmids. MOs are taken up by more cells over a physically
Protein Expression larger area of the brain than seen by GFP expression from
in the Tectum co- electroporated plasmids. Figure 4 shows two typical examples
of right tectal lobes ~24 h after electroporation of a GFP plasmid
vector and lissamine-tagged control MO. The animals were
injected with 0.1 mM lissamine-tagged control MO and 0.5 μg/μl
turboGFP plasmid under the control of the Gal4/UAS expression
system described in Bestman et al. [29]; the electrode spanned the
tectum as shown in Fig. 1b and electroporated with 3, 1.6 ms, and
40 V pulses with each polarity.
The confocal stacks in Fig. 4 show that the majority of the
tectal cells have taken up the MO (magenta signal), and relatively
few cells express the GFP (green). The right side of the CNS of the
tadpole is lightly outlined in Fig. 4a. The right tectal lobe is shown
in the bright-field image (Fig. 4b) and in the flattened confocal
image of a 50 μm stack taken ~20 μm below the surface of the skin
(Fig. 4c). The image in Fig. 4d is a flattened 180 μm stack through
the right tectal lobe, with magnified views of cell bodies (Fig. 4d
(i), (ii)) and distal processes (Fig. 4d (iii–vi)). These images show
that the MO signal in the tectum is extensive and that the GFP-
expressing cells also contain MO. The differences in the number of
cells expressing GFP and the MO may be due to the different size
and charge of MOs and plasmid DNA. Adjustment of the DNA to
MO ratio may increase the correlation between distribution of
Morpholino Studies in Xenopus Brain Development 167
Fig. 4 Examples of MO uptake and fluorescent protein expression in two electroporated tectal lobes. The tadpoles
were electroporated 24 h prior to image acquisition with 0.1 mM lissamine-tagged MO (magenta) and 0.5 μg/
μl turboGFP (green). (a) A tadpole head. The right side of the brain is outlined and the bracket indicates the
tectum, which is magnified in (b). (b) A bright-field image of the right tectal lobe with the plane of focus about
60 μm below the surface of the skin. (c) An image of a flattened 50 μm confocal stack taken about 20 μm
below the surface of the skin showing the distribution of the lissamine-tagged control MO and turboGFP-
expressing cells in the right tectal lobe. Scale bar = 50 μm. (d) An image of a flattened 180 μm confocal stack
taken through the right tectal lobe of a stage 46 tadpole. The boxes in (d) are magnified to show the distribution
of MO in the cells. Scale bar = 50 μm. (i, ii) MO fills the soma of the turboGFP-expressing cell, as well as many
of its neighbors. The arrows indicate the position of the turboGFP-expressing cell. (iii–vi) We see that many
cells also show evidence of the MO signal extending >100 μm into the distal processes of the cells. Arrows
indicate the distal tips of tectal cells expressing turboGFP and show the lissamine MO signal. Panels (i)–(vi) are
50 μm square
3.6 Verification Control and knockdown verification experiments for MO use have
of Target Knockdown been outlined by Eisen and Smith [3] and Bedell et al. [27] and
and Analysis of also can be found on the GeneTools website (http://www.gene-
Morpholino Efficacy tools.com). We note here that the methods suggested in these
and Specificity resources have been largely developed for experiments where MOs
168 Jennifer E. Bestman and Hollis T. Cline
4 Notes
References
1. Robu ME, Larson JD, Nasevicius A, Beiraghi 7. Draper BW, Morcos PA, Kimmel CB (2001)
S, Brenner C et al (2007) p53 activation by Inhibition of zebrafish fgf8 pre-mRNA splicing
knockdown technologies. PLoS Genet 3:e78 with morpholino oligos: a quantifiable method
2. Hardy S, Legagneux V, Audic Y, Paillard L for gene knockdown. Genesis 30:154–156
(2010) Reverse genetics in eukaryotes. Biol 8. Morcos PA (2007) Achieving targeted and
Cell 102:561–580 quantifiable alteration of mRNA splicing with
3. Eisen JS, Smith JC (2008) Controlling mor- Morpholino oligos. Biochem Biophys Res
pholino experiments: don’t stop making anti- Commun 358:521–527
sense. Development 135:1735–1743 9. Kloosterman WP, Lagendijk AK, Ketting RF,
4. Summerton JE (2007) Morpholino, siRNA, Moulton JD, Plasterk RH (2007) Targeted
and S-DNA compared: impact of structure and inhibition of miRNA maturation with morpho-
mechanism of action on off-target effects adn linos reveals a role for miR-375 in pancreatic
sequence specificity. Curr Top Med Chem islet development. PLoS Biol 5:e203
7:651–660 10. Choi WY, Giraldez AJ, Schier AF (2007)
5. Chen CM, Chiu SL, Shen W, Cline HT (2009) Target protectors reveal dampening and bal-
Co-expression of Argonaute2 enhances short ancing of Nodal agonist and antagonist by
hairpin RNA-induced RNA interference in miR-430. Science 318:271–274
Xenopus CNS neurons in vivo. Front Neurosci 11. Bruno IG, Jin W, Cote GJ (2004) Correction
3:63 of aberrant FGFR1 alternative RNA splicing
6. Lund E, Sheets MD, Imboden SB, Dahlberg JE through targeting of intronic regulatory ele-
(2011) Limiting ago protein restricts RNAi and ments. Hum Mol Genet 13:2409–2420
microRNA biogenesis during early development 12. Heasman J, Kofron M, Wylie C (2000) Beta-
in Xenopus laevis. Genes Dev 25:1121–1131 catenin signaling activity dissected in the early
Morpholino Studies in Xenopus Brain Development 171
Xenopus embryo: a novel antisense approach. chronological survey of the development from
Dev Biol 222:124–134 fertilized egg till the end of metamorphosis.
13. Tandon P, Showell C, Christine K, Conlon FL North-Holland Publishing Co, Amsterdam,
(2012) Morpholino injection in Xenopus. p 243
Methods Mol Biol 843:29–46 27. Bedell VM, Westcot SE, Ekker SC (2011)
14. S-i O, Mann F, Boy S, Perron M, Harris WA Lessons from morpholino-based screening in
(2002) Lipofection strategy for the study of zebrafish. Brief Funct Genomics 10:181–188
Xenopus retinal development. Methods 28: 28. Kos R, Tucker RP, Hall R, Duong TD,
411–419 Erickson CA (2003) Methods for introducing
15. Ando H, Okamoto H (2006) Efficient trans- morpholinos into the chicken embryo. Dev
fection strategy for the spatiotemporal control Dyn 226:470–477
of gene expression in zebrafish. Mar Biotechnol 29. Bestman JE, Lee-Osbourne J, Cline HT
(NY) 8:295–303 (2012) In vivo time-lapse imaging of cell pro-
16. Sasagawa S, Takabatake T, Takabatake Y, liferation and differentiation in the optic tec-
Muramatsu T, Takeshima K (2002) Improved tum of Xenopus laevis tadpoles. J Comp Neurol
mRNA electroporation method for Xenopus 520:401–433
neurula embryos. Genesis 33:81–85 30. Sauka-Spengler T, Barembaum M (2008)
17. Eide FF, Eisenberg SR, Sanders TA (2000) Gain- and loss-of-function approaches in the
Electroporation-mediated gene transfer in free- chick embryo. Methods Cell Biol 87:237–256
swimming embryonic Xenopus laevis. FEBS 31. Bestman JE, Cline HT (2008) The RNA bind-
Lett 486:29–32 ing protein CPEB regulates dendrite morpho-
18. Bestman JE, Ewald RC, Chiu SL, Cline HT genesis and neuronal circuit assembly in vivo.
(2006) In vivo single-cell electroporation for Proc Natl Acad Sci USA 105:20494–20499
transfer of DNA and macromolecules. Nat 32. Chiu S-L, Chen C-M, Cline HT (2008) Insulin
Protoc 1:1267–1272 receptor signaling regulates synapse number,
19. Falk J, Drinjakovic J, Leung K, Dwivedy A, dendritic plasticity, and circuit function in vivo.
Regan A et al (2007) Electroporation of Neuron 58:708–719
cDNA/Morpholinos to targeted areas of 33. Shen W, McKeown CR, Demas JA, Cline HT
embryonic CNS in Xenopus. BMC Dev Biol (2011) Inhibition to excitation ratio regulates
7:107 visual system responses and behavior in vivo.
20. Haas K, Jensen K, Sin WC, Foa L, Cline HT J Neurophysiol 106(5):2285–2302
(2002) Targeted electroporation in Xenopus 34. Sharma P, Cline HT (2010) Visual activity reg-
tadpoles in vivo–from single cells to the entire ulates neural progenitor cells in developing
brain. Differentiation 70:148–154 Xenopus CNS through musashi1. Neuron
21. Haas K, Sin W-C, Javaherian A, Li Z, Cline HT 68:442–455
(2001) Single-cell electroporation for gene 35. Schwartz N, Schohl A, Ruthazer ES (2011)
transfer in vivo. Neuron 29:583–591 Activity-dependent transcription of BDNF
22. Javaherian A, Cline HT (2005) Coordinated enhances visual acuity during development.
motor neuron axon growth and neuromuscu- Neuron 70:455–467
lar synaptogenesis are promoted by CPG15 in 36. Ewald RC, Van Keuren-Jensen KR, Aizenman
vivo. Neuron 45:505–512 CD, Cline HT (2008) Roles of NR2A and
23. Ruthazer ES, Li J, Cline HT (2006) NR2B in the development of dendritic arbor
Stabilization of axon branch dynamics by syn- morphology in vivo. J Neurosci 28:850–861
aptic maturation. J Neurosci 26:3594–3603 37. Zhao Y, Ishibashi S, Amaya E (2012) Reverse
24. Osterele A (2011) P-1000 & P-97 pipette genetic studies using antisense morpholino
cookbook, 2011. In:Rev G (ed) Sutter oligonucleotides. Methods Mol Biol 917:
Instrument Company 143–154
25. Koster RW, Fraser SE (2001) Tracing trans- 38. Rana AA, Collart C, Gilchrist MJ, Smith JC
gene expression in living zebrafish embryos. (2006) Defining synphenotype groups in
Dev Biol 233:329–346 Xenopus tropicalis by use of antisense morpho-
26. Nieuwkoop PD, Faber J (1956) Normal table lino oligonucleotides. PLoS Genet 2:
of Xenopus laevis (Daudin); a systemical and 1751–1772
Part V
Zebrafish Protocols
Chapter 12
Abstract
Whole-mount in situ hybridization is the preferred method for detecting transcript distributions in whole
embryos, tissues, and organs. We present here a sensitive fluorescent in situ hybridization method for
colocalization analysis of different transcripts in whole embryonic zebrafish brains. The method is based
on simultaneous hybridization of differently hapten-labeled RNA probes followed by sequential rounds
of horseradish peroxidase (POD)-based transcript detection. Sequential detection involves enhancement of
fluorescent signals by tyramide signal amplification (TSA) and effective inactivation of the antibody–POD
conjugate prior to the following detection round. We provide a detailed description of embryo preparation,
hybridization, antibody detection, POD–TSA reaction, and mounting of embryos for imaging. To achieve
high signal intensities, we optimized key steps of the method. This includes improvement of embryo
permeability by hydrogen peroxide treatment and efficacy of hybridization and TSA–POD reaction by
addition of the viscosity-increasing polymer dextran sulfate. The TSA–POD reaction conditions are further
optimized by application of substituted phenol compounds as POD accelerators and use of highly efficient
bench-made tyramide substrates. The obtained high signal intensities and cellular resolution of our method
allows for co-expression analysis and generation of three-dimensional models. Our protocol is tailored to
optimally work in zebrafish embryos, but can surely be modified for application in other species as well.
Key words Fluorescent in situ hybridization, FISH, Tyramide signal amplification, TSA, Peroxidase,
Zebrafish
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_12, © Springer Science+Business Media, LLC 2014
175
176 Gilbert Lauter et al.
Fig. 1 Two-color whole-mount FISH using enhanced tyramide signal amplification. Lateral views of dlx2a
and lhx1a expression in a 28-hpf zebrafish forebrain with anterior oriented to the left. Transcript signals are
visualized in cyan and magenta as indicated in the panels. Single-channel detection and the overlay of both
channels in the same confocal plane are shown. (a) Expression of dlx2a was visualized by using a DNP-labeled
RNA probe together with the DyLight 633-tyramide. The TSA reaction was allowed to proceed for 20 min in the
presence of 0.15 mg/ml 4-iodophenol. (b) Expression of lhx1a was visualized by using a DIG-labeled RNA
probe together with the FAM-tyramide. The TSA reaction was allowed to proceed for 30 min in the presence of
0.15 mg/ml 4-iodophenol. (c) Overlay of both single channels shows the expression of dlx2a and lhx1a in the
developing forebrain with areas of overlap indicated in yellow. Scale bars = 50 μm
2 Materials
3 Methods
3.5 Fluorogenic The TSA reaction can be significantly enhanced by the use of a
Reaction POD accelerator. Use 4-iodophenol or vanillin at a concentration
of 0.15 mg/ml and 0.45 mg/ml, respectively (see Note 8).
1. For 1 ml reaction buffer, combine 500 μl borate buffer pH 8.5
(200 mM), 40 μl 50 % dextran sulfate, 10 μl of 10 % Tween-
20, 6 μl of 0.5 % H2O2, and an appropriate accelerator and
adjust the volume with water to 1 ml (see Note 9).
2. Addition of 4-iodophenol will result in the appearance of a
cloudy smear. Mix the reaction buffer well by pipetting up and
down until the dextran sulfate has dispersed and the solution
appears opaque.
3. Dilute the desired tyramide with the reaction buffer and mix
well by pipetting. Use 6 μl of DyLight 633-tyramide or 4 μl
of FAM-tyramide stock solution per 1 ml reaction buffer
(see Note 10).
4. Rinse embryos twice with 100 mM borate buffer pH 8.5 con-
taining 0.1 % Tween-20. Remove supernatant borate buffer as
closely as possible and add 90 μl of the tyramide solutions to
the embryos. Gently mix by pipetting using a cut tip. Incubate
for the desired length of time in the dark at RT without agitation
(see Note 11).
5. To stop the TSA reaction, wash for four times with PBST. For
each washing step, fill the entire tube with PBST. After invert-
ing the tubes for several times, wait until the embryos are at
the bottom and then remove excess buffer carefully. From this
step onward, embryos should be protected from light.
6. To inactivate POD activity of the first antibody, incubate
samples in 100 mM glycine-HCl pH 2.0 for 10 min followed by
four 5-min washes in PBST under agitation (see Note 12).
7. For the second antibody detection round, repeat the steps
described under Subheading 3.4 using sheep-anti-DIG-POD
Fab fragments (1:500).
8. Repeat steps 1–5 for fluorogenic detection (see Note 13).
4 Notes
Acknowledgments
References
1. Abellan A, Medina L (2009) Subdivisions and 8. Tautz D, Pfeifle C (1989) A non-radioactive in
derivatives of the chicken subpallium based on situ hybridization method for the localization
expression of LIM and other regulatory genes of specific RNAs in Drosophila embryos reveals
and markers of neuron subpopulations during translational control of the segmentation gene
development. J Comp Neurol 515(4):465–501 hunchback. Chromosoma 98(2):81–85
2. Puelles L, Kuwana E, Puelles E, Bulfone A, 9. Hauptmann G, Gerster T (1994) Two-color
Shimamura K, Keleher J, Smiga S, Rubenstein whole-mount in situ hybridization to verte-
JL (2000) Pallial and subpallial derivatives in brate and Drosophila embryos. Trends Genet
the embryonic chick and mouse telencephalon, 10(8):266
traced by the expression of the genes Dlx-2, 10. Hauptmann G, Gerster T (1996) Multicolour
Emx-1, Nkx-2.1, Pax-6, and Tbr-1. J Comp whole-mount in situ hybridization to
Neurol 424(3):409–438, doi:10.1002/1096- Drosophila embryos. Dev Genes Evol 206(4):
9861(20000828)424:3<409::AID- 292–295
CNE3>3.0.CO;2-7 [pii] 11. Hauptmann G, Gerster T (2000) Multicolor
3. Diez-Roux G, Banfi S, Sultan M, Geffers L, whole-mount in situ hybridization. Methods
Anand S, Rozado D, Magen A, Canidio E, Mol Biol 137:139–148, doi:1-59259-066-7-
Pagani M, Peluso I, Lin-Marq N, Koch M, 139 [pii] 10.1385/1-59259-066-7:139
Bilio M, Cantiello I, Verde R, De Masi C, 12. Jowett T, Lettice L (1994) Whole-mount in
Bianchi SA, Cicchini J, Perroud E, Mehmeti S, situ hybridizations on zebrafish embryos
Dagand E, Schrinner S, Nurnberger A, Schmidt using a mixture of digoxigenin- and
K, Metz K, Zwingmann C, Brieske N, Springer fluorescein-labelled probes. Trends Genet
C, Hernandez AM, Herzog S, Grabbe F, 10(3):73–74
Sieverding C, Fischer B, Schrader K,
Brockmeyer M, Dettmer S, Helbig C, Alunni 13. Hauptmann G (1999) Two-color detection of
V, Battaini MA, Mura C, Henrichsen CN, mRNA transcript localizations in fish and fly
Garcia-Lopez R, Echevarria D, Puelles E, embryos using alkaline phosphatase and beta-
Garcia-Calero E, Kruse S, Uhr M, Kauck C, galactosidase conjugated antibodies. Dev
Feng G, Milyaev N, Ong CK, Kumar L, Lam Genes Evol 209(5):317–321
M, Semple CA, Gyenesei A, Mundlos S, 14. Hauptmann G (2001) One-, two-, and three-
Radelof U, Lehrach H, Sarmientos P, Reymond color whole-mount in situ hybridization to
A, Davidson DR, Dolle P, Antonarakis SE, Drosophila embryos. Methods 23(4):359–372,
Yaspo ML, Martinez S, Baldock RA, Eichele G, doi:10.1006/meth.2000.1148
Ballabio A (2011) A high-resolution anatomi- S1046-2023(00)91148-4 [pii]
cal atlas of the transcriptome in the mouse 15. O’Neill JW, Bier E (1994) Double-label in situ
embryo. PLoS Biol 9(1):e1000582 hybridization using biotin and digoxigenin-
4. Shimogori T, Lee DA, Miranda-Angulo A, tagged RNA probes. Biotechniques 17(5):870,
Yang Y, Wang H, Jiang L, Yoshida AC, Kataoka 874, 875
A, Mashiko H, Avetisyan M, Qi L, Qian J, 16. Bobrow MN, Harris TD, Shaughnessy KJ, Litt
Blackshaw S (2010) A genomic atlas of mouse GJ (1989) Catalyzed reporter deposition, a
hypothalamic development. Nat Neurosci novel method of signal amplification.
13(6):767–775 Application to immunoassays. J Immunol
5. Suzuki-Hirano A, Ogawa M, Kataoka A, Methods 125(1–2):279–285, doi:0022-1759
Yoshida AC, Itoh D, Ueno M, Blackshaw S, (89)90104-X [pii]
Shimogori T (2011) Dynamic spatiotemporal 17. van de Corput MP, Dirks RW, van Gijlswijk RP,
gene expression in embryonic mouse thalamus. van Binnendijk E, Hattinger CM, de Paus RA,
J Comp Neurol 519(3):528–543 Landegent JE, Raap AK (1998) Sensitive
6. Ferran JL, Sanchez-Arrones L, Sandoval JE, mRNA detection by fluorescence in situ
Puelles L (2007) A model of early molecular hybridization using horseradish peroxidase-
regionalization in the chicken embryonic pre- labeled oligodeoxynucleotides and tyramide
tectum. J Comp Neurol 505(4):379–403. signal amplification. J Histochem Cytochem
doi:10.1002/cne.21493 46(11):1249–1259
7. Lauter G, Söll I, Hauptmann G (2013) 18. Kosman D, Mizutani CM, Lemons D, Cox WG,
Molecular characterization of prosomeric and McGinnis W, Bier E (2004) Multiplex detection
intraprosomeric subdivisions of the embryonic of RNA expression in Drosophila embryos.
zebrafish diencephalon. J Comp Neurol 521(5): Science 305(5685):846, doi:10.1126/sci-
1093–1118 ence.1099247 305/5685/846 [pii]
FISH in Zebrafish 185
19. Tessmar-Raible K, Steinmetz PR, Snyman H, 23. Denkers N, Garcia-Villalba P, Rodesch CK,
Hassel M, Arendt D (2005) Fluorescent Nielson KR, Mauch TJ (2004) FISHing for
two-color whole mount in situ hybridization in chick genes: triple-label whole-mount fluores-
Platynereis dumerilii (Polychaeta, Annelida), cence in situ hybridization detects simultane-
an emerging marine molecular model for evo- ous and overlapping gene expression in avian
lution and development. Biotechniques embryos. Dev Dyn 229(3):651–657.
39(4):460, 462, 464. doi:000112023 [pii] doi:10.1002/dvdy.20005
20. Clay H, Ramakrishnan L (2005) Multiplex 24. Lauter G, Söll I, Hauptmann G (2011)
fluorescent in situ hybridization in zebrafish Multicolor fluorescent in situ hybridization to
embryos using tyramide signal amplification. define abutting and overlapping gene expres-
Zebrafish 2(2):105–111. doi:10.1089/ sion in the embryonic zebrafish brain. Neural
zeb.2005.2.105 Dev 6(1):10, doi:1749-8104-6-10
21. Lauter G, Söll I, Hauptmann G (2011) Two- [pii]10.1186/1749-8104-6-10
color fluorescent in situ hybridization in the 25. Fischer AH, Jacobson KA, Rose J, Zeller R
embryonic zebrafish brain using differential (2008) Media for mounting fixed cells on
detection systems. BMC Dev Biol 11:43 microscope slides. CSH Protoc 2008:pdb ip52
22. Vize PD, McCoy KE, Zhou X (2009) 26. Liu G, Amin S, Okuhama NN, Liao G, Mingle
Multichannel wholemount fluorescent and LA (2006) A quantitative evaluation of peroxi-
fluorescent/chromogenic in situ hybridiza- dase inhibitors for tyramide signal amplification
tion in Xenopus embryos. Nat Protoc mediated cytochemistry and histochemistry.
4(6):975–983, doi:nprot.2009.69 [pii] Histochem Cell Biol 126(2):283–291.
10.1038/nprot.2009.69 doi:10.1007/s00418-006-0161-x
Chapter 13
Abstract
Genetic means to visualize and manipulate neuronal circuits in the intact animal have revolutionized
neurobiology. “Dynamic neuroanatomy” defines a range of approaches aimed at quantifying the architecture
or subcellular organization of neurons over time during their development, regeneration, or degeneration.
A general feature of these approaches is their reliance on the optical isolation of defined neurons in toto
by genetically expressing markers in one or few cells. Here we use the afferent neurons of the lateral line
as an example to describe a simple method for the dynamic neuroanatomical study of axon terminals in the
zebrafish by laser-scanning confocal microscopy.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_13, © Springer Science+Business Media, LLC 2014
187
188 Adèle Faucherre and Hernán López-Schier
2 Materials
3 Methods
3.1 Expressing 1. Prepare DNA for injection (see Fig. 1). To achieve mosaic
Membrane Red expression in single neurons, clone the cDNA encoding the
Fluorescent Protein in red fluorescent protein (e.g., mCherry or tdTomato) under
the Zebrafish Lateral the control of a neuronal promoter (e.g., HuC). For a better
Line Afferent Neurons visualization of neurites, the red fluorescent protein has to be
fused to a membrane-targeting sequence (e.g., CAAX domain
or first 20 codons of GAP43).
HuC:mem-TdTomato DNA
Brn3c:memEGFP
embryo
Fig. 1 Scheme of the injection and selection of the samples to image. Overview
of the injection strategy to label single lateralis neurons. Injection of cDNA coding
with HuC:mem-TdTomato into 1–2-cell Tg[Brn3c:mem-eGFP] host embryos
190 Adèle Faucherre and Hernán López-Schier
3.3 Neuronal-Arbor 1. Image stacks are rendered three-dimensional using the Imaris
Tracing (See Fig. 2) software. Under the file menu, chose open and select the
desired file. In order to accelerate the analysis process, resample
the dataset before loading it (see Note 5).
Fig. 2 (continued) A and B, respectively, after 10 min. The dendrite C is no more present. (e, f) Example of
tracing of the whole neuronal arbor at T1 (e) and T2 (f). (g) Example of data for four-dimensional quantification.
The dendrite length is given by the software in micrometers. To calculate the growth or retraction of a neurite,
values obtained at T2 are subtracted from the values obtained at T1 for each neurite. In this case, the dendrite
A has extended 4.1 μm whereas the dendrite B has retracted 2.45 μm. In the case of the dendrite C, which
was present at T1 but absent at T2, the distance of retraction is calculated from the branching point, here Y
Dynamic Neuroanatomy in Zebrafish 191
T1 T2
a b
c d
X X
Y
C
B B’
A
A’
e f
Begining point
Branching point
End point
Fig. 2 Neuronal-arbor tracing. (a, b) Neuronal arbor at time points T1 and T2. (c, d) Example of dendrite tracing.
X represents the beginning point. A, A′; B, B′; and C represent 3 end points and Y represents a branching point.
In this case, at T1 (c), 3 dendrites have been traced. At T2 (d), A′ and B′ represent the position of the dendrites
192 Adèle Faucherre and Hernán López-Schier
3.4 Four- 1. Select a terminal point. In the Statistics menu, display the
Dimensional Selection values and chose Pt distance (i.e., dendrite length)
Quantification (see Note 7). Copy the value of Pt distance and paste it in a
of Axonal Arbors new Excel file.
(See Fig. 3) 2. For the same neurite, follow the terminal point from one time
point to the following one and repeat step 1.
3. In the case of transient neurites, select the branching point and
repeat step 15.
4. To express the Neurites stability, calculate the percentage of
stable neurites (present throughout all the movie) versus the
percentage of transient neurites (neurites visible during less
than 6 time points).
5. Calculate the Neurites persistence by counting the percentage
of neurites present for only one, two, three, four, five, or six
time frames.
6. To calculate growth and retraction, subtract the position value
of a neurite at a specific time point of the value of the previous
time point. Positive values correspond to growth and negative
values to retraction. Decide of an arbitrarily absolute value that
reflects significant growth or retraction movements (in our
study, the arbitrarily absolute value reflecting significant move-
ment is 1.5 μm) (see Note 8).
7. Calculate the number of neurites that have achieved a defined
number of events. Neurites consistency is defined as the number
of events (growth or retraction) observed for each neurite.
“Consistent” corresponds to a neurite that has neither grown
nor retracted over the 6 time frames (i.e., no events). “One
event” corresponds to a neurite that has only grown or only
retracted over the 6 time frames. “Two events” correspond to a
Dynamic Neuroanatomy in Zebrafish 193
Fig. 3 Four-dimensional representation. (a, b) One color is attributed to each neurite and end points are duplicated
in order to visualize the positions of a specific tip over time. At T1 (a), the end points of dendrites A, B, and C have
been plotted. At T2 (b), A′ and B′ indicate the positions of the tip of the neurites A and B, respectively, after 10 min.
(c) Four-dimensional representation of the whole arbor after 1 h (six times 10 min). (d) Same as in (c) with the
maximal projection of the mem-tdTomato-expressing neuron and GFP-expressing hair cells
4 Notes
2. Select for analysis only those specimens with single red fluores-
cent neurons whose soma is localized within the lateralis affer-
ent ganglion, which can be marked using the HGn39D
transgenic line.
3. If more than one afferent neuron are expressing the red fluores-
cent protein, you can try to inject less DNA. Nevertheless, if
the neuromast is innervated by only one neuron and if the soma
of this neuron can be localized within the lateralis afferent
ganglion, the experiment can be carried away.
4. If none of the afferent neurons is expressing the red fluorescent
protein, you can try to inject more DNA. Check as well that
you are using a promoter/enhancer allowing expression in the
afferent neurons.
5. If the sample has moved during the time of the acquisition,
align the three-dimensional reconstructions of each time point
using the Spot/Correct drift function of Imaris.
6. It is very important to assign the starting point as Dendrite
Beginning Point in the Edit menu since every following calcu-
lation is based on the position of this point.
7. If, in the statistic menu of Filament tracer, there is no value for
the dendrite length, check that the starting point has been
assigned as the Dendrite Beginning Point. Check as well that
there is no discontinuity in the tracing of the neuronal arbor
reconstruction.
8. In the case of transient neurites, the value before appearance or
after disappearance is the distance between the beginning point
and the branching point.
Acknowledgements
We thank the generosity of C.B. Chien for the Tol2kit and advice
and dedicate this article to his memory. We also thank T. Zimmermann
and J. Swoger for advice on, respectively, confocal and SPIM micros-
copy. The original research that encouraged the development of
this methodology was supported by a grant from the European
Research Council (ERC-2007-StG SENSORINEURAL) and by
the Ministerio de Ciencia e Innovación of Spain to H.L. S.A.F. was
supported by an Intra-European Marie Curie Fellowship from the
European Union.
References
1. Arber S (2012) Motor circuits in action: speci- 2. Seelig JD, Jayaraman V (2011) Studying senso-
fication, connectivity, and function. Neuron rimotor processing with physiology in behaving
74:975–989 Drosophila. Int Rev Neurobiol 99:169–189
Dynamic Neuroanatomy in Zebrafish 195
3. Koch M (1999) The neurobiology of startle. 15. Pujol-Martí J, Baudoin JP, Faucherre A et al
Prog Neurobiol 59:107–128 (2010) Progressive neurogenesis defines late-
4. Piggott BJ, Liu J et al (2011) The neural circuits ralis somatotopy. Dev Dyn 239:1919–1930
and synaptic mechanisms underlying motor ini- 16. Collins RT, Linker C, Lewis J (2010) MAZe: a
tiation in C. elegans. Cell 147:922–933 tool for mosaic analysis of gene function in
5. Anikeeva P, Andalman AS et al (2011) zebrafish. Nat Methods 7:219–223
Optetrode: a multichannel readout for optoge- 17. Hans S, Kaslin J, Freudenreich D et al (2009)
netic control in freely moving mice. Nat Temporally-controlled site-specific recombina-
Neurosci 15:163–170 tion in zebrafish. PLoS One 4:e4640
6. Ahrens MB, Li JM et al (2012) Brain-wide 18. Luo L (2007) Single-neuron labeling using the
neuronal dynamics during motor adaptation in genetic MARCM method. CSH Protoc 2007:
zebrafish. Nature 485:471–477 pdb.prot4789
7. Moser EI, Kropff E, Moser MB (2008) Place 19. Livet J, Weissman TA, Kang H et al (2007)
cells, grid cells, and the brain’s spatial represen- Transgenic strategies for combinatorial expres-
tation system. Annu Rev Neurosci 31:69–89 sion of fluorescent proteins in the nervous sys-
8. Gahtan E, Baier H (2004) Of lasers, mutants, tem. Nature 450:56–62
and see-through brains: functional neuroanat- 20. Pan YA, Livet J, Sanes JR et al (2011)
omy in zebrafish. J Neurobiol 59:147–161 Multicolor brainbow imaging in zebrafish.
9. Friedrich RW, Jacobson GA, Zhu P (2010) CSH Protoc 2011(1):pdb.prot5546
Circuit neuroscience in zebrafish. Curr Biol 21. Raible DW, Kruse GJ (2000) Organization of
20:R371–R381 the lateral line system in embryonic zebrafish.
10. Faucherre A, Baudoin JP, Pujol-Martí J et al J Comp Neurol 421:189–198
(2010) Multispectral four-dimensional imag- 22. Metcalfe WK, Kimmel CB, Schabtach E (1985)
ing reveals that evoked activity modulates Anatomy of the posterior lateral line system in
peripheral arborization and the selection of young larvae of the zebrafish. J Comp Neurol
plane-polarized targets by sensory neurons. 233:377–389
Development 137:1635–1643 23. Swoger J, Muzzopappa M, López-Schier H
11. Simmich J, Staykov E, Scott E (2012) Zebrafish et al (2011) 4D retrospective lineage tracing
as an appealing model for optogenetic studies. using SPIM for zebrafish organogenesis studies.
Prog Brain Res 196:145–162 J Biophotonics 4:122–134
12. Santoriello C, Zon LI (2012) Hooked! 24. Kaufmann A, Mickoleit M, Weber M et al
Modeling human disease in zebrafish. J Clin (2012) Multilayer mounting enables long-
Invest 122:2337–2343 term imaging of zebrafish development in a
13. Shu X, Lev-Ram V, Olson ES et al (2011) Spiers light sheet microscope. Development 139:
Memorial lecture. Breeding and building molec- 3242–3247
ular spies. Faraday Discuss 149:63–77 25. Huisken J, Stainier DY (2009) Selective plane
14. Detrich HW 3rd (2008) Fluorescent proteins illumination microscopy techniques in devel-
in zebrafish cell and developmental biology. opmental biology. Development 136:
Methods Cell Biol 85:219–241 1963–1975
Chapter 14
Abstract
Zebrafishbrain.org is an online neuroanatomical atlas of the embryonic zebrafish. The atlas uses high-
resolution confocal images and movies of transgenic lines to describe different brain structures. This chapter
covers detail of materials and protocols that we employ to generate data for the atlas.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_14, © Springer Science+Business Media, LLC 2014
197
198 Katherine J. Turner et al.
2 Materials
2.4 Brain Dissection 1. Sylgard: 1 kit 184 from Dow Corning WPI Sylgard http://
Reagents and www.dowcorning.com/applications/search/products/
Solutions Details.aspx?prod=01064291&type=&country=GBR.
2. Saturated NaOH.
3. PBS.
2.5 Brain Dissection 1. Sylgard is an elastomer curing agent. The Sylgard kit contains
Sylgard Dishes two solutions that set to a hard rubberlike texture when mixed.
A layer of Sylgard in a glass petri dish makes a great dissecting
dish as you can pin the embryos using insect/minute pins to
the Sylgard, immobilizing them while you dissect. You also
minimize damage to forceps and dissecting needles.
2. Mix Part A liquid from the Sylgard 184 kit with Part B liquid
in a ration of 10:1 in a beaker. You will need 40–50 ml per
petri dish.
3. Thoroughly mix the two solutions together using a disposable
stirrer such as a tongue depressor. Be careful not to stir too vig-
orously so as not to create unwanted bubbles in the mixture.
4. After mixing, pour the Sylgard slowly into the glass petri dishes
laid on a flat, non-vibrating surface where they can remain undis-
turbed for several days. Fill each petri dish to 1/3–1/2 full.
5. After the Sylgard has settled for a few minutes, you will see that
bubbles have risen to the surface of the liquid. Burst these
using a Bunsen burner by passing the flame lightly close to the
surface of the Sylgard.
6. Put the petri dish lids on top to prevent dust settling on the
plates while they set.
7. Sylgard dishes need at least 24 h to set and improve if left for
several days. It is possible to speed up this process by heat
curing them at 60 °C for several hours in a mini oven or
incubator.
200 Katherine J. Turner et al.
2.6 Brain Dissection To remove the skin and eyes from zebrafish embryos, we use
Needles dissecting needles fashioned from fine tungsten wire. The
tungsten wire is clamped into a needle holder and cut using
strong scissors so it protrudes from the tip of the holder by about
1 cm. The wire at this point is blunt but can be sharpened to a
very fine sharp point using an electric current and saturated
sodium hydroxide (Fig. 1).
3 Methods
3.2 Brain Dissection Dissecting the skin and eyes off of zebrafish embryos prior to
antibody staining vastly improves the penetration of antibodies and
reduces auto-fluorescence of the skin common to many transgenic
lines. Although this technique requires some dexterity, with practice
it becomes routine and the results justify the effort (Fig. 2).
204 Katherine J. Turner et al.
Fig. 2 Dissection. (a) Pin embryo laterally to a Sylgard dish. (b) Remove eye using a sharpened tungsten nee-
dle. (c) Remove pins and flip the embryo so the other side now faces upwards. (d) Remove second eye using
sharpened tungsten needle. (e) Cut away the yolk and jaw from the ventral surface of the embryo. (f) At the
level of the otic vesicle using either a sharpened tungsten needle or fine forceps, grip a piece of skin and tease
away from the brain. (g) Pull this flap of skin rostrally, removing all of the skin from the surface of the brain. (h)
Close up showing completely dissected brain ready to be transferred into methanol for immunohistochemistry
or in situ hybridization
11. After all embryos are dissected and transferred to 1.5 ml tube.
Wash several times with PBTr, then transfer to MeOH (see
Subheading 3.1), and store at −20 °C until you want to start
the immunohistochemistry (see Note 3).
3.5 Cell Dyes Using a nuclear label in conjunction with a fluorescent immuno-
for Neuroanatomy staining can be very useful to delineate brain nuclei, neuropil, and
ventricles through the tissue (Fig. 4). Nuclear staining has also
been employed by the Driever lab to produce a 3D reference brain
for their ViBE-Z software. They have also used acetylated tubulin
immunohistochemistry with their reference brain [17, 18].
For nuclear staining, use SYTOX Orange or Green at a con-
centration of 1:10,000 or Topro3 at a concentration of 1:5,000
depending upon the wavelengths required. These dyes can be
added with the secondary antibody incubation. Staining with
nuclear dyes works best following room temperature incubation in
the dye so it is advisable to add 2 mM azide with the secondary
antibody/nuclear dye IB mixture to inhibit mold formation. The
dyes bleach very easily so minimize exposure to light and keep the
laser intensity on the confocal as low as possible when imaging, and
also, timely imaging following the staining process produces the
best results.
208 Katherine J. Turner et al.
Fig. 4 Nuclear dyes. Nuclear dyes show brain nuclei and also expose morpho-
logical boundaries between brain areas. Neuropil is not marked and is revealed
as dark spaces. In this confocal slice through the forebrain of this enhancer trap
line, we can see expression of GFP in the olfactory epithelia (Image generated by
Tim Geach and TAH)
Fig. 5 Mounting in glass rings. (a) On a dissecting microscope, place a 22 × 22 coverslip onto a slide. Smear
silicone grease on both sides of a glass ring, and press it onto the coverslip forming a watertight seal. Pipette
the embryo into the agarose and then onto the coverslip in the center of the glass ring. Orientate the embryo
so the surface of the embryo you wish to image is against the coverslip. (b) Fill the ring 2/3 full with agarose
and let it set. Once the agarose has set firm, pipette enough PBS onto the surface of the agarose to form a
convex meniscus over the top of the ring. Overfilling the ring with PBS means that no bubbles will be trapped
when you place the slide on top. (c) Take another slide and press down onto the top of the glass ring expelling
the excess PBS. Make sure the slide and the glass ring have formed a watertight seal with the silicone grease.
(d) The prep can now be inverted with the coverslip on top and slide on the bottom. Your embryo will now be
at the top of the prep just under the coverslip ready for imaging
3.7 Confocal If glass rings are not available, a similar mounting method can be
Mounting Without used where a well of silicone grease is constructed directly onto a
Glass Ring slide (Fig. 6). A syringe filled with silicone grease should be used
to squeeze the grease onto the slide. The embryo is mounted on a
coverslip as above in a large drop of agarose (the diameter and
depth of this drop should not exceed the width and depth of the
silicone grease well). The silicone grease well is filled with PBS to
form a convex meniscus. The coverslip with the embryo attached
is inverted and pressed gently down into the well expelling the
excess PBS. Make sure the coverslip forms a watertight seal with
the top of the silicone grease well. The embryo is now ready for
imaging. This method also has the advantage of allowing for a
small adjustment in the orientation of the sample.
3.8 Mounting For imaging of live embryos (particularly for timelapse), embryos
of Live Embryos can be mounted in a large epoxy resin chamber or well. This allows
for Confocal Imaging for constant gaseous exchange and thus improves the health of the
embryo. These chambers are easily made using Araldite or similar
Anatomical Dissection of Zebrafish Brain Development 211
Fig. 6 Mounting fixed embryos in silicone grease wells. (a) Using a syringe filled with silicone grease, pipe a
well onto a microscope slide. (b) On a coverslip orientate your embryo in a drop of 1 % agarose. The surface
of the embryo you wish to image should be touching the coverslip. Half fill the silicone grease well with PBS.
(c) Once the agarose is set, invert your coverslip so that the embryo is submerged in PBS inside the silicone
grease well. Lightly push down on the coverslip to create a sealed chamber
3.9 Mounting of Live An alternative to agarose for mounting live embryos is a compound
Embryos Using CyGEL called CyGEL Sustain. CyGEL is liquid at low temperatures and
Sustain changes from a sol to a gel at 23–24 °C. The sol–gel conversion
can be reversed, by simply placing the sample on ice for a few
seconds. This is particularly useful for short procedures that require
a fast and easy way of recovering the embryo undamaged after
manipulation, such as electroporation or Kaede photoconversion.
CyGEL Sustain is a compound commercialized by BioStatus
Limited (www.biostatus.com). They provide protocols on their
web page, but these are optimized for the growth of cells. Here we
provide a protocol adapted for zebrafish mounting:
1. Place vial of CyGEL Sustain on ice and make sure it is a sol.
2. Add E3 so that the final concentration will be 1× (8.4 μl of
60XE3 for 500 μl of CyGEL). Keep it on ice.
3. Prepare a chamber on a slide with silicone grease, or use a glass
ring as described in Subheading 3.6.
4. Warm the vial slightly using hands so that it is not too cold
(but not too much because it gels).
5. Pipette the anesthetized embryo into the CyGEL then refill
the pipette with the embryo and enough CyGEL to fill the sili-
cone chamber or glass ring.
6. Orientate embryo as required the consistency of CyGEL
quickly becomes dense, becoming a tight gel within minutes.
At that point, the embryo is ready for microscopy.
No additional liquid should be placed on the gel as it immedi-
ately becomes a sol again. If the embryo needs to be imaged using
Anatomical Dissection of Zebrafish Brain Development 213
3.10 Confocal Setup of the imaging system for optimal imaging depends very
Microscopy Setup much on the individual sample employed. High-NA, long working
distance objectives (water and/or glycerol immersion) are available
from most microscope manufacturers. These lenses are ideally
suited to imaging the zebrafish brain as they permit imaging
through the whole depth of the brain at high resolution. There are
many tradeoffs in the setup of the imaging parameters. For high-
quality three-dimensional reconstructions when imaging fixed,
antibody-stained preparations, it is best to aim for approximately
isometric voxels at resolutions approaching the limit of diffraction;
however, this should be traded off against the time taken and the
file size. For live imaging of fluorophores, it is best to minimize
bleaching of the (usually dim) fluorescent proteins. Thus
minimizing the dwell time at any particular voxel is advisable.
4 Notes
References
1. Norton W, Bally-Cuif L (2010) Adult zebraf- 11. Wilson SW, Ross LS, Parrett T et al (1990)
ish as a model organism for behavioural genet- The development of a simple scaffold of axon
ics. BMC Neurosci 11:90 tracts in the brain of the embryonic zebrafish,
2. Ahrens MB, Li JM, Orger MB et al (2012) Brachydanio rerio. Development
Brain-wide neuronal dynamics during motor 108(1):121–145
adaptation in zebrafish. Nature 485:471–477 12. Buckley K, Kelly RB (1985) Identification of a
3. Stewart A, Gaikwad S, Kyzar E et al (2012) transmembrane glycoprotein specific for secre-
Modeling anxiety using adult zebrafish: a concep- tory vesicles of neural and endocrine cells.
tual review. Neuropharmacology 62:135–143 J Cell Biol 100:1284–1294
4. Mathur P, Guo S (2010) Use of zebrafish as a 13. Hendricks M, Jesuthasan S (2007) Asymmetric
model to understand mechanisms of addiction innervation of the habenula in zebrafish.
and complex neurobehavioral phenotypes. J Comp Neurol 502:611–619
Neurobiol Dis 40:66–72 14. Miyasaka N, Morimoto K, Tsubokawa T et al
5. Prober DA, Rihel J, Onah AA et al (2006) (2009) From the olfactory bulb to higher
Hypocretin/orexin overexpression induces an brain centers: genetic visualization of second-
insomnia-like phenotype in zebrafish. ary olfactory pathways in zebrafish. J Neurosci
J Neurosci 26:13400–13410 29:4756–4767
6. Lein ES, Hawrylycz MJ, Ao N et al (2007) 15. Cerveny KL, Cavodeassi F, Turner KJ et al
Genome-wide atlas of gene expression in the (2010) The zebrafish flotte lotte mutant
adult mouse brain. Nature 445:168–176 reveals that the local retinal environment pro-
7. Hawrylycz MJ, Lein ES, Guillozet-Bongaarts motes the differentiation of proliferating pre-
AL et al (2012) An anatomically comprehen- cursors emerging from their stem cell niche.
sive atlas of the adult human brain transcrip- Development 137:2107–2115
tome. Nature 489:391–399
16. Collins RT, Linker C, Lewis J (2010) MAZe:
8. Rink E, Wullimann MF (2002) Connections
a tool for mosaic analysis of gene function in
of the ventral telencephalon and tyrosine
zebrafish. Nat Methods 7:219–223
hydroxylase distribution in the zebrafish brain
17. Ronneberger O, Liu K, Rath M et al (2012)
(Danio rerio) lead to identification of an
ViBE-Z: a framework for 3D virtual colocal-
ascending dopaminergic system in a teleost.
ization analysis in zebrafish larval brains. Nat
Brain Res Bull 57:385–387
Methods 9:735–742
9. Mueller T, Dong Z, Berberoglu MA et al (2011)
18. Meta R, Roland N, Alida F, Olaf R et al (2012)
The dorsal pallium in zebrafish, Danio rerio
Generation of high quality multi-view confocal
(Cyprinidae, Teleostei). Brain Res 1381:95–105
3D datasets of zebrafish larval brains suitable for
10. Wullimann MF, Rupp B, Reichert H (eds)
analysis using Virtual Brain Explorer (ViBE-Z)
(1996) Neuroanatomy of the zebrafish brain: a
software. Nat Protoc Exchange. doi:10.1038/
topological atlas. Basel, Switzerland: Birkhäuser
protex.2012.031
Verlag, p. 160
Part VI
Chicken Protocols
Chapter 15
Abstract
One of the first steps in studies of gene function is the spatiotemporal analysis of patterns of gene expres-
sion. Indirect immunohistochemistry is a method that allows the detection of a protein of interest by
incubating a histological section with an antibody or antiserum raised against the protein and then local-
izing this primary antibody with a tagged secondary antibody. To determine the cellular source of a protein
of interest, or if a specific antibody is not available, specific transcripts can be localized using in situ hybrid-
ization. A histological section is incubated with a labeled RNA probe that is complementary to the target
transcript; after hybridization with the target transcript, the labeled RNA probe can be identified with an
antibody. Here we describe materials and methods used to perform basic indirect immunohistochemistry
and in situ hybridization on frozen sections through the developing chicken brain, emphasizing controls
and potential problems that may be encountered.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_15, © Springer Science+Business Media, LLC 2014
217
218 Richard P. Tucker and Qizhi Gong
2 Materials
Fig. 1 Schematic illustration of an immunohistochemistry incubation chamber, viewed from above (top ) and
the side (bottom ). The chamber should have a flat bottom and either a securely fitting lid or a lid fashioned
from plastic wrap. As the slides rest directly on the moist paper towel, care should be taken not to let the slides
touch each other, and the “puddle” of antibody should not extend to the very edge of the slide
3 Methods
3.1 Cryosectioning 1. Remove the embryo from the egg by cracking the egg into a
small bowl containing a small volume of PBS (room tempera-
ture). After trimming put the whole embryo (embryonic day
[E]4–E7) or the head (E8–E16) into a 50 mL conical centri-
fuge tube with ice-cold 4 % paraformaldehyde in PBS (pH 7.6).
Cap the tube and place it on ice on a rotary shaker for 1–6 h,
and then store the tube at 4 °C overnight (see Note 3).
2. Pour off and dispose of the fixative appropriately and add ice-
cold PBS to the tube. Cap the tube and place on a rotary shaker
in an ice bath for 30 min. Pour off the PBS and repeat twice
with fresh PBS.
3. Add sucrose to the third PBS rinse to a final concentration of
20–25 %. Cap the tube and place on rotating shaker until the
sucrose has gone into solution. The tissues should “float” on
the sucrose. Store the tube with floating tissues at 4 °C over-
night (see Note 4).
4. Put the sucrose infiltrated (cryoprotected) tissues into a petri
dish or onto a piece of Parafilm©, and trim them with a fresh
razor blade such that the trimmed surface will match the ori-
entation of the sections to be cut in the cryostat. Transfer the
trimmed tissues to a “puddle” of embedding compound to
minimize the presence of any sucrose solution, and then trans-
fer the tissue to an embedding mold partially filled with embed-
ding medium. With forceps orient the tissue so that the cut
surface is resting along the bottom of the mold (see Note 5).
Float the mold in a shallow slurry of 2-methylbutane and
crushed dry ice, taking care not to let the 2-methylbutane
come into direct contact with the embedding medium. When
the embedding medium is solid white, transfer the mold with
the frozen tissue to dry ice (see Note 6).
5. Cut sections with a cryostat using appropriate protocols, and
collect the sections on pre-subbed slides (see Note 7). Allow the
sections to air-dry for 4–6 h, and then either proceed with the
protocol or store the slides at −20 °C to −70 °C (see Note 8).
Immunohistochemistry and In Situ Hybridization in the Developing Chicken Brain 223
rinse, put the slides into a Coplin jar with nuclear stain (Hoechst)
diluted to the 1× working concentration in PBS for 1 min.
10. Coverslip carefully, without introducing air bubbles, using
either the PBS/glycerol wet mount or a permanent mounting
medium suited for fluorescence microscopy (e.g.,
Fluoromount-G) and appropriately sized glass coverslips (see
Note 11).
11. One or more control incubations should also be run. If the anti-
body is well characterized, it may be sufficient to incubate a sec-
tion in the blocking solution overnight and then treat it with the
secondary antibody the following day. Such “secondary anti-
body only” controls will reveal background fluorescence or non-
specific binding of the secondary antibody. When illustrating
this type of control (Fig. 2), be sure to use the same camera
settings that were used when imaging the experimental sections.
If a polyclonal antiserum is used, one should run a control that
includes similarly diluted pre-immune serum.
3.3 In Situ In situ hybridization is a multistep procedure that can take 3–4
Hybridization days. For better planning, make all solutions ahead of time. Frozen
sections can be prepared days to weeks ahead and stored at −20 °C.
DIG-labeled RNA probes need to be made and the quality of the
probe evaluated before starting in situ hybridization.
3.4 Preparation For this protocol we used riboprobes generated against tenascin-C
of DIG-Labeled to match the immunohistochemistry described above.
Riboprobes
1. cDNAs of tenascin-C (GENE ID: 396440 TNC, 2,761–
3,179 bp) were obtained by RT-PCR using primers, 5′- acag-
gactaccattgacctctctg -3′ and 5′- atcagtgccagcattaatggtagc -3′,
and cloned into pCRII-TOPO. Orientation and the sequence
were confirmed by sequencing (see Note 12).
2. 10 μg of cDNA-containing plasmids were linearized either at
the 5′ end for an antisense probe or at the 3′ end of the cDNA
for a control sense probe, with appropriate restriction endo-
nucleases (Fig. 3). The restriction digest should be done over-
night to ensure completeness (see Notes 13 and 14).
3. Take out 1/20 of the reaction to run on an agarose gel to con-
firm that the restriction digest is complete.
4. Purify the restriction digest reactions with phenol extraction:
Bring the volume to 100 μL with ddH2O in a 1.5 mL tube. Add
100 μL of phenol/chloroform/isoamyl alcohol (25:24:1);
vortex for 1 min and centrifuge at 16,000 × g for 5 min; carefully
remove the top aqueous layer and add it to a new tube; add an
equal volume of chloroform to the aqueous layer and vortex for
1 min and centrifuge at 16,000 × g for 5 min. Again, collect the
top aqueous layer and transfer to a new tube.
Immunohistochemistry and In Situ Hybridization in the Developing Chicken Brain 225
Fig. 2 Examples of indirect immunohistochemistry and in situ hybridization on adjacent sections through the
E12 chicken brain. The primary antibody recognizes tenascin-C, and the antisense riboprobe recognizes
tenascin-C transcripts. (a) Anti-tenascin-C labels the optic fiber layer (ofl) and inner plexiform layer (ipl) of the
E12 retina. gcl, ganglion cell layer; inl, inner nuclear layer; rpe, retina pigment epithelium. (b) An adjacent sec-
tion through the E12 retina subjected to in situ hybridization with a tenascin-C antisense probe. The dark
reaction product is found in the gcl and inl. (c). An adjacent section incubated with a control sense probe,
illustrating the specificity of the hybridization signal in b. The rpe is dark due to the presence of melanosomes
and not the target transcript. Scale bar: 100 μm (a–c). (d) Anti-tenascin-C immunoreactivity is found around
the nucleus of Edinger-Westphal (EW) and oculomotor nucleus (ON). AQ, cerebral aqueduct. (e) The tenascin-C
antisense probe hybridizes in large cells within the EW and ON as well as in the ependymal layer (el). (f). The
control section incubated with a sense probe shows that the reaction products seen in E are specific.
(g). The tenascin-C antibody labels the region underlying the ependymal layer (el) of the optic tectum as well
as the stratum griseum periventriculare (SGP). VT ventricle of the optic tectum, SGC stratum griseum centrale.
(h) A secondary antibody only control shows the absence of background fluorescence in the same region in an
adjacent section. (i) An adjacent section incubated with a tenascin-C antisense probe. The reaction product is
found in the el and SGC. Scale bar: 500 μm (d–i)
226 Richard P. Tucker and Qizhi Gong
Fig. 3 Steps for making tenascin-C riboprobes. A cDNA fragment of tenascin-C is cloned into the pCRII-TOPO
plasmid with an Sp6 promoter at its 5′ end. Restriction enzyme Spe I and EcoRV sites are located at different
ends of the cloned cDNA. To make antisense probes, plasmids are digested with Spe I and transcribed with T7
RNA polymerase. To make control sense probes, plasmids are digested with EcoR V and transcribed with Sp6
RNA polymerase. DIG-labeled UTPs are used to incorporate DIG (indicated by black dots) into the riboprobes
Template DNA 1 μg
5× transcription buffer 4 μL
DIG RNA labeling mix 2 μL
0.1 M DTT 1 μL
RNA polymerase 2 μL
RNase inhibitor 0.5 μL
RNase-free water to 20 μL
Immunohistochemistry and In Situ Hybridization in the Developing Chicken Brain 227
RNase inhibitor 1 μL
DNase 1 μL
3.5 In Situ 1. Dry sections with cold air using a hair dryer for up to 1 min
Hybridization: Day 1. (see Note 16).
Pre-hybridization 2. All pre-hybridization and subsequent washing steps are done
and Hybridization: in a vertical slide mailer that holds five slides. Complete immer-
RNase-Free sion of tissue sections requires 15 mL of solution for each
mailer.
3. Place slides into a slide mailer and postfix with 4 % paraformal-
dehyde in PBS for 15 min.
4. Rinse in DEPC-treated PBS three times, 1 min each.
5. Digest sections with proteinase K at room temperature for
6–12 min (see Note 17).
6. Inactivate the proteinase K by incubating the sections in 4 %
paraformaldehyde in PBS for 10 min at room temperature.
7. Rinse in DEPC-treated PBS three times, 1 min each.
8. Treat the sections with 0.2 N HCl for 10 min at room tem-
perature (see Note 18).
9. Rinse in DEPC-treated PBS three times, 1 min each.
10. Measure 15 mL of 0.1 M triethanolamine–HCl into a 15 mL
tube. Add 37.5 μL of acetic anhydride drop by drop. Mix by
inverting and pour into the mailer immediately.
11. Transfer slides into the mailer and incubate sections for 10 min
at room temperature.
12. Wash in DEPC-treated PBS three times, 1 min each.
13. Dehydrate sections in increasing concentrations of ethanol
(50 %, 75 %, 95 %, 100 %, 100 %) for 2 min each. Sections are
subsequently air-dried for 2 min.
14. Add 400 μL of hybridization solution to two 1.5 mL tubes.
Heat the hybridization solution at 85 °C for 10 min.
228 Richard P. Tucker and Qizhi Gong
3.6 In Situ 1. Carefully remove the coverslips by dipping the slides in and
Hybridization: Day 2. out of 2× SSC (see Note 22).
Post-hybridization 2. Incubate in 2× SSC and 50 % formamide at 60 °C for 30 min.
Washes and
3. Transfer sections to TNE buffer at 37 °C for 10 min.
Immunological
Detection: 4. Replace the TNE buffer with TNE plus RNaseA (20 μg/mL)
and incubate at 37 °C for 30 min.
Non-RNase-Free
5. Wash with TNE for 10 min at 37 °C.
6. Further wash the sections in 2× SSC at 60 °C for 30 min.
7. Proceed with high-stringency washes in 0.2× SSC twice for
30 min at 60 °C (see Note 23).
8. Let the sections stay cool at room temperature for 5 min.
9. Wash with Buffer 1 for 5 min at room temperature.
10. Incubate with 1 % blocking reagent for 1 h at room
temperature.
11. Briefly rinse the sections with Buffer 1 plus 0.1 % Tween 20
(B1-T). Put 300 μL of alkaline phosphatase-conjugated anti-
DIG antibody (1:1,000 with B1-T) onto each slide. Cover the
sections with coverslips and place the slides into a humidified
chamber (e.g., a flat-bottomed box with a moist paper towel as
in Fig. 1, but with the sections coverslipped to reduce the vol-
ume of antibody solution).
12. Incubate overnight at 4 °C.
3.7 In Situ 1. Remove the coverslips by dipping the slides in B1-T. Wash in
Hybridization: Day 3. B1-T three times 10 min each at room temperature.
Signal Detection: 2. Precondition with Buffer 2 (B2) for 5 min.
Non-RNase-Free
3. Replace with 20 mL of B2 with 400 μL of NBT/BCIP. The
NBT/BCIP solution is light sensitive. Keep the reaction in the
dark.
Immunohistochemistry and In Situ Hybridization in the Developing Chicken Brain 229
4 Notes
25. Mounting should be done with a quick wash with water and
using an aqueous mount. Do not dehydrate with xylene as the
treatment will result in the formation of crystals.
Acknowledgments
We would like to thank Huaiyang Chen for her help in the design
of the riboprobe and the cloning of the cDNA, and Chenghao
Qian for his technical help with the in situ hybridization.
References
1. Coons AH, Creech HJJ, Jones RN, Berliner E 9. Chiquet M, Fambrough DM (1984) Chick
(1942) The demonstration of pneumococcal myotendinous antigen. I. A monoclonal anti-
antigen in tissues by the use of fluorescent body as a marker for tendon and muscle mor-
antibody. J Immunol 45:159–170 phogenesis. J Cell Biol 98:1926–1936
2. Beutner EH, Witebsky E (1963) Studies on 10. Iezzoni JC, Kang JH, Bucana CD, Reed JA,
organ specificity. Xv. Immunohistologic evalu- Brigati DJ (1993) Rapid colorimetric detec-
ation of reactions produced by thyroid autoan- tion of epidermal growth factor receptor
tibodies. J Immunol 91:204–209 mRNA by in situ hybridization. J Clin Lab
3. Akam ME (1983) The location of Ultrabithorax Anal 7:247–251
transcripts in Drosophila tissue sections. 11. Bodkin DK, Knudson DL (1985) Assessment
EMBO J 2:2075–2084 of sequence relatedness of double-stranded
4. Hafen E, Levine M, Garber RL, Gehring WJ RNA genes by RNA-RNA blot hybridization.
(1983) An improved in situ hybridization J Virol Methods 10:45–52
method for the detection of cellular RNAs in 12. Herrington CS, McGee JO (1994)
Drosophila tissue sections and its application Discrimination of closely homologous human
for localizing transcripts of the homeotic genomic and viral sequences in cells and tis-
Antennapedia gene complex. EMBO J sues: further characterization of Tmt.
2:617–623 Histochem J 26:545–552
5. Garner CC, Tucker RP, Matus A (1988) 13. Herrington CS, Anderson SM, Graham AK,
Selective localization of messenger RNA for McGee JO (1993) The discrimination of high-
cytoskeletal protein MAP2 in dendrites. risk HPV types by in situ hybridization and the
Nature 336:674–677. doi:10.1038/336674a0 polymerase chain reaction. Histochem J
6. Wilkinson DG (1998) In situ hybridization: a 25:191–198
practical approach, 1999/01/30th edn. 14. Herrington CS, Graham AK, Flannery DM,
Oxford University Press, Oxford, p 224 Burns J, McGee JO (1990) Discrimination of
7. Zeller R, Rogers M, Haramis AG, Carrasceo closely homologous HPV types by nonisotopic
AS (2001) In situ hybridization to cellular in situ hybridization: definition and derivation
RNA. Current protocols in molecular biol- of tissue melting temperatures. Histochem J
ogy/edited by Frederick M. Ausubel … [et al.] 22:545–554
Chapter 14, Unit 14 13, doi:10.1002/ 15. Evans MF, Aliesky HA, Cooper K (2003)
0471142727.mb1403s55 Optimization of biotinyl-tyramide-based in
8. Chiquet-Ehrismann R, Tucker RP (2011) situ hybridization for sensitive background-
Tenascins and the importance of adhesion free applications on formalin-fixed, paraffin-
modulation. Cold Spring Harb Perspect Biol embedded tissue specimens. BMC Clin Pathol
3, doi:10.1101/cshperspect.a004960 3:2. doi:10.1186/1472-6890-3-2
Chapter 16
Abstract
Tissue transplantation is an important approach in developmental neurobiology to determine cell fate, to
uncover inductive interactions required for tissue specification and patterning as well as to establish tissue
competence and commitment. Avian species are among the favorite model systems for these approaches
because of their accessibility and relatively large size. Here we describe two culture techniques used to gener-
ate quail-chick chimeras at different embryonic stages and methods to distinguish graft and donor tissue.
Key words Chick, Brain, Neural plate, Neural tube, Quail, Transplantation
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_16, © Springer Science+Business Media, LLC 2014
235
236 Andrea Streit and Claudio D. Stern
2 Materials
transmitted light base and for in ovo work a cold light source
(fiber optics) for illumination from the top. Fertile hens’ or quails’
eggs are incubated in a humidified incubator at 38 °C until they
have reached the stage desired; staging of host and donor embryos
is performed according to Hamburger and Hamilton [25]. All
solutions are diluted from autoclaved stock solutions in distilled
water immediately before use; beakers for salines are autoclaved
before use.
2.1 Preparing Chick Operations on primitive streak to early somite stage (HH3+-8) chick
Hosts for New Culture host embryos are performed in modified New culture [26, 27]; at
this stage embryos are fragile and difficult to manipulate in ovo, and
survival rate in ovo is poor. On the other hand, in New culture
embryos can only be grown for 24–36 h even in an expert’s hands.
In addition to the above materials, this method requires:
1. Pyrex baking dish about 5 cm deep with 2 l capacity.
2. Watch glasses about 5–7 cm diameter.
3. Rings cut from glass tubing (approx. 27 mm outer diameter,
24 mm inner diameter and 3–4 mm deep; obtained from a
local glass blower).
4. 35 mm plastic dishes with lids (bacteriological grade).
5. Plastic box with lid for incubating culture dishes.
6. Pannett-Compton saline is prepared from two stock solutions,
which can be kept at 4 °C if autoclaved. Solution A: 121 g
NaCl, 15.5 g KCl, 10.42 g CaCl2 ⋅ 2H2O, 12.7 g MgCl2 ⋅ 6H2O,
H2O to 1 l. Solution B: 2.365 g Na2HPO4 · 2H2O, 0.188 g
NaH2PO4 · 2H2O, H2O to 1 l. To prepare working solution
just before use, mix (in order) 120 ml A, 2,700 ml H2O, and
180 ml B. Do not mix concentrated stocks of A and B.
2.2 Preparing Chick After HH8-9, operations are performed in ovo; under perfect
Hosts for In Ovo conditions embryos can be grown for a long time, even until hatch-
Operation ing. Collect the following materials in addition to those listed at
the beginning of the methods section.
1. Scalpel with No. 3 handle and No. 11 blades.
2. Plasticine or foam (from packaging) to make a ring for resting
eggs on their side.
3. PVC tape to seal the eggs.
4. 5 ml syringe with 21G needle (for removing albumen).
5. 1 ml syringe with 27G (or finer) needle (for ink injection).
6. 1 ml syringe with 21G needle (for antibiotics).
7. Paper tissues.
8. Indian ink (Pelikan Fount India or Winsor and Newton;
diluted 1:10 in saline).
238 Andrea Streit and Claudio D. Stern
2.3 Preparing The following materials are required to harvest quail embryos:
Quail Donors
1. Glass Petri dish (10 or 15 cm diameter, depending on the
number of embryos to be collected).
2. Spoon spatula for collecting embryos.
3. Glass Pasteur pipette cut at the shoulder and fire polished
(for embryo transfer).
4. Rubber teats.
5. 500 ml Tyrode’s saline.
6. Dissecting microscope with transmitted light base.
2.4 Grafting To dissect quail and chick tissue for grafting, the following materi-
als are required:
1. 35 mm Sylgard-coated dish for dissecting (this should never
come into contact with fixative).
2. Entomological insect pins (A1; steel) for pinning out embryos
on the Sylgard dish.
3. Insect pins mounted on Pasteur pipettes or tungsten needles
(Goodfellow; 100 μm diameter, mounted on aluminum
holders or glass rods using sealing wax; sharpen by repeated
exposure of the tip to a very hot Bunsen flame).
4. 30G needles mounted on 1 ml syringes.
5. P20 Gilson pipette and yellow tips.
In addition, for in ovo transplantation in older embryos, the
following materials are needed:
6. Micro-knife (e.g., micro-feather microsurgery knives for eye
surgery 15E blade angle).
7. Aspirator tube (Sigma A5177).
8. 50 μl borosilicate glass capillaries (for trypsin injection) pulled
to fine injection needles using an electrode puller, tips broken
off (puller settings need to be determined; needles should be
fine enough to avoid fluid uptake by capillary forces but large
enough to deliver small amounts of trypsin by air pressure).
Quail-Chick Chimeras 239
2.5 Fixing Embryos Embryos are fixed several hours or days after transplantation; they
and Analyzing Results can be analyzed by in situ hybridization or immunostaining to label
specific tissues or cell types, followed by labelling with quail-specific
antibodies to detect the graft by in situ hybridization using chick-
and quail-specific probes or by histological sectioning followed by
Feulgen and Rossenbeck staining to reveal nucleoli [21]. Depending
on the analysis, different fixatives are used. Fixing requires:
1. Petri dish for collecting embryos (for in ovo).
2. 3.5 or 10 mm Sylgard dish for pinning out embryos.
3. Insect pins (see above).
4. 7 ml glass vials and phosphate-buffered saline (PBS).
5. For whole-mount in situ hybridization, embryos are fixed in
4 % PFA, 1 mM EGTA in PBS, and 0.1 % Tween-20. Embryos
are stored in methanol.
6. For Feulgen staining and in situ hybridization on sections
[28], embryos are fixed in Zenker’s fixative: 50 g HgCl2, 25 g
K2Cr2O7, 10 g of Na2SO4 × 10 H2O in 1 l distilled water; before
use, add 5 ml glacial acetic acid to 100 ml of the solution: or
fixed in Carnoy’s fixative: 50 % ethanol, 11.1 % formaldehyde,
10 % glacial acetic acid [24, 28].
The most important point for further analysis is the detection
of quail tissue. While traditionally Feulgen staining has been used to
reveal the difference between chick and quail nucleoli [22, 29, 30],
now quail-specific antibodies are the favorite method. The mono-
clonal mouse antibody QCPN (Developmental Studies Hybridoma
Bank) labels all quail cells, while QN is specific for neurites [23].
We generally perform QCPN staining after whole-mount in situ
hybridization; this procedure requires:
1. Glass vials.
2. Pasteur pipettes.
3. Rubber teats.
4. Rocking platform.
5. PBS, blocking buffer: 1 % goat serum, 0.5 % Triton X-100 in
PBS.
6. Anti mouse IgG-HRP coupled (Jackson).
7. 100 mM Tris–HCl pH 7.4.
8. 50 mg/ml 3,3′-diaminobenzidine (DAB) in 100 mM Tris–
HCl pH 7.4.
9. 0.3 % H2O2 in 100 mM Tris–HCl pH 7.4.
240 Andrea Streit and Claudio D. Stern
3 Methods
3.1 Preparing Quail To dissect the donor tissue, quail embryos are first removed from
Donor Embryos for the egg and cleaned using the following steps.
Grafting
1. Remove quail eggs from incubator. Using fine scissors, gently
tap the shell near the blunt end of the egg to penetrate the
shell. Use the tip of the scissors to cut off a small cap of shell;
avoid damaging the yolk.
2. Pour thin egg white into waste; use the scissors to help and cut
through the rather thick albumen if required (see Note 1).
3. Once most albumen is removed, turn the yolk by stroking it
very gently with the sides of the scissors to make the embryo
become visible on top of the yolk.
4. Using the scissors make four cuts into the vitelline membrane
around the embryo; make sure that all the cuts meet (see Note 2).
5. Using the spoon, pick up the square of embryo and membrane
including a little yolk; try to collect as little yolk as possible.
6. Transfer the embryo including yolk and membrane into the
large Petri dish containing Tyrode’s saline under a dissecting
microscope by sliding it carefully off the spoon. Using fine for-
ceps, turn the square of yolk/membrane/embryo so you can
see the embryo.
7. Once all donor embryos have been collected in the Petri dish,
separate the embryos from adhering yolk. Work at low magni-
fication; use two pairs of forceps to pick up a corner of the
vitelline membrane with one and slowly but steadily fold it
back, steadying the yolk with the other. Make sure that the
membrane and embryo remain submerged in saline. The
embryo will remain attached to the membrane. If not, peel off
the membrane completely, and then use the forceps gently to
remove the embryo from the underlying yolk.
8. Using the wide-mouth Pasteur pipette, pick up the embryo,
with (better) or without membrane, and transfer to a 10 cm
dish with clean saline. To clean the embryo use a fire-polished
Pasteur pipette and gently blow saline over it; this will
remove yolk particles. The embryos are now ready for dissec-
tion and grafting and can be kept for 1–2 h before proceed-
ing further.
Quail-Chick Chimeras 241
3.2 Preparing Chick At primitive streak and neural plate stages, operations in avian
Hosts for New Culture embryos are most easily performed in New culture. The method
described below is based on New’s original technique [27] modi-
fied by Stern and Ireland [26]. This modified culture method uses
rings cut from glass tubes, instead of rings bent from glass rods and
35 mm plastic dishes instead of glass watch glasses resting inside a
large glass Petri dish. The rings cut from tubing generate a slightly
rough surface that grips the vitelline membrane and therefore
allows easy transfer of the culture into the plastic dish. To set up
the cultures, proceed as follows (Fig. 1):
1. Remove eggs from the incubator.
2. Fill the large Pyrex dish with about 1.5 l of Pannett-Compton
saline; the volume should be large enough that eggs yolks are
submerged completely.
3. To open an egg, tap its blunt egg with the coarse forceps and
carefully remove pieces of the shell. Discard the thick albumen
into the waste bucket, assisted with the coarse forceps. Collect
the thin albumen in a small beaker (see Note 3).
4. Carefully tip the yolk into the Pyrex dish containing saline, tak-
ing care not to damage the membrane on the edges of the
shell. Carefully turn the yolk with the side of the coarse forceps
so the embryo is facing upwards. Now place a watch glass and
a glass ring into the dish.
5. Cut the vitelline membrane enveloping the yolk just below the
equator using small scissors; you can use one pair of forceps to
push the yolk around gently while continuing to cut all the way
around its circumference (see Note 4).
6. Using both pairs of fine forceps, peel the vitelline membrane
with the embryo attached slowly but very steadily off the yolk.
Use one pair of forceps to pull the edge of the membrane
slightly upwards (about 25–30° angle from the yolk surface)
and the other to hold the yolk down. Do not pull tangentially
along the yolk: this may detach the embryo from the mem-
brane. Do not stop during this process. The embryo should
come off with the membrane (see Note 5).
7. Turn the vitelline membrane with the inner face containing the
embryo pointing upwards and slide it, preserving its orientation,
onto the watch glass. Place the ring over it so that the mem-
brane protrudes around the ring and the embryo sits in its
center. Remove the watch glass, ring, and embryo from the
dish; tilt the assembly gently to pour off some saline while
steadying the ring with one finger (see Note 6).
8. Dry the bottom of the watch glass on some tissue. Using fine
forceps, carefully wrap the lose edges of the vitelline membrane
242 Andrea Streit and Claudio D. Stern
Fig. 1 Setting up New culture. (a) Set up. (b) Instruments. (c) Opening eggs. (d) Removing albumen. (e) Yolks
in Pyrex dish. (f) Cutting the vitelline membrane at the equator. (g) Removing membrane and embryo from the
yolk. (h) Membrane with embryo facing upwards on watch glass. (i) Placing glass ring on the membrane.
(j) Culture assembled on ring and watch glass. (k) Removing assembly form Pyrex dish. (l) Cleaning the culture.
(m) Primitive streak stage embryo on watch glass. (n) Embryo after removal of a piece of neural plate.
(o) Setting up the culture in a Petri dish with albumen. (p) Finished New culture
over the edge of the ring, all the way around its circumference.
Pull the membrane slightly so its bottom is smooth and free
from wrinkles, but be careful not to pull so tight that it breaks
(see Note 7).
9. Using the fire-polished Pasteur pipette, rinse the outside of the
ring to remove yolk particles. If there is a lot of egg albumen
remaining under the membrane, lift the ring gently and use the
Quail-Chick Chimeras 243
Pasteur pipette to remove it. Clean the yolk over and around
the embryo using clean saline; be careful not to dislodge the
embryo form the membrane. Damaged embryos do not grow
well or normal. If there is a lot of vitelline membrane inside the
ring, trim off the excess with the fine scissors while lifting the
edges with fine forceps. At this stage embryos are ready for
transplantation and can be kept on the bench for some time;
make sure they remain well submerged under saline and there
is sufficient saline on the watch glass. If keeping them for a few
hours, place them on a wet tissue and cover with a large glass
or plastic plate/dish.
10. To finalize the cultures after transplantations, work under the
microscope; carefully remove any remaining saline, both inside
and outside the ring. Drying helps the graft and host tissue to
heal faster. During culture the embryo and the inside of the
ring must remain dry.
11. Pour some thin albumen (about 2–3 mm thick layer) on the
bottom of a 35 mm Petri dish. Using fine forceps, slide the
ring with vitelline membrane off the watch glass, and transfer
it to the dish; lower it onto the albumen making sure that no
air is trapped underneath. Press the ring lightly onto the bot-
tom of the dish using two forceps to allow it to adhere.
12. Remove the excess albumen if its level comes close to the edge
of the ring. At this point, remove any remaining liquid from
inside the ring using a fire-polished Pasteur pipette. The vitel-
line membrane should be slightly dome shaped; this will help to
drain off any fluid that accumulates during culture (see Note 8).
13. To seal the Petri dish, wet the inside of the lid with a thin film of
albumen all around the edge, discard the excess, and place onto
the bottom part. Press lid down slightly to seal (see Note 9).
14. Place the dish in a plastic box containing a piece of wet tissue,
seal the box, and place it into an incubator at 38 °C.
3.3 Preparing Chick Later-stage embryos are generally grown in ovo, which allows
Hosts for In Ovo embryos to grow for long periods, even until hatching. Eggs must
Culture be incubated lying on their side, so the yolk turns with the embryo
facing upwards to make it accessible for manipulation. To prepare
hosts for in ovo operations, use the following procedure (Fig. 2):
1. Remove the eggs from the incubator, place one egg onto the
egg rest, and clean with 70 % ethanol; be careful not to rotate
the egg.
2. Hold the 5 ml syringe with 21G needle nearly vertical, and
insert the needle into the blunt end of egg until you feel the
shell at the bottom. Remove about 1 ml egg albumen and
discard. This lowers the embryo away from the top of the shell.
244 Andrea Streit and Claudio D. Stern
Fig. 2 In ovo culture. (a) Eggs are incubated on their side and placed on egg rest. Circle labels the blunt end of
the egg. (b) Blunt end is used to remove albumen with 5 ml syringe. (c) Inserting syringe. (d) Scoring the win-
dow. (e) Windowed egg. (f) Windowed egg with Vaseline border surrounding the window. (g) Embryo before ink
injection. (h) Ink injection using a 1 ml syringe. (i) Ink injection: Embryo is clearly visible. (j) Embryo after ink
injection. (k) Removing albumen to lower the embryo. (l) Eggs sealed with tape
3.4 Grafting The procedure below describes orthotopic, isochronic neural plate
Procedure: Quail grafts from quail donors into chick hosts at HH3+/4. The same
Neural Plate into Chick method can be applied for heterotopic and heterochronic grafts or
Hosts in New Culture transplantation of other tissues.
1. To prepare the host follow the procedure described under
Subheading 3.1 until step 9.
2. Replace one of the eyepieces of the microscope with an eyepiece
containing a graticule; a protractor, to measure angles, is par-
ticularly useful.
3. Place a host embryo from Subheading 3.1, step 9 (kept on a
watch glass) under the microscope and center the graticule on
the node.
246 Andrea Streit and Claudio D. Stern
3.5 Grafting This section describes orthotopic and isochronic neural tube grafts
Procedures: Quail in embryos older than HH9; as with transplantations in younger
Neuroectoderm into embryos described above, the same techniques can be used for
Chick Host In Ovo heterotopic and heterochronic experiments. First, prepare the
donor embryo, and then turn to the host.
Quail-Chick Chimeras 247
1. To prepare the quail donor, pin out the embryo dorsal side up
on a Sylgard dish in Tyrode’s saline and place under a dissect-
ing microscope with transmitted light base.
2. Using tungsten needles or a micro-knife, make a longitudinal
incision into the ectoderm dorsal to the neural tube on both of
its sides.
3. Replace the Tyrode’s with trypsin solution; working at high mag-
nification peel the ectoderm away from the neural tube using the
back of a 30G needle. In the same way, gently scrape any loosely
attached cells (neural crest depending on stage) off the neural
tube and free it from the adjacent tissues (see Note 16).
4. Progressively separate the neural tube from the underlying
notochord using a micro-knife to push it from side to side,
allowing the Trypsin to penetrate; when completely detached,
cut the neural tube transversely at its anterior and posterior
ends to free it.
5. Remove the excised neural tube using a Gilson pipette set to
3–5 μl and place into a 35 mm Petri dish containing saline with
5 % serum (see Note 17). Graft can be kept on ice until use.
6. Prepare the chick host by following steps 1–7 in Subheading 3.2.
Use a mounted insect pin make a small hole into the vitelline
membrane just over the area to be operated. The hole should
be as small as possible.
7. Replace the drop of Tyrode’s saline with trypsin solution and
follow steps 2–4 above to excise the same section of the neural
tube as in the quail donor.
8. Remove the excised neural tube using a Gilson pipette set to
3–5 μl and replace the trypsin solution with fresh Tyrode’s
saline twice.
9. Pick up the graft using a Gilson pipette; rinse in Tyrode’s saline
without serum before transferring it to the host.
10. Using a Gilson pipette and working under low magnification,
transfer the graft into the saline bubble over the host embryo.
11. Use a mounted insect pin or 30G needle to place the graft into
the hole made by removal of the host neural tube. Preserve
anterior-posterior and dorsoventral orientation (see Note 18).
12. Once the transplant is in position, carefully remove the saline
using a Pasteur pipette while observing under low magnifica-
tion. If needed, reposition the graft using a mounted pin.
13. Finish the egg by following steps 8–13 in Subheading 3.2.
3.6 Detecting As outlined above, grafted embryos can be analyzed in various ways
Quail Tissue depending on the question; these include whole-mount or section in
situ hybridization and tissue- or cell-specific immunohistochemistry.
248 Andrea Streit and Claudio D. Stern
4 Notes
12. Antibiotics are generally only required for long culture peri-
ods. Normally embryos survive well for 2–3 days without this.
13. Low survival rates can be due to a number of factors. The most
common problems are dehydration, damage to critical blood
vessels, and infection. Working in a drop of saline helps to alle-
viate dehydration. To avoid infection ensure you use clean
solutions; instruments can be cleaned periodically while work-
ing in distilled H2O and ethanol (make sure it evaporates
before using to operate), while tungsten needles are flamed
periodically to keep them sharp.
14. Be careful not to make any holes in the vitelline membrane.
15. Drying the area surrounding the graft greatly improves heal-
ing; be careful not to suck up the graft into the capillary.
16. Be patient; the trypsin works almost by itself, and it generally
sufficient to push the adjacent tissues away using your instru-
ments. Avoid going too deep and cut a hole into the endo-
derm; this will make ink and yolk leak out.
17. This inactivates trypsin; this is important because the tissue
should be exposed to proteolytic enzymes for as short as pos-
sible to avoid disintegration.
18. If you encounter difficulties to preserve orientation, mark one
end of the neural tube with a small crystal of carmine powder.
19. Make sure the pH is properly adjusted to 7.4; the reaction is
pH sensitive.
20. DAB is carcinogenic; make sure to wear appropriate protective
clothing (lab coat, gloves) and consult the local health and
safety regulations for inactivation and disposal.
Acknowledgment
References
1. Stern CD (2005) Neural induction: old prob- 3. Spemann H, Mangold H (1924) Über
lem, new findings, yet more questions. Induktion von Embryonalanlagen durch
Development 132:2007–2021 Implantation artfremder Organisatoren.
2. Waddington CH (1934) Experiments on Wilhelm Roux’ Arch Entwicklungsmech Org
embryonic induction. Part I: the competence 100:599–638
of the extra-embryonic ectoderm. Part II: 4. Cobos I, Shimamura K, Rubenstein JL,
experiments on coagulated organisers in the Martinez S, Puelles L (2001) Fate map of the
chick. Part III: A note on inductions by chick avian anterior forebrain at the four-somite
primitive streak transplanted to the rabbit stage, based on the analysis of quail-chick
embryo. J Exp Biol 11:211–227 chimeras. Dev Biol 239:46–67
Quail-Chick Chimeras 251
Abstract
The mouse is the most commonly used vertebrate model for the analysis of gene function because of the
well-established genetic tools that are available for loss-of-function studies. However, studies of gene func-
tion during development can be problematic in mammals. Many genes are active during different stages of
development. Absence of gene function during early development may cause embryonic lethality and thus
prevent analysis of later stages of development. To avoid these problems, precise temporal control of gene
silencing is required.
In contrast to mammals, oviparous animals are accessible for experimental manipulations during
embryonic development. The combination of accessibility and RNAi-based gene silencing makes the
chicken embryo a powerful model for developmental studies. Depending on the time window during
which gene silencing is attempted, chicken embryos can be used for RNAi in ovo or cultured in a domed
dish for easier access during ex ovo RNAi. Both techniques allow for precise temporal control of gene
silencing during embryonic development.
Key words Neural development, Chicken embryo, Artificial miRNA, RNA interference,
Electroporation, Gene silencing
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_17, © Springer Science+Business Media, LLC 2014
253
254 Irwin Andermatt and Esther T. Stoeckli
2 Materials
3 Methods
3.1 Windowing Eggs To obtain access to the developing embryo, a window is cut into
the eggshell (Fig. 1). Independent of the time of electroporation,
eggs are windowed at the second or third day of incubation (E2
and E3, respectively; see Note 1).
1. Incubate eggs at 38.5 °C and 45 % humidity (see Notes 2–4).
2. Let embryos develop until they have reached the desired stage
for the experimental manipulation. Staging of the embryo is
done according to Hamburger and Hamilton [14]; see Note 5.
3. About 20 min before windowing, place the egg on its side to
allow the embryo to reposition on top of the egg yolk.
4. Wipe the eggshell using facial tissue and 70 % ethanol to avoid
contamination (see Note 6).
5. Place a strip of Scotch tape to cover the area of the planned
window. This will prevent pieces of eggshell from falling onto
the embryo when cutting the window.
6. Use a scalpel to drill a hole into the corner of the intended
window and into the blunt end of the egg (see Fig. 1 and
Note 7).
7. Remove ~3 ml of albumen by pushing the needle of the syringe
at an angle greater than 45° into the hole at the blunt end of
the egg. This will avoid damage to the egg yolk (see Note 8).
8. Use a paint brush to seal the hole at the blunt end of the egg
with melted paraffin.
9. Cut a window into the eggshell. To avoid damaging the
embryo, carefully hold scissors horizontally.
10. Seal the window with a coverslip and paraffin. Use a brush to
apply melted paraffin to the edges of the window and carefully
press a coverslip onto the hot paraffin (alternatively, use Scotch
258 Irwin Andermatt and Esther T. Stoeckli
Fig. 1 Egg windowing. (a) After the egg is cleaned with 70 % ethanol, a strip of sticky tape is put on the top of
the egg, and two holes are drilled into the edge of the presumptive window and the blunt end of the egg
(arrowheads). (b) The needle is inserted at an angle greater than 45° in order not to damage the egg yolk.
Removal of about 3 ml of albumen will detach the embryo and the blood vessels from the eggshell. (c) The hole
at the blunt end is sealed (arrowhead ), and a window is cut into the shell starting at the previously drilled hole.
(d) With a brush melted paraffin is applied to the edges of the window and immediately covered with a cover-
slip (open arrows )
tape to seal the window; see Note 9). As paraffin cools down
quickly, carefully pressing a soldering iron on the coverslip will
remelt the paraffin below the glass and lead to proper sealing.
11. Put the windowed egg in the incubator at 38.5 °C and 45 %
humidity until further use (see Notes 3 and 4).
3.2 In Ovo 1. Clean the working space with 70 % ethanol and autoclave your
Electroporation working tools (see Note 6).
2. Prepare capillaries to make injection needles using the glass
needle puller (see Note 10).
3. Remove the coverslip or the tape covering the window. To
remove the coverslip briefly press the hot soldering iron onto
the glass.
4. To get direct access to the embryo, carefully remove the extra-
embryonic membranes with forceps and spring scissors (Fig. 2).
5. Break off the tip of the previously pulled needles to obtain a
diameter of 5–7 μm. Plug the needle into the polyethylene
tubing and fill the tip of the needle with your injection mix
(see Note 10).
RNAi in Brain Development 259
Fig. 2 (a, b) Schematic drawing of in ovo electroporation of the caudal neural tube. (a) Injection of dsRNA or
miRNA into the central canal of the spinal cord. (b) Electroporation with anode and cathode positioned parallel
to the body axis. (c) Depending on the positioning of the electrodes, electroporation can be spatially controlled.
(d) Successful unilateral transfection of EGFP in the right hemisegment is visualized on a cryosection of a HH25
embryo. (e) Unilateral in ovo injection and electroporation of an E3 chicken embryo. Using forceps, the extra-
embryonic membranes are removed to obtain access to the spinal cord. The injection mix is injected into the
central canal of the lumbar region of the spinal cord. The injection volume is controlled by mouth. The maximal
injection volume is achieved when the blue solution reaches both the ventricle and the tail (arrowheads ). For
unilateral electroporation the electrodes are placed laterally in parallel to the body axis of the embryo. Make
sure not to touch major blood vessels to avoid fatal bleeding. (f) For dorsal or ventral electroporation HH20
embryos are easy to handle because their body is more detached from the egg yolk and slightly tilted to the
side compared to the younger embryo shown in (e). Note that the lumbar region is tilted to the side and there-
fore you see more lateral parts of the spinal cord. Use the forceps to pull the extraembryonic membranes
sidewards to position the embryo and stabilize it in the desired position for injection and electroporation with
anode and cathode positioned dorsally and ventrally, respectively. For dorsal electroporation the polarity would
be reversed. Scale bar: 2 mm
Fig. 3 Ex ovo culture. (a) The embryo is transferred from the egg into a domed dish at E2.5 and kept at 38.5 °C.
(b and c) Ex ovo culture of a HH34 embryo (E8) ready for cerebellar injection and electroporation
3.3 Ex Ovo Culture 1. Incubate fertilized eggs for 2.5 days at 38.5 °C and at least
of Chicken Embryos 45 % humidity (see Notes 4 and 17).
2. Position the eggs on the side for 20 min to allow for the
embryo to position on top of the egg yolk.
3. Wipe the egg with 70 % ethanol and crack it on a sharp edge.
Transfer the whole egg content into the domed dish without
destroying the egg yolk (Fig. 3).
4. Cover the culture with a lid from a petri dish to minimize evap-
oration and keep the ex ovo cultures in the incubator until
further use.
3.4 Ex Ovo Injection Here we describe injection and electroporation of the developing
and Electroporation cerebellum of embryos at E8 as an example and as previously
described [3]. Other areas of the brain can be targeted with the
same parameters as a starting point.
1. Use autoclaved tools and clean the work space with 70 %
ethanol.
2. Stage the embryos according to Hamburger and Hamilton [14].
RNAi in Brain Development 261
Fig. 4 Ex ovo electroporation. (a and b) Using spring scissors and forceps, a small hole is cut into the extraem-
bryonic membranes in order to get access to the desired injection site. In the example shown here, the injec-
tion would be planned into the eye. (c) For injections into the developing cerebellum, the blood vessels on the
back of the head can be used as landmarks. (d) The tweezer electrodes are placed parallel to the head for
electroporation. Importantly, electrodes should not touch the embryo. Scale bar in (d): 2 mm. (e) and (f) Two
examples of successful electroporation of the cerebellum. Coronal sections are shown with EGFP-positive cells
(green ) indicating successful transfection. Axonin-1/contactin-2 is a marker for parallel fibers, axons of
granule cells (red ). V ventricle, PS pial surface
3. Remove the lid and cut a small hole of 3–4 mm diameter into
the extraembryonic membranes above the head, where the
injection is planned (Fig. 4).
4. Depending on the age of the embryo and the injection site, it
may be required to fix the head of the embryo with a spatula
bent to a hook by placing it underneath the neck.
5. Use a glass capillary connected to a piece of tubing to inject
the mix (see Notes 10–13) into the cerebellum (Fig. 4c). To
target all the cerebellar layers, insert the glass capillary first into
the ventricular zone and apply constant pressure while pulling
the needle out (see Note 18).
6. Put a few drops of sterile PBS on the injection site and place
the platelet electrodes on either side of the head of the embryo.
Avoid touching embryonic tissue with the electrodes to pre-
vent damage. Use six pulses of 40 V and 99 ms duration with
1 s interpulse intervals (Fig. 4d; see Note 16).
7. Cover the dish and put it back to the incubator until the desired
stage for analysis is reached.
262 Irwin Andermatt and Esther T. Stoeckli
3.6 Phenotype Depending on the target gene and its function, the methods of
Analysis phenotype analysis will vary. It is not within the scope of this
chapter to list methods for immunohistochemistry, axonal tracing
[1, 19], or analysis of cell migration. No matter what kind of
analysis you will use, keep in mind that phenotypes can only be
assessed by comparison of your experimental group with two
control groups (see Note 20).
4 Notes
Acknowledgments
We thank Evelyn Avilés and Nicole Wilson for their help in Figs. 2
and 4. Work in the laboratory of E.S. is supported by a grant from
the Swiss National Science Foundation.
References
1. Pekarik V, Bourikas D, Miglino N, Joset P, 5. Baeriswyl T, Mauti O, Stoeckli ET (2008)
Preiswerk S, Stoeckli ET (2003) Screening for Temporal control of gene silencing by in ovo
gene function in chicken embryo using RNAi electroporation. Methods Mol Biol
and electroporation. Nat Biotechnol 442:231–244
21:93–96 6. Mauti O, Baeriswyl T, Stoeckli ET (2008)
2. Chesnutt C, Niswander L (2004) Plasmid- Gene silencing by injection and electropora-
based short-hairpin RNA interference in the tion of dsRNA in Avian embryos. CSH Protoc
chicken embryo. Genesis 39:73–78 2008:pdb.prot5094
3. Baeriswyl T, Stoeckli ET (2008) Axonin-1/ 7. Luo J, Redies C (2005) Ex ovo electropora-
TAG-1 is required for pathfinding of granule tion for gene transfer into older chicken
cell axons in the developing cerebellum. embryos. Dev Dyn 233:1470–1477
Neural Dev 3:7 8. Mauti O, Sadhu R, Gemayel J, Gesemann M,
4. Wilson NH, Stoeckli ET (2011) Cell type spe- Stoeckli ET (2006) Expression patterns of
cific, traceable gene silencing for functional plexins and neuropilins are consistent with
gene analysis during vertebrate neural develop- cooperative and separate functions during neu-
ment. Nucleic Acids Res 39:e133 ral development. BMC Dev Biol 6:32
266 Irwin Andermatt and Esther T. Stoeckli
9. Das RM, van Hateren NJ, Howell GR, Farrell of the chick embryo. 1951. Dev Dyn
ER, Bangs FK, Porteous VC, Manning EM, 195:231–272
McGrew MJ, Ohyama K, Sacco MA et al 15. Rao M, Baraban JH, Rajaii F, Sockanathan
(2006) A robust system for RNA interference S (2004) In vivo comparative study of
in the chicken using a modified microRNA RNAi methodologies by in ovo electropor-
operon. Dev Biol 294:554–563 ation in the chick embryo. Dev Dyn
10. Bourikas D, Pekarik V, Baeriswyl T, Grunditz 231:592–600
A, Sadhu R, Nardó M, Stoeckli ET (2005) 15. Stepanek L, Stoker AW, Stoeckli E, Bixby JL
Sonic hedgehog guides commissural axons (2005) Receptor tyrosine phosphatases guide
along the longitudinal axis of the spinal cord. vertebrate motor axons during development.
Nat Neurosci 8:297–304 J Neurosci 25:3813–3823
11. Domanitskaya E, Wacker A, Mauti O, 16. Katahira T, Nakamura H (2003) Gene silenc-
Baeriswyl T, Esteve P, Bovolenta P, Stoeckli ing in chick embryos with a vector-based small
ET (2010) Sonic hedgehog guides post- interfering RNA system. Dev Growth Differ
crossing commissural axons both directly and 45:361–367
indirectly by regulating Wnt activity. J Neurosci 17. Dai F, Yusuf F, Farjah GH, Brand-Saberi B
30:11167–11176 (2005) RNAi-induced targeted silencing of
12. Wilson NH, Stoeckli ET (2013) Sonic hedge- developmental control genes during chicken
hog regulates its own receptor on post-crossing embryogenesis. Dev Biol 285:80–90
commissural axons in a Glypican-dependent 18. Sato F, Nakagawa T, Ito M, Kitagawa Y,
manner. Neuron in press Hattori M-A (2004) Application of RNA
13. Niederkofler V, Baeriswyl T, Ott R, Stoeckli interference to chicken embryos using small
ET (2010) Nectin-like molecules/SynCAMs interfering RNA. J Exp Zool A Comp Exp
are required for post-crossing commissural Biol 301:820–827
axon guidance. Development 137:427–435 19. Perrin FE, Stoeckli ET (2000) Use of lipo-
14. Hamburger V, Hamilton HL (1992) A philic dyes in studies of axonal pathfinding in
series of normal stages in the development vivo. Microsc Res Tech 48:25–31
Part VII
Mouse Protocols
Chapter 18
Abstract
During development, the mouse brain is progressively divided into functionally distinct compartments.
Numerous neuronal and glial cell types are subsequently generated in response to various inductive signals.
Each cell expresses a unique combination of genes encoding proteins from transcription factors to
neurotransmitters that define its role in brain function. To understand these important and highly sophis-
ticated processes, it is critical to accurately locate the various proteins and cells that produce them. In this
chapter, we introduce the techniques of immunohistochemistry, which detects the localization of specific
proteins, and RNA in situ hybridization, which enables the visualization of specific mRNAs.
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_18, © Springer Science+Business Media, LLC 2014
269
270 Jinling Liu and Aimin Liu
2 Materials
2.1 Cryosection 1. Dissection tools: student quality iris scissors (Fine Science
Preparation Tools), Dumont forceps (Fine Science Tools), and spoon (Fine
Science Tools).
2. 6 cm petri dishes.
3. 24-well tissue culture plates (see Note 1).
4. Stereomicroscope, such as Nikon SMZ645.
5. 4 % paraformaldehyde (PFA): Prepare 16 % stock by adding
32 g PFA powder and 100 μl 5 N NaOH in 150 ml pre-
warmed distilled, deionized water (ddH2O, at ~65 °C) (see
Note 2). Once PFA is dissolved, add 20 ml 10× PBS and adjust
the volume to 200 ml with ddH2O. Filter through Whatman
In Situ and Immunostaining in Mouse 271
2.3 Synthesis 1. A plasmid with the promoters for viral RNA polymerases (T3,
of RNA Probes T7, and Sp6) flanking the multiple cloning sites. Store at
−20 °C.
2. Restriction endonucleases: Store at −20 °C.
3. Horizontal DNA electrophoresis apparatus and power supply.
4. Agarose.
272 Jinling Liu and Aimin Liu
17. 10× DIG-labeling mix (Roche): Store at −20 °C (see Note 10).
18. RNase inhibitor (Promega, 40 U/μl): Store at −20 °C.
19. RNA polymerase (Roche T3, T7, or Sp6, 20 U/μl): Store at
−20 °C.
20. DNaseI (RNase-free): Store at −20 °C.
2.4 RNA In Situ The amount of reagents in this section is enough for processing
Hybridization five slides in a 50 ml Coplin jar and should be adjusted according
to the number of slides and size of the container used.
2.5 RNA In Situ All containers and reagents need to be RNase-free on day 1.
Hybridization Day 1:
1. Coplin jars: 50 ml.
Hybridization of
Cryosections on the 2. Diethylpyrocarbonate (DEPC): Store at 4 °C (see Note 8).
Slides 3. Magnetic stir plate.
4. DEPC-H2O (400 ml): Add 400 μl DEPC to 400 ml ddH2O.
Mix o/n on a magnetic stir plate at room temperature and
autoclave at 121 °C for 25 min (see Note 9).
5. DEPC-PBS (400 ml): Add 400 μl DEPC to 400 ml PBS. Mix
o/n on a magnetic stir plate at room temperature and auto-
clave at 121 °C for 25 min (see Note 9).
6. 4 % PFA in DEPC-PBS (80 ml): Thaw two 10 ml aliquots of
16 % PFA (see Subheading 2.1 for 16 % PFA preparation) and
dilute each with 30 ml DEPC-PBS. Store at 4 °C.
7. 0.25 % acetic anhydride in 0.1 M TEA-HCl (40 ml): Add
0.742 g triethanolamine-HCl and 360 μl 5 N NaOH to
39.8 ml DEPC-H2O to make TEA-HCl. Add 100 μl acetic
anhydride right before use (see Note 11).
8. 20 μg/ml proteinase K (proK; Roche #03115879001):
Dissolve one vial of proK (100 mg) in 5 ml DEPC-H2O to
make 20 mg/ml stock. Aliquot and store at −20 °C. Thaw an
aliquot before use and add 40 μl into 40 ml DEPC-PBS (see
Note 12).
9. 70 % ethanol in DEPC-H2O (40 ml): 28 ml 100 % ethanol,
12 ml DEPC-H2O.
10. 95 % ethanol in DEPC-H2O (40 ml): 38 ml 100 % ethanol,
2 ml DEPC-H2O.
11. Hybridization solution (40 ml): Mix the following in a clean
50 ml tube, aliquot, and store at −20 °C (see Note 13). 20 ml
formamide (deionized, aliquot in 50 ml tubes, and store at
−20 °C); 8 ml 50 % dextran sulfate; 400 μl 100× Denhardt’s
(VWR, aliquot in 1.5 ml tubes and store at −20 °C); 1 ml yeast
tRNA (10 mg/ml, aliquot in 1.5 ml tubes, store at −20 °C);
2.4 ml 5 M NaCl; 800 μl 1 M Tris–HCl (pH 8.0); 400 μl
0.5 M EDTA; 400 μl 1 M NaPO4; 4 ml 10 % sarkosyl; 2.6 ml
DEPC-H2O.
274 Jinling Liu and Aimin Liu
2.6 RNA In Situ Starting from day 2, reagents do not need to be RNase-free.
Hybridization Day 2:
1. 20× saline-sodium citrate (SSC) buffer: Mix the following in
Post-hybridization
an appropriate container: 800 ml ddH2O; 175.3 g NaCl;
Washes and Antibody 88.2 g sodium citrate. Adjust pH to 7.0 with a few drops of
Incubation 12 N HCl. Adjust the volume to 1 l with ddH2O. Aliquot and
autoclave at 121 °C for 25 min.
2. 5× SSC (40 ml): 10 ml 20× SSC, 30 ml ddH2O.
3. 2× SSC (40 ml): 4 ml 20× SSC, 36 ml ddH2O.
4. 0.1× SSC (40 ml): 0.2 ml 20× SSC, 39.8 ml ddH2O.
5. High-stringency wash buffer (120 ml) (make fresh): 60 ml for-
mamide, 12 ml 20× SSC, 48 ml ddH2O.
6. PBT (350 ml, enough for both days 2 and 3): Add 350 μl
Tween 20 to 350 ml PBS in a 500 ml bottle. Mix well by shak-
ing vigorously. Alternatively, 10 % Tween 20 stock can be made
in ddH2O in advance and stored at room temperature.
7. RNase buffer (400 ml) (see Note 14): 40 ml 5 M NaCl; 4 ml
1 M Tris–HCl, pH 7.5; 4 ml 0.5 M EDTA, pH 8.0; 352 ml
ddH2O.
8. 10 mg/ml RNase A stock (DNase-free): Add 10 ml 0.01 M
NaOAc (pH 5.2) to a vial of 100 mg RNase A. Heat at 100 °C
for 15 min. Cool to room temperature. Add 1 ml 1 M Tris–
HCl (pH 7.5) and aliquot. Store at −20 °C (see Note 15). On
the day of experiment, thaw one aliquot and make 20 μg/ml
RNase A in RNase buffer by adding 80 μl 10 mg/ml RNase A
stock to 40 ml RNase buffer.
9. Alkaline phosphatase-conjugated anti-Digoxigenin antibody
(Fab fragments from sheep; Roche). Store at 4 °C.
10. A humidified slide incubation chamber: See Subheading 2.2.
2.7 RNA In Situ 1. NTMT (80 ml) (make fresh; see Note 16): 8 ml 1 M Tris–HCl
Hybridization Day 3: 9.5; 4 ml 1 M MgCl2; 1.6 ml 5 M NaCl; 800 μl 10 % Tween
Color Reaction 20; 40 mg levamisole; adjust to 80 ml with ddH2O.
2. Levamisole: make 50 mg/ml stock in ddH2O, aliquot, and
store in 1.5 ml tubes at −20 °C.
3. BM purple (Roche). Store at 4 °C.
4. Water-based mounting medium, such as Mount Quick aq.
(SPI). Store at 4 °C.
5. Coverslips: VWR micro cover glass, 25 × 50 mm.
In Situ and Immunostaining in Mouse 275
3 Methods
3.1 Mouse Brain 1. Dissect embryos in ice-cold PBS in a 6 cm dish (see Note 17).
Cryosection 2. Cut the embryos at the shoulder level and transfer the heads
Preparation into a 24-well plate with a spoon.
3. Fix the embryos in 4 % PFA on a nutator at 4 °C for 1 h for
IHC or o/n for RISH.
4. Rinse the embryos with PBS and then wash the embryos in
PBS on a nutator o/n at 4 °C.
5. Immerse the embryos in 30 % sucrose o/n at 4 °C, on a nuta-
tor (see Note 18).
6. Change the 30 % sucrose and incubate for another 2–3 h for
further infiltration at 4 °C, on a nutator.
7. Transfer the embryos into a disposable embedding mold and
immerse the embryos in O.C.T. compound for 1 h at 4 °C
(see Note 19).
8. Position the samples at desired orientation and freeze them on
dry ice or, alternatively, in a paperboard box floating on the
surface of liquid nitrogen. Wait for 5 min (see Note 20).
9. Transfer the frozen O.C.T. block containing the brain samples to
the cryostat and wait at least 1 h so that the temperature of the
block can reach the optimal cutting temperature (see Note 21).
10. Cut 10 μm sections using a cryostat (see Note 22). Collect sec-
tions on Superfrost Plus slides.
11. Dry the slides in a micro slide holder for 1 h at room tempera-
ture. Store them in a slide box at −80 °C (see Note 23).
3.2 Immunostaining 1. Remove sections from −80 °C freezer and dry slides in a micro
on Mouse Brain slide holder at room temperature for about 45 min (see Note 24).
Sections 2. Place the slides in a Coplin jar and incubate the sections with
blocking buffer for 1 h.
3. Take slides out of blocking buffer; wipe the backside (the one
without sections) and edges with paper towel. Place them with
the front side (the one with sections) up on the pipettes of the
humidified slide incubation chamber (Fig. 1; see Note 25).
276 Jinling Liu and Aimin Liu
Fig. 2 Immunostaining of brain sections. Shown are two coronal sections of an E12.5 brain incubated with a
monoclonal antibody against Pax6 and a Cy3-conjugated secondary antibody. (a) Pax6 is weakly expressed in
the dorsal region and strongly expressed in the intermediate region in the hindbrain. (b) Pax6 is expressed in
the diencephalon (Di), but not the mesencephalon (mes). Dashed line demarcates the boundary between the
diencephalon and mesencephalon
3.3 Synthesis of RNA 1. Clone the cDNA (or part of the cDNA if the full-length cDNA
Probes for RISH is longer than 1,500 bps; see Note 31) of the gene of interest
into a plasmid containing the promoters for the viral RNA
polymerases (T3, T7, and Sp6; such as pBluescript).
In Situ and Immunostaining in Mouse 277
Incubate at 37 °C for 2 h.
5. Run 1 μl in 1 % agarose gel for 20 min to 1 h to check the yield
of the probe.
6. DNase I treatment (optional):
Add 1 μl DNaseI and 1 μl RNase inhibitor to the RNA probe
and incubate at 37 °C for 15 min to remove the template.
7. Add 180 μl DEPC-H2O, 20 μl 3 M NaOAc, and mix well.
Then add 600 μl 100 % ethanol, mix well, and leave at −80 °C
for 30 min.
8. Centrifuge at top speed (>14,000 × g) in a tabletop centrifuge
for 15 min at 4 °C (see Note 32).
9. Discard the supernatant and rinse the pellet with 70 % ethanol
once without disturbing the pellet.
10. Discard 70 % ethanol and dry the pellet for 5 min. Dissolve the
RNA probe in 40–50 μl DEPC-H2O and store at −80°C.
278 Jinling Liu and Aimin Liu
3.4 RNA In Situ Day 1: hybridization of cryosections (all steps are carried out in Coplin
Hybridization of jars unless otherwise specified; avoid RNase contamination)
Mouse Brain Sections
1. Dry slides at room temperature for about 45 min
(see Note 24).
2. Postfix slides in 4 % PFA in DEPC-PBS for 10 min
(see Note 33).
3. Wash with DEPC-PBS, 2 × 5 min.
4. Drain excess DEPC-PBS and incubate for 6 min in 20 μg/ml
proteinase K in DEPC-PBS (see Note 34).
5. Drain and wash with DEPC-PBS for 5 min.
6. Refix in 4 % PFA for 5 min, then wash 5 min in DEPC-PBS
(see Note 35).
7. Acetylate sections with acetic anhydride in 0.1 M TEA-HCl for
10 min (see Note 36).
8. Wash in DEPC-PBS for 5 min; dehydrate in 70 % ethanol for
5 min and 95 % ethanol for 2 min. Air dry for 30 min to 2 h.
9. Add 2 μl appropriate probe (approx. 1 μg) to 1 ml hybridiza-
tion solution and heat at 80 °C for 2 min (see Note 37).
10. Place slides horizontally in a humidified slide incubation cham-
ber. Cover sections with 200 μl of hybridization solution with
the probe and lower parafilm coverslips over sections avoiding
bubbles (see Note 38).
11. Seal the slide incubation chamber carefully and hybridize at
55 °C in a hybridization oven overnight (16–18 h).
Day 2: post-hybridization washes and antibody incubation (all
steps are carried out in Coplin jars; RNase-free environment is not
required)
1. Dip slides gently in a Coplin jar filled with 5× SSC to let the
coverslips float off the slides (see Note 39).
2. Incubate the sections in high-stringency wash at 65 °C in a
Coplin jar for 30 min (see Note 40).
3. Wash in RNase buffer at 37 °C, 3 × 10 min.
4. Wash in RNase buffer with 20 μg/ml RNase A at 37 °C for
30 min (see Note 41).
5. Wash in RNase buffer at 37 °C for 15 min.
6. Repeat high-stringency wash (as in step 2) at 65 °C, 2 × 20 min.
7. Wash in 2× SSC, then 0.1× SSC for 15 min each at 37 °C.
8. Wash with PBT for 15 min.
9. Take the slide out of PBT; wipe the backside and edges of the
slide with paper towel. Place slides horizontally in a humidified
slide incubation chamber and block for 1 h with 10 % goat
serum in PBT (300 μl per slide).
In Situ and Immunostaining in Mouse 279
10. Pour the blocking buffer onto paper towels. Wipe the backside
and edges of the slide with paper towel. Incubate with
AP-conjugated anti-Digoxigenin antibody (diluted 1/5,000
in PBT with 1 % goat serum, 320 μl per slide) at 4 °C over-
night in the same humidified chamber.
Day 3: color reaction
1. Pour the antibody onto paper towels. Place slides in a Coplin
jar and wash in PBT for five times, 1 h each.
2. Wash 2 × 10 min in freshly prepared NTMT buffer.
3. Wipe the backside and edges of the slide with paper towel.
Place the slides horizontally in a humidified slide incubation
chamber and incubate o/n to several days in BM purple solu-
tion (300 μl per slide) supplemented with 0.5 mg/ml levami-
sole in the dark (see Note 42).
4. Observe periodically the progress of the color reaction under a
microscope. If the staining is not ready, reapply BM purple
solution and incubate for longer time (see Note 43).
5. When the staining is ready, i.e., when the signal is strong and
the background staining just begins to show, place slides back
into a Coplin jar. Wash slides in PBS for 2–5 min and dip briefly
in ddH2O (see Note 18).
6. (Optional) Counterstain the sections with nuclear fast red until
the sections turn slightly pink. Usually it takes 2–3 min.
7. Wash the excess nuclear fast red in slow-running tap water.
8. Wipe the backside and edges of the slide with paper towel.
Apply mounting medium to the slides and put coverslips on
(see Note 44).
9. Observe the staining under a microscope and take photos with
a color camera. Some examples are shown in Fig. 3.
10. The mounted sections can be stored at 4 °C.
4 Notes
Fig. 3 RNA in situ hybridization of brain sections. Shown are two sections of E12.5 brains hybridized with a
Digoxigenin-labeled RNA probe against Otx2. (a) A coronal section through the forebrain region shows Otx2
expression in ventral telencephalon (tel) and the retinal pigment epithelium of the eye, but not in ventral dien-
cephalon (di). (b) A sagittal section shows that Otx2 is expressed in the mesencephalon (mes) and diencephalon
(di), but not in the metencephalon (met) and pons. The arrowhead points to the boundary between diencephalon
and mesencephalon. Dashed lines demarcate the boundary between the midbrain and hindbrain, known as the
isthmus (is)
Acknowledgments
We would like to thank Dr. Simeone for providing the RNA in situ
probe for Otx2. The monoclonal antibody against Pax6 developed
by Dr. Jessell was obtained from the Developmental Studies
In Situ and Immunostaining in Mouse 283
References
1. Kandel E, Schwartz J, Jessell T (2000) Principles 3. Komminoth P, Merk FB, Wolfe HJ, Roth J
of neural science, 4th edn. McGraw-Hill, Health (1992) Comparison of 35S-and digoxigenin-
Professions Division, New York, NY. ISBN labeled RNA and oligonucleotide probes for
0-8385-7701-6 in situ hybridization. Histochemistry 98:
2. Joseph G. Gall, Mary Lou Pardue (1969) 217–228
Formation and detection of RNA-DNA hybrid 4. Wilcox JN (1993) Fundamental principles of in
molecules in cytological preparations*. Proc situ hybridization. J Histochem Cytochem
Natl Acad Sci USA 63:378–383 41:1725–1733. doi:10.1177/41.12.8245419
Chapter 19
Abstract
In utero electroporation is a rapid and powerful technique to study the development of many brain regions.
This approach presents several advantages over other methods to study specific steps of brain development
in vivo, from proliferation to synaptic integration. Here, we describe in detail the individual steps necessary
to carry out the technique. We also highlight the variations that can be implemented to target different
cerebral structures and to study specific steps of development.
Key words Electroporation, In utero, Brain, Embryo, Neuronal development, Gain and loss of func-
tion, Transfection, Mouse
1 Introduction
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_19, © Springer Science+Business Media, LLC 2014
285
286 Emilie Pacary and François Guillemot
Table 1
The different brain structures that can be
targeted by in utero electroporation
Table 2
Developmental events that can be studied using in utero
electroporation and corresponding references
2 Materials
2.1 DNA Injection 1. DNA solution: Purify plasmid DNA with an endotoxin-free
Maxi-prep kit. Prepare plasmid DNA solution to desired con-
centrations in water and add Fast Green (final concentration
of 0.05 %; Sigma) (see Note 1).
2. Needles: Pull borosilicate glass capillaries (1.0 mm O.D. × 0.58 mm
I.D.) (Harvard Apparatus) using a micropipette puller.
3. Microloader tips (Eppendorf).
3 Methods
3.2 Surgery Before 1. Anesthetize the pregnant mouse with isoflurane in oxygen
DNA Injection and carrier (oxygen 2 l/min) using an anesthetic induction cham-
Electroporation ber. Wait until the animal loses righting reflex.
2. Transfer the animal to a “presurgery” mask. Place a drop of
eye gel on each eye to prevent corneal ulceration of the eyes
while it is under general anesthesia. Use an electric razor to
shave the hair of the abdomen. Clean the shaved area once
with chlorhexidine to collect flying hair.
3. Transfer the animal to a second mask in the surgical area. Place
the mouse with its back on the heating pad. Start the surgery
when the pedal reflex has been lost.
4. Put on mask and sterile gloves. Cover the animal with a sterile
drape (with a small hole over the abdomen) to prevent the tis-
sues and instruments from being contaminated by the areas of
skin that have not been shaved and disinfected. Clean the
shaved area at least three times with chlorhexidine. Use a dif-
ferent sterile cotton bud each time. Use a scalpel to make a
vertical incision along the midline (~1 in. long) through the
skin. Using scissors, make a similar incision of the muscle of
the abdomen along the linea alba (white line composed mostly
of collagen connective tissue).
5. Choose the most accessible embryos, place the ring forceps
between two embryos, and carefully pull the embryonic chain
out of the abdominal cavity (see Note 2). From this point on,
keep the embryos hydrated with sterile pre-warmed PBS.
3.3 Injection of DNA 1. Pinch off the tip of the needle with a forceps.
and Electroporation 2. Start with one of the most lateral embryos, making it easier to
keep track of which embryos were electroporated. Manipulate
the position of the embryo inside the amniotic sac using the ring
forceps and stabilize the head of the embryos between the rings.
Squeeze gently to push up the embryo closer to the uterine wall.
3. With the other hand, take the capillary holder and insert the
needle carefully into the middle of the hemisphere to target
the lateral ventricle or into the desired structure (see Note 3).
Press the pedal to inject the DNA solution (see Note 4). The
location of the DNA solution is monitored with Fast Green
(see Note 5).
4. Place the electrodes on the sides of the embryo head to target
the desired area (see Note 6).
5. Apply electrical pulses (see Notes 7 and 8).
All the embryos in a pregnant mouse can be electroporated,
usually with the same DNA construct to avoid confusion when
harvesting electroporated embryos or pups (see Note 9). However,
290 Emilie Pacary and François Guillemot
3.4 Surgery Post 1. After electroporating the embryos, add PBS into the abdomi-
Electroporation nal cavity and use the ring forceps to replace the uterine horn
in its original location.
2. Suture the abdomen wall and skin with Vicryl absorbable sutures.
3. Place the animal in a recovery chamber until it wakes up (usually
5–10 min) and then transfer in a cage placed on a heating pad.
3.5 Post-surgery Check the behavior of the mice to assess pain, suffering, or distress
and weigh the animals 24 and 48 h after the surgery. If needed,
analgesics can be administered to minimize pain and discomfort.
4 Notes
References
1. Chen F, LoTurco J (2012) A method for S (2009) Development of the mouse amygdala
stable transgenesis of radial glia lineage in rat as revealed by enhanced green fluorescent pro-
neocortex by piggyBac mediated transposi- tein gene transfer by means of in utero electro-
tion. J Neurosci Methods 207(2):172–180 poration. J Comp Neurol 513(1):113–128
2. Iguchi T, Yagi H, Wang CC, Sato M (2012) A 12. Nakahira E, Yuasa S (2005) Neuronal genera-
tightly controlled conditional knockdown sys- tion, migration, and differentiation in the
tem using the Tol2 transposon-mediated tech- mouse hippocampal primoridium as revealed
nique. PLoS One 7(3):e33380 by enhanced green fluorescent protein gene
3. Pacary E, Haas MA, Wildner H, Azzarelli R, transfer by means of in utero electroporation.
Bell DM, Abrous DN, Guillemot F (2012) J Comp Neurol 483(3):329–340
Visualization and genetic manipulation of 13. Navarro-Quiroga I, Chittajallu R, Gallo V,
dendrites and spines in the mouse cerebral Haydar TF (2007) Long-term, selective gene
cortex and hippocampus using in utero expression in developing and adult hippo-
electroporation. J Vis Exp (65):e4163. doi: campal pyramidal neurons using focal in
10.3791/4163 utero electroporation. J Neurosci 27(19):
4. Saito T, Nakatsuji N (2001) Efficient gene trans- 5007–5011
fer into the embryonic mouse brain using in vivo 14. Garcia-Frigola C, Carreres MI, Vegar C,
electroporation. Dev Biol 240(1):237–246 Herrera E (2007) Gene delivery into mouse
5. Pacary E, Heng J, Azzarelli R, Riou P, Castro retinal ganglion cells by in utero electropora-
D, Lebel-Potter M, Parras C, Bell DM, Ridley tion. BMC Dev Biol 7:103
AJ, Parsons M, Guillemot F (2011) Proneural 15. Petros TJ, Rebsam A, Mason CA (2009) In
transcription factors regulate different steps of utero and ex vivo electroporation for gene
cortical neuron migration through Rnd- expression in mouse retinal ganglion cells. J
mediated inhibition of RhoA signaling. Vis Exp (31):e1333. doi:10.3791/1333
Neuron 69(6):1069–1084 16. Punzo C, Cepko CL (2008) Ultrasound-
6. Shimogori T, Ogawa M (2008) Gene applica- guided in utero injections allow studies of the
tion with in utero electroporation in mouse development and function of the eye. Dev Dyn
embryonic brain. Dev Growth Differ 237(4):1034–1042
50(6):499–506 17. Borrell V, Yoshimura Y, Callaway EM (2005)
7. Tabata H, Nakajima K (2001) Efficient in Targeted gene delivery to telencephalic inhibi-
utero gene transfer system to the developing tory neurons by directional in utero electro-
mouse brain using electroporation: visualiza- poration. J Neurosci Methods 143(2):
tion of neuronal migration in the developing 151–158
cortex. Neuroscience 103(4):865–872 18. Castro DS, Martynoga B, Parras C, Ramesh V,
8. Niwa M, Kamiya A, Murai R, Kubo K, Gruber Pacary E, Johnston C, Drechsel D, Lebel-
AJ, Tomita K, Lu L, Tomisato S, Jaaro-Peled Potter M, Garcia LG, Hunt C, Dolle D, Bithell
H, Seshadri S, Hiyama H, Huang B, Kohda K, A, Ettwiller L, Buckley N, Guillemot F (2011)
Noda Y, O’Donnell P, Nakajima K, Sawa A, A novel function of the proneural factor Ascl1
Nabeshima T (2010) Knockdown of DISC1 in progenitor proliferation identified by
by in utero gene transfer disturbs postnatal genome-wide characterization of its targets.
dopaminergic maturation in the frontal cortex Genes Dev 25(9):930–945
and leads to adult behavioral deficits. Neuron 19. Rouaux C, Arlotta P (2010) Fezf2 directs the
65(4):480–489 differentiation of corticofugal neurons from
9. Bai J, Ramos RL, Paramasivam M, Siddiqi F, striatal progenitors in vivo. Nat Neurosci
Ackman JB, LoTurco JJ (2008) The role of 13(11):1345–1347
DCX and LIS1 in migration through the lat- 20. Gelman DM, Martini FJ, Nobrega-Pereira S,
eral cortical stream of developing forebrain. Pierani A, Kessaris N, Marin O (2009) The
Dev Neurosci 30(1–3):144–156 embryonic preoptic area is a novel source of
10. Remedios R, Huilgol D, Saha B, Hari P, cortical GABAergic interneurons. J Neurosci
Bhatnagar L, Kowalczyk T, Hevner RF, Suda 29(29):9380–9389
Y, Aizawa S, Ohshima T, Stoykova A, Tole S 21. Bonnin A, Torii M, Wang L, Rakic P, Levitt P
(2007) A stream of cells migrating from the (2007) Serotonin modulates the response of
caudal telencephalon reveals a link between the embryonic thalamocortical axons to netrin-1.
amygdala and neocortex. Nat Neurosci Nat Neurosci 10(5):588–597
10(9):1141–1150 22. Kataoka A, Shimogori T (2008) Fgf8 controls
11. Soma M, Aizawa H, Ito Y, Maekawa M, Osumi regional identity in the developing thalamus.
N, Nakahira E, Okamoto H, Tanaka K, Yuasa Development 135(17):2873–2881
In Utero Electroporation in Mouse 293
23. Vue TY, Bluske K, Alishahi A, Yang LL, King C, Sklar P, Sive H, Tsai LH (2011)
Koyano-Nakagawa N, Novitch B, Nakagawa Y Common DISC1 polymorphisms disrupt
(2009) Sonic hedgehog signaling controls tha- Wnt/GSK3beta signaling and brain develop-
lamic progenitor identity and nuclei specifica- ment. Neuron 72(4):545–558
tion in mice. J Neurosci 29(14):4484–4497 34. Elias LA, Wang DD, Kriegstein AR (2007)
24. Kawauchi D, Taniguchi H, Watanabe H, Saito Gap junction adhesion is necessary for radial
T, Murakami F (2006) Direct visualization of migration in the neocortex. Nature
nucleogenesis by precerebellar neurons: 448(7156):901–907
involvement of ventricle-directed, radial fibre- 35. Cubelos B, Sebastian-Serrano A, Beccari L,
associated migration. Development 133(6): Calcagnotto ME, Cisneros E, Kim S, Dopazo
1113–1123 A, Alvarez-Dolado M, Redondo JM, Bovolenta
25. Nishiyama J, Hayashi Y, Nomura T, Miura E, P, Walsh CA, Nieto M (2010) Cux1 and Cux2
Kakegawa W, Yuzaki M (2012) Selective and regulate dendritic branching, spine morphol-
regulated gene expression in murine Purkinje ogy, and synapses of the upper layer neurons of
cells by in utero electroporation. Eur J the cortex. Neuron 66(4):523–535
Neurosci 36(7):2867–2876 36. Fang WQ, Ip JP, Li R, Ng YP, Lin SC, Chen Y,
26. Okada T, Keino-Masu K, Masu M (2007) Fu AK, Ip NY (2011) Cdk5-mediated phos-
Migration and nucleogenesis of mouse precer- phorylation of Axin directs axon formation
ebellar neurons visualized by in utero electro- during cerebral cortex development.
poration of a green fluorescent protein gene. J Neurosci 31(38):13613–13624
Neurosci Res 57(1):40–49 37. Yi JJ, Barnes AP, Hand R, Polleux F, Ehlers
27. Alvarez-Maya I, Navarro-Quiroga I, Meraz- MD (2010) TGF-beta signaling specifies
Rios MA, Aceves J, Martinez-Fong D (2001) axons during brain development. Cell
In vivo gene transfer to dopamine neurons of 142(1):144–157
rat substantia nigra via the high-affinity neuro- 38. Elias GM, Elias LA, Apostolides PF, Kriegstein
tensin receptor. Mol Med 7(3):186–192 AR, Nicoll RA (2008) Differential traffick-
28. Willett RT, Greene LA (2011) Gata2 is ing of AMPA and NMDA receptors by
required for migration and differentiation of SAP102 and PSD-95 underlies synapse
retinorecipient neurons in the superior collicu- development. Proc Natl Acad Sci USA
lus. J Neurosci 31(12):4444–4455 105(52):20953–20958
29. Fukuchi-Shimogori T, Grove EA (2001) 39. Trimbuch T, Beed P, Vogt J, Schuchmann S,
Neocortex patterning by the secreted signaling Maier N, Kintscher M, Breustedt J, Schuelke
molecule FGF8. Science 294(5544): M, Streu N, Kieselmann O, Brunk I, Laube G,
1071–1074 Strauss U, Battefeld A, Wende H, Birchmeier
30. Lange C, Huttner WB, Calegari F (2009) C, Wiese S, Sendtner M, Kawabe H,
Cdk4/cyclinD1 overexpression in neural stem Kishimoto-Suga M, Brose N, Baumgart J,
cells shortens G1, delays neurogenesis, and Geist B, Aoki J, Savaskan NE, Brauer AU,
promotes the generation and expansion Chun J, Ninnemann O, Schmitz D, Nitsch R
of basal progenitors. Cell Stem Cell 5(3): (2009) Synaptic PRG-1 modulates excitatory
320–331 transmission via lipid phosphate-mediated
31. Weimer JM, Yokota Y, Stanco A, Stumpo DJ, signaling. Cell 138(6):1222–1235
Blackshear PJ, Anton ES (2009) MARCKS 40. Garcia-Marques J, Lopez-Mascaraque L
modulates radial progenitor placement, prolif- (2013) Clonal Identity Determines Astrocyte
eration and organization in the developing cere- Cortical Heterogeneity. Cereb Cortex 23(6):
bral cortex. Development 136(17):2965–2975 1463–1472
32. Nguyen L, Besson A, Heng JI, Schuurmans C, 41. Subramanian L, Sarkar A, Shetty AS,
Teboul L, Parras C, Philpott A, Roberts JM, Muralidharan B, Padmanabhan H, Piper M,
Guillemot F (2006) p27kip1 independently Monuki ES, Bach I, Gronostajski RM,
promotes neuronal differentiation and migra- Richards LJ, Tole S (2011) Transcription fac-
tion in the cerebral cortex. Genes Dev tor Lhx2 is necessary and sufficient to suppress
20(11):1511–1524 astrogliogenesis and promote neurogenesis in
33. Singh KK, De Rienzo G, Drane L, Mao Y, the developing hippocampus. Proc Natl Acad
Flood Z, Madison J, Ferreira M, Bergen S, Sci USA 108(27):E265–E274
Chapter 20
Abstract
Cre-mediated recombination has become a powerful tool to confine gene deletions (conditional knock-
outs) or overexpression of genes (conditional knockin/overexpression). By spatiotemporal restriction of
genetic manipulations, major problems of classical knockouts such as embryonic lethality can be circum-
vented. Furthermore Cre-mediated recombination has broad applicability in the analysis of the cellular
behavior of subpopulations and cell types as well as for genetic fate mapping. This chapter will give an
overview about applications for the Cre/LoxP system and their execution.
Key words Cre recombinase, Transgenesis, Conditional knockout, Conditional knockin, CreERT2,
Flpe recombinase, MADM, Split-Cre, Brainbow
1 Introduction
After the first gene knockout (KO) in the mouse was obtained by
Thomas and Capecchi using site-directed mutagenesis of the
HPRT gene in 1987 [1], the in vivo function of many genes has
been analyzed using this technique. Nevertheless the KO approach
has two main restrictions. First of all, genes whose inactivation is
embryonically lethal cannot be analyzed for their function in late
development and adulthood. Secondly, KOs of genes which have a
function in multiple tissues and/or cell types are difficult to ana-
lyze, since phenotypes might be combinations of multiple distinct
defects and therefore quite complex to dissect.
In 1994 Gu et al. described for the first time the use of the
Cre/Lox recombination system to induce a gene knockout in mice
[2]. This technology made it possible to conditionally knockout
genes solely in subsets of cells (i.e., in a cell-type- or tissue-specific
manner), where Cre recombinase is expressed. Two years before,
the technique had been already used to conditionally overexpress
the SV40 large tumor antigen in mice [3]. The key principle of the
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9_20, © Springer Science+Business Media, LLC 2014
295
296 Claudius F. Kratochwil and Filippo M. Rijli
2 Materials
2.1 Reporter Mice 1. ROSA26 LacZ reporter line [30]; available at Jackson
Laboratories:
# 003504 B6.129S4-Gt(ROSA)26Sor tm1Sor/J.
2. ROSA26 tdTomato reporter line [31]; available at Jackson
Laboratories:
# 007914 B6.Cg-Gt(ROSA)26Sortm14(CAG-tdTomato)Hze/J.
3. ROSA26 ZsGreen reporter line [31]; available at Jackson
Laboratories:
# 007906 B6.Cg-Gt(ROSA)26Sor tm6(CAG-ZsGreen1)Hze/J.
4. Brainbow reporter lines [29]; six transgenic lines available at
Jackson Laboratories:
# 007901 B6.Cg-Tg(Thy1-Brainbow1.0)HLich/J.
# 007910 B6;CBA-Tg(Thy1-Brainbow1.0)LLich/J.
# 007911 B6.Cg-Tg(Thy1-Brainbow1.1)MLich/J.
# 007921 B6.Cg-Tg(Thy1-Brainbow2.1)RLich/J.
# 013731 STOCK Gt(ROSA)26Sor tm1(CAG-Brainbow2.1)Cle/J.
# 017492 B6.129P2-Gt(ROSA)26Sortm1(CAG-Brainbow2.1) Cle/J.
5. MADM mice [25, 26, 32, 33]; several transgenic lines are
available at Jackson Laboratories, the following are the most
widely used:
# 013749 STOCK Tg(ACTB-EGFP,-tdTomato)11Luo/J.
# 013751 STOCK Tg(ACTB-tdTomato,-EGFP)11Luo/J.
# 017932 STOCK Tg(ACTB-EGFP*)10Luo/J.
# 017923 STOCK Tg(ACTB-EGFP*,-tdTomato)10Luo/J.
# 017912 STOCK Gt(ROSA)26Sor tm6(ACTB-EGFP*,-tdTomato)Luo/J.
# 017921 STOCK Gt(ROSA)26Sortm7(ACTB-EGFP*)Luo/J.
298 Claudius F. Kratochwil and Filippo M. Rijli
3 Methods
3.1 Applications of To create a conditional knockout, the gene of interest is flanked with
the Cre/LoxP System recognition sites (LoxP sites; Locus of crossing [x-ing]-over of
bacteriophage P1) for the bacteriophage P1 Cre recombinase [35]
(Fig. 1). If Cre recombinase is present, the sequence flanked by two
LoxP sites (the “floxed” sequence) is excised (if Lox sites have the
same orientation) or inverted (if Lox sites have opposing orientation)
(Fig. 1). It is also possible to recombine between two different plas-
mids or chromosomes (interchromosomal recombination), which is,
for example, used in the MADM system (see Subheading 3.7). The
excision of genomic fragments gives the possibility to perturb gene
function either (a) completely, by removing the whole gene or the
start codon; (b) partly, by removing certain parts/exons of the gene
or by truncating it; or by (c) changing its expression pattern and
levels by excising/replacing/modifying promoter or enhancer ele-
ments. In a similar way, Cre-mediated recombination can be used to
overexpress genes, by excision of an intervening transcriptional ter-
mination sequence, flanked by LoxP sites, that prevents the tran-
scription of the target gene [3] (Fig. 2b). Since these constructs
might result in low levels of leaking readthrough transcription, espe-
cially if many copies are present (e.g., when using gene transfer by
Cre/Lox in Mouse Development 299
Fig. 1 Distinct genomic outcomes of Cre/LoxP-mediated recombination. (a) If two Lox sites have opposing direction-
ality, the sequence in between is inversed upon Cre-mediated recombination. The recombination is reversible. (b) If
Lox sites have the same orientation, the sequence in between is recombined out, resulting in a circular fragment and
the sequence lacking this fragment. (c) If LoxP sites are present on two chromosomes, recombination can also occur
interchromosomally, albeit at a lower frequency than if the LoxP sites are on the same chromosome. (d) The LoxP
site is 34 base pair (bp) long. Two palindromic 13 bp sequences, containing the Cre binding site (boxed), flank an
asymmetric 8 bp sequence. Arrows indicate the sites of cleavage during recombination. The underlined base pairs
are mutated in the most commonly used variants LoxN and Lox2722 (modified from [36])
3.2 Designing Cre/LoxP is a binary system. On the one hand, the gene of interest
Constructs has to be flanked by LoxP sites (“floxed”) (Fig. 1). On the other hand
recombinase expression in the cell or tissue of interest is provided
300 Claudius F. Kratochwil and Filippo M. Rijli
Fig. 2 Three examples for applications using the Cre/LoxP system. (a) The classic approach to generate a
conditional knockout is to place LoxP sites in introns either flanking the exon, which contains the ATG, or exons
that contain functionally important domains. Using this strategy, the probability to interfere with gene expres-
sion before Cre recombination is reasonably smaller than if LoxP sites are placed in promoter regions. (b) To
overexpress genes usually an intervening sequence is placed between promoter and target gene, which blocks
transcription. Upon Cre-mediated recombination this sequence is removed, and the gene starts to be expressed
under the control of the upstream promoter. (c) To reduce the risk of leaking, recently the use of double-
inverted Lox sites became an alternative approach (flip-excision switch, or FLEX). Here, the gene of interest is
placed in inverted or antisense orientation. Cre mediates the inversion of the sequence in the presence of a
pair of Lox sites in inverted orientation. The use of a second pair of Lox sites also in reverse orientation, though
incompatible with the first pair, eventually results in an irreversible sense configuration, since both sites lack
at the end a partner site, allowing stable transcription
Fig. 3 Two approaches to obtain tissue-/cell-type-specific Cre expression. (a) A tissue-specific enhancer is
combined with a minimal promoter to drive tissue-specific Cre expression, where the enhancer is active. (b)
Another approach is site-directed recombination into a bacterial artificial chromosome (BAC) or into the
genome of embryonic stem cells
3.3 Mouse Mating Once Cre KI (i.e., inserted at specific loci) or Cre-expressing trans-
Schemes genic mice have been created, they need to be crossed to mice
carrying LoxP site-bearing conditional alleles, in order to generate
double heterozygotes for Cre and the floxed locus. Since most
laboratories dispose of multiple Cre drivers, it is more space effi-
cient to generate a few double heterozygous males for the Cre-
expressing line and the conditional allele and mate them to
homozygous conditional mutant females, which can be readily
maintained as a pool. However, it should be noted that in this type
of crossings, double heterozygous Cre/conditional allele and/or
homozygous conditional mutant specimen in the absence of Cre-
mediated excision usually displays a wild-type-like phenotype, thus
serving as controls. It may be useful to additionally cross a floxed
conditional reporter line into the background of Cre/conditional
allele double heterozygotes. In many projects this will ease the
analysis, as it will allow to (1) have a direct readout of the cells in
which the gene has been knocked out, (2) directly analyze cellular
behavior between heterozygotes and homozygotes, and (3) select
tissue or cells using methods such as fluorescent-activated cell sort-
ing (FACS). Conditional Cre-inducible reporter mice in which the
reporter genes are inserted in housekeeping gene loci are available
from the Jackson Laboratories. These lines provide stable and con-
stitutive reporter expression once activated by Cre-mediated exci-
sion. In particular, three KIs in the ROSA26 locus are the most
frequently used reporter lines since they are highly sensitive to Cre-
mediated activation, which express either ß-galactosidase (LacZ)
[30], ZsGreen or tdTomato [31], but additional reporter lines
expressing other fluorescent proteins are also available (e.g., [43]).
The choice depends on the application. ß-Galactosidase catalyzes
the transformation of X-gal into an insoluble blue enzymatic product
302 Claudius F. Kratochwil and Filippo M. Rijli
Fig. 4 Mating scheme to analyze gene function by conditional knockout. The use of pools for the conditional
knockout (with or without reporter) eases the analysis if multiple Cre drivers are used to test gene function in
different tissues. The offspring contains animals displaying mutant (Cre tg/+, Gene fl/fl ) and control (Cre tg/+,
Gene fl/+, and Gene fl/fl ) phenotypes
Fig. 5 Genotyping by PCR to detect gene deletion. (a) The targeted allele can be detected by primer pair 1 (F1
and R1). (b) The floxed allele can be detected by the same primers, yielding a longer amplicon. (c) A distinct
primer pair (F2 and R1) detects the Cre-mediated recombination. The non-recombined locus usually cannot be
detected, as the amplicon will be too long. (d) Theoretical result of a PCR with different genotypes in Cre-
positive (Cre+) and Cre-negative (Cre−) tissues
1 reaction 20 reactions
®
GoTaq Green Master Mix, 2× 4 μl 80 μl
Primer Mix, 10 μM for each primer 0.4 μl 8 μl
Reporter expression
Activity promoter 1 (coexpressing area) Activity promoter 2
b
Promoter 1 Cre Promoter 2 Flpe
Fig. 6 Tools to generate intersectional recombination. (a) In the Split-Cre system, Cre is divided into two inac-
tive fragments expressed under the control of two distinct promoters. In the tissue, or cell type, where both
promoters are active, the two fragments are co-expressed resulting in a functional Cre. This triggers a recom-
bination event, e.g., the Cre-mediated activation of a reporter gene. (b) The Cre/Flpe system uses two different
recombinases recognizing distinct target sites. When both recombinases are active, a specific reporter gene is
expressed (e.g., GFP) which is not activated by either one of the recombinases alone
Cre/Lox in Mouse Development 307
3.7 MADM System The MADM (mosaic analysis with double markers) is an adapta-
tion in mouse of the conceptually similar MARCM (mosaic analy-
sis with a repressible cell marker) used in Drosophila [27]. Hereby,
rare interchromosomal recombination (Fig. 1c) during mitosis
results in sparse labeling of cells with one of two fluorescent
reporter genes (e.g., GFP or RFP). In differentiated cells inter-
chromosomal recombination can occur as well but can only result
in the expression of both (GFP and RFP) or no reporter (Fig. 7).
Because of the low frequency of recombinations during mitosis,
this allows (1) to follow the behavior of clonally related (i.e., gen-
erated from the same progenitor) cells [45] as well as (2) to sparsely
generate mutant cells in the background of a heterozygous mouse
labeled by a fluorescent marker (e.g., RFP).
Before recombination no fluorescent marker is expressed in
MADM mice, because the N-terminus sequence of either one of
the two fluorescent markers (separated by a LoxP site) is followed
by the C-terminus of the other marker (e.g., N-RFP-LoxP-C-GFP
and N-GFP-LoxP-C-RFP, respectively), resulting in nonfunctional
fluorescent proteins. After Cre-induced recombination, N-terminus
and C-terminus can be reconstituted to functional fluorescent pro-
teins (e.g., RFP and GFP in the old versions of MADM and GFP
and tdTomato in the new versions (Fig. 8)). Since recombination
can be tightly controlled by the use of tissue- or cell-specific Cre
drivers as well as by tamoxifen-inducible CreERT2 drivers for tempo-
ral induction, the lineage of single cells can be traced during devel-
opment and adulthood. Furthermore MADM can be used to
selectively knockout genes in labeled cells by introducing (by natu-
ral recombination) a mutant allele between the centromere and the
MADM transgene, while the second chromosome solely carries the
wild-type allele (besides the complementary MADM transgene/
knockin). In case of a X-segregation (sister chromatids segregating
into different daughter cells) in G2 phase of mitosis, two different
cells with different colors (red and green) and genotypes (homozy-
gote mutant and homozygote wild-type allele) are generated, while
a Z-segregation would result in two heterozygous sister cells (one
unlabeled and one labeled with both fluorescent markers) (Fig. 7).
In contrast to classical conditional knockouts, MADM
conditional knockouts have several advantages (Fig. 9). First of all,
non-cell-autonomous defects can be almost excluded, because just
a small proportion of cells carries the homozygous knockout
(which can be titered up or down by the use of a CreERT2 driver).
Secondly it has clear advantages, when the cellular character and
behavior (cell shape, cell migration, axon guidance) have to be
308 Claudius F. Kratochwil and Filippo M. Rijli
Fig. 7 The MADM system. The MADM system consists of two knockins, which are chimeric genes constituted
by the N-terminus and C-terminus of two distinct fluorescent proteins (e.g., RFP and GFP). Before Cre-mediated
recombination, the chimeric N-RFP/C-GFP or N-GFP/C-RFP alleles result in two nonfunctional proteins. During
the S phase of the mitosis, sister chromatids duplicate and pair. If an interchromosomal recombination hap-
pens, the correspondingly recombined sister chromatids segregate during the G2 phase into different daughter
cells (X-segregation). Two cells of different colors (i.e., expressing either red or green fluorescence) are then
generated. In case each sister chromatids segregate into the same cell, a double-labeled and colorless cell is
produced. A recombination during G1 or G0 (differentiated cell) phase results as well in a colorless and in a
double-labeled cell. If additionally a mutant allele is recombined onto a MADM-bearing chromosome (while the
other MADM containing chromosome has the wild-type allele), X-segregation results in the generation of sib-
ling mutant and wild-type cell, labeled with different fluorescent markers, while Z-segregation or recombina-
tion in G1 or G0 phase does not change the genotype of the cell
Fig. 8 MADM cassettes. To increase variability in the choice of the second reporter/gene, the old cassettes (a)
containing GFP and RFP have been replaced by one cassette containing an ATG and the C-terminus of GFP and
a second cassette containing N-GFP and an ATG-less tdTomato (b). If another gene except tdTomato should be
overexpressed (GeneX ), only one transgenic line has to be created making the system more universal
Fig. 9 Advantages of conditional knockouts and MADM conditional knockouts. While classical knockouts give
little information about the role of the knockedout gene in different tissues, the use of conditional knockouts
allows the analysis of gene function in selected cell subpopulations or tissues. The MADM system allows fur-
thermore to trace the lineage of single cells with different genotypes (homozygous knockout and wild type,
respectively) in an environment of heterozygous cells
chr.11 and only between the MADM insertion locus and the
centromere.
Recently the system has been further optimized by using con-
structs with tdTomato, a red fluorescent protein brighter than RFP,
as well as by making the choice of the second gene to be recom-
bined more flexible (e.g., by substituting a second fluorescent
reporter gene with a GeneX to be overexpressed) (Fig. 8) [26].
3.8 Brainbow An elegant approach to analyze the behavior of single cells in a tis-
System sue, especially for the analysis of connectivity patterns in neural
circuits, came from the Lichtman group [29]. By the use of
different, incompatible Lox sites (LoxN, Lox2722, and LoxP), these
authors generated mice, which, in a stochastic manner, are induced
to express only one fluorescent protein (YFP, CFP, or RFP) as a
final recombination event (Figs. 10 and 11).
Four different brainbow versions that stochastically express
different kinds of fluorescent proteins exist (Fig. 11a, b), and the
corresponding transgenic mouse lines are available from Jackson
310 Claudius F. Kratochwil and Filippo M. Rijli
Fig. 10 The brainbow system. Recombination of the Brainbow 1.1 constructs can result in three possible
outcomes. A recombination of the LoxN site results in the transcriptional activation of RFP. Because the other
two Lox sites (Lox2722 and LoxP ) lack a second site because they have been placed 3′ of the LoxN site, no
further recombination can occur. If the first recombination happens between the Lox2272 sites, the result is
expression of YFP; if it happens between the LoxP sites, it results instead in expression of CFP
4 Notes
Fig. 11 Overview of brainbow constructs and principle of “multicolor” brainbow. (a, b) Four brainbow con-
structs have been generated (a) with different possible outcomes (b). While Brainbows 1.0 and 1.1 recombine
only once, Brainbows 2.0 and 2.1 continue to recombine changing the cassette expression pattern after every
recombination event. (c) Due to multiple insertions more than three different colors are possible. Three inserts
would give ten distinguishable colors. Here YFP channel would be set to green and CFP channel to blue, giving
a similar principle as the R(ed)G(reen)B(lue) color model of screen displays
Acknowledgements
References
1. Thomas KR, Capecchi MR (1987) Site- 10. Siegal ML, Hartl DL (1996) Transgene
directed mutagenesis by gene targeting in Coplacement and high efficiency site-specific
mouse embryo-derived stem cells. Cell recombination with the Cre/loxP system in
51(3):503–512 Drosophila. Genetics 144(2):715–726
2. Gu H, Marth JD, Orban PC, Mossmann H, 11. Werdien D, Peiler G, Ryffel GU (2001) FLP
Rajewsky K (1994) Deletion of a DNA poly- and Cre recombinase function in Xenopus
merase beta gene segment in T cells using cell embryos. Nucleic Acids Res 29(11):E53–3
type-specific gene targeting. Science 12. Dong J, Stuart GW (2004) Transgene manip-
265(5168):103–106 ulation in zebrafish by using recombinases.
3. Lakso M, Sauer B, Mosinger B, Lee EJ, Methods Cell Biol 77:363–379
Manning RW, Yu SH, Mulder KL, Westphal 13. Gilbertson L (2003) Cre-lox recombination:
H (1992) Targeted oncogene activation by Creative tools for plant biotechnology. Trends
site-specific recombination in transgenic Biotechnol 21(12):550–555
mice. Proc Natl Acad Sci USA 14. Guo F, Gopaul DN, van Duyne GD (1997)
89(14):6232–6236 Structure of Cre recombinase complexed with
4. Sauer B (1998) Inducible gene targeting in DNA in a site-specific recombination synapse.
mice using the Cre/lox system. Methods Nature 389(6646):40–46
14(4):381–392 15. Nakano M, Odaka K, Ishimura M, Kondo S,
5. O’Neal KR, Agah R (2007) Conditional tar- Tachikawa N, Chiba J, Kanegae Y, Saito I
geting: inducible deletion by cre recombinase. (2001) Efficient gene activation in cultured
Methods Mol Biol 366:309–320 mammalian cells mediated by FLP
6. Kim JC, Dymecki SM (2009) Genetic fate- recombinase-expressing recombinant adenovi-
mapping approaches: new means to explore rus. Nucleic Acids Res 29(7):E40
the embryonic origins of the cochlear nucleus. 16. Buchholz F, Angrand P-O, Stewart AF (1998)
Methods Mol Biol 493:65–85 Improved properties of FLP recombinase
7. Wilson TJ, Kola I (2001) The LoxP/CRE sys- evolved by cycling mutagenesis. Nat
tem and genome modification. Methods Mol Biotechnol 16(7):657–662
Biol 158:83–94 17. Rodríguez CI, Buchholz F, Galloway J,
8. Brault V, Besson V, Magnol L, Duchon A, Sequerra R, Kasper J, Ayala R, Stewart AF,
Hérault Y (2007) Cre/loxP-mediated chro- Dymecki SM (2000) High-efficiency deleter
mosome engineering of the mouse genome. mice show that FLPe is an alternative to Cre-
Handb Exp Pharmacol 178:29–48 loxP. Nat Genet 25(2):139–140
9. Feil S, Valtcheva N, Feil R (2009) Inducible 18. Feil R, Wagner J, Metzger D, Chambon P
Cre mice. Methods Mol Biol 530:343–363 (1997) Regulation of Cre recombinase activity
Cre/Lox in Mouse Development 313
by mutated estrogen receptor ligand-binding 32. Luo L (2007) Fly MARCM and mouse
domains. Biochem Biophys Res Commun MADM: genetic methods of labeling and
237(3):752–757 manipulating single neurons. Brain Res Rev
19. Feil R, Brocard J, Mascrez B, LeMeur M, 55(2):220–227
Metzger D, Chambon P (1996) Ligand- 33. Potter CJ, Tasic B, Russler EV, Liang L, Luo L
activated site-specific recombination in mice. (2010) The Q system: a repressible binary sys-
Proc Natl Acad Sci USA 93(20):10887–10890 tem for transgene expression, lineage tracing,
20. Edwards WF, Young DD, Deiters A (2009) and mosaic analysis. Cell 141(3):536–548
Light-activated Cre recombinase as a tool for the 34. Zhang Y, Buchholz F, Muyrers JP, Stewart AF
spatial and temporal control of gene function in (1998) A new logic for DNA engineering
mammalian cells. ACS Chem Biol 4(6):441–445 using recombination in Escherichia coli. Nat
21. Hirrlinger J, Scheller A, Hirrlinger PG et al Genet 20(2):123–128
(2009) Split-cre complementation indicates 35. Sauer B, Henderson N (1988) Site-specific
coincident activity of different genes in vivo. DNA recombination in mammalian cells by
PLoS One 4(1):e4286 the Cre recombinase of bacteriophage P1.
22. Hirrlinger J, Requardt RP, Winkler U, Wilhelm Proc Natl Acad Sci USA 85(14):5166–5170
F, Schulze C, Hirrlinger PG (2009) Split- 36. Santoro SW, Schultz PG (2002) Directed evo-
CreERT2: temporal control of DNA recombi- lution of the site specificity of Cre recombinase.
nation mediated by split-Cre protein fragment Proc Natl Acad Sci USA 99(7):4185–4190
complementation. PLoS One 4(12):e8354 37. Schnütgen F, Doerflinger N, Calléja C,
23. Wang P, Chen T, Sakurai K, Han B-X, He Z, Wendling O, Chambon P, Ghyselinck NB
Feng G, Wang F (2012) Intersectional cre (2003) A directional strategy for monitoring
driver lines generated using split-intein medi- Cre-mediated recombination at the cellular
ated split-cre reconstitution. Sci Rep 2:497 level in the mouse. Nat Rev Neurosci
24. Farago AF, Awatramani RB, Dymecki SM 21(5):562–565
(2006) Assembly of the brainstem cochlear 38. Zheng B, Sage M, Sheppeard EA, Jurecic V,
nuclear complex is revealed by intersectional Bradley A (2000) Engineering mouse chromo-
and subtractive genetic fate maps. Neuron somes with Cre-loxP: range, efficiency, and
50(2):205–218 somatic applications. Mol Cell Biol
25. Zong H, Espinosa JS, Su HH, Muzumdar 20(2):648–655
MD, Luo L (2005) Mosaic analysis with dou- 39. Visel A, Minovitsky S, Dubchak I, Pennacchio
ble markers in mice. Cell 121(3):479–492 LA (2007) VISTA Enhancer Browser–a data-
26. Tasic B, Miyamichi K, Hippenmeyer S, Dani base of tissue-specific human enhancers.
VS, Zeng H, Joo W, Zong H, Chen-Tsai Y, Nucleic Acids Res 35:D88–D92
Luo L (2012) Extensions of MADM (mosaic 40. Engström PG, Fredman D, Lenhard B (2008)
analysis with double markers) in mice. PLoS Ancora: a web resource for exploring highly
One 7(3):e33332 conserved noncoding elements and their asso-
27. Lee T, Luo L (1999) Mosaic analysis with a ciation with developmental regulatory genes.
repressible cell marker for studies of gene func- Genome Biol 9(2):R34
tion in neuronal morphogenesis. Neuron 41. Visel A, Blow MJ, Li Z et al (2009) ChIP-seq
22(3):451–461 accurately predicts tissue-specific activity of
28. Hippenmeyer S, Youn YH, Moon HM, enhancers. Nature 457(7231):854–858
Miyamichi K, Zong H, Wynshaw-Boris A, Luo 42. Yee SP, Rigby PW (1993) The regulation of
L (2010) Genetic mosaic dissection of Lis1 myogenin gene expression during the embry-
and Ndel1 in neuronal migration. Neuron onic development of the mouse. Genes Dev
68(4):695–709 7(7A):1277–1289
29. Livet J, Weissman TA, Kang H, Draft RW, Lu 43. Srinivas S, Watanabe T, Lin CS, William CM,
J, Bennis RA, Sanes JR, Lichtman JW (2007) Tanabe Y, Jessell TM, Costantini F (2001) Cre
Transgenic strategies for combinatorial expres- reporter strains produced by targeted insertion
sion of fluorescent proteins in the nervous sys- of EYFP and ECFP into the ROSA26 locus.
tem. Nature 450(7166):56–62 BMC Dev Biol 1:4
30. Soriano P (1999) Generalized lacZ expression 44. Hope IA, Struhl K (1987) GCN4, a eukary-
with the ROSA26 Cre reporter strain. Nat otic transcriptional activator protein, binds as a
Genet 21(1):70–71 dimer to target DNA. EMBO J
31. Madisen L, Zwingman TA, Sunkin SM et al 6(9):2781–2784
(2010) A robust and high-throughput Cre 45. Espinosa JS, Luo L (2008) Timing neurogen-
reporting and characterization system for the esis and differentiation: insights from quantita-
whole mouse brain. Nat Neurosci tive clonal analyses of cerebellar granule cells.
13(1):133–140 J Neurosci 28(10):2301–2312
INDEX
Simon G. Sprecher (ed.), Brain Development: Methods and Protocols, Methods in Molecular Biology, vol. 1082,
DOI 10.1007/978-1-62703-655-9, © Springer Science+Business Media, LLC 2014
315
BRAIN DEVELOPMENT: METHODS AND PROTOCOLS
316 Index
Formaldehyde ................... 6, 7, 22, 25, 40, 52, 72, 101, mCherry ................................................ 58, 59, 65, 189
122, 123, 130, 131, 138, 203, 229, 231, mCitrine ........................................................ 58, 59, 65
239, 248 Microinjection ................................................. 143–153
Fruit fly ..................................................................... 12 Microinjector ................................... 149, 157, 189, 288
micro RNA (mRNA) ......................... 29, 130, 143, 145,
G 146, 149, 150, 177, 182, 262, 265, 269, 270,
Gal80.. .............................................. 38, 80–83, 85–87 281, 282
Gal4/UAS ..................................... 57, 80, 87, 160, 166 Microscope ................................... 5, 20, 40, 62, 73, 82,
Gastropod ............................................... 117, 118, 124 100, 124, 130, 148, 157, 179, 188, 202, 217,
Gene silencing ................................................. 253–265 236, 262, 269, 288
Genotyping .................... 15, 28, 60, 63, 72, 82, 84, 88, Microscope slides .................... 8, 11, 13, 15, 16, 23, 26,
89, 281, 298, 302–304, 307–308 28, 30, 62, 64, 83, 87, 100, 105, 159, 160, 165,
GFP..... ............... 13, 41, 47, 48, 54, 76, 77, 80, 85, 86, 166, 179, 182, 202, 211, 219, 288
88, 92, 146, 150, 157, 166, 189, 190, 193, 201, Minutien pins .................................................. 8, 23, 28
206–208, 236, 290, 296, 305–309 Mitotic recombination ............................................... 79
Glass capillary .......................................... 152, 157, 261 Molluscs .......................................................... 117–124
Glass needles............................ 148, 149, 152, 256, 258 Monoclonal antibody ......................... 62, 217, 276, 282
Glia..... .......................... 3, 57, 67, 81, 86, 99–112, 269 Morpholino ..................... 143, 146, 147, 150, 155–170
Glutaraldehyde .................................................... 6, 207 Mosaic(s) .................... 5, 67, 79, 80, 86, 144, 145, 188,
Graft........................ 236, 239, 240, 243, 245–247, 250 189, 296, 307
Grasshopper....................................................... 99–112 Mosaic analysis with a repressible cell marker
(MARCM) ........ 5, 38, 67, 79–92, 188, 296, 307
I Mosaic analysis with double marker (MADM) ......... 188,
295–298, 307–309
ImageJ......................................... 66, 83, 104, 202, 213
Mounting medium ....................... 8, 10, 11, 16, 54, 83,
Immunostaining ....................3–16, 62, 84–85, 99–112,
120, 123, 136, 207, 208, 219, 224, 230, 274,
129–140, 207, 239, 248, 275–276, 290
279, 282, 288
Incubator ............................. 63, 67, 82, 151, 165, 166,
Mouse ................................. 11, 13, 62, 72, 76, 88, 102,
190, 199, 203, 222, 237, 240, 241, 243, 245,
192, 201, 207, 217, 219, 239, 253, 254,
256, 258, 260–263, 303
269–282, 285–291, 295–312
In ovo. ............. 237–239, 243–247, 254–260, 262–264
Insects. .......... 20, 37, 99, 102, 199, 238, 239, 246, 247 N
In situ hybridization ........................... 19–34, 129–140,
175–183, 204, 217–233, 236, 239, 240, 247, Neural plate ............................................ 235, 236, 241,
248, 254, 262, 269–282, 311 242, 245–246
In utero ................................................... 285–291, 300 Neural stem cell ....................................... 20, 37, 71–77
In vivo imaging................................................ 165–166 Neuroblast .................. 3, 20, 21, 29, 31, 34, 37–55, 71,
76, 77, 80, 81, 86, 89
K Neuron............... 3, 38, 57, 81, 99, 124, 139, 169, 187,
197, 218, 236, 254, 269, 287
Knock-in ......................................... 285, 300, 307, 308
Normal goat serum .............. 7, 8, 61, 83, 119, 201, 271
Knock-out ...... 143, 254, 285, 295, 296, 298, 300, 302,
305, 307–309, 311 P
L Paraffin section ........................ 133, 134, 136–138, 270
Paraformaldehyde ........................... 6, 8, 61, 66, 83, 84,
LacZ.............................................. 29, 31, 81, 297, 301 101, 109, 119, 130, 139, 177, 198, 218, 222,
Larva... ...................................... 49, 73, 88, 90, 92, 118 227, 229, 270, 288
LexA...................................................................81, 86 PCR. See Polymerase chain reaction (PCR)
Lophotrochozoa ...................................................... 117 Phalloidin ........................................ 118, 119, 121, 124
LoxP.... .................................... 296–302, 307, 309, 310 Phenotype .................... 5, 81, 145, 146, 165–166, 168,
262, 265, 295, 301, 302
M
Phi-C31................................................................... 145
MADM. See Mosaic analysis with double marker Pipette.......................8, 12, 13, 15, 16, 28, 33, 52, 62,
(MADM) 63, 67, 72, 74–76, 83, 84, 87, 91, 92, 100–102,
MARCM. See Mosaic analysis with a repressible cell 105, 109, 133, 139, 147, 149, 153, 157,
marker (MARCM) 159, 163, 165, 167, 200, 201, 209–212, 236,
BRAIN DEVELOPMENT: METHODS AND PROTOCOLS
Index
317
238–240, 242, 243, 245–247, 249, 271, 272, SPIM. See Selective Plane Illumination Microscopy
275, 281 (SPIM)
Polyclonal antibody ................................................. 217 Split-Cre .......................................................... 297, 306
Polymerase chain reaction (PCR) .............. 87, 224, 262, Stereo microscope.............................. 23, 25, 26, 28, 34
298, 302–304
Proteinase K (PK) ................... 130, 135, 140, 177, 178, T
180, 201, 205, 221, 227, 231, 232, 273, 278, Tadpole .......................................... 132, 139, 156, 157,
282, 298, 303 159–167, 170
Tamoxifen ...................................... 296, 298, 305, 307,
Q
311, 312
Quail ................................................................ 235–250 Tissue dissection ...................................................... 130
Tissue fixation............................................................ 38
R Transgenesis ............................... 58, 59, 63, 65, 66, 68,
Restriction enzyme ................. 144–145, 220, 226, 231, 85–87, 187, 188
277, 298 Transplantation .................................. 79, 144, 235–250
RNA... ................ 19–34, 131, 132, 134, 136, 139, 147, Tyramide signal amplification (TSA) .............. 21, 23, 26,
153, 155, 156, 176–178, 180, 182, 218, 220, 33, 34, 50, 175–183
224, 226, 227, 230–232, 254, 269–282
V
interference ........................................................ 156
probe .................................. 20, 132, 134, 139, 176, Viratome ................................. 118–123, 248, 288, 290
177, 180, 182, 218, 224, 226, 227, 231, 232,
270–273, 276–277, 280 X
RNAse .................... 131, 149, 150, 153, 155, 220, 221, Xenopus ...........................129–140, 143–153, 155–170
226–229, 231, 232, 273, 274, 277, 278,
281, 282 Y
S YFP.......................................................... 157, 309–311