PCR Methods and Protocols 2nd Edition 2023
PCR Methods and Protocols 2nd Edition 2023
PCR
Methods and Protocols
Second Edition
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Second Edition
Edited by
Lucília Domingues
CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
Editor
Lucı́lia Domingues
CEB - Centre of Biological Engineering
University of Minho
Braga, Portugal
Cover illustration by Carlos E. Costa (CEB – Centre of Biological Engineering, University of Minho).
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 1 New York Plaza, New York, NY 10004, U.S.A.
Preface
With 40 years since its official discovery and 51 years since its theoretical proposition, the
polymerase chain reaction (PCR) has revolutionized the fields of biotechnology, medicine,
food microbiology, environmental microbiology, industry, and science, in general. The
concept is so perfectly simple that the elemental scheme remains unchanged since its
foundation. There are very few inventions that can compete with the importance of PCR.
PCR is still today a fundamental tool in current scientific research and its importance has
been recently disclosed to the general public with its widespread use during the COVID-19
pandemic that began in 2019. Being such a relevant technique with wide-range applications,
significant literature exists on the basics of PCR. Still, the specificities for its application in
diverse areas of the biotechnology and bioengineering field are mostly dispersed and are
preferentially found in the health area. PCR is a powerful and flexible tool in modern
biotechnology and the continued development of this technology is still expanding its
wide range of applications.
This new edition of PCR Methods and Protocols maintains the focus of the first edition
on PCR application specificities to the biotechnology and bioengineering field with updated
content on recently developed cutting-edge methodologies and novel applications. While
the previous edition almost exclusively covered end-point PCR, this volume is balanced with
real-time PCR and with fresh applications in the biotechnology and bioengineering field, in
particular in the food sector, in which a growing trend for the use of this technology is
observed. Topics such as detection of foodborne microbial contaminants, toxins, and
allergens are included as well as food authentication. Two protocols involving high-
resolution melting assays are illustrated for the detection of foodborne pathogens and
fungi detection in plant matrices. Relevant applications in biotechnology are emphasized
with protocols for accurate absolute quantification of bacterial populations in mixed cultures
and for gene expression quantification from pathogenic bacterial biofilms. Applications in
synthetic biology for the assessment of recombination efficiency in minicircle production
and quantification of plasmid copy number are also included. More recently developed PCR
techniques like digital PCR protocols were incorporated in this new edition highlighting the
applications for SARS-Cov-2 detection and surveillance from sewage samples and food
herbal spices and products authentication. Emulsion PCR coupled with denaturing gradient
gel electrophoresis is described in the context of microbial diversity studies. New develop-
ments for end-point PCR like the use of disruptors for PCR improvement are included as
well as novel applications such as the use of mitochondrial DNA D-loop amplification and
sequencing for species differentiation in milk. Highly used end-point PCR applications from
the previous edition were kept and updated like long fragment PCR and megaprimer
applications in the synthesis of fusion genes, colony PCR, inverse PCR for site-directed
mutagenesis, and degenerate PCR.
It is amazing as such a straightforward methodology like PCR has evolved and expanded
its applications over 40 years. The ongoing development of PCR technology has enabled
PCR to continue to play an indispensable role in the biotechnology and bioengineering
field. The trend is the development of new PCR technologies and applications with digitali-
v
vi Preface
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 Digital PCR: A Partitioning-Based Application for Detection
and Surveillance of SARS-CoV-2 from Sewage Samples. . . . . . . . . . . . . . . . . . . . . . 1
Bhumika Prajapati, Dalipsingh Rathore, Chaitanya Joshi,
and Madhvi Joshi
2 Digital PCR: A Tool to Authenticate Herbal Products and Spices . . . . . . . . . . . . . 17
Abhi P. Shah, Tasnim Travadi, Sonal Sharma, Ramesh Pandit,
Chaitanya Joshi, and Madhvi Joshi
3 Emulsion Polymerase Chain Reaction Coupled with Denaturing
Gradient Gel Electrophoresis for Microbial Diversity Studies . . . . . . . . . . . . . . . . . 31
Maria-Eleni Dimitrakopoulou, Dimosthenis Tzimotoudis,
and Apostolos Vantarakis
4 Real-Time PCR High-Resolution Melting Assays for the Detection
of Foodborne Pathogens. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
Prashant Singh and Frank J. Velez
5 High-Throughput Real-Time qPCR and High-Resolution Melting
(HRM) Assay for Fungal Detection in Plant Matrices . . . . . . . . . . . . . . . . . . . . . . . 53
Filipe Azevedo-Nogueira, Sara Barrias, and Paula Martins-Lopes
6 Multiplex Real-Time PCR for the Detection of Shiga Toxin-Producing
Escherichia coli in Foods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
Ana Costa-Ribeiro, Sarah Azinheiro, Foteini Roumani,
Marta Prado, Alexandre Lamas, and Alejandro Garrido-Maestu
7 DNA Isolation from Cocoa-Derived Products and Cocoa Authentication
by TaqMan Real-Time PCR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
Ana Caroline De Oliveira, Yordan Muhovski, Herve Rogez,
and Frédéric Debode
8 Quantitative Real-Time PCR for the Detection of Allergenic Species
in Foods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Joana Costa, Caterina Villa, and Isabel Mafra
9 Accurate Absolute Quantification of Bacterial Populations in Mixed
Cultures by qPCR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
Ângela Lima, Lúcia G. V. Sousa, and Nuno Cerca
10 Real-Time PCR Method for Assessment of ParA-Mediated
Recombination Efficiency in Minicircle Production . . . . . . . . . . . . . . . . . . . . . . . . . 117
Cláudia P. A. Alves, Duarte Miguel F. Prazeres,
and Gabriel A. Monteiro
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253
Contributors
ix
x Contributors
Abstract
The wastewater-based surveillance of SARS-CoV-2 has emerged as a potential tool for cost-effective,
simple, and long-term monitoring of the pandemic. Since the COVID-19 pandemic, several developed
countries have incorporated the national wastewater surveillance program into their national policies related
to pandemic management. Various research groups have utilized the approach of real-time quantitative
reverse transcription PCR (RT-qPCR) for the quantification of SARS-CoV-2 from environmental samples
like sewage water. However, detection and quantification using RT-qPCR relies on standards and is known
to have lesser tolerance to inhibitors present in the sample. Unlike RT-qPCR, digital PCR (dPCR) offers an
absolute and sensitive quantification without a need reference and offers higher tolerance to inhibitors
present in the wastewater samples. Additionally, the accuracy of detection increases with the presence of rare
target copies in the sample. The methodology herein presented comprises the detection and quantification
of SARS-CoV-2 from sewer shed samples using the dPCR approach. The main features of the process
include virus concentration and absolute quantification of the virus surpassing the substantial presence of
inhibitors in the sample. This chapter presents the optimized PEG and NaCl-based protocol for virus
concentration followed by nucleic acid extraction and quantification using CDC-approved N1 + N2 assay.
The protocol uses MS2 bacteriophage as a process recovery or internal control.
The methodology herein described highlights the importance of digital PCR technologies for environ-
mental surveillance of important emerging pathogens or pandemics.
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_1,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
1
2 Bhumika Prajapati et al.
2 Materials
n × 0:56 mL = y mL
y mL × 10 μL=mL = z μL
where n = number of samples to be processed
y = calculated volume of buffer AVL
z = volume of carrier RNA-buffer AVE to add to buffer AVL
Gently mix by inverting the tube 10 times. To avoid foam-
ing, do not vortex.
5. 0.5–10, 10–100, and 100–1000 μL micropipettes with their
suitable filter tips (pipette tips with aerosol barriers for prevent-
ing cross-contamination are recommended).
6. Refrigerated centrifuge.
7. Ethanol (96–100%).
8. 1.5 mL microcentrifuge tubes.
4 Bhumika Prajapati et al.
2.5 Digital PCR Mix 1. QIAcuity nanoplate with 26,000 nanopartitions with sealing
Preparation film (Qiagen, see Note 1).
2. Primers and probe cocktail (10 μM).
3. Sterile PCR strips or plate.
4. One-step viral RNA probe assay (Qiagen or equivalent).
5. DNase, RNase-free Eppendorf tubes (1.5 mL).
6. RNAout: Solution containing surfactant like Tween-20 in
nuclease-free water.
7. Mini centrifuge.
8. 0.5–10, 10–100, and 100–1000 μL micropipettes with their
suitable filter tips (pipette tips with aerosol barriers for prevent-
ing cross-contamination are recommended).
9. PCR workstation.
10. QIAcuity digital PCR system (Qiagen; see Note 2).
3 Methods
3.1 Wastewater The wastewater samples from an in-flow of eight different STPs
Sample Collection across Ahmedabad city in Gujarat, India, were collected in the
morning hours once on a weekly basis using the grab sampling
method. Collect the samples in 250 mL sterile polypropylene bot-
tles, and transport them to the lab on the same day by maintaining
appropriate cooling and sterile conditions. The sampling sites
should be selected on the basis of the catchment area and minimal
liquid discharge (MLD) capacity of each plant [11]. The entire
workflow for the process of SARS-CoV-2 detection from wastewa-
ter samples is presented in Fig. 1 [12].
3.2 Filtration, 1. Collect the sample in 250 mL sterile polypropylene bottles, and
Centrifugation, and transport it to the lab on the same day by maintaining appro-
Precipitation priate conditions. Each sample should be labeled giving infor-
mation regarding the name of the collector, the date, time, and
exact geographical location with co-ordinates.
2. On the same day, open the collected sample in the biosafety
cabinet, and transfer 30 mL of wastewater samples to a 50 mL
centrifuge tube, and centrifuge at 1800 × g for 40 min at
10–14 °C temperature.
Digital PCR for Wastewater-Based Surveillance of SARS-CoV-2 5
Fig. 1 Overview of process workflow. The entire process workflow includes sample collection with different
types of samples, sampling methods such as grab or composite sampling, sample/virus concentration using
PEG-8000 and NaCl, viral RNA extraction using spin column-based kit, absolute quantification using digital
PCR assay, and data analysis
3.3 RNA Extraction 1. Perform the RNA extraction from the concentrated pellet
from Concentrated using a commercially available spin column-based kit. In the
Pellet current study, QIAamp viral RNA mini kit (Qiagen) has been
used as per the protocol mentioned by the manufacturer with
slight modifications [12]. Take 300 μL of concentrated sample
from the above procedure using a sterile filter tip for RNA
isolation, and add 300 μL nuclease-free water (NFW) in a
separate tube which will act as negative control (NC). Add
10 μL of MS2 bacteriophage (see Note 3) as internal process
recovery control for each sample and controls.
6 Bhumika Prajapati et al.
3.4 Primer and Probe The dPCR assay for quantifying the viral RNA consists of a set of
for Digital PCR primers and a set of fluorescent probes. The assay quantitatively
detects the SARS-CoV-2 nucleic acid from the upper and lower
respiratory clinical and environmental specimen (nasopharyngeal or
oropharyngeal swabs, sputum, lower respiratory aspirates, bronch-
oalveolar lavage, environmental samples, etc.). The cocktail of
N1 + N2 primers and probes targets the genomic regions
(N1 and N2) of the SARS-CoV-2 viral genome [13]. The primer-
probe mix has been acquired commercially. The two probes are
coupled with FAM as a reporter dye and use ZEN™ quenchers for
enhanced sensitivity (sequence provided in Table 1 [12, 14]). Dur-
ing the PCR amplification, the (5′-3′) exonuclease activity of DNA
polymerase will degrade the probes that are hybridized to the target
sequence. When the fluorophore (FAM or HEX or ROX) dye
molecules are released from the probes and thus are no longer in
close proximity to the quencher, they can be detected and quanti-
fied by the imaging analysis steps in the dPCR.
Digital PCR for Wastewater-Based Surveillance of SARS-CoV-2 7
Table 1
Primers and probe sequence used in the protocol herein described for the detection of SARS-CoV-2
3.5 Digital PCR Mix 1. Before setting up the final reaction mixture for the dPCR assay,
Preparation for enough precautions should be taken to avoid cross-
Wastewater Samples contamination of the sample and reagents. All reagents includ-
ing PCR master mix, enzymes, primer-probe mix, and
nuclease-free water should be properly thawed before use.
Wipe the entire PCR hood surface area and micropipettes
with the RNAout (carryover RNA-degrading solution) to
degrade or remove any contaminant RNA from the surface
and pipettes (see Note 5).
2. After thawing the reagents, vials should be gently inverted 2–3
times and spun down using microcentrifuge to ensure proper
mixing of all enzymes and components in the reagent vials. The
master mix preparation and sample/template addition area
should be separated to avoid aerosol-based contamination (see
Note 6).
3. Prepare the PCR master mix in sterile 1.5 μL of Eppendorf
tubes as mentioned below. The QIAcuity viral probe kit con-
tains a 4× concentrated PCR master mix, which is optimized
for microfluidic use in the QIAcuity nanoplate.
4. Make the mixture of all components as mentioned in Table 2
(see Note 7) except for the RNA sample or positive control.
Vortex the mixture well, and dispense appropriate volumes of
the reaction mixture into the wells of a standard 24-well,
26,000 partitions PCR nanoplate. Carry the plate to the desig-
nated area for template addition to prevent contamination. The
PCR along with samples and controls should be prepared at
least in duplicates (see Note 8).
5. Dilute the positive control RNA to appropriate dilutions (1:50
or 1:100) using nuclease-free water. Critical: Samples that are
not sufficiently diluted will result in a saturation of the micro-
chambers with the target RNA that will preclude calculating
8
Table 2
PCR mixture composition
Bhumika Prajapati et al.
Component Mix 1 (for N1 + N2 assay) Mix 1 (for MS2 assay) Mix N Final concentration
Nanoplate type 26,000 partitions 26,000 partitions 26,000 partitions (24-well) –
(24-well) (24-well)
4× one-step viral RT-PCR master mix 10 μL 10 μL 10 μL × N 1×
100× multiplex reverse transcription mix 0.4 μL 0.4 μL 0.4 μL × N 1×
10× primer-probe mix 2 μL (N1N2 assay mix) 1 μL (MS2 assay mix) 2 μL (N1N2)/1 μL MS2 × N 0.4 μM forward primer
0.4 μM reverse primer
0.2 μM probe
Template RNA/PC/NC 7 μL 7 μL 7 μL × N 100–200 ng
Nuclease-free water 20.6 μL 21.6 μL Up to 40 μL × N –
Total reaction volume 40 μL 40 μL 40 μL 1×
Digital PCR for Wastewater-Based Surveillance of SARS-CoV-2 9
3.6 Software Setup 1. Before starting the experiment, it is always recommended to set
and Thermal Cycling up experimental design in the software by following the guide-
Conditions lines provided by the manufacturer.
2. Open the software suite, and assign a reaction mix name con-
taining N1 + N2 assay and MS2 assay in QIAcuity software
suite. Assign appropriate wells with sample annotations such as
positive control, unknown, NTC and NC.
3. Assign the respective sample numbers in the plate layout prior
in the software to avoid any mistakes. Set the thermal cycling
program with a heated lid as mentioned in Table 3.
4. In the software suite, select the imaging tab, and capture the
appropriate channel (based on the dye or probes), exposure
duration, and gain value for fluorescent signal imaging. The
current assay targets the N1 + N2 gene and the probe is labeled
with FAM dye; thus a green channel with default exposure
duration and gain value should be selected.
Table 3
PCR amplification profile of the digital PCR system herein described
Priming
Nanoplates micropartitions are filled with the input volume by
plunging of elastic top seal and the input wells which creates a
peristaltic pressure that pumps the input well liquid into the micro-
channel and partitions. Subsequently, the connecting channels
between the partitions are closed by a pressure-controlled rolling
process.
Thermocycling
The thermal cycling step performs the polymerase chain reaction in
the QIAcuity thermal cycler with high speed and precise tempera-
ture control of the various cycling steps.
Imaging
The image acquisition of all wells is the final step in the dPCR
system. The microfluidics partitions that have one or few copies
of target molecule inside can emit fluorescence land are brighter
than those without the target.
3.7 Result 1. Post-run analysis can be easily performed and exported from
Interpretation and the QIAcuity software suite. After the run, view the file, and
Data Analysis observe the threshold line in the 1D scatter plot (Fig. 2) which
separates the negative and positive partitions. There should be
good separation among positive and negative partitions for
getting output copies in a precise manner (see Note 10).
2. Baseline thresholds for each well/sample can be adjusted man-
ually by clicking into the respective wells and adjusting the
recalculation tab.
3. Save the results of the run and remove the plate from the
instrument.
4. Analyze each well for valid partitions. The valid partition indi-
cates the total analyzable volume of the PCR mixture. It is
recommended to achieve around 25,000 valid partitions for
each well.
5. The obtained copy numbers/μL should be reviewed as per the
dMIQE guideline and final (total) copy numbers of tar-
get genes should be calculated using the following formula:
Digital PCR for Wastewater-Based Surveillance of SARS-CoV-2 11
Fig. 2 Representative image of 1D scatter plot: 1D scatter plot indicates the separation of negative and
positive partitions based on the fluorescence intensity. The fluorescence amplitude threshold is represented
by the red line. Positive partitions are seen above the red line, while negative are seen below the line. B1,
positive control; B2,C1, wastewater samples
4 Notes
13
14 Bhumika Prajapati et al.
Fig. 3 Representative image of signal map: The figure represents the signal map of the N1 + N2 target gene,
which is captured in the green channel with FAM as a fluorescent molecule. (a) Positive control. (b) Negative
control. (c) Wastewater samples. The higher copies in positive control indicate more green fluorescent wells,
while samples contain very few target copies, thus showing few wells with green fluorescence. A negative
control should not show any wells with fluorescence
Acknowledgments
References
1. Guidance for surveillance of SARS-CoV-2 var- using droplet digital PCR for environmental
iants: interim guidance (2021) World Health samples. Appl Microbiol 1(1):74–88
Organization 8. Ahmed W, Smith WJM, Metcalfe S, Jackson G,
2. D’Aoust PM, Graber TE, Mercier E, Choi PM, Morrison M et al (2022) Compari-
Montpetit D, Alexandrov I, Neault N et al son of RT-qPCR and RT-dPCR platforms for
(2021) Catching a resurgence: increase in the trace detection of SARS-CoV-2 RNA in
SARS-CoV-2 viral RNA identified in wastewa- wastewater. ACS ES & T Water 2(11):
ter 48 h before COVID-19 clinical tests and 1871–1880
96 h before hospitalizations. Sci Total Environ 9. Ma D, Straathof J, Liu Y, Natalie HNM (2022)
770:145319 Monitoring SARS-CoV-2 RNA in wastewater
3. Baldovin T, Amoruso I, Fonzo M, Buja A, with RT-qPCR and chip-based RT-dPCR:
Baldo V, Cocchio S et al (2021) SARS-CoV- sewershed-level trends and relationships to
2 RNA detection and persistence in wastewater COVID-19. ACS ES & T Water 2(11):
samples: an experimental network for COVID- 2084–2093
19 environmental surveillance in Padua, 10. Huggett JF (2020) The digital MIQE guide-
Veneto Region (NE Italy). Sci Total Environ lines update: minimum information for publi-
760:143329 cation of quantitative digital PCR experiments
4. Chik AHS, Glier MB, Servos M, Mangat CS, for 2020. Clin Chem 66:1012–1029
Pang X, Qiu Y et al (2021) Comparison of 11. Kumar M, Joshi M, Patel AK, Joshi CG (2021)
approaches to quantify SARS-CoV-2 in waste- Unravelling the early warning capability of
water using RT-QPCR: results and implica- wastewater surveillance for COVID-19: a tem-
tions from a collaborative inter-laboratory poral study on SARS-CoV-2 RNA detection
study in Canada. J Environ Sci 107:218–229 and need for the escalation. Environ Res 196:
5. K€astner A, Lücker P, Sombetzki M, Ehmke M, 110946
Koslowski N, Mittmann S et al (2022) SARS- 12. Kumar M, Patel A, Shah A, Raval J, Rajpara N,
CoV-2 surveillance by RT-qPCR-based pool Joshi M et al (2020) The first proof of the
testing of saliva swabs (lollipop method) at capability of wastewater surveillance for
primary and special schools—a pilot study on COVID-19 in India through the detection of
feasibility and acceptability. PLoS One 17(9): the genetic material of SARS-CoV-2. Sci Total
e0274545 Environ 746:141326
6. Jenney A, Chibo D, Batty M, Druce J, 13. Natarajan A, Han A, Zlitni S, Brooks EF, Vance
Melvin R, Stewardson A et al (2022) Surveil- SE, Wolfe M et al (2021) Standardized preser-
lance testing using salivary RT-PCR for SARS- vation, extraction and quantification techni-
CoV-2 in managed quarantine facilities in ques for detection of fecal SARS-CoV-2 RNA.
Australia: a laboratory validation and imple- Nat Commun 12:5753
mentation study. Lancet Reg Health West Pac 14. Centers for Disease Control and Prevention.
26:100533 2019-Novel Coronavirus (2019-nCoV) real-
7. Kokkoris V, Vukicevich E, Richards A, time rRT-PCR panel primers and probes
Thomsen C, Hart MM (2021) Challenges
Chapter 2
Abstract
Authentication of herbal products and spices is experiencing a resurgence using DNA-based molecular
tools, mainly species-specific assays and DNA barcoding. However, poor DNA quality and quantity are the
major demerits of conventional PCR and real-time quantitative PCR (qPCR), as herbal products and spices
are highly enriched in secondary metabolites such as polyphenolic compounds. The third-generation digital
PCR (dPCR) technology is a highly sensitive, accurate, and reliable method to detect target DNA
molecules as it is less affected by PCR inhibiting secondary metabolites due to nanopartitions. Therefore,
it can be certainly used for the detection of adulteration in herbal formulations. In dPCR, extracted DNA is
subjected to get amplification in nanopartitions using target gene primers, the EvaGreen master mix, or
fluorescently labeled targeted gene-specific probes. Here, we describe the detection of Carica papaya
(CP) adulteration in Piper nigrum (PN) products using species-specific primers. We observed an increase
in fluorescence signal as the concentration of target DNA increased in PN-CP blended formulations (mock
controls). Using species-specific primers, we successfully demonstrated the use of dPCR in the authentica-
tion of medicinal botanicals.
Key words Absolute quantification, Digital PCR, Herbal products, Species-specific PCR assay, Third-
generation PCR
1 Introduction
The demand and supply chains of the herbal market are expanding
with industrialization and globalization. However, a bottleneck is
created, which leads to an increased incidence of economically
motivated adulteration in herbal products and spices [1]. As DNA
is a stable biomolecule, it is unaffected by the environmental and
physiological parameters of the plant life cycle which makes it very
suitable to be used for authentication methods with a universal,
reliable, and reproducible approach [1, 2]. Regulatory guidelines
and various pharmacopeia advocate the inclusion of DNA-based
methods such as species-specific PCR assays and DNA barcoding to
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_2,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
17
18 Abhi P. Shah et al.
Fig. 1 Overview of digital PCR workflow. The steps in the dPCR workflow are as follows: (1) prepare the PCR
mixture; (2) load the PCR mixture containing the template DNA onto the QIAcuity nanoplate and seal it;
(3) partitioning of template DNA, amplification of targeted DNA, and detection of the target DNA by the QIAcuity
digital PCR system; and (4) data analysis after adjusting the threshold line
Digital PCR for Authentication of Herbal Products and Spices 19
2 Materials
2.2 DNA Extraction, 1. DNA extraction can be done either with manual CTAB-based
Quantification, and modified methods [8–11] or using commercially available
Dilution plant DNA extraction kit-based methods. The necessary mate-
rials/reagents should be prepared in accordance with that.
2. DNA quantification can be performed using Qubit fluorome-
ter with Qubit dsDNA BR (broad-range) assay kit or Qubit
dsDNA HS (high-sensitivity) assay kit, as directed by the man-
ufacturer, or with a nanodrop spectrophotometer, QIAxpert,
or a UV/VIS spectrophotometer (see Note 2). A DNA quanti-
fication requirement should be prepared in accordance with it.
3. 1 M stock solution of Tris–HCl (pH 8.0): For preparing 1 M
stock solution of Tris–HCl, add about 500 mL water to a 1 L
graduated cylinder or a glass beaker, weigh 121.14 g Tris base,
and transfer to the cylinder. Mix and adjust pH with HCl.
Make up the final volume to 1 L with water. Store at 4 °C.
4. 0.5 M stock solution of EDTA (pH 8): For preparing 0.5 M
stock solution of EDTA, add about 500 mL water to a 1 L
graduated cylinder or a glass beaker, weigh EDTA, and transfer
to the cylinder. Add NaOH pellets to dissolve the EDTA and
adjust the pH. Make up the final volume to 1 L with water.
Store at 4 °C.
5. Low TE (Tris-EDTA) buffer: 10 mM Tris–HCl (pH 8.0),
0.1 mM EDTA (pH 8.0). For preparing low TE, add 10 mL
of 1 M stock solution of Tris–HCl (pH 8.0) and 0.2 mL of
0.5 M stock solution of EDTA (pH 8) to a 1 L graduated
cylinder or a glass beaker, and make up the final volume to
1 L with water. Store at 4 °C.
20 Abhi P. Shah et al.
Table 1
Primers used for the authentication of Piper nigrum (PN) and Carica papaya (CP) with the protocol
described herein
3 Methods
3.1 Preparation of 1. Grind the sample materials [here, dried Carica papaya
Blended Formulations (CP) seed and Piper nigrum (PN) berries] with liquid nitrogen
in a mortar and pestle (see Note 4).
2. For preparing a blended formulation (mock control) contain-
ing 75% (w/w) of CP and 25% (w/w) PN, weigh 75 mg of CP
seed powder and 25 mg of PN barriers powder in a single
1.5 mL microcentrifuge tube.
3. For preparing a blended formulation containing 50% (w/w) of
CP and PN, weigh 50 mg of CP seed powder and 50 mg of PN
barriers powder in another 1.5 mL microcentrifuge tube.
4. For preparing a blended formulation containing 25% (w/w) of
CP and 75% (w/w) of PN, weigh 25 mg of CP seed powder
and 75 mg of PN barriers powder in a third 1.5 mL
microcentrifuge tube.
5. After weighing, homogenize each prepared mixture.
3.2 DNA Extraction, 1. Extract DNA either with manual CTAB-based modified meth-
Quantification, and ods [8–11] or using commercially available plant DNA extrac-
Dilution tion kit-based methods (see Note 5).
2. Quantify the extracted DNA by any method that is described in
Subheading 2.2.
3. After quantification, dilute the extracted DNA using low TE
according to the desired final concentration. Here, we have
diluted the extracted DNA of each blended formulation using
low TE to obtain a final concentration of 0.25 ng/μL.
3.3 Digital PCR Setup Precautions should be taken while setting up digital PCR at all
stages to prevent contamination of the sample and reagent. To
avoid cross-contamination, DNA extraction and PCR setup should
be performed in different rooms (see Note 6). Preparation of the
reaction mixture for PCR should be performed in a laminar flow
hood or PCR cabinet:
1. All reagents should be thawed and mixed by repeatedly
inverting the tubes or, for small volumes, by flicking the tube
and spinning briefly in a centrifuge, prior to preparing the
reaction mixture. For the selection or design of primers, refer
to Note 7.
2. Prepare the PCR mixture in 1.5 mL microcentrifuge tubes, as
shown in Table 2. To ensure homogeneous distribution of the
template across the partitions on the plate, circular DNA such
as plasmids or long DNA templates (>20 kb) such as genomic
DNA extracted from plants and animals must be digested with
22 Abhi P. Shah et al.
Table 2
Components of PCR mixture (see Note 10)
For authentication of PN
Mixture for N and CP (mixture for N
Mixture for one reaction reactions reactions)
Component Nanoplate with 26,000 Nanoplate with Nanoplate with 26,000
partitions (24-well) 26,000 partitions partitions (24-well)
(24-well)
3× EvaGreen PCR 13.3 μL 13.3 × N μL 13.3 × N μL
Master Mix
(QIAGEN)
10 μM forward 1.6 μL 1.6 × N μL 1.6 × N μL
primer
10 μM reverse 1.6 μL 1.6 × N μL 1.6 × N μL
primer
Restriction Up to 1 μL (final concentration Up to 1 μL × N 1 μL EcoRI (1 U/μL), i.e.,
enzyme in reaction system: 0.025 U/μL reaction
(optional) 0.025–0.25 U/μL) volume
Template DNA Variablea (in μL) Variablea × N 2 μL (0.25 ng/μL)
Nuclease-free Variablea (in μL) Variablea × N 20.5 μL
water (NFW)
Final volume 40 μL 40 × N μL 40 × N μL
See Notes 10 and 11
a
a restriction enzyme (see Note 8). The final volume of the PCR
mixture is calculated by multiplying the total number of reac-
tions required for the dPCR experiment by the volume of each
reaction. To account for pipetting variations, PCR mixtures can
be made in excess, i.e., 10–12% more than required. It is not
required to keep samples on ice during reaction setup or when
programming the QIAcuity instrument because of the
hot-start polymerase in the dPCR Master Mix. Mix the master
mix thoroughly, centrifuge it, and dispense equal aliquots into
each well of the PCR plate or each tube of PCR strips.
3. Add template DNA (here we have used 2 μL of 0.25 ng/μL
DNA of each blended formulation, i.e., a total of 0.5 ng of
DNA) to each reaction tube as needed [make sure that negative
template control (NTC) does not receive any template] (see
Note 9). For each sample, prepare at least duplicate reactions
(see Note 9).
4. Seal the PCR plate or PCR strips and vortex it for proper
mixing. Then, centrifuge the PCR plate or PCR strips.
Digital PCR for Authentication of Herbal Products and Spices 23
3.4 PCR Thermal 1. Operate the QIAcuity digital PCR system and software accord-
Cycle and Run Set Up ing to the manufacturer’s recommendations.
in QIAcuity Digital PCR 2. Set the thermal cycling program as follows: initial heat activa-
System tion at 95 °C for 2 min, followed by 40 cycles of 95 °C for 15 s,
58 °C for 30 s, and 72 °C for 40 s, and a final cooling step at
40 °C for 5 min (see Note 12).
3. In the Imaging tab of QIAcuity software, select the channel,
exposure duration, and gain value for fluorescent signal imag-
ing depending on the dye or probes used (see Note 13). We
chose a green channel with the default exposure duration and
gain value for the EvaGreen-based reaction system.
4. Place the QIAcuity nanoplate into the QIAcuity digital PCR
system and start the dPCR program.
5. To start the PCR run, click on the “Run” icon located on the
plate’s pane in the QIAcuity software.
3.5 Data Collection 1. After the run is completed, you can adjust the threshold line to
and Analysis obtain a good separation between negative and positive parti-
tions using automatic thresholding.
2. When the positive and negative partitions are not well sepa-
rated, automatic thresholding fails, and the threshold line must
be adjusted manually. The threshold line can be adjusted man-
ually by clicking into the different wells and adjusting the
threshold line vertically. After adjusting the threshold, the
analysis can be updated by selecting “recalculate” (Fig. 2,
Table 3).
3. Save the results of the run and remove the plate from the
instrument.
4. The obtained copy numbers/μL should be reviewed and as per
the dMIQE guideline [6]. Further, the final (total) copy num-
bers of targeted DNA should be calculated using the following
24 Abhi P. Shah et al.
Fig. 2 Digital PCR assay of Piper nigrum (PN) and Carica papaya (CP) blended formulations. Digital PCR assay
of 75% CP + 25% PN, 50% CP + 50% PN, 25% CP + 75% PN blended formulations with CP- and PN-specific
primers. The fluorescence amplitude threshold is represented by the red line. Above the threshold, positive
partitions were seen, and below the threshold, negative partitions. NTC = no template control. (a) Digital PCR
assay using CP primers. (b) Digital PCR assay using PN primers
Table 3
Copy numbers of Piper nigrum (PN) and Carica papaya (CP) determined by digital PCR with the
protocol described herein
Obtained copy Total copy numbers (obtained Total valid Positive Negative
Target Samplea numbers/μL copy numbers/μL × 20 μL) partitions partitions partitions
CP 75% CP 698.4 13,968 25,474 12,010 13,464
50% CP 389.55 7,791 25,332 7,609 17,723
25% CP 322.4 6,448 25,480 6,495 18,985
PN 25% PN 9.2 184 25,232 219 25,013
50% PN 107.85 2,157 25,463 2,387 23,076
75% PN 137.95 2,759 25,439 3,011 22,428
Total valid partitions and valid positive and valid negative partitions are also described in the table
a
Digital PCR assay of 75% CP + 25% PN, 50% CP + 50% PN, 25% CP + 75% PN blended formulations with CP- and
PN-specific primers
4 Notes
References
Abstract
Emulsion PCR-DGGE is a molecular biology technique used to amplify and analyze DNA fragments. This
technique combines two processes, emulsion PCR and denaturing gradient gel electrophoresis (DGGE), to
enhance the specificity and yield of the amplification process and to separate the amplified fragments based on
their melting behavior. In the emulsion PCR step, a high-quality DNA template is mixed with the PCR
reagents and droplet generator oil to create an oil-in-water emulsion. The emulsion is then subjected to
thermal cycling to amplify the target DNA fragments. The amplified fragments are recovered from the
droplets and purified to remove any impurities that may interfere with downstream applications. In the
DGGE step, the purified amplicon is loaded onto a DGGE apparatus, where the DNA fragments are separated
and visualized based on their melting behavior. This method allows for the concurrent amplification and
separation of multiple DNA fragments, thereby enhancing the resolution and sensitivity of the analysis. It is
widely used in environmental and medical microbiology research, as well as in other fields that require the
identification and characterization of microorganisms, such as the study of microbial diversity in soil, water, and
other natural environments, as well as in the human gut microbiome and other medical samples.
Key words Emulsion PCR, DGGE, Polyacrylamide gel, DNA extraction, Omics technologies, Digi-
tal PCR
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_3,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
31
32 Maria-Eleni Dimitrakopoulou et al.
Fig. 1 Principle of emulsion PCR-DGGE in comparison to conventional PCR-DGGE (food sample is presented) [5]
This article was published in Current Research in Food Science, 4, Dimitrakopoulou, M. E., Panteleli, E., &
Vantarakis, A, Improved PCR-DGGE analysis by emulsion-PCR for the determination of food geographical
origin: A case study on Greek PDO “avgotaracho Mesolonghiou,” 746–751, Copyright Elsevier (2021).
Emulsion PCR-DGGE 33
2 Materials
Table 1
Purpose of specific reagents required for DGGE protocol
Reagent Purpose
Polyacrylamide gel Matrix used to separate DNA fragments based on their melting behavior
Gel apparatus Equipment used to cast and run the polyacrylamide gel
Electrophoresis buffer Buffer used to run the gel and maintain a stable pH during electrophoresis
Loading buffer Solution used to visualize the DNA fragments during electrophoresis
Template DNA The sample of DNA to be separated by DGGE
Denaturants Substances used to create a denaturant gradient within the gel matrix to
separate DNA fragments based on melting behavior
Power supply Used to provide the electrical current for electrophoresis
UV transilluminator Used to visualize the DNA fragments after the electrophoresis is complete
4. TEMED.
5. Staining (GelStar nucleic acid).
6. 50×TAE buffer.
3 Methods
3.1 DNA Extraction An optimized protocol for DNA extraction should be employed on
the specific sample under examination. The selection of a commer-
cial kit for DNA extraction should be accompanied by strict adher-
ence to the manufacturer’s protocols. For example, commercial kits
such as DNeasy PowerFood Microbial Kit and DNeasy mericon
Food Kit are suitable for DNA extraction from food samples. Post-
extraction, it is crucial to ensure the integrity of the DNA sample by
assessing its degradation, which can be determined through elec-
trophoresis on a 0.8% (w/v) agarose gel, with a threshold of 60 °C
for thermal degradation. Additionally, the purity and concentration
of the extracted DNA should be verified using a nanodrop spectro-
photometer (see Note 3).
3.2 Emulsion PCR 1. Introduce into the droplet generator plate a combination of
Amplification DNA template, master mix, and droplet generator oil (see
Notes 4, 12 and 18).
2. Prepare the PCR mixture in 50 μL final volume containing
100 ng of template DNA, 0.2 μM primers, 200 μM deoxyribo-
nucleotide triphosphate (dNTP), and 2.5 μL of 10× reaction
buffer A with Mg+2. In parallel, add an increased concentration
of Taq-polymerase (20×) in accordance with literature
Emulsion PCR-DGGE 35
3.3 Emulsion PCR- 1. Set up the purring apparatus, making sure that the valve con-
DGGE necting the two wells of the gradient maker is closed.
3.3.1 Preparation of 2. Thoroughly mix the acrylamide solutions (20% with either
Polyacrylamide Gel for 40% or 60% or 80%), formamide and urea with the APS and
DGGE Analysis TEMED (see Notes 1, 2, 7, 19, 21 and 25).
3. As quickly as possible, pour the higher concentration solution
into the front chamber (the one closest to the outlet valve), and
pour the lower concentration solution into the back chamber.
4. Open the valve between the wells on the gradient chamber
(bring to horizontal position), and begin the mixing and the
pouring of the gel.
5. To promote even mixing, reduce the speed of the stir bar as the
gel pours (one should just barely be able to see the mixing in
the front chamber). If the stirring speed is too great, the higher
concentration denaturant will flow back into the other
chamber.
6. Allow the gel to sit for at least 2 h.
7. Prepare the stacking gel to load above the already prepared
acrylamide gel.
8. Pour stacking gel as quickly as possible.
36 Maria-Eleni Dimitrakopoulou et al.
3.3.2 Assembling the D- 1. Clean off a set of glass plates (dish soap only).
Code (Gel Running) System 2. Lock plates into place with supports, ensuring they are flush.
3. Carefully remove the comb from the gel, along with any excess
gel from around the wells.
4. With the tall glass facing out and short glass facing in, snap
both assemblages into the holder.
5. Fill D-Code system with approximately 7 L of 1×TAE (140 mL
50×TAE and 6860 mL Milli-Q water) until fill line is reached.
6. Place the holder down into the buffer, ensuring that it is
properly in place.
7. Place the top of the D-Code system on top of the apparatus.
Verify that all electrical connections are created.
8. Plug in electrical cord and turn power switch on.
9. Set temperature to 60 °C and turn on the heater and pump.
Make sure that the buffer level rises and remains above the
negative electrode wire (see Note 11).
3.3.3 Loading the Gel 1. Turn off power and remove top of D-Code system.
2. Prepare samples by combining equal amounts of template
(at least 300 ng) with 5× loading buffer in 500 μL PCR tubes
(see Note 8 and 20).
3. Using corresponding gel loading tips, load samples by placing
the pipettor tip half-way into the well and expunging sample
with extreme care (see Note 22).
4. Load the standard into every fourth lane.
5. Replace top to the D-Code system, ensuring all electrical con-
nections are made.
6. Plug in D-Code and plug it into both the electrical outlet.
7. Turn on D-Code system and set temperature to 60 °C.
8. Set PowerPack300 (with DCODE chip) to appropriate vault
(see Note 9).
9. Turn on the pump only after samples are pulled into the gel
(approximately 15 min see Note 23).
10. Let gel run (see Note 10).
3.3.4 Staining and 1. Drain the excess buffer from the top of the gel and dismantle
Imaging the Gel the apparatus.
2. With the upmost of caution, remove the supports and spacers.
3. With extreme care, use one of the spacers as a lever to pry off
the short glass plate, leaving the gel on the tall glass plate.
4. Soak the tall plate (with gel) in staining (see Note 26), and place
on rocker (slow mode) for 30 min.
Emulsion PCR-DGGE 37
4 Notes
References
1. Shao K, Ding W, Wang F, Li H, Ma D, Wang H emulsion-PCR for the determination of food
(2011) Emulsion PCR: a high efficient way of geographical origin: a case study on Greek
PCR amplification of random DNA libraries in PDO “avgotaracho Mesolonghiou”. Curr Res
aptamer selection. PLoS One 6(9):1–7 Food Sci [Internet] 4:746–751. https://doi.
2. Verma V, Gupta A, Chaudhary VK (2020) org/10.1016/j.crfs.2021.10.005
Emulsion PCR made easy. BioTechniques 6. Iacumin L, Cecchini F, Vendrame M, Comi G
69(1):65–69 (2020) Emulsion pcr (ePCR) as a tool to
3. Ercolini D (2004) PCR-DGGE fingerprinting: improve the power of dgge analysis for microbial
novel strategies for detection of microbes in population studies. Microorganisms 8(8):1–11
food. J Microbiol Methods 56(3):297–314 7. Ampe F, Ben Omar N, Moizan C, Wacher C,
4. El Sheikha AF (2019) Molecular detection of Guyot JP (1999) Polyphasic study of the spatial
mycotoxigenic fungi in foods: the case for distribution of microorganisms in Mexican
using PCR-DGGE. Food Biotechnol [Internet] pozol, a fermented maize dough, demonstrates
33(1):54–108. https://doi.org/10.1080/ the need for cultivation-independent methods
08905436.2018.1547644 to investigate traditional fermentations. Appl
5. Dimitrakopoulou ME, Panteleli E, Vantarakis A Environ Microbiol 65(12):5464–5473
(2021) Improved PCR-DGGE analysis by
Chapter 4
Abstract
Real-time PCR high-resolution melting assays are a method for the identification of single nucleotide
polymorphisms (SNPs). The assay is performed by amplifying a short DNA fragment using a specific primer
pair flanking a target SNP in the presence of a high-resolution melting dye. The HRM analysis of amplicons
groups the samples based on the differences in the melting temperature and the shape of the melt curves,
facilitating a convenient genotyping of samples. This chapter describes the steps and considerations of real-
time PCR HRM assay standardization.
Key words Genotyping, Melting, Pre-melt, Post-melt, Protocol, Saturating dye, SNP
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_4,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
41
42 Prashant Singh and Frank J. Velez
Fig. 1 A high-resolution analysis plot showing the pre-melt region with 100% fluorescence, a post-melt region
with 0% fluorescence, and the melt-phase region showing the difference in the melting behavior of the
amplicons
2 Materials
2.1 DNA Purification There are a wide range of published protocols and commercially
available DNA isolation kits. These kits vary in their ability to
remove impurities from DNA and the length of time and effort to
complete the protocol steps [e.g., DNeasy PowerFood Microbial
Kit (QIAGEN, Valencia, CA, USA); PrepMan™ Ultra Sample
Preparation Reagent (Applied Biosystems, Foster City, CA, USA);
Extracta DNA Prep for PCR (Quanta Biosciences, Beverly, MA,
USA); and InstaGene™ Matrix (Bio-Rad Laboratories, Hercules,
CA)] (see Note 1).
44 Prashant Singh and Frank J. Velez
3 Methods
3.1 Primer Design Primer3 is a freely available web-based tool that can be used for
designing primers for HRM assays [14]. The Primer3 “target”
feature is useful for designing SNP-specific primer pairs. Further,
NCBI’s primer designing tool can be used to check the specificity of
the designed primer pairs. When considering the design of the
HRM primer, it is recommended to target a small-sized amplicon
(60–150 bp) (see Note 9), e.g., O26-32F: 5′-GTG GCA CTG
GTT CTT TTG GT-3′ and O26-118R: 5′-TTT CAT CCC TGC
TAA ATA TTC G-3′ [6].
4 Notes
Fig. 2 Effect of (a) 2 mM, (b) 3 mM, and (c) 3.5 mM MgCl2 concentrations on HRM genotyping assay. Addition
of optimum concentration of MgCl2 to HRM assay facilitate resolution of genotypes
Fig. 3 (a) A non-sigmodal amplification plots obtained using 2× LightCycler® 480 high-resolution melting
master mix. Sample showed a non-sigmoidal amplification curve and high Cq values. (b) Sigmodal amplifica-
tion plots obtained using Apex green master mix for the same sample set
poor Cq-value [7, 17]. Our group has extensively worked with
LightCycler® 480 high-resolution melting master mix (Roche
Diagnostics, Indianapolis, IN, USA) for the development of
HRM-based SNP detection assays [6, 7, 17, 18]. Data from
our studies demonstrate the LightCycler® 480 high-resolution
melting master mix’s ability to differentiate target SNP; how-
ever, for two of our studies, the master mix generated data with
poor Cq-value, making presence/absence calls difficult
(Fig. 3). In such cases the presence/absence calls based on
presence of target genotype in the HRM analysis can be con-
sidered. As initially mentioned, each polymerase is unique; each
mix has its optimum activation temperature and extension
temperatures. Therefore, it is crucial to follow the manufac-
turer’s recommendation for these two steps.
16. The HRM data for the target is analyzed by determining the
pre-melt and post-melt regions. The amplicons of the same
genetic makeup group together in the analysis.
17. HRM assay is a method for the identification of a target SNP
located in a small amplicon, and one limitation of HRM assays
is its inability to identify all the SNP which can be present in the
High-Resolution Melting (HRM) Assays 51
References
1. Wittwer CT, Reed GH, Gundry CN et al 9. Keikha M, Karbalaei M (2021) High resolution
(2003) High-resolution genotyping by ampli- melting assay as a reliable method for diagnos-
con melting analysis using LCGreen. Clin ing drug-resistant TB cases: a systematic review
Chem 49:853–860. https://doi.org/10. and meta-analysis. BMC Infect Dis 21:989.
1373/49.6.853 https://doi.org/10.1186/s12879-021-
2. Erali M, Voelkerding KV, Wittwer CT (2008) 06708-1
High resolution melting applications for clini- 10. Rezaei F, Haeili M, Fooladi AI, Feizabadi MM
cal laboratory medicine. Exp Mol Pathol 85: (2017) High resolution melting curve analysis
50–58. https://doi.org/10.1016/j.yexmp. for rapid detection of streptomycin and etham-
2008.03.012 butol resistance in Mycobacterium tuberculosis.
3. Gundry CN, Vandersteen JG, Reed GH et al Maedica (Bucur) 12:246–257
(2003) Amplicon melting analysis with labeled 11. Monis PT, Giglio S, Saint CP (2005) Compar-
primers: a closed-tube method for differentiat- ison of SYTO9 and SYBR Green I for real-time
ing homozygotes and heterozygotes. Clin polymerase chain reaction and investigation of
Chem 49:396–406. https://doi.org/10. the effect of dye concentration on amplification
1373/49.3.396 and DNA melting curve analysis. Anal Bio-
4. Tong SYC, Giffard PM (2012) Microbiological chem 340:24–34. https://doi.org/10.1016/
applications of high-resolution melting analy- j.ab.2005.01.046
sis. J Clin Microbiol 50:3418–3421. https:// 12. Singh P, Pfeifer Y, Mustapha A (2016) Multi-
doi.org/10.1128/JCM.01709-12 plex real-time PCR assay for the detection of
5. Liu Y, Singh P, Mustapha A (2018) High- extended-spectrum β-lactamase and carbape-
resolution melt curve PCR assay for specific nemase genes using melting curve analysis. J
detection of E. coli O157:H7 in beef. Food Microbiol Methods 124:72–78. https://doi.
Control 86:275–282. https://doi.org/10. org/10.1016/j.mimet.2016.03.014
1016/j.foodcont.2017.11.025 13. Singh P, Mustapha A (2014) Development of a
6. Singh P, Cubillos G, Kirshteyn G, Bosilevac JM real-time PCR melt curve assay for simulta-
(2020) High-resolution melting real-time neous detection of virulent and antibiotic resis-
PCR assays for detection of Escherichia coli tant Salmonella. Food Microbiol 44:6–14.
O26 and O111 strains possessing Shiga toxin https://doi.org/10.1016/j.fm.2014.04.014
genes. LWT 131:109785. https://doi.org/10. 14. Untergasser A, Cutcutache I, Koressaar T et al
1016/j.lwt.2020.109785 (2012) Primer3—new capabilities and inter-
7. Velez FJ, Bosilevac JM, Delannoy S, et al. faces. Nucleic Acids Res 40:e115–e115
(2022) Development and validation of high- 15. Jana M, Adriana V, Eva K (2020) Evaluation of
resolution melting assays for the detection of DNA extraction methods for culture-
potentially virulent strains of Escherichia coli independent real-time PCR-based detection
O103 and O121. Food Control 109095. of Listeria monocytogenes in cheese. Food Anal
https://doi.org/10.1016/j.foodcont.2022. Methods 13:667–677. https://doi.org/10.
109095 1007/s12161-019-01686-2
8. Riahi A, Kharrat M, Lariani I, Chaabouni- 16. Singh P, Liu Y, Bosilevac JM, Mustapha A
Bouhamed H (2014) High-resolution melting (2019) Detection of Shiga toxin-producing
(HRM) assay for the detection of recurrent Escherichia coli, stx1, stx2 and Salmonella by
BRCA1/BRCA2 germline mutations in Tuni- two high resolution melt curve multiplex real-
sian breast/ovarian cancer families. Familial time PCR. Food Control 96:251–259.
Cancer 13:603–609. https://doi.org/10. https://doi.org/10.1016/j.foodcont.2018.
1007/s10689-014-9740-5 09.024
52 Prashant Singh and Frank J. Velez
17. Sharma L, Watts E, Singh P (2020) High reso- 18. Velez FJ, Bosilevac JM, Singh P (2021) Valida-
lution real-time PCR melting curve assay for tion of high-resolution melting assays for the
identification of top five Penaeidae shrimp spe- detection of virulent strains of Escherichia coli
cies. LWT 133:109983. https://doi.org/10. O26 and O111 in beef and pork enrichment
1016/j.lwt.2020.109983 broths. Food Control 128:108123. https://
doi.org/10.1016/j.foodcont.2021.108123
Chapter 5
Abstract
Crop producers are under great pressure to produce more and better food items. Effective control of crop
pathogens is fundamental to guaranteeing food safety and reducing economic losses. Therefore, their
identification throughout the production chain is of utmost interest. To achieve this goal, genomic analysis
tools are currently being developed allowing to control crop production more effectively.
Genomic analysis in some samples is difficult, mostly due to the sample’s intrinsic characteristics, i.e., high
levels of phenols, fatty acids (e.g., oleaginous fruits, such as olives), and carbon hydrates (e.g., honey),
among others. Additionally, some samples yield very low DNA recovery with high content of contaminants,
imposing protocol improvements to overcome these difficulties.
Here we present protocols focused on qPCR and HRM to detect the presence of fungal pathogens
collected from plant-derived samples.
1 Introduction
Filipe Nogueira-Azevedo and Sara Barrias contributed equally with all other contributors.
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_5,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
53
54 Filipe Azevedo-Nogueira et al.
2 Materials
3 Methods
3.1 qPCR These assays must account for DNA concentration (see Notes 1–4),
primer concentration (see Notes 5–7), annealing temperature, and
3.1.1 Experimental
temperature cycle (see Note 8). Our experience shows us that good
Design of qPCR
results are in the range of 20 cycles (Ct, cycle threshold) or lower
and using 50 ng of DNA and 2 μM of primers per reaction (see
Note 8). Also, the product must be checked on gel electrophoresis
to confirm the absence of nonspecific bands and/or primer dimers.
If nonspecific bands and/or primer dimers are observable, further
optimizations are required.
To assess the specificity of the reaction, positive controls should
always be used, and the comparison of the melting peaks can rule
out unspecific amplicons.
qPCR and HRM Assay for Fungal Detection 55
3.1.2 Standard Curve The target amplicon obtained by amplification with the designed
Design primers for qPCR is cloned into a plasmid-transformed vector, and
positive colonies are selected by blue-white screening and con-
firmed by Sanger sequencing.
The standard curve method is used to compare qPCR results to
the curve, to determine copy number or concentration of the
targeted amplicon (see Note 9). The qPCR is followed by a melting
curve stage, to ensure assay specificity.
Serial dilutions of the target amplicon, with a known copy
number, are obtained, starting with the determination of the
more concentrated curve point that is based on the following
parameters:
(a) The weight of the plasmid with insert.
(b) The number of copies of the target sequence in each curve
point.
(c) The quantity of DNA used in the qPCR standard curve
reaction.
1. Calculate the mass of the plasmid with the insert (mtp). Thus,
the number of base pairs of the transformed plasmid (plasmid
length (np) and insert sequence length (nt)) must be known
and multiplied by the mean dsDNA base pair weight
(1.096 × 10-21 g) as shown in formula 1:
Table 1
qPCR mix for the standard curve
Quantity
Component Concentration (μL)a
Xpert Fast MasterMix Provided 10
Primer forward 10 μM 0.2
GCCAACAAATAAACGCCACT [7]
Primer reverse 10 μM 0.2
GACTTATTCGGTGACGTGCC [7]
Ultrapure H2O 7.6
Plasmid DNA Adjusted per standard 2
curve point
a
Per reaction
Table 2
qPCR temperature cycle
3.1.3 Quantitative PCR The samples were tested after the standard curve was designed,
Assay following the same temperature regime (Table 2) and reaction
conditions (Table 3) as used to obtain the standard curve.
To perform the qPCR assay, follow the instructions below:
1. Pipette 5 μL (50 ng) of each sample into a pre-established well
in a reaction plate. Perform triplicates of each sample, and add a
negative (non-template) control and positive controls (posing
as references) to the plate.
2. Prepare the qPCR mix according to Table 3.
3. Briefly vortex the mix (see Note 11).
4. Add 15 μL of qPCR mix to each sample.
5. Seal the reaction plate with the optical adhesive film, using the
appropriate accessory included in the MicroAmp™ plate box.
qPCR and HRM Assay for Fungal Detection 57
Table 3
qPCR mix for sample quantification
6. Spin the reaction plate and confirm that the liquid is at the
bottom of the wells.
7. Open the StepOne™ software, and set up the experiment using
the following steps:
1. Experiment Properties
– Name the experiment.
– Experiment Type: Quantitation—standard curve.
– Select Reagents: Other.
– Select Ramp Speed: Standard (~2 h to complete a run).
2. Plate Setup
– Define targets and samples.
– Name the target.
– Select “SYBRGreen” as the reporter.
– Add and name the samples.
– Select “ROX” as the passive reference.
– Assign targets and samples.
– Setup the plate layout following the order of the samples
in the optical plate.
– Assign the chosen target to all samples.
– Select task U (unknown) for every sample to be analyzed
and task N (negative) for the negative (non-template)
control.
3. Run Method
– Set the thermal cycler conditions as followed.
– Reaction Volume Per Well: 20 μL.
– Thermal profile as shown in Table 2.
58 Filipe Azevedo-Nogueira et al.
Fig. 1 Amplification of fungal samples in olive samples and comparison to a standard curve. (a) A standard
curve with an indication of the standard curve points (red) and fungi-infected olive samples (blue). (b) The
melting curve ensures reaction specificity, where olive samples infected by the target fungus have a distinct
melt peak (blue arrow) as compared to negative (black arrow). (Adapted from [7])
8. Save the experiment, load the reaction plate into the instru-
ment, and select “START RUN” on the navigation panel.
9. When the reaction is finished, save the file to a desired location.
10. Review the amplification plot: fluorescence levels must exceed
the threshold between cycles 8 and 35, and an exponential
increase in fluorescence should be observed.
11. Finally, obtain the cycle threshold (C(t)) values.
Reaction specificity was defined by melting peak analysis
(Fig. 1). Triplicates were performed, and medium C(t) was
obtained and substituted in formula 4 to obtain the copy number
of the target sequence in each sample.
Table 4
qPCR mix for HRM assay
5. Seal the reaction plate with the optical adhesive film, using the
appropriate accessory included in the MicroAmp™ plate box.
6. Spin the reaction plate and confirm that the liquid is at the
bottom of the wells.
7. Open the StepOne™ software, and set up the experiment using
the following steps:
1. Experiment Properties
– Name the experiment.
– Experiment Type: Quantitation—standard curve.
– Select Reagents: Other.
– Select Ramp Speed: Standard (~2 h to complete a run).
2. Plate Setup
– Define targets and samples.
– Name the target.
– Select “MeltDoctor” as the reporter.
– Add and name the samples.
– Select “None” on the passive reference.
– Assign targets and samples.
– Setup the plate layout following the order of the samples
in the optical plate.
– Assign the chosen target to all samples.
– Select task U for every sample to be analyzed and task N
for the negative control.
3. Run Method
– Set the thermal cycler conditions as indicated in Table 5.
– -Reaction Volume Per Well: 20 μL.
60 Filipe Azevedo-Nogueira et al.
Table 5
HRM temperature cycle
8. Save the experiment, load the reaction plate into the instru-
ment, and select “START RUN” on the navigation panel.
9. When the reaction is terminated, save the file to the desired
location.
10. Review the amplification plot: fluorescence levels must exceed
the threshold between cycles 8 and 35, and an exponential
increase in fluorescence should be observed.
11. Open the High-Resolution Melt Software v3.0 to perform
high-resolution melting analysis of the data and review the
variants.
12. Adjust the pre-melt and post-melt regions in the Derivative
Melt Curves plot to optimize the variant calls. Select all the
wells, and set the pre-melt and post-melt regions as close as
possible to the melting transition region, including the
melting peak.
13. Click Analyze, and then save the changes.
Good results allow us to discriminate variants by analysis of two
HRM plots (aligned melt curve plot and difference plot) as pre-
sented in Fig. 2.
4 Notes
Fig. 2 Representative HRM plots that distinguish two fungal species with the same set of primers. With the
same conditions of amplification, we observe that fungal species are discriminated by (a) melting tempera-
ture, where one fungal species (red) presents an amplicon with a lower melting temperature than the other
(green) and (b) where both species present distinct curve profiles that group the two variants. Therefore, this
shows that the assay can distinguish fungal samples
References
1. Bansal S, Singh A, Mangal M et al (2017) Food to olive oil and wine authenticity. Food Res Int
adulteration: sources, health risks, and detection 103:170–181
methods. Crit Rev Food Sci Nutr 57:1174– 6. Pereira L, Martins-Lopes P (2015) Vitis vinifera
1189 L. single-nucleotide polymorphism detection
2. Umesha S, Manukumar HM (2018) Advanced with high-resolution melting analysis based on
molecular diagnostic techniques for detection of the UDP-glucose:flavonoid 3- O -glucosyltrans-
food-borne pathogens: current applications and ferase gene. J Agric Food Chem 63:9165–9174
future challenges. Crit Rev Food Sci Nutr 58: 7. Azevedo-Nogueira F, Gomes S, Lino A et al
84–104 (2021) Real-time PCR assay for Colletotrichum
3. Martins-Lopes P, Gomes S, Pereira L et al acutatum sensu stricto quantification in olive
(2013) Molecular markers for food traceability. fruit samples. Food Chem 339:127858
Food Technol Biotechnol 51:198–207 8. White TJ, Bruns TD, Lee SB et al (1990) Ampli-
4. Druml B, Cichna-Markl M (2014) High resolu- fication and direct sequencing of fungal ribo-
tion melting (HRM) analysis of DNA - its role somal RNA genes for phylogenetics. In: PCR –
and potential in food analysis. Food Chem 158: protocols and applications – a laboratory man-
245–254 ual. Academic Press, pp 315–322
5. Pereira L, Gomes S, Barrias S et al (2018) Apply-
ing high-resolution melting (HRM) technology
Chapter 6
Abstract
Shiga toxin-producing Escherichia coli (STEC) is a group of human foodborne pathogens transmitted to
humans through the consumption of different types of food. Their detection is mainly performed by
targeting specific serogroups by classical microbiological methods and, later, by molecular typing with
different techniques. The application of multiplex real-time PCR (qPCR) can significantly improve the
turnaround time of the existing methodologies as in one single run it is possible to detect and characterize
specific microorganisms. In the present chapter, a pentaplex qPCR assay is described for the identification of
STEC which may also be applied for the rapid screening of these pathogens in different types of foods. The
assay targets the most important virulence factors of these microorganisms, the genes stx1, stx2, and eae,
along with the rfbE gene which encodes for the “O157” antigen as this is the most prevalent serogroup
among all STEC, as well as an internal amplification control to rule out false-negative results due to qPCR
inhibition.
Key words qPCR, Multiplex, Shiga toxin-producing E. coli, Internal amplification control
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_6,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
63
64 Ana Costa-Ribeiro et al.
2 Materials
2.1 Media (See Note 1. Nutrient broth: 10.0 g tryptone, 5.0 g meat extract, 5.0 NaCl,
1) 1000 mL Milli-Q water, pH 7.2 ± 0.2.
2. Tryptic soy agar: 15.0 g tryptone, 5.0 g papainic digest of
soybean meal, 5.0 g NaCl, 15.0 g bacteriological agar,
1000 mL Milli-Q water, pH 7.3 ± 0.2.
2.3 Equipment 1. Real-time PCR thermocycler with at least five different chan-
nels for the detection of different fluorophores. In the
described assay, a QuantStudio™ 5 (Applied Biosystems™,
Foster City, CA, USA) was used. Reactions may be run in
0.1 mL PCR tubes/strips or 96-well plates sealed with optical
quality adhesive film or plastic lids.
2. Centrifuge with a speed range of 300–16,000 × g. Also must
have a rotor suitable for 1.5–2 mL tubes.
3. Thermomixer comfort (Eppendorf AG, Germany). Similar
devices may be used to incubate and shake simultaneously.
4. Qubit™ 4 fluorometer (Invitrogen™, Carlsbad, CA, USA).
Suitable 0.5 mL tubes must also be acquired.
STEC Detection by Multiplex qPCR 65
Table 1
Multiplex STEC qPCR primers and probes
3 Methods
3.1 Bacterial 1. Pick one well isolated colony from a TSA plate and resuspend it
Enrichment in 1–4 mL of nutrient broth. Incubate at 37 ± 1 °C overnight.
2. After incubation, take an aliquot (1–2 mL) for DNA extraction.
3.3 DNA 1. Remove Qubit™ reagents from the refrigerator and allow
Quantification them to reach room temperature. It is recommended to per-
form the assay at room temperature (18–28 °C). Temperature
fluctuations may influence the accuracy of the assay.
2. Prepare the standards and sample reactions. For standards
transfer 190 μL of Qubit™ 1× dsDNA HS working solution
to two Qubit™ 0.5 mL compatible tubes (label the tubes only
in the cap and not in the walls as it can interfere with the
reading of the sample). Add 10 μL of Standard #1 (0 ng/ μL)
in one tube, and 10 μL of Standard #2 (10 ng/μL) in another
one. For the samples, 1–20 μL may be measured, and the
working solution will be added to make a final volume of
200 μL. The amount of sample used depends on the sample
volume available and the DNA concentration of the sample. In
this protocol, 1 μL is used.
3. Mix the standards and samples vigorously using a vortex for 5 s
and incubate in the dark for at least 2 min. Reactions are stable
for 3 h in the dark.
4. Read the standards in the equipment to perform the calibra-
tion, and then proceed with the samples indicating in the
equipment the volume of sample used in the reaction. This is
necessary for the equipment to calculate the DNA concentra-
tion in the original sample. In the case the equipment indicates
“concentration too high,” perform a 1/ 10 dilution of the
original sample, and repeat the process from step 2 of Sub-
heading 3.3. In the case the equipment indicates “concentra-
tion too low,” increase the volume of the sample in step 2 of
Subheading 3.3. If the problem persists, perform again the
DNA isolation.
3.4 STEC Multiplex 1. The assay described herein targets three different STEC viru-
qPCR Assay lence markers, namely, stx1, stx2, and eae, along with the gene
rfbE which encodes for the “O157” antigen (this serogroup is
the most commonly reported STEC in most countries world-
wide) and an IAC. The sequences of all the primers and probes
are provided in Tables 1 and 2. See Notes 6 and 7.
2. Perform tenfold serial dilutions from the original DNA extract
in TE 1×, e.g., 10 μL of DNA and 90 μL of TE 1×.
3. Considering the number of dilutions to be tested, prepare
enough reaction mixture to test each one in technical dupli-
cates (ideally triplicates), along with negative control (Milli-Q
water) and at least one extra reaction having some excess vol-
ume. In Table 3 the volumes required for one reaction are
presented.
4. To perform a pentaplex assay, a qPCR thermocycler with five
filters, or channels, is needed. The thermal profile run is sum-
marized in Table 3.
STEC Detection by Multiplex qPCR 67
Table 2
Reagent concentrations and volumes for one STEC multiplex qPCR assay
Final
Primer/probe Volume/rxn (μL) concentration (nM)
NZYSupreme 12.5 –
stx1-P3F 0.1 400
stx1-P3R 0.1 400
stx1-P3P 0.05 200
stx2-P3F 0.1 400
stx2-P3R 0.1 400
stx2-P3P 0.05 200
eae-P3F 0.1 400
eae-P3R 0.1 400
eae-P3P 0.05 200
O157-rfbE-F 0.1 400
O157-rfbE-R 0.1 400
O157-rfbE-P 0.05 200
IAC F 0.2 100
IAC R 0.2 100
IAC P 0.2 100
IAC DNA 1.0 7 × 102a
H2O 1.0 –
Template DNA 3b –
Final volume 25 –
Primer volumes indicated are calculated based on a working solution of 100 μM except
for the IAC in which 10 μM stocks were used. Final concentrations may vary depending
on specific Master Mixes
a
Copies/μL, the volume may vary depending on the DNA stock concentration
b
To reduce pipetting errors, avoid pipetting volumes below 2 μL
Table 3
Thermal profile for multiplex qPCR
Fig. 1 (a) stx1 amplification plot, (b) stx2 amplification plot, (c) eae amplification plot, (d) rfbE amplification
plot, (e) IAC amplification plot, (f) simultaneous multiplex amplification
Table 4
qPCR result interpretation
stx1/ stx2 eae rfbE IACa
Gene
Positive Negative Positive Negative Positive Negative Positive Negative
a
Certain samples with a high concentration of target DNA may result in no amplification of the IAC. “P,” pathogenic.
“I,” inconclusive, inhibition in the IAC, the test must be repeated
Fig. 2 (a) Dynamic range of stx1, (b) dynamic range of stx2, (c) dynamic range of eae, (d) dynamic range of
rfbE, (e) standard curves generated from the dynamic range, used to calculate the multiplex qPCR amplifica-
tion efficiency
4 Notes
1. If desired to apply the assay for food analysis, the media speci-
fied in the ISO 13136 may be used [2]; these are modified TSB
supplemented with novobiocin (mTSBn): 17 g enzymatic
digest of casein, 3 g enzymatic digest of soy, 2.5 g D(+)
70 Ana Costa-Ribeiro et al.
6. Different types of IAC may be designed and used [13, 14], and
commercial products are also available. These may be identified
as internal positive control (IPC). In the current chapter, the
noncompetitive IAC described by Garrido-Maestu et al. was
selected [15, 16]. Any chimeric DNA accommodating the
primers and probe described herein will be suitable; thus, no
particular DNA sequence was provided. The IAC included in
this protocol is expected to provide a Cq of 30–33 in
non-inhibited negative samples; if inhibition is observed, this
can typically be overcome by performing a ½–1/10 dilution of
the DNA in Milli-Q water of TE 1×.
7. Primer and probe design may be performed with different
software. Primer3 is an online free tool [17] where this task
can be performed automatically or manually specified attend-
ing to specific requirements such as length, GC %, or Tm
among others with particular caution when designing hydroly-
sis probes to avoid placing a G in the 5′ end, where the
fluorophore is attached, due to the high natural quenching
capacity of this nucleotide [18, 19]. As input for the design,
the sequence of the desired gene may be used, e.g., obtained
from NCBI’s Refseq, or several may be retrieved from the same
database and aligned and the consensus sequence of a con-
served region may be used; free tools to perform this visualiza-
tion and alignment are also available, for instance, CLC
Sequence Viewer [20].
The inclusivity and exclusivity of newly designed primers
should be tested in silico prior to their acquisition and later by
in vitro testing. This task may be initially performed by a
BLAST search (https://blast.ncbi.nlm.nih.gov/Blast.cgi) and
later through an in silico PCR [21]. For in vitro studies, the
optimized assay must be used (see Notes below) against a panel
of target and nontarget strains [22].
The optimal primers and probes concentration must be
optimized for optimal performance. The optimization typically
begins with the primers using an intercalating dye such as SYBR
Green I and testing different relative concentrations of the
forward and reverse primer in the range of 100–500 nM.
Once determined the optimal primer concentration, one may
perform the same step for the probe where concentrations in
the range 100–300 nM typically lead to good results.
8. Attention must be paid to the selection of fluorophores. The
ones specified in Table 1 are a suitable combination that works
well in the qPCR instrument that was used. It is advised to
confirm that the fluorophores selected do not have overlapping
emission peaks. Oligonucleotide suppliers may have alternative
dyes and quenchers equivalent to the ones indicated in Table 1
which may be more economic (e.g., Cy®5 for TYE665 or
72 Ana Costa-Ribeiro et al.
References
1. Kim SO, Kim SS (2021) Bacterial pathogen 3. Feng P, Weagant SD, Jinneman K (2011) Diar-
detection by conventional culture-based and rheagenic Escherichia coli. http://www.fda.
recent alternative (Polymerase Chain Reaction, gov/Food/ScienceResearch/
isothermal amplification, Enzyme Linked LaboratoryMethods/Bacteriolo
Immunosorbent Assay, bacteriophage amplifi- gicalAnalyticalManualBAM/ucm070080.htm
cation, and gold nanoparticle aggregation) 4. Garrido-Maestu A, Chapela M-J, Peñaranda E
methods in food samp. J Food Saf 41:1–12 et al (2015) Re-evaluation of enhanced qPCR
2. ISO/TS (2012) Microbiology of food and ani- prevalidated method for next-day detection of
mal feed – Real-time polymerase chain reaction Salmonella spp., Shigella spp., Escherichia coli
(PCR)-based method for the detection of O157 and Listeria monocytogenes. Food Bio-
food-borne pathogens – horizontal method technol 29:317–335
for the detection of Shiga toxin-producing 5. Bundidamorn D, Supawasit W, Trevanich S
Escherichia coli (STEC) and the determination (2021) Taqman® probe based multiplex
of O157
STEC Detection by Multiplex qPCR 73
RT-PCR for simultaneous detection of Listeria controls for diagnostic PCR assays. J Clin
monocytogenes, Salmonella spp. and Shiga Microbiol 42:1863–1868
toxin-producing Escherichia coli in foods. 15. Garrido-Maestu A, Azinheiro S, Carvalho J
LWT 147:111696 et al (2019) Combination of immunomagnetic
6. Villamizar-Rodrı́guez G, Lombó F (2017) separation and real-time recombinase polymer-
Multiplex Detection of Food-Borne ase amplification (IMS-qRPA) for specific
Pathogens. In: Domingues L (ed) PCR: meth- detection of Listeria monocytogenes in smoked
ods and protocols. Springer New York, salmon samples. J Food Sci 84(7):1881–1887
New York, pp 153–162 16. Garrido-Maestu A, Azinheiro S, Carvalho J
7. Dhital R (2021) Detection of virulence and et al (2018) Development and evaluation of
extended spectrum β-lactamase genes in Salmo- loop-mediated isothermal amplification, and
nella by multiplex high-resolution melt curve Recombinase Polymerase Amplification meth-
real-time PCR assay. J Appl Microbiol 132(3): odologies, for the detection of Listeria mono-
2355–2367 cytogenes in ready-to-eat food samples. Food
8. Müştak İB, Müştak HK (2022) Detection and Control 86:27–34
differentiation of Salmonella Enteritidis and 17. Untergasser A, Cutcutache I, Koressaar T et al
Salmonella Typhimurium by multiplex quanti- (2012) Primer3-new capabilities and inter-
tative PCR from different poultry matrices. Br faces. Nucleic Acids Res 40:e115–e115
Poult Sci 63:171–178 18. Mackay IM (2004) Real-time PCR in the
9. Bundidamorn D, Supawasit W, Trevanich S microbiology laboratory. Clin Microbiol Infect
(2018) A new single-tube platform of melting 10:190–212
temperature curve analysis based on multiplex 19. Biassoni R, Raso A (eds) (2020) Quantitative
real-time PCR using EvaGreen for simulta- real-time PCR methods and protocols.
neous screening detection of Shiga toxin- Humana, New York, NY
producing Escherichia coli, Salmonella spp. 20. CLC Bio-Qiagen (2016) CLC Sequence
and Listeria monocytogenes in food. Food Con- Viewer
trol 94:195–204
21. Bikandi J, San Millán R, Rementeria A et al
10. Vitullo M, Grant KA, Sammarco ML et al (2004) In silico analysis of complete bacterial
(2013) Real-time PCRs assay for serogrouping genomes: PCR, AFLP–PCR and endonuclease
Listeria monocytogenes and differentiation from restriction. Bioinformatics 20:798–799
other Listeria spp. Mol Cell Probes 27:68–70
22. Kralik P, Ricchi M (2017) A basic guide to real
11. Raymaekers M, Smets R, Maes B et al (2009) time PCR in microbial diagnostics: definitions,
Checklist for optimization and validation of parameters, and everything. Front Microbiol 8:
real-time PCR assays. J Clin Lab Anal 23: 1–9
145–151
23. Costa-Ribeiro A, Azinheiro S, Fernandes SP,
12. Bustin SA, Benes V, Garson JA et al (2009) The Lamas A, Prado M, Salonen LM, Garrido-
MIQE guidelines: minimum information for Maestu A (2023) Evaluation of covalent
publication of quantitative real-time PCR organic frameworks for the low-cost, rapid
experiments. Clin Chem 55:611–622 detection of Shiga Toxin-producing Escheri-
13. Deer DM, Lampel KA, González-Escalona N chia coli in ready-to-eat salads. Anal Chim
(2010) A versatile internal control for use as Acta 341357. https://doi.org/10.1016/j.aca.
DNA in real-time PCR and as RNA in real-time 2023.341357
reverse transcription PCR assays. Lett Appl 24. Garrido-Maestu A, Azinheiro S, Carvalho J
Microbiol 50:366–372 et al (2020) Optimized sample treatment,
14. Hoorfar J, Malorny B, Abdulmawjood A et al combined with real-time PCR, for same-day
(2004) Practical considerations in design of detection of E. coli O157 in ground beef and
internal amplification controls for diagnostic leafy greens. Food Control 108:106790
PCR assays MINIREVIEW practical considera-
tions in design of internal amplification
Chapter 7
Abstract
Cocoa (Theobroma cacao L.) is an international commodity used as an ingredient in the manufacturing of
chocolate making its authentication a key issue in the cocoa chain. Various molecular techniques have been
increasingly applied for quality requirements. These issues highlight the need for techniques that allow the
extraction and detection of cocoa DNA from highly processed cocoa products and chocolate. The applica-
bility of real-time PCR to highly processed cocoa-derived products for authentication purposes depends on
the possibility of extracting high-quality and amplifiable DNA and further developing efficient PCR tests.
This methodology herein describes the use of a classical CTAB method providing DNA suitable for
TaqMan real-time PCR amplification. Real-time PCR is a simple and fast method, with a high potential
application in a wide range of food products. The main features of this technique are focused on two DNA
targets, one located in the nuclear genome (vicilin-li PCR test) and a second one based on chloroplast DNA
(lipids PCR test), which successfully passed the performance criteria considering the specificity, sensitivity,
efficiency of amplification, robustness, and applicability in processed cocoa-derived products and chocolate.
Key words Chloroplast DNA, Chocolate, DNA extraction, Nuclear DNA, Theobroma cacao, qPCR
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_7,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
75
76 Ana Caroline De Oliveira et al.
2 Materials
2.1 General Supplies 1. Sterile DNase-free microtubes of 1.5 mL and 2.0 mL.
2. Micropipettes of variable volume and their respective tips
(0.1–2.5 μL; 0.5–10 μL; 10–100 μL; 100–1000 μL).
3. Water purification system (Milli-Q, Millipore).
4. Sterile nuclease-free ultrapure water.
Cocoa-Derived Products Authentication 77
3 Methods
3.1 DNA Extraction 1. DNA is extracted using a CTAB-based protocol [5]. Prepare
CTAB solutions, use within 3 months, and store capped in the
refrigerator at 4 °C.
2. For the DNA extraction from the cocoa-derived and chocolate
samples, prepare the samples as described (see Note 1):
For raw cocoa beans:
(a) Freeze dry the sample material for 24 h.
(b) Grind sample material with a mortar and pestle, and weigh
200 mg of it into a 2.0 mL microtube.
78 Ana Caroline De Oliveira et al.
Table 1
Primers and probes used for cocoa detection in processed cocoa-derived products
Oligonucleotide Amplicon
Target NCBI acc. no. designation Sequence (5′-3′) size (bp)
Lipids XM007027212 Forward primer TCCCTTATTCCCA 86
(chloroplast ATTTTCTCCTT
target) Reverse primer AGATTGGGAGAG
TTCGGTTTCTAA
Probe (FAM-TAMRA CCAAGAAACAAAA
labeled) CCCGAAACGCCA
Vicilin-like XM018124728 Forward primer CCCAAACTACTAA 77
seed storage AGCCCTCATCT
protein Reverse primer GCTTTGCCCATGC
(nuclear TTTCG
target) Probe (FAM-TAMRA CTTCCCTCCCCT
labeled) CACCTCAAAAACTCAA
3.2 TaqMan Real- The reaction components are assembled in a final volume of 25 μL
Time PCR as described below:
1. Keep all reaction components on ice (see Note 5).
2. Mix and prepare master mix in 1.5 mL microcentrifuge tubes,
following the proportions indicated in Table 2. Prepare the
reaction mix for each target separately. The final volume of
the master mix depends on the number of reactions required
for each real-time PCR (see Note 6).
80 Ana Caroline De Oliveira et al.
Table 2
Components of real-time PCR
3.3 Thermocycling 1. Incubate the 96-well plate containing the DNA and the master
mix in a Real-Time PCR System.
2. Run the program with the standard or fast cycling:
(a) Standard cycling: 10 min at 95 °C (initial denaturation)
and 40 cycles of 15 s at 95 °C (denaturation) and 1 min at
60 °C (annealing and extension).
(b) Fast cycling: 20 s at 95 °C (initial denaturation) and
40 cycles of 3 s at 95 °C (denaturation) and 30 s at 60 °
C (annealing and extension).
Cocoa-Derived Products Authentication 81
3.4 Program and Run 1. Operate the Real-Time PCR System and software according to
the manufacturer’s recommendations. Define the experiment
properties including the set reaction volume to 25 μL and the
amplification program. Use FAM-TAMRA as a detector/
quencher.
2. Briefly, launch the software, and open a new plate template
window to denote well locations on the 96-well plate for the
controls and experimental samples. Define the plate wells with
sample names and targets. Save the template window with the
recorded data as a run file.
3. Load the 96-well plate in the instrument. Select the “Start
Run” button to begin the actual run and determine Cq (cycle
threshold) values for each sample.
3.5 Data Collection 1. After the run is completed, select baseline (see Note 10), and
and Analysis place the threshold line at the exponential phase of amplifica-
tion (see Note 11). Remove the plate from the instrument and
save the results of the run. When the analysis button is selected
in the software, the results will be analyzed automatically if the
standards and testing sample information were recorded as
noted in the previous section. The obtained Cq values should
be thoroughly reviewed, and interpretation of the results must
be made taking into account the acquired data in the internal
validation performed in each laboratory for a given detection
limit (see Note 12).
4 Notes
References
Abstract
Food allergy is an increasing challenge to public health, with widespread global distribution. With no cure
for this pathology, the food-allergic individuals are forced to adopt food eviction measurements, relying on
label information to avoid consuming the offending foods. To safeguard these individuals, the analytical
methods based on real-time PCR approaches are currently faced as excellent tools to verify labeling
compliance, aiding industry and regulatory agencies to efficiently manage food allergen control programs.
Therefore, this chapter intends to describe a protocol of real-time PCR to analyze allergenic food species.
For method development, the main steps to be considered are (i) in silico sequence analysis and primer/
hydrolysis probe design, (ii) preparation of calibrators (model foods containing the allergenic ingredient),
(iii) efficient DNA extraction from complex food matrices, (iv) amplification by real-time PCR with
hydrolysis probe (90–200 bp) targeting a highly specific DNA region (allergen-encoding gene),
(v) sequencing PCR products for identity confirmation, and (vi) validation and application to commercial
foods. Herein, a real-time PCR approach for the detection and quantification of cashew nut as an allergenic
food is described as an example protocol, including all the steps for method development and validation.
Key words Food allergen, Real-time PCR, Detection, Quantification, Allergen-encoding genes
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_8,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
85
86 Joana Costa et al.
Fig. 1 Schematic representation of a real-time PCR assay targeting Ana o 3 gene from cashew nut
2 Materials
3 Methods
3.1 In Silico Analysis 1. Consult the NCBI database, and select regions (gene or mRNA
sequences) for the potential discrimination of the target species
(Anacardium occidentale) (see Note 8). Look for local simila-
rities with orthologue genes/sequences using the BLASTn
algorithm (see Note 9).
2. Design primers and hydrolysis probe, either manually or using
primer designing tools, such as Primer-BLAST (https://www.
ncbi.nlm.nih.gov/tools/primer-blast/) (see Note 10). Check
the properties of primers and hydrolysis probe (physicochemi-
cal parameters, absence of hairpins, 3′-complementary and self-
annealing) using specific algorithms (e.g., OligoCalc: Oligonu-
cleotide Properties Calculator, http://biotools.nubic.north
western.edu/OligoCalc.html). Verify the specificity of the
designed primers and hydrolysis probe with target and nontar-
get sequences using the software Primer-BLAST (see Note 11).
3. Order primers (Ana 3-F, AGGTACGTGAAGCAGGAGGT
CCA and Ana3-R, GCTGCAGCTGCCTCACCATTTG) and
hydrolysis probe (Ana3-P, FAM-AAAGCTTGAGGGAATGC
TGCCAGGAGTT -BHQ1) [11] synthesis in specialized
outsourced facilities (e.g., Eurofins Genomics, Ebersberg,
Germany), which might take up to 1 week, depending on the
selected production facility.
Real-Time PCR for Allergenic Food Detection 91
3.2 Preparation of Although some reference materials have been proposed by the
Model Foods Standard Reference Materials Program of the National Institute
of Standards and Technology (NIST, Gaithersburg, USA) for
food allergen analysis, they are still in a development phase, and
none of them can mimic real foods. Therefore, for method devel-
opment, the preparation of reference/model foods is a crucial step.
Herein, it is provided an example of a model food based on biscuits
containing known amounts of cashew nut [11]:
1. Prepare a dough matrix using the following ingredients and
recipe, namely, 225 g of wheat flour (Triticum aestivum),
100 g of sugar, 90 g of butter, and 50 mL of milk.
2. Ground cashew nut into a fine powder of approximately
0.3 mM of diameter in a laboratory knife mill.
3. Prepare the model reference foods with concentrations
of 100,000 mg/kg, 50,000 mg/kg, 10,000 mg/kg,
5000 mg/kg, 1000 mg/kg, 500 mg/kg, 100 mg/kg,
50 mg/kg, 10 mg/kg, 5 mg/kg, 1 mg/kg, (w/w) of cashew
nut in the dough. Add 30 g of cashew nut to 270 g of dough to
prepare the first model mixture of 10% (100,000 mg/kg).
Prepare the following concentrations by serial additions of
dough until the level of 0.0001% (1 mg/kg).
4. Divide each mixture into two portions. Immediately store one
portion at -20 °C to serve as raw model foods, while the
second portion should be oven-baked at 180 °C for 20 min
to simulate the production of biscuits. After cooling, ground
the biscuits to a fine powder of approximately 0.3 mM of
diameter with a laboratory knife mill, and store at -20 °C
until DNA extraction.
3.3 DNA Extraction The selection of an appropriate protocol to extract DNA from
foods with high purity and yield is a critical step for the successful
development of real-time PCR methods. Firstly, the DNA extrac-
tion protocol should be selected according to the composition of
the food(s) to be analyzed. This might require testing several DNA
extraction protocols. The NucleoSpin Food Kit (Macherey-Nagel,
Düren, Germany) has proved to be appropriate for most food
matrices, thus being the recommended protocol for the detection
of allergenic species. Herein, it is described its protocol according
to manufacturer’s instructions with minor alterations (see Note 5):
1. Weigh approximately 200 mg of material (raw dough or
grounded (model) food) in a 2.0 mL sterile reaction tube.
Add 550 μL of CF buffer (preheated at 65 °C) to each tube
and 10 μL of proteinase K (10 mg/mL) solution, vortex vigor-
ously, and incubate for 1 h at 65 °C in the thermomixer
(~1000 rpm). Vortex occasionally (every 15–20-min interval)
during incubation.
92 Joana Costa et al.
3.5 Qualitative PCR 1. Prepare the PCR mix for a total volume of 25 μL by mixing all
the components needed for the specified reaction. Add
PCR-grade water (adjust the volume considering the total
amount of remaining reagents), 2.5 μL of buffer 10×, 2.0 μL
of 10 mM of dNTP, 3.0 μL MgCl2 (3.0 mM), 7.0 μL of each
primer (Ana3-F/Ana3-R) (280 nM), and 0.2 μL of SuperHot
Taq DNA polymerase (1.0 U).
2. Add 23 μL of the reaction mix and 2 μL of DNA extract
(100–200 ng) in each 0.2 mL reaction tube. Include a positive
(DNA from target species) and a negative (no DNA template)
control in the PCR amplification.
3. Set the temperature program in the thermal cycler, which was
previously optimized [11]: initial denaturation at 95 °C for
5 min; 40 cycles at 95 °C for 30 s, 65 °C (Ana3-F/ Ana3-R)
for 30 s, and 72 °C for 30 s; and a final extension at 72 °C for
5 min.
4. Prepare a 1.5% agarose gel stained with GelRed 1× (Biotium
Inc., Hayward, CA, USA) to visualize the amplicons. Mix
20 μL of the PCR product with 4 μL of loading buffer, apply
to gel wells, and run electrophoresis using SGTB 1× (Grisp,
Porto, Portugal) for 25–30 min at 200 V. For each gel, use a
DNA marker (e.g., DNA 100 bp marker, Bioron GmbH,
Römerberg, Germany) according to the size of expected PCR
products. Add 4 μL of loading buffer to unstained DNA mar-
kers and load to each gel.
5. Visualize the agarose gel with a UV light tray and the Gel Doc
EZ Imager using the GelRed dye protocol. Register a digital
image with Image Lab software version 5.2.1 and analyze the
results.
3.6 DNA Sequencing It is highly recommended to confirm the size and sequence of the
amplified PCR products, especially if primers are previously
designed in mRNA sequences [10] (see Note 8). Therefore, the
amplicons should be analyzed by Sanger sequencing that can be
easily outsourced. Prior to sequencing, it is recommended a clean-
ing step of PCR products, which can be performed according to the
following steps:
1. Go to Subheading 3.5 “Qualitative PCR,” and execute the
described steps to obtain the PCR products from the template
species (e.g., cashew nut, pistachio nut) (see Note 15).
2. Use a PCR Purification Kit to remove the interferents that are
present in the amplified PCR products (e.g., remaining com-
ponents of amplification reaction like primers, dNTP, enzyme).
Perform amplicon purification according to manufacturers’
instructions (see Note 16).
94 Joana Costa et al.
3. Place the purified PCR products into two sterile reaction tubes,
and add primer-F or primer-R (e.g., Ana3-F or Ana3-R) to the
respective tube (this will ensure sequencing both strands in the
opposite directions of each target amplicon). Tag each tube,
and send them to a specialized research facility (Eurofins Geno-
mics, Ebersberg, Germany) for direct Sanger sequencing (see
Note 17).
4. Verify the quality of the electropherograms using an appropri-
ate software (e.g., FinchTV 1.4 software, Geospiza Inc.,
https://digitalworldbiology.com/FinchTV). Analyze the
PCR electropherograms and align the high-resolution/quality
product sequences with the consensus sequence. For sequence
alignment, use a sequence alignment tool/software like BIOE-
DIT v7.2 (Biological Sequence Alignment Editor, https://
bioedit.software.informer.com/versions/) or MEGA (Molec-
ular Evolutionar y Genetics Analysis, https://www.
megasoftware.net/) (see Note 18).
5. Critically analyze the sequencing data, assessing the size and
nucleotide identity, and compare it with the consensus
sequence.
3.7 Real-Time PCR When performing a real-time PCR run, the components of
with Hydrolysis Probe the reaction mixture, namely, the amounts of primers/probe and
template DNA, should be previously adjusted, as well as the tem-
perature program that should be appropriate for each primer/
probe set (see Note 19). The following steps were previously
evaluated and defined when developing a real-time PCR approach
for the detection of Ana o 3-encoding gene [11] using the CFX96
Real-Time PCR System and respective software (Subheading 2.3)
(see Note 20):
1. Open the startup wizard of Bio-Rad CFX Manager 3.1, and
define the program of temperatures (protocol total duration
<100 min): initial step at 95 °C for 5 min, 50 cycles at 95 °C
for 15 s and 65 °C for 45 s, with the collection of fluorescence
signal at the end of each cycle (Fig. 2a) (see Notes 21 and 22).
2. Prepare the plate, by selecting the correct fluorophore (FAM),
the plate type (white for white strips/plate), and the unit
(define “percent” if using model foods). Set the number of
samples and respective replicates (three to four replicates per
sample by run are recommended), and define their place in the
plate (Fig. 2b) (see Note 23). Define the standards and provide
correct concentrations of the target species. Unknown samples
or blind mixtures should be defined as “Unknown.” A negative
control (no DNA template) per gene target should be added to
each run.
Real-Time PCR for Allergenic Food Detection 95
Fig. 2 Real-time PCR temperature program for the amplification of cashew nut targeting the Ana o 3-encoding
gene (a); example of a real-time PCR plate defining the calibrators as standards and the blind samples as
unknown samples (negative control included) (b)
96 Joana Costa et al.
3.8 Real-Time PCR 1. Perform the real-time PCR assays using the same conditions as
Validation described in Subheading 3.7, running the calibrators (model
mixtures of biscuits containing known amounts of cashew nut)
to construct the calibration curve, along with blind samples
(as theoretical unknown samples). Use three to four replicates
per blind sample and per standard.
2. Perform all the calculations necessary to determine the LOD,
LOQ, dynamic range, precision (coefficient of variation
(CV) or relative repeatability standard deviation (RSDr)), true-
ness (bias), and robustness of the method, as well as other
important parameters [22, 23] (see Note 26).
Real-Time PCR for Allergenic Food Detection 97
Fig. 3 Real-time amplification curves (a) and respective calibration curve (b) targeting Ana o 3 gene using
reference model mixtures of biscuits containing known amounts of cashew nut (100,000 to 10 mg/kg) (n = 4
replicates per run)
4 Notes
Funding
This work was funded by national funds (FCT, Fundação para a
Ciência e Tecnologia) through project Hypoallergen (PTDC/
BAA-AGR/4005/2021) and the strategic funding of UIDB/
50006/2020|UIDP/50006/2020 from FCT/MCTES and the
European Union (EU) through European Regional Development
Fund (FEDER funds through NORTE-01-0145-FEDER-
000052). This work was also supported by project SYSTEMIC
which received funding from national research funding parties in
a joint action of JPI HDHL, JPI-OCEANS, and FACCE-JPI
launched in 2019 under the ERA-NET ERA-HDHL (n°
696295). JC and IM thank FCT for funding through the Individ-
ual Call to Scientific Employment Stimulus (2021.03583.CEE-
CIND/CP1662/CT0012 and 2021.03670.CEECIND/
CP1662/CT0011, respectively).
References
1. Michelet M, Balbino B, Guilleminault L, Reber 4. Holzhauser T (2018) Protein or no protein?
LL (2021) IgE in the pathophysiology and Opportunities for DNA-based detection of
therapy of food allergy. Eur J Immunol 51(3): allergenic foods. J Agric Food Chem 66(38):
531–543. https://doi.org/10.1002/eji. 9889–9894. https://doi.org/10.1021/acs.
202048833 jafc.8b03657
2. Sicherer SH, Sampson HA (2018) Food 5. Costa J, Fernandes TJR, Villa C, Oliveira
allergy: a review and update on epidemiology, MBPP, Mafra I (2017) Advances in food aller-
pathogenesis, diagnosis, prevention, and man- gen analysis. In: Spizzirri G, Cirillo G (eds)
agement. J Allergy Clin Immunol 141(1): Food safety: innovative analytical tools for
41–58. https://doi.org/10.1016/j.jaci.2017. safety assessment. Wiley, Hoboken, NJ, pp
11.003 305–360
3. Regulation (EU) No 1169/2011 of 6. Holzhauser T, Johnson P, Hindley JP,
25 October 2011 on the provision of food O’Connor G, Chan C-H, Costa J et al (2020)
information to consumers, amending Regula- Are current analytical methods suitable to ver-
tions (EC) No 1924/2006 and (EC) No ify VITAL® 2.0/3.0 allergen reference doses
1925/2006 of the European Parliament and for EU allergens in foods? Food Chem Toxicol
of the Council, and repealing Commission 145:111709. https://doi.org/10.1016/j.fct.
Directive 87/250/EEC, Council Directive 2020.111709
90/496/EEC, Commission Directive 1999/ 7. Costa J, Amaral JS, Grazina L, Oliveira MBPP,
10/EC, Directive 2000/13/EC of the Mafra I (2017) Matrix-normalised real-time
European Parliament and of the Council, PCR approach to quantify soybean as a poten-
Commission Directives 2002/67/EC and tial food allergen as affected by thermal proces-
2008/5/EC and Commission Regulation sing. Food Chem 221:1843–1850. https://
(EC) No 608/2004., L304 doi.org/10.1016/j.foodchem.2016.10.091
Real-Time PCR for Allergenic Food Detection 103
8. Costa J, Mafra I, Kuchta T, Oliveira MBPP the novel single-tube nested real-time PCR for
(2012) Single-tube nested real-time PCR as a the detection of walnut allergens in sponge
new highly sensitive approach to trace hazel- cakes. Food Res Int 54(2):1722–1729.
nut. J Agric Food Chem 60(33):8103–8110. https://doi.org/10.1016/j.foodres.2013.
https://doi.org/10.1021/jf302898z 09.047
9. Costa J, Oliveira MBPP, Mafra I (2013) Novel 18. Köppel R, Velsen-Zimmerli F, Bucher T
approach based on single-tube nested real-time (2012) Two quantitative hexaplex real-time
PCR to detect almond allergens in foods. Food PCR systems for the detection and quantifica-
Res Int 51(1):228–235. https://doi.org/10. tion of DNA from twelve allergens in food. Eur
1016/j.foodres.2012.12.006 Food Res Technol 235(5):843–852. https://
10. Costa J, Silva I, Villa C, Mafra I (2023) A novel doi.org/10.1007/s00217-012-1806-8
single-tube nested real-time PCR method to 19. Waiblinger H-U, Boernsen B, Geppert C,
quantify pistachio nut as an allergenic food: Demmel A, Peterseil V, Koeppel R (2017)
influence of food matrix. J Food Compost Ring trial validation of single and multiplex
Anal 115:105042. https://doi.org/10.1016/ real-time PCR methods for the detection and
j.jfca.2022.105042 quantification of the allergenic food ingredi-
11. Costa J, Villa C, Grazina L, Mafra I (2022) ents mustard, celery, soy, wheat and rye. J Con-
Single-tube nested real-time PCR versus nor- sum Prot Food Saf 12(1):55–72. https://doi.
malised real-time PCR for the quantification of org/10.1007/s00003-016-1063-z
allergenic cashew nut in foods: impact of ther- 20. Waiblinger H-U, Boernsen B, N€aumann G,
mal processing and matrix. Food Chem 397: Koeppel R (2014) Ring trial validation of single
1 3 3 7 7 8 . h t t p s :// d o i . o r g / 1 0 . 1 0 1 6 / j . and multiplex real-time PCR methods for the
foodchem.2022.133778 detection and quantification of the allergenic
12. Fernandes TJR, Costa J, Oliveira MBPP, Mafra food ingredients sesame, almond, lupine and
I (2018) Exploiting 16S rRNA gene for the Brazil nut. J Verbr Lebensm 9(3):297–310.
detection and quantification of fish as a poten- https://doi.org/10.1007/s00003-014-
tial allergenic food: a comparison of two real- 0868-x
time PCR approaches. Food Chem 245:1034– 21. Waiblinger H-U, Geppert C, Bartsch D,
1041. https://doi.org/10.1016/j.foodchem. Neumann K, Hochegger R, Peterseil V et al
2017.11.068 (2022) Collaborative trial validation of a new
13. Fernandes TJR, Costa J, Oliveira MBPP, Mafra multiplex real-time PCR to sensitively detect
I (2018) A new real-time PCR quantitative allergenic nuts in food. J Consum Prot Food
approach for the detection of shrimp crusta- Saf 17(3):265–277. https://doi.org/10.
ceans as potential allergens. J Food Compost 1007/s00003-022-01385-x
Anal 72:7–14. https://doi.org/10.1016/j. 22. Bustin SA, Benes V, Garson JA, Hellemans J,
jfca.2018.05.012 Huggett J, Kubista M et al (2009) The MIQE
14. Villa C, Costa J, Gondar C, Oliveira MBPP, guidelines: minimum information for publica-
Mafra I (2018) Effect of food matrix and ther- tion of quantitative real-time PCR experi-
mal processing on the performance of a nor- ments. Clin Chem 55(4):611–622. https://
malised quantitative real-time PCR approach doi.org/10.1373/clinchem.2008.112797
for lupine (Lupinus albus) detection as a poten- 23. ENGL (2015) Definition of minimum perfor-
tial allergenic food. Food Chem 262:251–259. mance requirements for analytical methods of
https://doi.org/10.1016/j.foodchem.2018. GMO testing. European network of GMO
04.079 laboratories, Joint Research Center, EURL.
15. Villa C, Costa J, Mafra I (2019) Detection and Available online at http://gmo-crl.jrc.ec.
quantification of milk ingredients as hidden europa.eu/doc/MPR%20Report%20Applica
allergens in meat products by a novel specific tion%2020_10_2015.pdf. Accessed Dec 2022
real-time PCR method. Biomol Ther 9(12): 24. Rodrı́guez A, Rodrı́guez M, Córdoba JJ,
804. https://doi.org/10.3390/biom912 Andrade MJ (2015) Design of primers and
0804 probes for quantitative real-time PCR
16. Villa C, Costa J, Mafra I (2023) Detection and methods. In: Basu C (ed) PCR primer design,
quantification of white and black sesame as vol 1275. Springer, New York, pp 31–56
potential allergenic ingredients in processed 25. Primer Design Tips. Available from: https://
foods: a comparative gene marker study. Food www.thermofisher.com/pt/en/home/life-sci
Control 145:109449. https://doi.org/10. ence/oligonucleotides-primers-probes-genes/
1016/j.foodcont.2022.109449 custom-dna-oligos/oligo-design-tools.html.
17. Costa J, Oliveira MBPP, Mafra I (2013) Effect Accessed Dec 2022
of thermal processing on the performance of
Chapter 9
Abstract
Quantitative PCR (qPCR) is a well-established technique that allows to accurately quantify nucleic acids or
proteins, being widely used in several types of biological samples for bacterial load quantification. However,
there are many recent studies that do not consider the potential pitfalls involved in key experimental qPCR
stages, namely, those related to the extraction and purification of genomic DNA and to the thermal
amplification process, that can lead to biased results in mixed cultures. Herein, we outline a proper protocol
for bacterial quantification by qPCR, addressing how to overcome the main issues in that methodology.
Key words qPCR, DNA extraction, Exogenous control, Bacterial populations, Calibration curves,
Reaction efficiency
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_9,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
105
106 Ângela Lima et al.
2 Materials
2.2 Genomic DNA 1. Commercial silica-based gDNA extraction kit (see Note 3).
Extraction 2. Ice.
3. DNase-free water.
4. Silica beads (see Note 4).
5. 2 mL microcentrifuge tubes with caps.
6. 2 mL collection tubes.
7. Vortex.
8. Centrifuge.
9. Microtube cell disrupter (see Note 5).
Proper Bacteria Quantification by qPCR 107
2.3 qPCR Material 1. Any commercial qPCR SYBR master mix (see Notes 6 and 7).
2. DNase-free water.
3. Primers (see Notes 8 and 9).
4. qPCR 96-well plates, low profile.
5. qPCR plate seals.
6. qPCR instrument: thermocycler with fluorescence detector.
3 Methods
3.1 Preparation of 1. Grow the target bacterial species for the required incubation
Bacterial Cultures for conditions until the needed cell density is achieved.
gDNA Calibration 2. Prepare pure cultures of each target bacterial species. Adjust the
Curves initial bacterial concentrations to a certain optical density
(OD), and confirm the bacterial concentration through
colony-forming units (CFUs) (see Note 10), by making
ten-fold serial dilutions of the adjusted suspension and plating
these dilutions on the appropriate solid medium. Incubate the
plates under adequate conditions and count the CFUs.
3. Prepare at least four dilutions from the initial concentration (see
Note 11), adjusted in the previous step, with PBS 1× in 2 mL
collection tubes. The volume of each dilution should be
900 μL; therefore, if the expected concentration values of the
targets are between 1 × 106 CFU/mL and 1 × 109 CFU/mL,
the bacterial cultures should be prepared with concentrations
between ~1.11 × 106 and ~ 1.11 × 109 CFU/mL, since the
final volume will be 1 mL (900 μL of target species +100 μL of
exogenous control).
4. Adjust the bacterial concentration of the exogenous control to
a certain OD, and confirm the bacterial concentration through
CFUs (see Note 10), by making tenfold serial dilutions of the
adjusted suspension and plating these dilutions on solid
medium. Incubate the plates under adequate conditions and
count the CFUs.
5. Add 100 μL of the exogenous control (see Note 12) with a
concentration of 1 × 109 CFU/mL to 900 μL of each prepared
concentration of the target bacteria, in a 2 mL collection tube.
The final concentration of the exogenous control will be
1 × 108 CFU/mL.
6. Centrifuge the 2 mL collection tubes at 16,000 × g for 10 min.
Completely remove the supernatant with a pipette tip and
freeze the pellets at -20 °C overnight (see Note 13).
108 Ângela Lima et al.
3.2 gDNA Extraction 1. Defrost the bacterial pellets prepared in Subheading 3.1, at
Using a Commercial room temperature.
Kit 2. Resuspend the bacterial pellets in the appropriate volume of the
lysis buffer that is supplied in the kit. Transfer the resuspended
bacteria to a microcentrifuge tube containing approximately
0.4 g of silica glass beads (see Note 14).
3. To lyse the cells, put the tubes with beads in a cell disrupter,
using 2 × 3 cycles of 30 s at maximum × g. Keep the samples on
ice between cycles, for 1 min.
4. Centrifuge the tubes at a maximum of 10,000 × g for 30 s.
5. Avoiding the glass beads, transfer the supernatant to a clean
2 mL collection tube provided.
6. Add 100 μL of an ethanol or isopropanol solution (or any other
solution that precipitates non-DNA organic and inorganic
material) to the supernatant, and vortex for 5 s. Incubate at
4 °C for 5 min.
7. Centrifuge the tubes at 10,000 × g for 1 min.
8. Avoiding the pellet, transfer the entire volume of supernatant
to a clean 2 mL collection tube (provided) (see Note 15).
9. Add 900 μL of a highly concentrated salt solution (washing
buffer 1) to the supernatant, and vortex for 5 s (see Note 16).
10. Add the maximum volume possible to the column (see Note
17) and centrifuge at 10,000 × g for 30 s. Discard the flow-
through, add the remaining supernatant to the silica column,
and centrifuge again at 10,000 × g for 30 s.
11. Add 300 μL of an ethanol-based wash solution (washing buffer
2), and centrifuge at 10,000 × g for 30 s (see Note 18).
12. Discard the flow-through. Centrifuge at 10,000 × g for 1 min.
13. Place the silica column in a new 2 mL collection tube (see Note
19).
14. Add 50 μL of DNase-free water to the center of the white filter
membrane.
15. Centrifuge at 10,000 × g for 30 s and discard the silica column.
16. The gDNA is now ready for downstream applications (see Note
20).
3.3 qPCR Efficiency 1. For each target bacterial species, dilute one gDNA sample
extracted from a pure culture, at least four ten-fold dilutions
in DNase-free water (see Note 21).
2. For each target species, prepare one reaction master mix
solution to a final individual qPCR of 10 μL (see Note 22), as
described in Table 1.
Proper Bacteria Quantification by qPCR 109
Table 1
Description of the master mix preparation for a set of primers (see
Note 28)
3.4 qPCR Calibration 1. Start by extracting gDNA from the samples prepared as
Curve Construction for described in Subheading 3.1, using the method described in
Each Target Species Subheading 3.2.
2. Dilute all the DNA samples at least 1:10 (see Note 31), in
DNase-free water.
3. Repeat the steps 2–6 described in Subheading 3.3.
4. Add an inter-run calibrator to the qPCR 96-well plate (see
Note 32).
5. Normalize the results of the cycle threshold using the equation
ΔCT = 2ðCTtarget species - CTcontrol species Þ (see Note 33).
6. Construct the calibration curve “Relative quantification
(ΔCT),” as depicted in Fig. 1a.
3.5 Absolute 1. After collection of the polymicrobial samples, add the exoge-
Quantification of nous bacterial control, as described at the step 5, of the
Bacterial Species Subheading 3.1.
Concentration in
Polymicrobial Samples
110 Ângela Lima et al.
Fig. 1 Quantification of bacterial species concentration in polymicrobial samples. (a) calibration curves for
each species plotted with the relative quantification (ΔCT) vs bacterial concentration (CFU/mL). (b) bacterial
species concentration (CFU/mL) in a polymicrobial sample determined using the calibration curves
4 Notes
30. Quantify the samples with primers designed for the target
species and with primers for the exogenous control species.
31. The DNA extracted should be diluted at least 1:10, with the
purpose of decreasing the qPCR inhibitors concentration in
the extracted samples.
32. This inter-run calibrator, added on the qPCR plate, serves to
avoid the fluorescence variations between the qPCR runs to be
performed. It can be a bacterium genome, a plasmid, or a
cDNA molecule at a known concentration.
33. This equation, however, makes several assumptions including
that the efficiency of PCR is close to one and that the efficiency
of the primers for target species is similar to the efficiency of the
primers for control species [21].
Acknowledgments
References
1. Benes V, Castoldi M (2010) Expression 6. Bustin SA (2010) Why the need for qPCR
profiling of microRNA using real-time quanti- publication guidelines ? – the case for MIQE.
tative PCR, how to use it and what is available. Methods 50:217–226. https://doi.org/10.
Methods 50:244–249. https://doi.org/10. 1016/j.ymeth.2009.12.006
1016/j.ymeth.2010.01.026 7. Cotto A, Looper JK, Mota LC, Son A (2015)
2. Yu J, Wang HM, Zha MS et al (2015) Molecu- Quantitative polymerase chain reaction for
lar identification and quantification of lactic microbial growth kinetics of mixed culture sys-
acid bacteria in traditional fermented dairy tem. J Microbiol Biotechnol 25:1928–1935.
foods of Russia. J Dairy Sci 98:5143–5154. https://doi.org/10.4014/jmb.1503.03090
https://doi.org/10.3168/jds.2015-9460 8. Leigh Greathouse K, Sinha R, Vogtmann E
3. Rocha D, Santos C, Pacheco L (2015) Bacte- (2019) DNA extraction for human micro-
rial reference genes for gene expression studies biome studies: the issue of standardization.
by RT-qPCR: survey and analysis. Antonie Van Genome Biol 20:1–4. https://doi.org/10.
Leeuwenhoek 108:685–693. https://doi.org/ 1186/s13059-019-1843-8
10.1007/s10482-015-0524-1 9. Costea PI, Zeller G, Sunagawa S et al (2017)
4. Ståhlberg A, Thomsen C, Ruff D, Åman P Towards standards for human fecal sample pro-
(2012) Quantitative PCR analysis of DNA, cessing in metagenomic studies. Nat Biotech-
RNAs, and proteins in the same single cell. nol 35:1069–1076. https://doi.org/10.
Clin Chem 58:1682–1692. https://doi.org/ 1038/NBT.3960
10.1373/clinchem.2012.191445 10. Magalhães AP, França Â, Pereira MO, Cerca N
5. Smith CJ, Osborn AM (2009) Advantages and (2019) RNA-based qPCR as a tool to quantify
limitations of quantitative PCR (Q-PCR)- and to characterize dual-species biofilms. Sci
based approaches in microbial ecology. FEMS Rep 9:1–12. https://doi.org/10.1038/
Microbiol Ecol 67:6–20. https://doi.org/10. s41598-019-50094-3
1111/j.1574-6941.2008.00629.x 11. Marotz C, Amir A, Humphrey G et al (2017)
DNA extraction for streamlined metagenomics
Proper Bacteria Quantification by qPCR 115
of diverse environmental samples. BioTechni- red wine using real time PCR. Front Microbiol
ques 62:290–293. https://doi.org/10.2144/ 7:1–9. https://doi.org/10.3389/fmicb.2016.
000114559 00831
12. O’Connell GC, Chantler PD, Barr TL (2017) 17. Rosca A, Castro J, Sousa L et al (2022) In vitro
High interspecimen variability in nucleic acid interactions within a biofilm containing three
extraction efficiency necessitates the use of species found in bacterial vaginosis
spike-in control for accurate qPCR-based mea- (BV) support the higher antimicrobial toler-
surement of plasma cell-free DNA levels. Lab ance associated with BV recurrence. J Antimi-
Med 48:332–338. https://doi.org/10.1093/ crob Chemother 77:2183–2190
LABMED/LMX043 18. Coyne KJ, Handy SM, Demir E et al (2005)
13. Cerca N, Lima Â, França A (2022) Accurate Improved quantitative real-time PCR assays for
qPCR quantification in polymicrobial commu- enumeration of harmful algal species in field
nities requires assessment of gDNA extraction samples using an exogenous DNA reference
efficiency. J Microbiol Methods 194:1–4. standard. Limnol Oceanogr Methods 3:381–
https://doi.org/10.1016/j.mimet.2022. 391. https://doi.org/10.4319/lom.2005.
106421 3.381
14. Bustin SA, Benes V, Garson JA et al (2009) The 19. Taskin B, Gozen AG, Duran M (2011) Selec-
MIQE guidelines: minimum information for tive quantification of viable Escherichia coli bac-
publication of quantitative real-time PCR teria in biosolids by quantitative PCR with
experiments. Clin Chem 55:611–622. propidium monoazide modification. Appl
https://doi.org/10.1373/CLINCHEM. Environ Microbiol 77:4329–4335. https://
2008.112797 doi.org/10.1128/AEM.02895-10
15. Svec D, Tichopad A, Novosadova V et al 20. Scarsella E, Zecconi A, Cintio M, Stefanon B
(2015) How good is a PCR efficiency estimate: (2021) Characterization of microbiome on
recommendations for precise and robust qPCR feces, blood and milk in dairy cows with differ-
efficiency assessments. Biomol Detect Quantif ent milk leucocyte pattern. Animals 11:1–14.
3:9–16. https://doi.org/10.1016/j.bdq. https://doi.org/10.3390/ani11051463
2015.01.005 21. Livak KJ, Schmittgen TD (2001) Analysis of
16. Longin C, Guilloux-benatier M, Alexandre H relative gene expression data using real-time
(2016) Design and performance testing of a quantitative PCR and the 2-ΔΔCT method.
DNA extraction assay for sensitive and reliable Methods 25:402–408. https://doi.org/10.
quantification of acetic acid bacteria directly in 1006/meth.2001.1262
Chapter 10
Abstract
The in vivo intramolecular recombination of a parental plasmid allows excising prokaryotic backbone from
the eukaryotic cassette of interest, leading to the formation of, respectively, a miniplasmid and a minicircle.
Here we describe a real-time PCR protocol suitable for the determination of recombination efficiency of
parental plasmids with multimer resolution sites (MRS). The protocol was successfully applied to purified
DNA samples obtained from E. coli cultures, allowing a more reproducible determination of recombination
efficiency than densitometry analysis of agarose gels.
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_10,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
117
118 Cláudia P. A. Alves et al.
2 Materials
11. Pipettes.
12. Sterile filter pipette tips.
13. Real-time qPCR system.
14. Benchtop minicentrifuge.
15. Commercial qPCR kit (see Note 8).
3 Methods
3.1 Primer Design 1. Identify the multimer resolution sites (MRS) in the parental
plasmid (see Fig. 1).
2. Design primers in the locations identified in point 1 (regions
from the prokaryotic backbone and eukaryotic cassette flanking
the MRS). The use of primer design software is recommended
(e.g., Primer 3, https://bioinfo.ut.ee/primer3/ [15, 16]).
This allows users to set the desired melting temperature and
constraints to minimize the probability of primer-dimer forma-
tion. Ideally, primer sequences should have a length of 20–24
nucleotides, a GC content of about 40–60%, and a melting
temperature close to 60 °C [17, 18] (see Note 10).
ng
Molecules Concentration μL × 6:022 × 10
23
=
μL DNA size ðbpÞ × 1 × 109 × 660
3. Calculate the volume of pure DNA sample to prepare a solu-
tion with 1 × 109 molecules per μL using:
1 × 109 molecules
μL × Final volume ðμLÞ
Volume of pure sample ðμLÞ =
molecules
Initial concentration μL
120 Cláudia P. A. Alves et al.
Fig. 1 Representation of the parental plasmid, with an indication of the regions that originate the miniplasmid,
which contains the prokaryotic backbone, and the minicircle, which contains the eukaryotic cassette. These
regions are flanked by multimer resolution sites (MRS). Four primers are designed to anneal to the sequences
upstream and downstream of the MRS. Primer sequences should be oriented to form pairs and amplify
amplicons containing each MRS, being indicated by their annealing sites in the parental plasmid, miniplasmid,
and minicircle. Amplicons expected by real-time PCR using this strategy are also presented
4. Ensure that all the following steps are performed with sterile
filter pipette tips.
5. Prepare tenfold dilutions with between 1 × 109 and 1 × 104
molecules per μL, by mixing the appropriate volume of sample
(calculated as described in step 3) with the appropriate volume
of PCR-grade water. Keep the solution in ice (see Note 11).
6. Prepare PCR-grade water containing 150,000 cells/μL by
making serial dilutions of the bacterial culture of
non-transformed cells (see Note 5 and 12).
7. Thaw qPCR reagents (commercial qPCR mixture and primers)
in ice.
8. Prepare qPCR in 200 μL PCR tubes by mixing the following
components: 2 μL of DNA standard (parental plasmid, mini-
plasmid, or minicircle), 2 μL of PCR-grade water containing
150,000 cells per μL, 4.4 μL of PCR-grade water, 0.8 μL of
10 μM forward primer, 0.8 μL of 10 μM reverse primers, and
10 μL of commercial qPCR mix. Keep reactions in ice.
Real-Time PCR Method for Recombination Efficiency Evaluation 121
Table 1
Description of the real-time PCR program used for quantification [14]
Target Signal
temperature Hold Slope acquisition Analysis
Step description Cycles (°C) (min:s) (°C/s) mode mode
Polymerase 1 95 02:00 20 – –
activation
Amplification 30 95 00:10 20 – Quantification
55 00:05 20 –
72 00:07 20 Single
Coolinga 1 40 00:30 20 – –
a
Cooling can be performed after the amplification cycles or only at the end, if a program for melting temperature analysis
is included after amplification
4 Notes
1. This protocol was developed using the E. coli BW2P strain [3]
and pMINILi-CVG plasmid [4, 14, 21].
The strain was constructed by engineering the genome of
an E. coli strain optimized for L-arabinose uptake (E. coli
BW27783, CGSC#: 12119, obtained from the Coli Genetic
Stock Center, Yale, USA) [3]. Modifications consisted of the
knockout of the recA gene and disruption of the endA gene by
insertion of an optimized cassette for L-arabinose expression,
containing a D-glucose-repressible promoter (PBAD) [3]. Nev-
ertheless, other systems are described in which ParA resolvase is
expressed from the parental plasmid [5, 12] or, alternatively,
from a helper plasmid [3]. In the latter case, however, an
additional contaminating plasmid molecule will be present.
The model plasmid (pMINILi-CVG) [4, 14, 21] used
presented multimer resolution sites (MRS) flanking the pro-
karyotic and eukaryotic backbone. These sequences have a
length of 133 bp, are identical, and do not suffer modifications
during the recombination process [11]. Since MRS sequences
are present in parental plasmids (two MRS), minicircles (one
MRS), and miniplasmids (one MRS), primers that anneal at
MRS will not be able to distinguish the molecules. The
pMINILi-CVG (4563 bp) plasmid was previously obtained
after successive modifications of the pMINI plasmid, which
was derived from the commercial pVAX1™/LacZ (Invitrogen,
Carlsbad, CA) [3, 22, 23]. Plasmid pMINI was modified by
(i) site-directed mutagenesis to include PvuII restriction sites in
the prokaryotic backbone, (ii) removal of remaining sequences
of a PBAD/AraC-parA cassette, and (iii) cloning of the VEGF
(vascular endothelial growth factor) gene. The prokaryotic
backbone of pMINILi-CVG harbors six PvuII recognition
sites and consists of a pMB1-derived replication origin and
the kanamycin resistance gene. The plasmid’s eukaryotic cas-
sette comprises the CMV (human cytomegalovirus)
immediate-early promoter, a VEGF-GFP (green fluorescent
protein) gene fusion, and the BGH (bovine growth hormone)
polyadenylation signal. Prokaryotic and eukaryotic sequences
on the plasmid are separated by MRS [4, 14, 21].
The protocol applied for the production of the minicircle
used in the development of the real-time PCR method
described here can be found in [21].
2. Pure miniplasmid is needed as a standard to construct the
corresponding calibration curve. To obtain a strain trans-
formed with the miniplasmid, first, perform cultures for mini-
circle production as described in [21]. Use a sample collected at
the end of the bacterial exponential growth to perform a
Real-Time PCR Method for Recombination Efficiency Evaluation 125
Table 2
Sequence and properties of oligonucleotides used in the real-time PCR program
Oligonucleotides information
Melting
temperature
Primer Sequence (5′-3′) (°C) %GC
MP1 GCCTTTTGCTCACATGTTCTTGC 59 48
MP2 ATTCCGGTTCGCTTGCTGTC 59 55
MC1 ACTAGTCAATAATCAATGTCAACGCG 57 38
MC2 CGGTGGGCTCTATGGCTTCTAATG 60 54
Primers combinations for real-time PCR
Target DNA molecule Primer forward Primer reverse Amplicon length
expected (bp)
Parental plasmid MP1 MC1 199
Miniplasmid MP1 MP2 223
Minicircle MC2 MC1 201
Combinations used to amplify fragments from parental plasmid, miniplasmid, and minicircle target molecules are also
indicated
Real-Time PCR Method for Recombination Efficiency Evaluation 127
Fig. 2 Representation of a calibration curve. Quantification cycles obtained for two series of tenfold dilution
samples are plotted in function of the log10 of target molecules per reaction
Fig. 3 Representative results of analysis of parental plasmid recombination. Samples collected during
minicircle production (0 h, 1 h, and 2 h) were purified and run on an agarose gel before (non-digested) and
after enzymatic restriction. Recombination efficiency was calculated by densitometry analysis of linearized
bands on the agarose gel and by real-time PCR using pure samples. MC minicircle, MP miniplasmid, PP
parental plasmid, Lin linear, sc supercoiled, MWM molecular weight marker
Acknowledgement
References
1. Mairhofer J, Grabherr R (2008) Rational vec- 9. Smith MCM, Brown WRA, McEwan AR,
tor design for efficient non-viral gene delivery: Rowley PA (2010) Site-specific recombination
challenges facing the use of plasmid DNA. Mol by φC31 integrase and other large serine
Biotechnol 39:97–104. https://doi.org/10. recombinases. Biochem Soc Trans 38:388–
1007/s12033-008-9046-7 394. https://doi.org/10.1042/BST0380388
2. Hardee C, Arévalo-Soliz L, Hornstein B, 10. Stark WM (2014) The serine recombinases.
Zechiedrich L (2017) Advances in non-viral Microbiol Spectr 2:MDNA3–0046–2014.
DNA vectors for gene therapy. Genes 8:65. https://doi.org/10.1128/microbiolspec.
https://doi.org/10.3390/genes8020065 MDNA3-0046-2014
3. Šimčı́ková M, Alves CPA, Brito L et al (2016) 11. Thomson JG, Yau Y-Y, Blanvillain R et al
Improvement of DNA minicircle production (2009) ParA resolvase catalyzes site-specific
by optimization of the secondary structure of excision of DNA from the Arabidopsis
the 5′-UTR of ParA resolvase. Appl Microbiol genome. Transgenic Res 18:237–248.
Biotechnol 100:6725–6737. https://doi.org/ https://doi.org/10.1007/s11248-008-
10.1007/s00253-016-7565-x 9213-4
4. Alves CPA, Šimčı́ková M, Brito L et al (2016) 12. Mayrhofer P, Blaesen M, Schleef M, Jechlinger
Development of a nicking endonuclease- W (2008) Minicircle-DNA production by site
assisted method for the purification of minicir- specific recombination and protein-DNA inter-
cles. J Chromatogr A 1443:136–144. https:// action chromatography. J Gene Med 10:1253–
doi.org/10.1016/j.chroma.2016.03.035 1269. https://doi.org/10.1002/jgm.1243
5. Jechlinger W, Azimpour Tabrizi C, Lubitz W, 13. Kubista M (2014) Prime time for qPCR – rais-
Mayrhofer P (2004) Minicircle DNA immobi- ing the quality bar. Eur Pharm Rev 19:63–67
lized in bacterial ghosts: in vivo production of 14. Alves CPA, Prazeres DMF, Monteiro GA
safe non-viral DNA delivery vehicles. J Mol (2021) Recombination efficiency measurement
Microbiol Biotechnol 8:222–231. https:// by real-time PCR: a strategy to evaluate ParA-
doi.org/10.1159/000086703 mediated minicircle production. Anal Biochem
6. Chen Z-Y, He C-Y, Ehrhardt A, Kay MA 628:114285. https://doi.org/10.1016/j.ab.
(2003) Minicircle DNA vectors devoid of bac- 2021.114285
terial DNA result in persistent and high-level 15. Koressaar T, Remm M (2007) Enhancements
transgene expression in vivo. Mol Ther 8:495– and modifications of primer design program
500. https://doi.org/10.1016/S1525-0016 Primer3. Bioinformatics 23:1289–1291.
(03)00168-0 https://doi.org/10.1093/bioinformatics/
7. Kay MA, He C-Y, Chen Z-Y (2010) A robust btm091
system for production of minicircle DNA vec- 16. Untergasser A, Cutcutache I, Koressaar T et al
tors. Nat Biotechnol 28:1287–1289. https:// (2012) Primer3 – new capabilities and inter-
doi.org/10.1038/nbt.1708 faces. Nucleic Acids Res 40:e115–e115.
8. Darquet A-M, Cameron B, Wils P et al (1997) https://doi.org/10.1093/nar/gks596
A new DNA vehicle for nonviral gene delivery: 17. Nolan T, Huggett J, Sanchez E (2013) Good
supercoiled minicircle. Gene Ther 4:1341– practice guide for the application of quantita-
1349. https://doi.org/10.1038/sj.gt. tive PCR (qPCR). LGC
3300540
Real-Time PCR Method for Recombination Efficiency Evaluation 131
18. Svec D, Tichopad A, Novosadova V et al 21. Alves CPA, Šimčı́ková M, Brito L et al (2018)
(2015) How good is a PCR efficiency estimate: Production and purification of supercoiled
recommendations for precise and robust qPCR minicircles by a combination of in vitro endo-
efficiency assessments. Biomol Detect Quantif nuclease nicking and hydrophobic interaction
3:9–16. https://doi.org/10.1016/j.bdq. chromatography. Hum Gene Ther Methods
2015.01.005 29:157–168. https://doi.org/10.1089/hgtb.
19. Bustin SA, Benes V, Garson JA et al (2009) The 2018.046
MIQE guidelines: minimum information for 22. Azzoni AR, Ribeiro SC, Monteiro GA, Pra-
publication of quantitative real-time PCR zeres DMF (2007) The impact of polyadenyla-
experiments. Clin Chem 55:611–622. tion signals on plasmid nuclease-resistance and
https://doi.org/10.1373/clinchem.2008. transgene expression. J Gene Med 9:392–402.
112797 https://doi.org/10.1002/jgm.1031
20. Forootan A, Sjöback R, Björkman J et al 23. Šimčı́ková M, Prather KLJ, Prazeres DMF,
(2017) Methods to determine limit of detec- Monteiro GA (2014) On the dual effect of
tion and limit of quantification in quantitative glucose during production of pBAD/AraC-
real-time PCR (qPCR). Biomol Detect Quantif based minicircles. Vaccine 32:2843–2846.
12:1–6. https://doi.org/10.1016/j.bdq. https://doi.org/10.1016/j.vaccine.2014.
2017.04.001 02.035
Chapter 11
Abstract
Quantitative PCR (qPCR) is one of the most used techniques to quantify gene expression in bacterial
biofilms due to its easiness, sensitivity, and robustness. However, several practical aspects need to be
considered to obtain accurate and reliable results. Here, we describe a detailed and optimized protocol to
quantify mRNA transcripts from bacterial biofilms using qPCR, including pieces of advice to improve RNA
quality, which ultimately increases the accuracy, consistency, and relevance of gene expression data.
Key words Bacteria, Biofilms, Sample collection, RNA isolation, DNase treatment, RNA quality,
Complementary DNA synthesis, qPCR
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_11,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
133
134 Angela França and Nuno Cerca
Fig. 1 Schematic representation of the main factors impacting RNA quality and, consequently, the accuracy of
gene expression quantification. RNA quality has direct implications on the amount of RNA that is converted into
cDNA, which has a direct impact on qPCR quantification. Hence, care shall be taken to obtain high-quality RNA
for gene expression assays
besides the elements listed above, the quality of the RNA isolated is
one of the most critical aspects to consider in gene expression assays
[6–9]. Because of the natural regulation of mRNAs in response to
environmental stimuli [10], mRNA degrades quickly, and, thus,
diligent processes are essential to maintain the profile of mRNA
molecules being produced under the testing conditions. Overall, to
obtain representative gene expression data, it is important to be
aware of the good practices that support the isolation of high-
quality RNA. In this chapter, we describe a detailed and optimized
two-step protocol (cDNA synthesis and qPCR run performed sep-
arately) for gene expression quantification of biofilm samples by
qPCR, providing tips to improve RNA quality and, ultimately, data
reproducibility, accuracy, and relevance.
2 Materials
2.2 DNase Treatment 1. RNase-free DNase I kit (that includes enzyme, buffer
and EDTA).
2. Filter tips/RNase-free tips.
3. Thermal block (for 37 and 65 °C incubations).
5. PCR-grade water.
6. Thermal cycler with fluorescence detection module (qPCR).
3 Methods
3.1 Sample Sample collection is one of the most crucial steps in gene expression
Collection assays [11], as all the subsequent steps depend on initial sample
quality. During sample collection changes in gene expression profile
can occur due to specific (enzymatic) and nonspecific (mRNA life-
span) RNA degradation or due to transcriptional induction. Thus,
immediate stabilization is essential. For that, there are several com-
mercially available stabilizing solutions. If it is not possible to use
them, collect and process samples immediately:
1. Place biofilm plates on ice and remove the spent culture
medium.
2. Carefully wash biofilms once with phosphate-buffered saline
(PBS) to remove planktonic cells.
3. Immediately stabilize your sample by adding an
RNA-stabilizing solution into each well (see Note 1). From
then on, the RNA isolation procedure can be performed at
room temperature (see Note 2).
4. Harvest biofilm cells by scraping the bottom of the plate with a
tip or with the help of a 1 mL insulin syringe plunger.
5. Pool at least ten biofilms together (see Note 3).
6. Collect 1 mL of the pooled biofilm suspension, and centrifuge
following the stabilizing solution manufacturer’s indications
(see Note 3).
3.2 RNA Isolation RNA isolation is considered to be the second critical step when
performing gene expression assays [11]. There are several commer-
cially available kits for RNA isolation, and, depending on the lyses
method (chemical, enzymatic, or mechanical) and the isolation
principle (organic or columns), different kits will yield RNA with
varying quality levels [1, 2, 12]. It has been also demonstrated that
bacterial structures, such as Gram-positive or Gram-negative struc-
tures, along with the phenotype, planktonic or biofilm, can impact
the efficiency of the kit used [1, 2]. Herein, we present a protocol
that combines the efficacy of mechanical and chemical lysis with the
quickness of column-based isolation kits. As tested before, this
Biofilm Cells Gene Expression Quantification by qPCR 137
protocol can be used with numerous column-based kits [3] and has
shown to be efficient in isolating high-quality RNA from a varied
array of samples, ranging from single biofilms formed by Staphylo-
coccus epidermidis [13, 14] and Gardnerella vaginalis [15] to com-
plex consortia associated with bacterial vaginosis [16] and
Staphylococcus aureus-Pseudomonas aeruginosa mixed biofilms
[17, 18]:
1. After stabilizing and centrifuging your sample, discard the
supernatant, and suspend the pellet in 500 μL of the lysis
buffer, previously supplemented with β-mercaptoethanol
(optional) and in 500 μL of molecular-grade phenol (optional)
(see Note 4). If phenol is not used, then add the volume of lysis
buffer indicated by the manufacturer.
2. Transfer the bacterial suspension into tubes with acid-washed
glass beads (see Note 5).
3. Place the tubes in a cell disruptor for 35 s at 6.5 m/s. To cool
the samples down, incubate the tubes on ice for 5 min. Repeat
this step 2 times more (see Note 6).
4. Centrifuge the lysate at 16,000 g for 2 min to remove cell
debris and bring the beads down.
5. Transfer the supernatant into a 2 mL RNase-free tube. Avoid
aspirating beads that may clog the RNA binding membrane,
reducing RNA yield.
6. Add an equal volume of 70% ethanol and mix by inverting the
tubes a couple of times (see Note 7).
7. Transfer the lysate into the RNA isolation column. Consider-
ing the maximum capacity of the isolation column, this step
may be repeated more than once.
8. Centrifuge the column at ≥10,000 g for 1 min (see Note 8).
Discard the flow-through and reuse the collection tube.
9. Add wash buffer 1 (see Note 9) and centrifuge at ≥10,000 g for
1 min. Discard the flow-through and reuse the collection tube.
10. Add wash buffer 2 (see Note 9) and centrifuge at ≥10,000 g for
1 min. Discard the flow-through and reuse the collection tube.
11. Add wash buffer 2 for a second time and centrifuge at
≥10,000 g for 2 min. Discard the flow-through and place the
column into a new collection tube.
12. Centrifuge the column (without any liquid) at ≥10,000 g for
3 min (see Note 10).
13. Transfer the column into a 1.5 mL RNase-free tube.
14. Add 50 μL of RNase-free water into the center of the mem-
brane (see Note 11), and incubate for 1 min at room
temperature.
15. Centrifuge the column at full speed for 2 min.
138 Angela França and Nuno Cerca
3.3 Genomic DNA The presence of genomic DNA (gDNA) has important repercus-
Degradation/Removal sions in gene expression quantification, resulting in the detection of
false positives and/or difficult-to-interpret data [12]. Thus, the
reduction of gDNA contamination to minimal levels is crucial.
This is most often achieved by the addition of DNase I due to its
easiness and because there is no need to use hazardous chemicals.
Importantly, when using DNase I, the enzyme needs to be inacti-
vated at the end because, while with reduced activity (1–2%) [19], it
can also degrade mRNA/DNA hybrid molecules produced during
cDNA synthesis or double-stranded DNA products formed during
the qPCR run. DNase can be inactivated/removed from the reac-
tion in several ways, but the most common is inactivation by heat.
Hence, we present a protocol for a DNase treatment followed by
the inactivation of the enzyme by heat:
1. Add the indicated quantity of DNase enzyme (see Note 12) and
the respective buffer, and mix gently but thoroughly by
performing up and down.
2. Incubate the tubes at 37 °C for 30 min so the DNase can act.
3. Add the indicated quantity of EDTA and mix gently but thor-
oughly by performing up and down (see Note 13).
4. Incubate the tubes for 10 min at 65 °C.
5. Immediately transfer RNA to ice or safely store it at -20 °C or
-80 °C, for short- or long-term storage, respectively.
3.4 RNA Quality Since RNA quality can greatly impact gene expression quantifica-
Assessment tion [1, 2, 7, 9], it is imperative to ensure acceptable RNA quality
among comparing samples. RNA quality term includes RNA quan-
tity, purity, and integrity, and, so far, there is not a single method
that can examine all indicators at once. Usually, the quantity and
purity of RNA samples are determined by spectrophotometry and
RNA integrity by agarose gels. As such, we present a protocol for
both methods.
3.4.1 RNA Quantity and As referred to above, RNA quantity and purity are easily deter-
Purity mined by spectrophotometry, more specifically using a NanoDrop,
where only 1 μL of the sample is required:
1. Initialize the equipment by selecting the nucleic acids option
and, then, RNA.
2. Thoroughly clean the pedestal of the upper and lower arms
with water.
Biofilm Cells Gene Expression Quantification by qPCR 139
3.4.2 RNA Integrity Agarose gels, either under denaturant or non-denaturant condi-
tions, are one of the simplest and cheapest methods to evaluate
mRNA molecules’ integrity (Fig. 2). As such, a protocol for an
agarose gel under non-denaturant conditions is presented:
1. Clean all material needed for electrophoresis, including tanks,
trays, and combs with 0.1% sodium dodecyl sulfate (SDS) or
with 0.1 M of sodium hydroxide (NaOH) and 1 M EDTA
solution. In the alternative, commercial RNase-degrading solu-
tions may be used. Thereafter, rinse with plenty of RNase-free
or ultrapure water and protect all material from dust.
2. Prepare a 1% RNase-free agarose gel with Tris-acetate-EDTA
(TAE) or Tris-borate-EDTA (TBE) buffer (see Note 16).
3. Let agarose cool down and add nucleic acid staining solution.
4. Pour the gel and let it solidify (protect the gel from dust).
Table 1
An example of cDNA synthesis master mix preparation. n, the total
number of samples to be analyzed
Table 2
An example of sample preparation for cDNA synthesis reaction, in 10 μL of
volume
RNase-free water 2
Master mix (prepared in step 1) 3
Total reaction volume 10
a
In the case of reverse sequence-specific primers, ~1 μL of each primer, including
reference genes, needs to be added. The quantity of water may be reduced, or absent,
to accommodate the volume necessary for all reverse primers
6. Thereafter, mix the RNA sample with the master mix, and
centrifuge the samples briefly to burst possible bubbles created
during sample preparation.
7. Follow the thermal cycler conditions indicated by the manu-
facturer to complete cDNA synthesis and RT inactivation.
8. The newly synthesized cDNA may be immediately used or
stored until further use. For short-term storage (2 days),
cDNA can be placed at 4 °C, and for long-term storage, it is
advisable to store cDNA at -20 or - 80 °C.
3.6 Quantitative PCR Quantitative PCR can be used for the absolute or relative quantifi-
Run cation of gene expression. Due to its easiness and accuracy, relative
quantification is the most widely used method. However, to work
properly, a normalization strategy needs to be used to account for
the variability introduced by both biological and technical varia-
tions. Normalization with a reference gene is the most popular and
comprehensive normalization strategy. In this optimized protocol,
we use reference genes as the normalization strategy and SYBR®
Green dye for amplification quantification:
1. Dilute your cDNA, as well as NRT controls, at least 100× in
ultrapure water (see Note 25).
2. When evaluating the qPCR efficiency, which shall be deter-
mined every time a new set of primers are being used or
when qPCR master mixes are altered, from the first cDNA
dilution, prepare two- to tenfold dilutions (depending on the
abundance of the target), to obtain standard curves with at least
5 points (see Note 26). Consider that each dilution shall be
analyzed in triplicates.
3. Prepare the qPCR master mix for each of the genes under
study, including the reference genes, by mixing the
142 Angela França and Nuno Cerca
Table 3
An example of qPCR master mix preparation for a set of primers/target
genes, in 10 μL of volume. This has to be repeated to all sets of primers/
target genes under study
4 Notes
Amplicon Efficiencya
Target gene Sequence 5′ ! 3′ size (bp) (%) Function
143
144 Angela França and Nuno Cerca
Acknowledgments
References
1. França A, Bento JC, Cerca N (2012) Variability Res Notes 4:572. https://doi.org/10.1186/
of RNA quality extracted from biofilms of 1756-0500-4-572
foodborne pathogens using different kits 3. França A, Freitas AI, Henriques AF, Cerca N
impacts mRNA quantification by qPCR. Curr (2012) Optimizing a qPCR gene expression
Microbiol 65:54–59. https://doi.org/10. quantification assay for S. epidermidis biofilms:
1007/s00284-012-0124-5 a comparison between commercial kits and a
2. França A, Melo LD, Cerca N (2011) Compari- customized protocol. PLoS One 7:e37480.
son of RNA extraction methods from biofilm https://doi.org/10.1371/journal.pone.
samples of Staphylococcus epidermidis. BMC 0037480
148 Angela França and Nuno Cerca
DNA-derived signals with ValidPrime. Nucleic recommendations for precise and robust qPCR
Acids Res 40:e51–e51. https://doi.org/10. efficiency assessments. Biomol Detect Quantif
1093/NAR/GKR1259 3:9–16. https://doi.org/10.1016/j.bdq.
25. Huggett JF, Novak T, Garson JA et al (2008) 2015.01.005
Differential susceptibility of PCR reactions to 29. Freitas AI, Lopes N, Oliveira F et al (2018)
inhibitors: an important and unrecognised Comparative analysis between biofilm forma-
phenomenon. BMC Res Notes 1:70. https:// tion and gene expression in Staphylococcus epi-
doi.org/10.1186/1756-0500-1-70 dermidis isolates. Future Microbiol 13:415–
26. Livak KJ, Schmittgen TD (2001) Analysis of 4 2 7 . h t t p s : // d o i . o r g / 1 0 . 2 2 1 7 / f m b -
relative gene expression data using real-time 2017-0140
quantitative PCR and the 2-ΔΔCT method. 30. Gaio V, Lopes N, Cerca N, França A (2021)
Methods 25:402–408. https://doi.org/10. codY and pdhA expression is induced in Staphy-
1006/meth.2001.1262 lococcus epidermidis biofilm and planktonic
27. Pfaffl MW, Pfaffl MW (2001) A new mathemat- populations with higher proportions of viable
ical model for relative quantification in real- but non-Culturable cells. Front Cell Infect
time RT-PCR. Nucleic Acids Res 29:e45. Microbiol 11. https://doi.org/10.3389/
https://doi.org/10.1093/nar/29.9.e45 FCIMB.2021.771666
28. Svec D, Tichopad A, Novosadova V et al
(2015) How good is a PCR efficiency estimate:
Chapter 12
Abstract
The determination of the number of plasmid copies in each cell of Lactococcus lactis is critical for the control
and regulation of the production of recombinant proteins and plasmids. This protocol describes a method
for the determination of the plasmid copy number per genome of L. lactis, which is based on the detection
by real-time quantitative PCR of the number of plasmid molecules and the number of chromosomes and
subsequently their ratio after calculating the amplification efficiency.
Key words Lactococcus lactis, Plasmid copy number, Real-time quantitative PCR
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_12,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
151
152 Sofia O. D. Duarte and Gabriel A. Monteiro
2 Materials
2.1 L. lactis PCN Prepare all solutions using PCR-grade water (prepared by filtering
Determination by Real- Milli-Q water with a 0.1 μm syringe filter and then autoclaved) and
Time Quantitative PCR analytical-grade reagents. Prepare and store all reagents at room
temperature (unless indicated otherwise). Filter tips should be used
while preparing all the dilutions and solutions. Diligently follow all
waste disposal regulations when disposing of waste materials:
1. Primer design:
(a) Appropriate primer design software (see Note 1).
2. Genomic and plasmid DNA standards preparation:
(a) Commercial kit for gDNA and pDNA purification.
(b) Microcentrifuge tubes.
(c) Filter tips.
(d) PCR-grade water.
(e) 0.5 M EDTA stock solution: Weigh the EDTA disodium
salt and add 800 mL of Milli-Q water. Adjust the pH to
8.0 with sodium hydroxide pellets (see Note 2). Adjust the
volume to 1 L using a volumetric flask.
(f) TAE 50×: 2 M Tris base, 50 mM EDTA disodium salt,
1 M acetic acid. Solubilize the three components in
800 mL of distilled water in a suitable container. Adjust
the volume to 1 L using a volumetric flask.
(g) TAE 1×: Dilute the TAE 50x solution using Milli-Q
water.
(h) 1% agarose gel: To do a 50 mL gel, add 0.5 g of agarose to
50 mL of TAE 1×. Microwave until totally dissolved (see
Note 3).
3. L. lactis cell growth:
(a) 15 mL tubes.
(b) M17 medium: Suspend 42.25 g in 1 L distilled water. Mix
until dissolved. Dispense into 15 mL tubes. Sterilize by
autoclaving at 121 °C for 15 min.
4. Real-time qPCR:
(a) Cooling block.
(b) Real-time qPCR appropriate capillaries and lids (see Note
4).
(c) Microcentrifuge tubes.
(d) Filter tips.
(e) PCR-grade water.
(f) SYBR Green mixture kit.
Quantification of Plasmid Copy Number in Lactococcus Lactis 153
3 Methods
3.1 L. lactis PCN Carry out all procedures at room temperature unless otherwise
Determination by Real- specified:
Time Quantitative PCR
1. Primer design: Design two sets of forward and reverse primers,
one specific to a sequence present in the plasmid and the other
specific to a conserved single-copy sequence present in the
L. lactis strain chromosome (see Note 5). Synthesize
(or order) the primers.
2. Genomic DNA standards preparation: Purify gDNA from an
overnight culture of the plasmid-free strain, using an appropri-
ate kit. Perform its quantification using a microvolume spec-
trophotometer and assess its quality by agarose gel
electrophoresis. Serially dilute the gDNA using PCR-grade
water in order to achieve the following masses: 1, 10,
100, 1000, and 10,000 pg per reaction (see Note 6).
3. Plasmid DNA standards preparation: Purify pDNA from an
overnight culture of the strain harboring the backbone plasmid
(see Note 7), using an appropriate kit. Perform its quantifica-
tion using a microvolume spectrophotometer and assess its
quality by agarose gel electrophoresis. Serially dilute the
pDNA using PCR-grade water in order to achieve the follow-
ing masses: 1, 10, 100, 1000, and 10,000 pg per reaction (see
Note 8).
4. Plasmid-free cell preparation: Grow overnight the plasmid-free
strain in a 15 mL tube containing 5 mL of M17 medium.
Calculate the necessary volume to spike each pDNA standard
with 1000 cells, diluting in PCR-grade water if necessary (see
Notes 9 and 10).
5. Sample preparation: Grow overnight each plasmid-harboring
strain in a 15 mL tube containing 5 mL of M17 medium.
Calculate the necessary volume for each reaction having 1000
cells, diluting in PCR-grade water if necessary (see Note 11).
6. Real-time qPCR: For each 20 μL reaction mixture, mix 2 μL of
10× SYBR Green mix, 1 μL of each primer (final concentration
0.5 μM), 1.6 μL of MgCl2 (final concentration 3 mM), and
10.4 μL of PCR-grade water, and the remaining volume was
completed with the samples (4 μL with 1000 cells containing
154 Sofia O. D. Duarte and Gabriel A. Monteiro
EgCPg
PCN = ð2Þ
EpCPp
Fig. 1 Example of plasmid and chromosome DNA standard curves using real-time qPCR equipment. The
plasmid standard curve was performed in triplicate, using four log serial dilutions of pDNA spiked with L. lactis
LMG19460 cells, while the chromosome standard curve was performed using five log serial dilutions of
non-spiked gDNA. The linear regression trendlines are represented. The equation for the plasmid standard
curve is y = -3.46x + 23.68, with a R2 of 0.996 and an efficiency of 94.68%. The equation for the
chromosome standard curve is y = -3.33x + 27.76, with a R2 of 0.997 and an efficiency of 99.77%. The
primers to quantify the pTRKH3 plasmid targeted the erythromycin resistance gene (erm) and had the
following sequences: forward primer, CTTCGTTATGATTTTACA, and reverse primer, CAATATCAACAATTCCAT
(amplified sequence with 190 bp). The primers designed to quantify the L. lactis LMG19460 chromosome
targeted the ferrous iron transport protein A ( feoA) gene and had the following sequences: forward,
TCAGACGCCGCTTGATGGAC, and reverse, AGTTCAAGAGGGTCGCCAAGTG (amplified sequence with 89 bp)
- 1
Eð%Þ = 10 slope - 1 × 100 ð3Þ
4 Notes
10. The pDNA standard curve reactions were spiked with 1000
L. lactis cells without plasmid, to ensure that the samples and
the pDNA standards have the same putative inhibitors. Since
the L. lactis genome of the cells used to spike the capillaries
could mislead the gDNA quantification, the gDNA standard
reactions should not be spiked with plasmid-free cells.
11. Prepare a dilution that allows to have 1000 cells in each 2 μL, in
order to keep a constant volume in each reaction.
12. If the SYBR Green mix is not a HotStart one, ensure that it is
the last component to be added to the reaction.
13. We recommend preparing each reaction mixture separately in
microcentrifuge tubes. Each tube should be thoroughly mixed
using a vortex and then spun in a tabletop microcentrifuge.
Next, each reaction could be transferred to the top part of the
capillary. Before closing the lid, the capillary should be spiked
in a tabletop microcentrifuge to ensure that all the reaction
volume moves to the bottom of the capillary.
14. The amplification parameters may need to be adjusted, consid-
ering the SYBR Green kit specificities and the primers melting
temperature.
15. Each parameter in the equation should be the average value of
at least three independent experiments.
Acknowledgments
References
1. Carapuça E, Azzoni AR, Prazeres DM, Mon- processes. Microb Cell Factories 7:6. https://
teiro GA, Mergulhao FJ (2007) Time-course doi.org/10.1186/1475-2859-7-6
determination of plasmid content in eukaryotic 3. Duarte SOD, Martins MC, Andrade SM, Pra-
and prokaryotic cells using real-time PCR. Mol zeres DMF, Monteiro GA (2019) Plasmid copy
Biotechnol 37(2):120–126 number of pTRKH3 in Lactococcus lactis is
2. Skulj M, Okrslar V, Jalen S, Jevsevar S, Slanc P, increased by modification of the repDE
Strukelj B, Menart V (2008) Improved determi- ribosome-binding site. Biotechnol J 14(8):
nation of plasmid copy number using quantita- e1800587. https://doi.org/10.1002/biot.
tive real-time PCR for monitoring fermentation 201800587. Epub 2019 May 20. PMID:
31009171
Chapter 13
Abstract
As a powerful tool, polymerase chain reaction (PCR) has been indispensable and widely used in a large array
of applications. In practice, many factors may affect the overall performance of a PCR. One such factor is the
stability of intramolecular secondary structure formed within single-stranded template. The higher the
stability of such a structure, the more likely it will have adverse effects on PCR performance. Traditionally,
chemical reagents believed to reduce the stability of nucleic acid secondary structures, such as DMSO and
betaine, have been used to mitigate their adverse effects on PCR performance. However, these reagents
have apparent downsides including increasing replication error rate, inhibiting polymerase activity, and
being ineffective against secondary structures of very high stabilities. Disruptors, a new class of oligonucle-
otide reagents, do not exhibit such downsides. They are specifically designed to target intramolecular
secondary structures only without any effect on the replication of other regions of the template. Their
effective concentration range for improving PCR performance is well tolerated by PCR. And they are very
effective in improving PCR performance on templates that are notoriously difficult to amplify by PCR even
in the presence of DMSO or betaine, e.g., the inverted terminal repeat of adeno-associated virus
(AAV-ITR). In this chapter, the application of disruptors in PCR is described with AAV-ITR as the example
template.
Key words Polymerase chain reaction (PCR), Intramolecular secondary structure, Disruptor, Free
energy (ΔG), Adeno-associated virus (AAV), Inverted terminal repeat (ITR)
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_13,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
159
160 Yong Ma and Minxue Zheng
Fig. 1 Diagram to illustrate the proposed mechanism of disruptor function. In the absence of disruptors, primer
binding and intramolecular secondary structure formation occur during temperature drop and annealing step.
Taq polymerase extends primer until encountering stable intramolecular secondary structure. Due to the
interference of such a structure, the following three outcomes may occur. First, polymerase stalls, resulting in
premature termination of elongation. Second, polymerase destabilizes the intramolecular secondary structure
through cleavage within its duplex region to generate truncated PCR product. Finally, polymerase continues
elongation to generate full-length PCR product at a low frequency. In the presence of disruptors, besides
primer binding and intramolecular secondary structure formation, the anchor of a disruptor binds to template
during temperature drop and annealing step. While polymerase extends primer, the effector of bound disruptor
unwinds the intramolecular secondary structure by strand displacement. And then polymerase continues
elongation to generate full-length product
Improve PCR Performance with Disruptors 161
Fig. 2 PCR amplification of the ITR sequence of a rAAV vector plasmid was successful in the presence of
disruptors but not DMSO or betaine. (a) The characteristic T-shaped hairpin structure of this AAV-ITR
sequence. Failed PCR amplification in the absence and presence of 2–15% DMSO (b) and 0.5–2 M betaine
(c) was shown by 3% agarose gel electrophoresis. (d) In contrast, the AAV-ITR was successfully amplified by
PCR in the presence of 2 μM disruptors
2 Materials
3 Methods
Fig. 3 The intramolecular secondary structures predicted by UNAFold web server for full-length AAV-ITR
amplicon (a) and trimmed AAV-ITR sequence (b). For the full-length amplicon, in addition to the T-shaped
hairpin structure (green triangle), the predicted structure contains another two standalone small hairpin
structures, which also contribute to the -33.51 kcal/mol free energy of the entire structure. To determine the
free energy of the T-shaped hairpin structure itself, the full-length amplicon is trimmed on both ends to leave
five nucleotides just outside of the T-shaped hairpin structure. As expected, only the same T-shaped hairpin
structure is predicted by the software for the trimmed sequence, and its free energy is calculated as -
32.49 kcal/mol
Fig. 4 The three parts of a disruptor exemplified with AAV-ITR sequence: effector
(red), anchor (green), and 3′ adduct. The AAV-ITR region complementary to both
anchor and effector sequences is marked in blue. The 3′ adduct can be any
chemical moiety that can prevent disruptor elongation by polymerase, such as
minor groove binders, inverted DNA nucleotides, 3-carbon spacers, etc.
Improve PCR Performance with Disruptors 167
3.4 Result Analysis 1. For real-time quantitative PCR, the software output files for
thermal cycler can be analyzed according to manufacturer’s
instructions.
2. For traditional PCR, routine agarose gel analysis can be used
for result analysis.
Improve PCR Performance with Disruptors 169
4 Notes
References
Abstract
Adulteration of dairy products, mainly through the substitution of high-quality milk for lower-quality milk,
results in the production of low-value products, raising health, social, and economic concerns. As such, the
development of methods to ensure dairy products’ safety and quality is of great concern for governments
and consumers. Although several methods have been developed for species differentiation in dairy pro-
ducts, their application and the establishment of reliable molecular markers for authentication purposes still
need to be improved. In this chapter, we describe a low-cost, sensitive, fast, and reliable PCR-based method
for mitochondrial D-loop DNA amplification for efficient detection of cattle milk in binary mixtures with
sheep milk, thereby allowing the authentication of processed dairy products.
Key words Species differentiation, Milk adulteration, Dairy authentication, DNA extraction and
purification, Food quality, Mitochondrial D-loop DNA
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_14,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
173
174 Marlene Baptista and Lucı́lia Domingues
2 Materials
7. Thermal cycler.
8. Agarose gel electrophoresis apparatus.
9. UV transilluminator with a gel documentation unit.
2.2 Supplies and 1. Milk samples stored at -20 °C (to prevent contamination and
Reagents degradation).
2. Clearing solution: 0.25 M EDTA, pH 8.0, 0.5% Triton X-100.
3. Extraction buffer: 10 mM Tris–HCl, pH 7.5, 150 mM NaCl,
2 mM EDTA, pH 8.0, and 1% SDS.
4. 5 M guanidine hydrochloride.
5. 20 mg/mL proteinase K.
6. Phenol-chloroform-isoamyl alcohol (25:24:1) pH 8.0.
7. Sterile ultrapure water.
8. Ethanol 100%.
9. Oligonucleotide primers: Ordered at a synthesis scale of
0.01 μmol and salt-free purification. Working solutions at
20 μM were prepared from a 100 μM stock solution to avoid
contamination of the stock and repeated freeze-thaw cycles.
10. Taq DNA polymerase: As an example, this protocol works
using the NZYTaq II DNA polymerase, supplied with a 10×
reaction buffer and a 50 mM MgCl2 solution (NZYTech,
Portugal).
11. dNTPs, 10 mM each.
12. 1X Tris-acetate-EDTA (TAE) buffer: 40 mM Tris, 20 mM
acetate, 1 mM EDTA, pH 8.5.
13. 1% (w/v) agarose gels (in 1× TAE buffer) with 0.05% (v/v)
GreenSafe Premium (NZYTech, Portugal), or equivalent.
14. Molecular weight marker, NZYDNA Ladder VII (NZYTech,
Portugal) or equivalent.
15. Gel Loading Dye, Purple or Blue (6X) (New England Biolabs,
USA), or equivalent.
16. QIAquick Gel Extraction Kit (Qiagen, Germany) or
equivalent.
3 Methods
3.1 Total DNA 1. Mix 1 mL of milk sample with 0.5 mL of clearing solution and
Extraction in Milk centrifuge at 12,500 × g for 5 min. Remove as much as possible
of the resulting cream pad and supernatant (see Note 1).
2. Lyse the pellet comprising the somatic cells by resuspending it
in 860 μL of extraction buffer, 100 μL of 5 M guanidine
hydrochloride, and 20 μL of 20 mg/mL proteinase K. Incubate
for 3 h or o/n (for higher efficiency) at 55 °C, 80 rpm.
176 Marlene Baptista and Lucı́lia Domingues
3.2 PCR 1. Prepare primers reported by [17] (Table 1), according to the
Amplification of description in step 9, Sect. 2.2.
Mitochondrial D-Loop 2. For the PCR amplification of the mitochondrial D-loop region
DNA from milk samples, prepare the following reaction mixture:
• 10× reaction buffer: 5 μL.
• 50 mM MgCl2: 4 μL.
• 10 mM dNTPs: 2.5 μL.
• 20 μM primer PRO: 2.5 μL.
• 20 μM primer PHE: 2.5 μL.
• 5 U/μL Taq polymerase: 0.25 μL.
• Template DNA: 200 ng.
• Sterile ultrapure water: up to 50 μL.
3. Perform the reaction in a thermal cycler as outlined in Table 2.
4. Load at least 5 μL of PCR product on 1% agarose gel in TAE
and run in the same buffer at 80 V for 90 min.
5. According to the number of bands visualized in the gel, it is
possible to infer the number of animal species present in the
milk sample. Moreover, the size of the bands could indicate the
species present by comparison with data deposited in the NCBI
for the mitochondrial D-loop region size (see Note 7).
Table 1
Primers for mitochondrial D-loop amplification
Table 2
PCR conditions for mitochondrial D-loop amplification
6. Excise the bands from the agarose gel, and purify the DNA
using the QIAquick Gel Extraction Kit, or equivalent. Send the
resulting purified fragments for sequencing (for instance, to
Eurofins Genomics) (see Note 8).
7. Perform sequence alignment with known mitochondrial
D-loop regions deposited in NCBI to identify the species.
4 Notes
Fig. 1 PCR products obtained from different binary mixtures of cow and ewe’s milk (CM and EM, respectively)
using primers PRO and PHE. MM, molecular marker; NTC, no template control. Lanes 1, 2, 3, 4, and 5 are from
mixtures containing 1% CM-99% EM, 5% CM-95% EM, 10% CM-90% EM, 25% CM-75% EM, and 50%
CM-50% EM of unpasteurized cow milk. Lanes 6, 7, 8, 9, and 10 are from mixtures containing identical
percentages of pasteurized cow milk
Acknowledgments
References
1. Lipkin E, Shalom A, Khatib H et al (1993) 9. Gonçalves J, Pereira F, Amorim A, van Asch B
Milk as a source of deoxyribonucleic acid and (2012) New method for the simultaneous
as a substrate for the polymerase chain reaction. identification of cow, sheep, goat, and water
J Dairy Sci 76:2025–2032 buffalo in dairy products by analysis of short
2. Liao J, Liu YF, Ku T et al (2017) Qualitative species- specific mitochondrial DNA targets. J
and quantitative identification of adulteration Agric Food Chem 60:10480–10485
of Milk powder using DNA extracted with a 10. Guo L, Qian JP, Guo YS et al (2018) Simulta-
novel method. J Dairy Sci 100:1657–1663 neous identification of bovine and equine DNA
3. Di Domenico M, Di Giuseppe M, Wicochea in milks and dairy products inferred from tri-
Rodrı́guez JD, Cammà C (2017) Validation of plex Taq man real-time PCR technique. J Dairy
a fast real-time PCR method to detect fraud Sci 101:6776–6786
and mislabeling in Milk and dairy products. J 11. Deng L, Li A, Gao Y et al (2020) Detection of
Dairy Sci 100:106–112 the bovine Milk adulterated in camel, horse,
4. López-Calleja I, González I, Fajardo V et al and goat Milk using duplex PCR. Food Anal
(2007) Quantitative detection of goats’ milk Methods 13:560–567. https://doi.org/10.
in sheep’s milk by real-time PCR. Food Con- 1007/s12161-019-01678-2
trol 18:1466–1473 12. Golinelli LP, Carvalho AC, Casaes RS et al
5. Lopparelli RM, Cardazzo B, Balzan S et al (2014) Sensory analysis and species-specific
(2007) Real-time TaqMan polymerase chain PCR detect bovine milk adulteration of frescal
reaction detection and quantification of cow (fresh) goat cheese. J Dairy Sci 97:6693–6699
DNA in pure water buffalo mozzarella cheese: 13. Abdel-Rahman S, Ahmed M (2007) Rapid and
method validation and its application on com- sensitive identification of buffalo’s, cattle’s and
mercial samples. J Agric Food Chem 55:3429– sheep’s milk using species-specific PCR and
3434. https://doi.org/10.1021/jf0637271 PCR–RFLP techniques. Food Control 18:
6. Rentsch J, Weibel S, Ruf J et al (2013) Inter- 1246–1249
laboratory validation of two multiplex quanti- 14. Plath A, Krause I, Einspanier R (1997) Species
tative real-time PCR methods to determine identification in dairy products by three differ-
species DNA of cow, sheep and goat as a mea- ent DNA-based techniques. Z Leb Unters
sure of milk proportions in cheese. Eur Food Forsch 205:437–441
Res Technol 236:217–227. https://doi.org/ 15. Baptista M, Cunha JT, Domingues L (2021)
10.1007/s00217-012-1880-y DNA-based approaches for dairy products
7. Cottenet G, Blancpain C, Golay PA (2011) authentication : a review and perspectives.
Simultaneous detection of cow and buffalo Trends Food Sci Technol 109:386–397.
species in milk from China, India, and https://doi.org/10.1016/j.tifs.2021.01.043
Pakistan using multiplex real-time PCR. J 16. Liu J, Ding X, Zeng Y et al (2016) Genetic
Dairy Sci 94:3787–3793. https://doi.org/10. diversity and phylogenetic evolution of Tibetan
3168/jds.2011-4195 sheep based on mtDNA D-loop sequences.
8. Bottero MT, Civera T, Nucera D et al (2003) A PLoS One 11:e0159308. https://doi.org/10.
multiplex polymerase chain reaction for the 1371/journal.pone.0159308
identification of cows’, goats’ and sheep’s 17. Wood NJ, Phua SH (1996) Variation in the
milk in dairy products. Int Dairy J 13:277– control region sequence of the sheep mito-
282. https://doi.org/10.1016/S0958-6946 chondrial genome. Anim Genet 27:25–33
(02)00170-X
Chapter 15
Abstract
Polymerase chain reaction (PCR) is a laboratory technique used to amplify a targeted region of DNA,
demarcated by a set of oligonucleotide primers. Long-range PCR is a form of PCR optimized to facilitate
the amplification of large fragments. Using the adapted long-range PCR protocol described in this chapter,
we were able to generate PCR products of 6.6, 7.2, 13, and 20 kb from human genomic DNA samples. For
some of the long PCRs, successful amplification was not possible without the use of PCR enhancers. Thus,
we also evaluated the impact of some enhancers on long-range PCR and included the findings as part of this
updated chapter.
Key words Long-range polymerase chain reaction (PCR), Long amplicons, PCR enhancers, PCR
additives, Agarose gel electrophoresis, DNA polymerase, Proofreading enzyme, Thermal cycling,
Primer design, Pharmacogenetics
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_15,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
181
182 Ping Siu Kee et al.
Fig. 1 Amplification of long PCR fragments from three pharmacogenes: CYP2A6 (left panel), CYP2D6 (middle
panel), and CYP2C19 (right panel). A–F represent different human genomic DNA samples. For sizing of PCR
products, GeneRuler 1 kb plus DNA ladder (Thermo Fisher Scientific, Baltics UAB, Lithuania) or KAPA Universal
Ladder (KAPA Biosystems, Roche Holdings, Inc., Basel, Switzerland) were used as markers
2 Materials
3 Methods
3.1 Primer Design The specificity and efficiency of a primer pair are one of the factors
contributing to a successful PCR, and they are heavily dependent
on two criteria: primer length and annealing temperature. For a
standard PCR, the ideal primer length ranges between 18 and
24 bases, with a suggested annealing temperature that is 54 °C or
higher and is usually a few degrees below the melting temperature
(Tm) [11]. For certain genes such as CYP2A6 and CYP2D6, the
presence of pseudogenes or genes with similar consensus sequences
further adds challenges to optimizing amplification. In such situa-
tions, primers that are highly specific are required.
2. The search will extract the targeted gene RefSeq for all available
species, each assigned with a unique gene ID. Locate and select
the Refseq of interest, which will bring the user to a subsequent
page with comprehensive information on the selected Refseq.
3. Go to the “Genomic regions, transcripts, and products” tab.
From the dropdown list, select the appropriate genome assem-
bly (see Note 11). After that, select the “FASTA” hyperlink
option, located next to the genome assembly.
4. Update the FASTA sequence shown to include 500 bp on both
sides of the gene. This can be done by changing the coverage of
genomic region numbers at the top right column labeled
“Change region shown.”
5. Download the FASTA sequence using the “Send to” option.
3.1.2 Obtaining Specific 1. Copy or upload the updated FASTA sequence onto an online
Primer Sequences (Fig. tool for primer design, Primer-BLAST; this tool can be
2 (See Note 12)) accessed at http://www.ncbi.nlm.nih.gov/tools/primer-
blast/ [13].
2. Specify the desired primer positions relative to the inserted
FASTA sequence. As we have allowed for 500-bp flanking
sequences on both sides of the gene, forward and reverse
primers can start anywhere within the 500-bp window. The
primers should be positioned in such a manner that they are at
least 150 bp away from the first and last exons to cover the
entire gene of interest.
Fig. 2 A simple guide to primer design using Primer-BLAST, a free Web-based tool. Circled numbers refer to
steps described in the text
Long PCR 185
3.2 PCR 1. Prepare all reaction mixes inside a horizontal laminar flow
hood. Also, it is important to have strictly separate areas for
pre- and post-PCR work to reduce the chances of sample
contamination with amplified products.
2. Irradiate the tools (e.g., pipettes and pipette tips) and plastic-
ware (e.g., microfuge tubes and PCR strip tubes) used for PCR
setup with ultraviolet light for about 15–20 min, in order to
minimize the risk of carryover contamination [14–16].
3. While waiting for the decontamination procedure to finish, set
up the PCR protocol in a thermal cycler (see Note 14).
4. Add, in a 0.7 mL microfuge tube (or larger if required), the
following reagents: reaction buffer, magnesium chloride,
dNTP mix, primers, enhancers (if necessary, see Note 8), and
enzyme mix (see Note 15).
5. Briefly flick the tube to mix all reaction components, and spin
down the mixture with a 5- to 10-s pulse in a microcentrifuge.
6. Divide the master mix into equal-volume aliquots, and transfer
these into individual PCR strip tubes (see Notes 16 and 17).
Add template DNAs (or PCR-grade water for no-template
controls; see Note 15). Always keep the tubes at 4 °C (on ice
or in a cold block) prior to thermal cycling, in order to mini-
mize unspecific amplification that may arise from residual Taq
polymerase activity at low temperatures. Some Taq polymerases
branded as “hot-start” have been found to be not entirely
devoid of enzymatic activity prior to heat activation [17].
186 Ping Siu Kee et al.
7. Close the strip tubes with their lids. Make sure that a tight seal
is formed to prevent evaporation of the reaction mixes during
thermal cycling.
8. Transfer the tubes into the thermal cycler and initiate the
cycling protocol. Note the positions in which the tubes are
placed within the thermal block (see Note 18).
9. At the end of the cycling protocol, remove the tubes from the
thermal cycler. Store the products at 4 °C until further analysis
or other downstream applications such as DNA sequencing.
3.3 Agarose Gel 1. On an electronic scale, weigh out the required amount of
Electrophoresis (See agarose gel powder (see Note 20).
Note 19) 2. Add the agarose powder into an adequate volume of 1× TAE
buffer in a glass bottle and swirl briefly to mix.
3. Heat the mixture in a microwave oven until it begins to boil.
The bottle should only be loosely capped to avoid a dangerous
build-up of pressure.
4. Remove the bottle from the microwave oven and gently swirl
the partially molten agarose mixture. Wear a pair of heatproof
gloves and safety glasses while doing this.
5. Replace the bottle in the microwave, and repeat steps 3 and
4 twice or until the agarose powder completely dissolves in the
buffer. At this point the cloudy mixture should turn into a clear
solution.
6. Allow the agarose solution to stand on the bench or in a water
bath until it has cooled to ~50 °C. Alternatively, hold the bottle
under cold running tap water, and gently swirl the molten
agarose solution to make sure that it is thoroughly cooled.
Do not overcool as the agarose solution will begin to solidify
(see Note 21).
7. Add an adequate amount of nucleic acid stain (see Note 22),
swirl to mix, and set aside the stained agarose solution to cool
further.
8. Place the gel tray, in its casting position, into the buffer cham-
ber. Pour the warm molten agarose solution onto the gel tray
and insert a well comb. Do not pour the agarose solution while
it is still hot as this may cause the gel tray to warp. Check for
and remove any bubbles inside the gel solution with a clean
pipette tip.
9. Once the gel solidifies, remove the comb, and add the TAE
buffer until it covers the entire gel to a depth of about 1 mm.
Make sure that all the wells are properly formed.
Long PCR 187
4 Notes
Table 1
The recommended range of concentrations for betaine, DTT, and trehalose as PCR enhancers
Recommended
PCR
Mechanism(s) of action Tested concentrations range of
enhancers
concentrations
Fig. 3 The effect of different amounts of input DNA on the CYP2D6 long-range
PCR, performed in 10 μL reactions, was ascertained. It is obvious that 40 ng/
10 μL of DNA resulted in greater amplification than 20 ng/10 μL. Increasing the
concentration to 80 ng/10 μL did not enhance the yield further; doubling it to
160 ng/10 μL was clearly detrimental. The samples used in these experiments
were human genomic DNAs
References
1. Mullis KB, Faloona FA (1987) Specific synthe- Protocols. Humana Press, Totowa, NJ, pp
sis of DNA in vitro via a polymerase-catalyzed 1–29
chain reaction. In: Methods in enzymology, vol 5. Long S (2022). Digital PCR: methods and
155. Academic Press, pp 335–350 applications in infectious diseases. Methods
2. Saiki RK, Gelfand DH, Stoffel S, Scharf SJ, (San Diego, Calif.)
Higuchi R, Horn GT, Mullis KB, Erlich HA 6. Zhu H, Zhang H, Xu Y, Laššáková S,
(1988) Primer-directed enzymatic amplifica- Korabečná M, Neužil P (2020) PCR past, pres-
tion of DNA with a thermostable DNA poly- ent and future. BioTechniques 69(4):317–325
merase. Science 239(4839):487–491 7. Green MR, Sambrook J (2019) Long and
3. Collins F, Galas D (1993) A new five-year plan accurate polymerase chain reaction (LA PCR).
for the US human genome project. Science Cold Spring Harb Protoc 2019(3):pdb-
262(5130):43–46 prot095158
4. Delidow BC, Lynch JP, Peluso JJ, White BA 8. Barnes WM (1994) PCR amplification of up to
(1993) Polymerase chain reaction. In: PCR 35-kb DNA with high fidelity and high yield
192 Ping Siu Kee et al.
Abstract
Megaprimer-based polymerase chain reaction (PCR) strategies allow the versatile and fast assembly and
amplification of a myriad of tailor-made or random DNA sequences readily available for conventional or
restriction-free (RF) cloning.
In this chapter, we present a megaprimer-based PCR protocol that enables the expeditious construction
of customized fusion genes ready for cloning into commercial expression plasmids. With the expanding use
of protein tag technology in the most diverse application fields, this protocol remains a versatile and
affordable solution for the synthesis and fusion of peptide tags/domains of interest.
Key words Megaprimers, PCR, Fusion genes, Molecular cloning, Restriction enzymes, Expression
plasmids
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_16,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
193
194 Tatiana Q. Aguiar et al.
Length Tm
Primer Sequence (5‘ → 3‘)a (bp) (°C)
P1 EmGFP _FW cggaattcatggtgagcaagggcgag 26 59
P2 TrCBM1Cel7A_RV1.1 CCACCACATTGACCGTAATGAGATTGAGTACCTTGAAAGTACAAGTTTTCcttgtacagctcg 70 56
tccatgc
P3 TrCBM1Cel7A_RV1.2 CCACCACATTGACCGTAATGAGATTGAGTTGGACCTGGAGAAGAACCAGTAGTA 142 56
GTAGCTGGTCTTCTAGTAGTAGTAGTACCTCTGTTACCACCTGGTGGACCTT
GAAAGTACAAGTTTTCcttgtacagctcgtccatgc
P4 TrCBM1Cel7A_RV2 ggggtaccTCAatgatgatgatgatgatgCAAACATTGAGAGTAGTATGGGTTCAAAACTTGACAAG 131 57
TAGTACCAGAAGCACAAACAGTTGGACCAGAGTAACCAATAccaccacattgaccgtaatgaga
T7 T7_FW GATCCCGCGAAATTAATACGACTCACTATAG 31 56
MR EGFP-NdeI-Car9- ccgctcgagTTAACGTTTACCCGGTTTTTTGAAACCACGAGCAGAGTC 81 56
XhoI_R catatgtttgtaaagttcatccatacccagggt
a
Simple lower and UPPER case refer to the sequences complementary to the EmGFP- and CBM1-coding regions, respectively. BOLD UPPER CASE refers to the sequence
encoding the CBM1 native linker. Underlined UPPER and lower case refer to the sequence encoding a TEV recognition site and a six histidine tag, respectively. The stop codon is
indicated in UPPER CASE ITALICS. The recognition sites for the selected restriction enzymes are indicated in lower case bold (EcoRI, gaattc; KpnI, ggtacc; NdeI, catatg; and
XhoI, ctcgag), and the nucleotides added upstream of them are in lower case italics
Megaprimer-Based PCR
195
196 Tatiana Q. Aguiar et al.
Fig. 1 Illustration of the megaprimer-based PCR strategy used to assemble the fusion genes EmGFP-
TrCBM1Cel7A (a) and EmGFP-NL-TrCBM1Cel7A (b), with indication of the location of primers and of all the
extra sequence elements added after each PCR. TEV, TEV protease recognition site; S, stop codon
2 Materials
Fig. 2 Illustration of the megaprimer-based PCR strategy used to assemble the fusion gene EGFP-Car9, with
indication of the location of primers and of all the extra sequence elements added after the PCR. S, stop codon
3 Methods
3.1 Primer Design Rational primer design is a critical step for the success of this
protocol, which requires prior knowledge of the DNA sequences
of the genes and elements that we desire to fuse, as well as the maps
of the expression vectors where we intend to clone the constructed
fusions:
1. Assemble the final base pair sequence of the desired fusion
genes, and optimize their codons according to the preferential
codon usage of the host where they are going to be expressed.
For the first case study, the codons of the sequences that were
fused to the EmGFP-coding gene were optimized for expres-
sion in the yeast P. pastoris using the online software GENEius
(Eurofins Genomics) (Fig. 3). For the second case study, the
same software was used to optimize the codons of the Car9-
coding sequence for expression in the bacterium E. coli (see
Note 1).
2. Select the appropriate enzymes for cloning. Analyze the multi-
ple cloning site of the selected expression vector and the restric-
tion map of each fusion gene (e.g., with the online NEBcutter
tool, New England Biolabs). Select two enzymes that do not
cut in the fusion genes, and allow their directional cloning into
the desired vector region in frame with the existing reading
frame (see Note 2). For the first case study, the enzymes EcoRI
and KpnI were selected to directionally clone the EmGFP-
TrCBM1Cel7A and EmGFP-NL-TrCBM1Cel7A fusions in frame
with the Saccharomyces cerevisiae α-factor signal peptide-coding
sequence in the commercial expression plasmid pPICZαA
(Invitrogen). For the second case study, the restriction
enzymes XbaI and XhoI were selected to directionally clone
the Car9 tag in frame with the EGFP-coding sequence in the
E. coli expression plasmid pETM20 (EMBL).
Megaprimer-Based PCR 199
Fig. 3 Sequences of the customized fusion genes EmGFP-TrCBM1Cel7A (a) and EmGFP-NL-TrCBM1Cel7A (b)
with the codons optimized for expression in P. pastoris. The EmGFP and CBM1 regions are highlighted in green
and gold, respectively. The CBM1 NL region is highlighted in gray. The TEV recognition site and the histidine
tag are highlighted in pink and blue, respectively. The stop codon is indicated in red, and the priming sites for
the designed primers are underlined and highlighted in bold
200 Tatiana Q. Aguiar et al.
3.2 Double-Step A two-step PCR was performed to synthesize the fusion genes
Primer Extension PCR: EmGFP-TrCBM1Cel7A and EmGFP-NL-TrCBM1Cel7A, in which
Case Study 1 the PCR products from the first PCR round served as template
for the second PCR (Table 2).
1. To obtain the PCR1 and PCR3 products (Table 2, Fig. 1), mix
the following components in PCR tubes:
• Template DNA—100–300 ng (in 1 μL)
• Primer P1 (25 μM)—1 μL
• Primer P2 for PCR1 and P3 for PCR3 (25 μM)—1 μL
• dNTPs (10 mM each)—1.5 μL
• 10× ThermoPol® Reaction Buffer—5 μL
• 100 mM MgSO4—1 μL
• Ultrapure H2O—39 μL
• Vent® DNA Polymerase—0.5 μL
Table 2
Primers, template DNA, and annealing temperature (Ta) used for the assembly and amplification of
the fusion genes EmGFP-TrCBM1Cel7A and EmGFP-NL-TrCBM1Cel7A in case study 1
Fig. 4 Final PCR products from the amplification of the fusion genes EmGFP-
TrCBM1Cel7 (A, PCR2 product) and EmGFP-NL-TrCBM1Cel7A (B, PCR4 product) in
case study 1 and EGFP-Car9 (C) in case study 2. MW, molecular weight
standards
Megaprimer-Based PCR 203
3.3 Standard PCR: A standard PCR was performed to synthesize the fusion gene
Case Study 2 EGFP-Car9:
1. To obtain the PCR product (Table 2, Fig. 2), mix the following
components in a PCR tube:
• Template DNA—100–300 ng (in 1 μL)
• Primer T7 (20 μM)—1.25 μL
• Primer MR (20 μM)—1.25 μL
• dNTPs (10 mM each)—1 μL
• 10× ThermoPol® Reaction Buffer—5 μL
• 100 mM MgSO4—1 μL
• Ultrapure H2O—39 μL
• Vent® DNA Polymerase—0.5 μL
2. Step up the thermal cycler using the following parameters:
3.4 Vector Cloning The final PCR products can be digested with the planned restric-
and Confirmation of tion enzymes and cloned directly into the expression vector or can
the Fusion Genes’ alternatively be cloned into an intermediate cloning vector, propa-
Sequence gated, excised by restriction digestion, and finally cloned into the
expression vector. Many convenient commercial systems for clon-
ing of PCR products are available, either for blunt-ended (e.g.,
pMOS®, GE healthcare) or A-ended products (e.g., pGEM®-T,
Promega). Thermostable DNA polymerases with proofreading
activity, such as Vent®, generate blunt-ended fragments, but these
can be modified using an A-tailing procedure (see Note 9) and thus
cloned in any type of PCR cloning vectors. However, direct cloning
is a faster and less expensive approach:
1. Digest overnight at 37 °C the purified PCR2 and PCR4 pro-
ducts from case study 1 with the cloning enzymes EcoRI-HF
and KpnI-HF and the purified PCR product from case study
2 with the cloning enzymes XbaI and XhoI, using the following
reaction mixture:
• Purified PCR product—25 μL
204 Tatiana Q. Aguiar et al.
4 Notes
Acknowledgments
References
1. Oliveira C, Aguiar TQ, Domingues L (2017) 5. Baptista SL, Romanı́ A, Oliveira C, Ferreira S,
Principles of genetic engineering. In: Rocha CMR, Domingues L (2021) Galactose
Pandey A, Teixeira JA (eds) Current develop- to tagatose isomerization by the L-arabinose
ments in biotechnology and bioengineering: isomerase from Bacillus subtilis: a biorefinery
foundations of biotechnology and bioengi- approach for Gelidium sesquipedale valorisa-
neering, 1st edn. Elsevier, Oxford tion. LWT-Food Sci Technol 151:112199
2. Li X, Jin J, Guo Z, Liu L (2022) Evolution of 6. Bandyopadhyay B, Peleg Y (2018) Facilitating
plasmid-construction. Int J Biol Macromol circular permutation using restriction free
209(Pt a):1319–1326 (RF) cloning. Protein Eng Des Sel 31(3):
3. Oliveira C, Sepúlveda G, Aguiar TQ, Gama 65–68
FM, Domingues L (2015) Modification of 7. Sonnendecker C, Zimmermann W (2019)
paper properties using carbohydrate-binding Domain shuffling of cyclodextrin glucano-
module 3 from the Clostridium thermocellum transferases for tailored product specificity and
CipA scaffolding protein produced in Pichia thermal stability. FEBS Open Bio 9:384–395
pastoris: elucidation of the glycosylation effect. 8. Binte Muhammad Jai HS, Dam LC, Tay LS,
Cellulose 22:2755–2765 Koh JJW, Loo HL, Kline KA, Goh BC (2020)
4. Freitas AI, Domingues L, Aguiar TQ (2022) Engineered lysins with customized lytic activ-
Bare silica as an alternative matrix for affinity ities against enterococci and staphylococci.
purification/immobilization of His-tagged Front Microbiol 11:574739
proteins. Sep Pur Technol 286:120448
Megaprimer-Based PCR 207
9. Costa CE, Møller-Hansen I, Romanı́ A, Teix- proteins. Appl Microbiol Biotechnol 102(1):
eira JA, Borodina I, Domingues L (2021) Res- 81–92
veratrol production from hydrothermally 14. Freitas AI, Domingues L, Aguiar TQ (2021)
pretreated Eucalyptus wood using recombi- Tag-mediated single-step purification and
nant industrial Saccharomyces cerevisiae strains. immobilization of recombinant proteins
ACS Synth Biol 10(8):1895–1903 toward protein-engineered advanced materials.
10. Baptista SL, Cunha JT, Romanı́ A, Domingues J Adv Res 36:249–264
L (2018) Xylitol production from lignocellu- 15. Aguiar TQ, Oliveira C, Domingues L (2017)
losic whole slurry corn cob by engineered Synthesis of fusion genes for cloning by
industrial Saccharomyces cerevisiae PE-2. Bior- megaprimer-based PCR. In: Domingues L
esour Technol 267:481–491 (ed) PCR: methods and protocols, Methods
11. Silva R, Aguiar TQ, Coelho C, Jiménez A, in molecular biology, vol 1620. Springer,
Revuelta JL, Domingues L (2019) Metabolic New York, NY
engineering of Ashbya gossypii for deciphering 16. Wan W, Wang DM, Gao XL, Hong J (2011)
the de novo biosynthesis of γ-lactones. Microb Expression of family 3 cellulose-binding mod-
Cell Factories 18:62 ule (CBM3) as an affinity tag for recombinant
12. Forloni M, Liu AY, Wajapeyee N (2019) Mega- proteins in yeast. Appl Microbiol Biotechnol
primer polymerase chain reaction (PCR)-based 91:789–798
mutagenesis. Cold Spring Harb Protoc 2019:6 17. Azevedo F, Pereira H, Johansson B (2017)
13. Oliveira C, Domingues L (2018) Guidelines to Colony PCR. In: Domingues L (ed) PCR:
reach high-quality purified recombinant methods and protocols, Methods in Molecular
Biology, vol 1620. Springer, New York, NY
Chapter 17
Abstract
The bacteria Escherichia coli and the yeast Saccharomyces cerevisiae are currently the two most important
organisms in synthetic biology. E. coli is almost always used for fundamental DNA manipulation, while yeast
is the simplest host system for studying eukaryotic gene expression and performing large-scale DNA
assembly. Yeast expression studies may also require altering the chromosomal DNA by homologous
recombination. All these studies require the verification of the expected DNA sequence, and the fastest
method of screening is colony PCR, which is direct PCR of DNA in cells without prior DNA purification.
Colony PCR is hampered by the difficulty of releasing DNA into the PCR mix and by the presence of PCR
inhibitors. We hereby present one protocol for E. coli and two protocols for S. cerevisiae differing in
efficiency and complexity as well as an overview of past and possible future developments of efficient
S. cerevisiae colony PCR protocols
Key words PCR, Colony, Yeast, Saccharomyces cerevisiae, Escherichia coli, Direct lysis
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_17,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
209
210 Humberto Pereira et al.
1.1 DNA Release The addition of preincubation with a yeast lytic enzyme such as
zymolyase or lyticase can improve efficiency [11, 12]. Lyticase usu-
ally refers to pure β-1,3-glucanase, while zymolyase is a mixture of
lytic enzymes. The factor targeted by this enzyme is the strong yeast
cell wall which is weakened or removed. The downside is the
enzyme cost and potentially the addition of phosphate in the incu-
bation buffer, which may lead to PCR inhibition by interaction with
the magnesium ions in the PCR buffer. Recombinant lyticase from
the bacteria Oerskovia xanthineolytica is easily produced by the
cultivation of cells harboring a plasmid carrying the glucanase
structural gene [13]. The resulting lyticase is cost-effective, but
PCR strategies should be designed with care since the resulting
enzyme is often contaminated with the expression plasmid and
E. coli chromosomal DNA. We previously used a protocol based
on homemade recombinant lyticase, but while effective, not ulti-
mately considered worth the extra work unless lyticase has some
other use in the laboratory.
A brief treatment of cells with sodium hydroxide [14] is a
method that has several potential targets. The authors suggest
that the modes of action could be increased cell wall permeability,
dissociation of DNA from bound proteins, or degradation of RNA.
Additionally, sodium hydroxide might neutralize intercalated PCR
inhibitors by denaturing DNA [15]. The addition of the strong
anionic detergent sodium dodecyl sulfate (SDS) alone [16] or in
combination with ethanol [17] or lithium acetate (LiAc) [10] has
also been described as a method for achieving PCR amplification
from whole yeast cells. SDS efficiently dissolves membrane lipids
but is also a potent PCR inhibitor [18]. The presence of SDS also
potentially eliminates DNA-protein interactions as SDS is used to
prevent gel shifts in the electrophoresis of DNA. Ethanol would
precipitate DNA as soon as it is liberated from the cells and may be a
way to selectively wash away inhibitors and concentrate DNA
[17]. LiAc is commonly used in yeast transformation [19], where
the mode of action may be to turn the cell wall more porous [20],
which probably improves cell lysis.
212 Humberto Pereira et al.
the yield of PCR in general [43] and also neutralize the negative
effects of sodium dodecyl sulfate (SDS) [44]. Several mono- and
disaccharides were recently reported to be effective PCR enhancers,
with sucrose surpassing trehalose and DMSO for the conditions
tested [45].
Relatively new PCR enhancers are nanoparticles from gold
(AuNPs) [46, 47], titanium dioxide (TiO2) [48], and graphene
oxide (GO) or reduced graphene oxide (rGO) [49]. The mode of
action of nanoparticles has not been elucidated in detail. Finally,
attempts have been made to combine several enhancers in an
attempt to find synergistic positive effects [50–52].
The PCR reagents can be altered in order to enhance PCR
specificity. This was observed when locked nucleic acid (LNA)-
modified primers were used instead of unmodified oligonucleotides
[53]. Replacing the canonical dNTPs with 2′-deoxynucleoside
5′-(alpha-P-seleno)-triphosphates (dNTPαSe) [54] was capable of
increasing PCR specificity by over 240-fold [55].
2 Materials
2.1 E. coli Colony 1. Water. PCR components and other solutions should be
PCR prepared using the best available water. We routinely use
double-deionized water with a specific conductance of
18.2 MΩ/cm at 25 °C.
2. 2× PCR master mix with DMSO (Table 1). We have found it
practical to prepare a two times concentrated PCR master mix
containing all components except PCR primers and template
DNA, as this minimizes pipetting errors and improves consis-
tency across PCR experiments. The PCR master mix can be
stored at -20 °C without a noticeable loss of efficiency. We
routinely include 1% DMSO in the final PCR mixture.
Table 1
Recipe for 1 mL twice concentrated PCR master mix containing 2% DMSO
suitable for colony PCR
Table 2
Recipe for 5× PCR compatible loading buffer
Component Volume
25% Ficoll 10 mL
Tartrazine food coloring 1 mL
Xylene cyanol 125 mg/mL 10 μL
2.3 PCR Using S. All the items listed on Subheading 2.1 plus:
cerevisiae LiAc
1. 1 M lithium acetate stock solution. The lithium acetate solu-
Permeabilized Cells
tion is prepared as a 1 M stock in water. Add 10.2 g lithium
acetate dihydrate (LiOAc*2H2O, Mw 102.02 g/mol) in
80 mL water and dissolve. Add water to 100 mL and autoclave.
2. SDS stock solution 20% (w/v). Add 10 g SDS to 40 mL
H2O. Heat to 60 °C to dissolve the SDS. Adjust pH to 7–8
using sodium hydroxide. Adjust volume to 50 mL with water.
Do not autoclave as SDS will precipitate.
3. LiOAc-SDS solution. Mix 75 μL water, 20 μL 1 M LiOAc, and
5 μL 20% (w/v) SDS for each DNA extraction. Aliquot 100 μL
in 1.5 mL microcentrifuge tubes.
Bacteria and Yeast Colony PCR 215
3 Methods
3.1 E. coli Colony This protocol can be used to amplify new constructs in E. coli
PCR transformants. We have found it efficient to use a three-primer
strategy using two vector-specific primers flanking the insertion
location of the insert and one gene-specific primer, usually one of
the primers used to amplify the insert (Fig. 1). The two vector-
specific primers should differ in the distance to the insertion site by
200–400 bp. Using this strategy, an empty clone will produce a
short PCR product corresponding to the distance between the
vector-specific primers, while one of two longer bands will arise
from a successful clone, depending on the orientation of a cloned
insert (see Note 1):
1. Prepare a 1× PCR master mix containing all PCR components
except template DNA. We use a homemade 2× PCR master
mix containing DNA polymerase, buffer, Mg2+, dNTPs, and
DMSO to which PCR primers are added to a final concentra-
tion of 1 μM and water. We prepare 110% of the theoretical
Fig. 1 Illustration of three-primer strategy for confirming cloning results. The annealing primer locations
(represented in purple) will be different depending on the outcome: (a) plasmid containing an insert with the
desired orientation; (b) plasmid containing an insert with the inverse orientation; (c) plasmid without insert
216 Humberto Pereira et al.
3.2 S. cerevisiae This protocol usually represents the best compromise between cost,
Colony PCR Using a work, and success rate and should probably be the first protocol
Microwave Oven tested for a laboratory wishing to implement S. cerevisiae colony
PCR. We have found it to be efficient for PCR products up to 2 kb,
with occasional success for products up to 3 kb in size:
1. Prepare 1× PCR master mix according to the same principles as
for the E. coli protocol (Subheading 3.1).
2. Pick a small, well-isolated colony with a sterile toothpick or a
sterile 200 μL pipette tip (see Notes 3 and 6).
3. Transfer part of the colony to the side of a PCR tube. The most
common mistake is to transfer too much cell material to the
tube. We usually swirl the toothpick on the inside of the tube.
4. Transfer the remaining cells on the toothpick to fresh solid or
liquid medium.
5. Incubate the tubes for 1–2 min at full power (800–1000 W)
using a stock domestic microwave oven.
Bacteria and Yeast Colony PCR 217
3.3 PCR Using S. This protocol may not qualify as colony PCR, as DNA is effectively
cerevisiae LiAc purified from the cells. However, this protocol is considerably less
Permeabilized Cells laborious than methods relying on any combination of glass beads,
phenol, and chloroform. In our hands, this protocol has succeeded
where the microwave oven protocol (Subheading 3.2) failed. This
protocol has given more stable results, especially in the hands of less
experienced workers. This protocol was first described by Lõoke
et al. [10]:
1. Prepare one tube of 100 μL LiOAc-SDS mix for each colony.
2. Transfer a small colony from a plate using a sterile toothpick
(see Note 6). The toothpick can also be used to inoculate liquid
or solid medium to preserve the clone.
3. Vortex the tubes briefly and incubate at >70 °C for 10 min (see
Note 8).
4. Add 300 μL of 96% ethanol and vortex briefly to
precipitate DNA.
5. Spin tubes at least 15,000 g in a microcentrifuge for 3–5 min to
precipitate DNA. The cells and cell debris will coprecipitate
with the DNA at this point.
6. Remove liquid by inverting the tubes.
7. Add 500 μL 70% ethanol to each tube.
8. Spin tubes like in step 5.
9. Remove liquid by inverting the tubes. Try to remove as much
of the liquid as possible in this step (see Note 9).
10. Resuspend the DNA in 100 μL TE buffer (see Note 10).
11. Spin down the cell debris for 1 min at top speed in a
microcentrifuge.
12. Use 1 μL of the supernatant for 20 μL of total PCR volume.
13. Transfer about half of the supernatant to a fresh tube and store
the DNA at -20 °C.
14. Run the PCR program and analyze 5–10 μL of the PCR
product by gel electrophoresis.
218 Humberto Pereira et al.
4 Notes
Acknowledgments
References
1. Güssow D, Clackson T (1989) Direct clone compartments for increased production of the
characterization from plaques and colonies by lipophilic natural products ginsenosides.
the polymerase chain reaction. Nucleic Acids Metab Eng 67:104–111
Res 17:4000 6. Wang S, Xu X, Lv X, Liu Y, Li J, Du G, Liu L
2. Gibson DG, Benders GA, Axelrod KC, (2022) Construction and Optimization of the
Zaveri J, Algire MA, Moodie M, Montague de novo Biosynthesis Pathway of Mogrol in
MG, Venter JC, Smith HO, Hutchison CA Saccharomyces Cerevisiae. Front Bioeng Bio-
(2008) One-step assembly in yeast of 25 over- technol 10:919526
lapping DNA fragments to form a complete 7. Pereira H, Azevedo F, Domingues L, Johans-
synthetic Mycoplasma genitalium genome. son B (2022) Expression of Yarrowia lipolytica
Proc Natl Acad Sci 105:20404–20409 acetyl-CoA carboxylase in Saccharomyces cerevi-
3. Pereira F, Azevedo F, Parachin NS, Hahn- siae and its effect on in-vivo accumulation of
H€agerdal B, Gorwa-Grauslund MF, Johansson Malonyl-CoA. Comput Struct Biotechnol J 20:
B (2016) Yeast Pathway Kit: A Method for 779–787
Metabolic Pathway Assembly with Automati- 8. Bonnet C, Rigaud C, Chanteclaire E,
cally Simulated Executable Documentation. Blandais C, Tassy-Freches E, Arico C, Javaud
ACS Synth Biol 5:386–394 C (2013) PCR on yeast colonies: an improved
4. Cataldo VF, Salgado V, Saa PA, Agosin E method for glyco-engineered Saccharomyces
(2020) Genomic integration of unclonable cerevisiae. BMC Res Notes 6:201
gene expression cassettes in Saccharomyces cer- 9. Sathe GM, O’Brien S, McLaughlin MM,
evisiae using rapid cloning-free workflows. Watson F, Livi GP (1991) Use of polymerase
Microbiologyopen 9:e978 chain reaction for rapid detection of gene inser-
5. Shi Y, Wang D, Li R, Huang L, Dai Z, Zhang X tions in whole yeast cells. Nucleic Acids Res 19:
(2021) Engineering yeast subcellular 4775
220 Humberto Pereira et al.
10. Lõoke M, Kristjuhan K, Kristjuhan A (2011) 23. Walsh PS, Metzger DA, Higuchi R (1991)
Extraction of genomic DNA from yeasts for Chelex 100 as a medium for simple extraction
PCR-based applications. BioTechniques 50: of DNA for PCR-based typing from forensic
325–328 material. BioTechniques 10:506–513
11. Ling M, Merante F, Robinson BH (1995) A 24. Jamal MAHM, Sharma SP, Chung H-J, Kim
rapid and reliable DNA preparation method for H-J, Hong S-T, Lee S (2017) Ultra-High Effi-
screening a large number of yeast clones by cient Colony PCR for High Throughput
polymerase chain reaction. Nucleic Acids Res Screening of Bacterial Genes. Indian J Micro-
23:4924–4925 biol 57:365–369
12. Kwiatkowski TJ Jr, Zoghbi HY, Ledbetter SA, 25. Liu D, Zeng X-A, Sun D-W, Han Z (2013)
Ellison KA, Chinault AC (1990) Rapid identi- Disruption and protein release by ultrasonica-
fication of yeast artificial chromosome clones tion of yeast cells. Innov Food Sci Emerg Tech-
by matrix pooling and crude lysate PCR. nol 18:132–137
Nucleic Acids Res 18:7191–7192 26. Kermekchiev MB, Kirilova LI, Vail EE, Barnes
13. Shen SH, Chrétien P, Bastien L, Slilaty SN WM (2009) Mutants of Taq DNA polymerase
(1991) Primary sequence of the glucanase resistant to PCR inhibitors allow DNA amplifi-
gene from Oerskovia xanthineolytica. Expres- cation from whole blood and crude soil sam-
sion and purification of the enzyme from ples. Nucleic Acids Res 37:e40
Escherichia coli. J Biol Chem 266(1991): 27. Wang Y, Prosen DE, Mei L, Sullivan JC,
1058–1063 Finney M, Horn PBV (2004) A novel strategy
14. Wang H, Kohalmi SE, Cutler AJ (1996) An to engineer DNA polymerases for enhanced
improved method for polymerase chain reac- processivity and improved performance
tion using whole yeast cells. Anal Biochem 237: in vitro. Nucleic Acids Res 32:1197–1207
145–146 28. Ghadessy FJ, Ong JL, Holliger P (2001)
15. Bourke MT, Scherczinger CA, Ladd C, Lee Directed evolution of polymerase function by
HC (1999) NaOH treatment to neutralize compartmentalized self-replication. Proc Natl
inhibitors of Taq polymerase. J Forensic Sci Acad Sci U S A 98:4552–4557
44:1046–1050 29. Baar C, d’Abbadie M, Vaisman A, Arana ME,
16. Akada R, Murakane T, Nishizawa Y (2000) Hofreiter M, Woodgate R, Kunkel TA, Holli-
DNA extraction method for screening yeast ger P (2011) Molecular breeding of poly-
clones by PCR. Biotechniques 28(2000): merases for resistance to environmental
668–670, 672, 674 inhibitors. Nucleic Acids Res 39:e51
17. Linke B, Schröder K, Arter J, Gasperazzo T, 30. Winship PR (1989) An improved method for
Woehlecke H, Ehwald R (2010) Extraction of directly sequencing PCR amplified material
nucleic acids from yeast cells and plant tissues using dimethyl sulphoxide. Nucleic Acids Res
using ethanol as medium for sample preserva- 17:1266
tion and cell disruption. BioTechniques 49: 31. Varadaraj K, Skinner DM (1994) Denaturants
655–657 or cosolvents improve the specificity of PCR
18. Rossen L, Nørskov P, Holmstrøm K, Rasmus- amplification of a G + C-rich DNA using genet-
sen OF (1992) Inhibition of PCR by compo- ically engineered DNA polymerases. Gene 140:
nents of food samples, microbial diagnostic 1–5
assays and DNA-extraction solutions. Int J 32. Henke W, Herdel K, Jung K, Schnorr D, Loen-
Food Microbiol 17:37–45 ing SA (1997) Betaine improves the PCR
19. Gietz RD, Schiestl RH (2007) High-efficiency amplification of GC-rich DNA sequences.
yeast transformation using the LiAc/SS carrier Nucleic Acids Res 25:3957–3958
DNA/PEG method. Nat Protoc 2:31–34 33. Hengen PN (1997) Optimizing multiplex and
20. Pham TA, Kawai S, Murata K (2011) Visuali- LA-PCR with betaine. Trends Biochem Sci 22:
zation of the synergistic effect of lithium ace- 225–226
tate and single-stranded carrier DNA on 34. Mytelka DS, Chamberlin MJ (1996) Analysis
Saccharomyces cerevisiae transformation. Curr and suppression of DNA polymerase pauses
Genet 57:233–239 associated with a trinucleotide consensus.
21. Harju S, Fedosyuk H, Peterson KR (2004) Nucleic Acids Res 24:2774–2781
Rapid isolation of yeast genomic DNA: Bust 35. Frackman S, Kobs G, Simpson D, Storts D
n’ Grab. BMC Biotechnol 4:8 (1998) Others, Betaine and DMSO: enhancing
22. Blount BA, Driessen MRM, Ellis T (2016) GC agents for PCR. Promega Notes 65:27–29
Preps: Fast and Easy Extraction of Stable Yeast 36. Kang J, Lee MS, Gorenstein DG (2005) The
Genomic DNA. Sci Rep 6:26863 enhancement of PCR amplification of a
Bacteria and Yeast Colony PCR 221
random sequence DNA library by DMSO and Enhancement in the efficiency of polymerase
betaine: application to in vitro combinatorial chain reaction by TiO 2 nanoparticles: crucial
selection of aptamers. J Biochem Biophys role of enhanced thermal conductivity. Nano-
Methods 64:147–151 technology. 21:255704
37. Hardjasa A, Ling M, Ma K, Yu H (2010) Inves- 49. Jia J, Sun L, Hu N, Huang G, Weng J (2012)
tigating the effects of DMSO on PCR fidelity Graphene enhances the specificity of the poly-
using a restriction digest-based method. J Exp merase chain reaction. Small 8:2011–2015
Microbiol Immunol (JEMI) 14:161–164 50. Musso M, Bocciardi R, Parodi S, Ravazzolo R,
38. Rees WA, Yager TD, Korte J, von Hippel PH Ceccherini I (2006) Betaine, dimethyl sulfox-
(1993) Betaine can eliminate the base pair ide, and 7-deaza-dGTP, a powerful mixture for
composition dependence of DNA melting. amplification of GC-rich DNA sequences. J
Biochemistry 32:137–144 Mol Diagn 8:544–550
39. Spiess A-N, Mueller N, Ivell R (2004) Treha- 51. Ralser M, Querfurth R, Warnatz H-J,
lose is a potent PCR enhancer: lowering of Lehrach H, Yaspo M-L, Krobitsch S (2006)
DNA melting temperature and thermal stabili- An efficient and economic enhancer mix for
zation of taq polymerase by the disaccharide PCR. Biochem Biophys Res Commun 347:
trehalose. Clin Chem 50:1256–1259 747–751
40. Desai UJ, Pfaffle PK (1995) Single-step purifi- 52. Zhang Z, Kermekchiev MB, Barnes WM
cation of a thermostable DNA polymerase (2010) Direct DNA amplification from crude
expressed in Escherichia coli. BioTechniques clinical samples using a PCR enhancer cocktail
19:780–782. 784 and novel mutants of Taq. J Mol Diagn 12:
41. Adlimoghadam A, Hedayati MH, Siadat SD, 152–161
Nejati HAM, Vandyousefi J, Norouzion D 53. Pan J, Zhang C, Teng Y, Zeng S, Chen S,
(2008) Optimization of PCR conditions for Liang D, Li Z, Wu L (2021) Detection of
detection of human brucellosis from human Spinal Muscular Atrophy Using a Duplexed
serum samples. Res J Microbiol 3:352–358 Real-Time PCR Approach With Locked
42. Lorenz TC (2012) Polymerase chain reaction: Nucleic Acid-Modified Primers. Ann Lab Med
basic protocol plus troubleshooting and opti- 41:101–107
mization strategies. J. Vis. Exp. 63:e3998 54. Hu B, Wang Y, Sun S, Yan W, Zhang C, Luo D,
43. Bachmann B, Lüke W, Hunsmann G (1990) Deng H, Hu LR, Huang Z (2019) Synthesis of
Improvement of PCR amplified DNA sequenc- Selenium-Triphosphates (dNTPαSe) for More
ing with the aid of detergents. Nucleic Acids Specific DNA Polymerization. Angew Chem
Res 18:1309 Int Ed Engl 58:7835–7839
44. Wilson IG (1997) Inhibition and facilitation of 55. Hu B, Wang Y, Li N, Zhang S, Luo G, Huang
nucleic acid amplification. Appl Environ Z (2021) Highly convenient and highly
Microbiol 63:3741–3751 specific-and-sensitive PCR using Se-atom
45. Sakhabutdinova AR, Chemeris AV, Garafutdi- modified dNTPs. Chem Commun 57:57–60
nov RR (2020) Enhancement of PCR effi- 56. Dallas-Yang Q, Jiang G, Sladek FM (1998)
ciency using mono- and disaccharides. Anal Avoiding false positives in colony PCR. Bio-
Biochem 606:113858 Techniques 24:580–582
46. Li H, Huang J, Lv J, An H, Zhang X, Zhang Z, 57. Lee AB, Cooper TA (1995) Improved direct
Fan C, Hu J (2005) Nanoparticle PCR: PCR screen for bacterial colonies: wooden
Nanogold-Assisted PCR with Enhanced Speci- toothpicks inhibit PCR amplification. Bio-
ficity. Angew Chem Int Ed 44:5100–5103 Techniques 18:225–226
47. Yang W, Li X, Sun J, Shao Z (2013) Enhanced 58. Lehti T, Westerlund-Wikström B (2013) Col-
PCR amplification of GC-rich DNA templates ony immunoblotting assay for detection of bac-
by gold nanoparticles. ACS Appl Mater Inter- terial cell-surface or extracellular proteins, Bio
faces 5:11520–11524 Protoc. 3:e888
48. Khaliq A, Sonawane RPJ, Sasi BK, Sahu BS,
Pradeep T, Das SK, Mahapatra NR (2010)
Chapter 18
Abstract
Inverse PCR is a powerful tool for the rapid introduction of desired mutations at desired positions in a
circular double-stranded DNA sequence. In this technique, custom-designed mutant primers oriented in
the inverse direction are used to amplify the entire circular template with incorporation of the required
mutation(s). By careful primer design, it can be used to perform such diverse modifications as the
introduction of point or multiple mutations, the insertion of new sequences, and even sequence deletions.
Three primer formats are commonly used, nonoverlapping, partially overlapping, and fully overlapping
primers, and here we describe the use of nonoverlapping primers for introduction of a point mutation. Use
of such a primer setup in the PCR, with one of the primers containing the desired mismatch mutation,
results in the amplification of a linear, double-stranded, mutated product. Methylated template DNA is
removed from the non-methylated PCR product by DpnI digestion, and the PCR product is then
phosphorylated by polynucleotide kinase treatment before being recircularized by ligation and transformed
to E. coli. This relatively simple site-directed mutagenesis procedure is of major importance in biology and
biotechnology where it is commonly employed for the study and engineering of DNA, RNA, and proteins.
Key words Site-directed mutagenesis, Inverse PCR, Nonoverlapping primers, Protein engineering
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_18,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
223
224 Diogo Silva et al.
Fig. 1 Illustration of primer design formats for inverse PCR with nonoverlapping (a), partially overlapping (b),
and fully overlapping (c) primers. The hatched sections show the overlapping regions of the primers
2 Materials
2.3 Template 1. Restriction enzyme DpnI (as supplied, typically 10–20 U/μL).
Removal and Product Store at -20 °C (see Note 10).
Recircularization 2. DpnI digestion buffer (as supplied with DpnI, typically 10×).
Store at -20 °C.
3. T4 DNA ligase buffer (as supplied with T4 DNA Ligase, typi-
cally 10×, ensure this contains 5–10 mM ATP and 50–100 mM
DTT) (see Note 11). Store at -20 °C.
4. T4 polynucleotide kinase (as supplied, typically 10 U/μL).
Store at -20 °C (see Note 12).
228 Diogo Silva et al.
2.4 Preparation of 1. E. coli XL1-Blue cells stored at -70 °C in 15% glycerol (see
Chemically Competent Note 14).
E. coli XL1-Blue 2. Transformation buffer: 3 g/L (10 mM) PIPES, 3.4 g/L
(30 mM) CaCl2, 18.6 g/L (250 mM) KCl, 9.9 g/L
(50 mM) MnCl2·4H2O. Mix all components except MnCl2
and adjust pH to 6.7 with 112 g/L (2 M) KOH. Add MnCl2
and mix, and sterilize solution through a 0.22 μm membrane.
3. 100% dimethyl sulfoxide (DMSO).
4. SOC (Super Optimal broth with Catabolite repression):
20 g/L bacto-tryptone, 5 g/L yeast extract, 0.6 g/L
(10 mM) NaCl, 0.75 g/L (2.5 mM) KCl, 0.95 g/L
(10 mM) MgCl2, 1.2 g/L (10 mM) MgSO4, 3.6 g/L
(20 mM) D-glucose.
3 Methods
3.1 iPCR Mutant 1. Primer Design. Inversed primers should anneal to opposite
Amplification strands of the plasmid, be nonoverlapping and aligned back
to back with apposing 5’ends. Ideally, the targeted mismatch
mutation should be located in the middle of the primer with
10–15 perfectly matched nucleotides on either side. Mutations
can be incorporated closer to the 5′ end, but at least ten
complementary nucleotides are required at the 3’end (see
Note 16). Normal considerations for PCR primer design
should be adhered to (see Note 17). Phosphorylation of pri-
mers is not required (see Note 2). For best results, at least
HPLC grade purification of primers is required; for primers
greater than 40 nucleotides in length, PAGE purification is
recommended (see Note 2).
2. PCR Setup (See Note 18). Add the following components in
the order given to a thin-walled PCR tube on ice: 13.4 μL
autoclaved ultrapure water (for a total final reaction volume of
20 μL), 4 μL of 5× concentrated Phusion HF buffer (1X buffer)
(see Note 4), 0.4 μL of 10 mM stock dNTP mix (200 μM of
each dNTP), 0.5 μL of each 20 μM primer stock (0.5 μM of
each primer) (see Note 2), 1 μL of 1 ng/μL plasmid template
stock (1 ng) (see Note 1), and 0.2 μL of 2 U/μL Phusion DNA
polymerase (0.4 U) (see Note 3). Gently mix, briefly centrifuge,
and immediately place in the thermal cycler. A negative control
reaction with all components except the primers, which are
substituted with an equal volume of water, should also be
set up.
3. PCR Cycle (See Note 19): 1 cycle at 98 °C for 2 min, 25 cycles
of denaturation, annealing, and extension at, respectively, 98 °
C for 20 s, the calculated primer annealing temperature for
20 s, and 72 °C for 2 min (~20 s/kb of the template). A final
extension is then carried out at 72 °C for 10 min before cooling
to 4–10 °C. The same conditions are used for the sample and
negative control.
4. Agarose Gel Confirmation. The results of the PCR are verified
by visualizing 5 μL of the sample and negative controls on a 1%
agarose gel using the following protocol. To 1 g of agarose, add
1X TAE buffer to 100 mL and boil until the agarose is
completely dissolved. When cooled to ~50–60 °C, pour into
the casting tray, insert comb, and leave until completely poly-
merized. Remove the comb, place the gel in the electrophoresis
chamber, and add 1× TAE buffer until the gel is covered with
230 Diogo Silva et al.
ligation). Swirl the tubes gently and incubate on ice for 30 min.
Swirl the tubes gently and heat-shock cells in a water bath at
42 °C for 45 s and immediately transfer to ice for 10 to 15 min.
Add 900 μL of fresh SOC medium and incubate for 1 h at 37 °
C, 1.1 × g. Centrifuge for 1 min at 5000×g, at room tempera-
ture, remove 850 μL of the supernatant, and gently resuspend
the pellet in the remaining solution. Spread-plate the remain-
ing ~150 μL solution on LB + ampicillin agar plates and incu-
bate overnight at 37 °C. A positive transformation control with
1 μL of 1 pg/μL pUC18 plasmid and a negative process control
with 1 μL of the PCR negative control should also be
carried out.
3. Select three transformant colonies and inoculate into 5 mL
LB + ampicillin medium (see Note 15) in a 15 mL Falcon
tube. Incubate at 37 °C, 1.1 × g overnight. No colonies should
be visible for the negative process control. The LB + ampicillin
plate for the positive transformation control should have at
least approximately 50 colonies.
4. Isolate plasmid from cultures with a commercial plasmid puri-
fication kit, and forward for sequencing of the insert in both
directions. > 80% of the sequences should contain the desired
mutation and no other undesired mutation (see Note 23).
4 Notes
22. Avoid using phospho-ligation mix volumes that are more than
10% of the competent cell volume as this leads to a reduced
transformation efficiency. Purify phospho-ligation mix by use
of a commercial DNA purification kit if transforming by
electroporation.
23. The insert sequence should contain the desired mutation only.
While it is not feasible to sequence the entire plasmid con-
struct, the use of a high-fidelity polymerase reduces the risk
of secondary mutations in the vector sequence. To ensure the
absence of such mutations, the insert sequence may be
re-cloned into the original non-PCR-amplified vector.
Acknowledgments
References
1. Burdick JP, Basi RS, Burns KS, Weers PMM 5. Sanguinetti M, Silva Santos LH, Dourron J,
(1865) The role of C-terminal ionic residues in Alamón C, Idiarte J, Amillis S, Pantano S,
self-association of apolipoprotein A-I. Biochim Ramón A (2022) Substrate recognition prop-
Biophys Acta Biomembr 2023:184098. erties from an intermediate structural state of
https://doi.org/10.1016/j.bbamem.2022. the UreA transporter. Int J Mol Sci 23:16039.
184098 https://doi.org/10.3390/ijms232416039
2. Dahhas MA, Alsenaidy MA (2022) Role of 6. Wang K-D, Dughbaj MA, Nguyen TTV,
site-directed mutagenesis and adjuvants in the Nguyen TQY, Oza S, Valdez K, Anda P,
stability and potency of anthrax protective anti- Waltz J, Sacco MA (2023) Systematic muta-
gen. Saudi Pharm J 30:595–604. https://doi. genesis of Polerovirus protein P0 reveals dis-
org/10.1016/j.jsps.2022.02.011 tinct and overlapping amino acid functions in
3. Wang L, Gu J, Zhao W, Wang M, Ng KR, Nicotiana glutinosa. Virology 578:24–34.
Lyu X, Yang R (2022) Reshaping the binding https://doi.org/10.1016/j.virol.2022.
pocket of cellobiose 2-epimerase for improved 11.005
substrate affinity and isomerization activity for 7. Collins T, De Vos D, Hoyoux A, Savvides SN,
enabling green synthesis of lactulose. J Agric Gerday C, Van Beeumen J, Feller G (2005)
Food Chem 70:15879–15893. https://doi. Study of the active site residues of a glycoside
org/10.1021/acs.jafc.2c06980 hydrolase family 8 xylanase. J Mol Biol 354:
4. Chen K, Zhang M, Gao B, Hasan A, Li J, 425–435. https://doi.org/10.1016/j.jmb.
Bao Y, Fan J, Yu R, Yi Y, Ågren H, Wang Z, 2005.09.064
Liu H, Ye M, Qiao X (2022) Characterization 8. Hutchison CA, Phillips S, Edgell MH,
and protein engineering of glycosyltransferases Gillam S, Jahnke P, Smith M (1978) Mutagen-
for the biosynthesis of diverse hepatoprotective esis at a specific position in a DNA sequence. J
cycloartane-type saponins in Astragalus mem- Biol Chem 253:6551–6560
branaceus. Plant Biotechnol J 21:698. https:// 9. Edgell MH, Hutchison CA 3rd, Sclair M
doi.org/10.1111/pbi.13983 (1972) Specific endonuclease R fragments of
238 Diogo Silva et al.
bacteriophage phiX174 deoxyribonucleic acid. 18. Ochman H, Gerber AS, Hartl DL (1988)
J Virol 9:574–582 Genetic applications of an inverse polymerase
10. Hutchison CA 3rd, Edgell MH (1971) Genetic chain reaction. Genetics 120:621–623
assay for small fragments of bacteriophage phi 19. Hemsley A, Arnheim N, Toney MD,
X174 deoxyribonucleic acid. J Virol 8:181– Cortopassi G, Galas DJ (1989) A simple
189 method for site-directed mutagenesis using
11. Reeves AR (2016) In Vitro Mutagenesis. the polymerase chain reaction. Nucleic Acids
Methods Protocol:1–796 Res 17:6545–6551
12. Ho SN, Hunt HD, Horton RM, Pullen JK, 20. Qi D, Scholthof KB (2008) A one-step
Pease LR (1989) Site-directed mutagenesis by PCR-based method for rapid and efficient
overlap extension using the polymerase chain site-directed fragment deletion, insertion, and
reaction. Gene 77:51–59 substitution mutagenesis. J Virol Methods
13. Das N, Ghosh Dhar D, Dhar P (2022) Editing 149:85–90. https://doi.org/10.1016/j.
the genome of common cereals (rice and jviromet.2008.01.002
wheat): techniques, applications, and industrial 21. Zheng L, Baumann U, Reymond JL (2004) An
aspects. Mol Biol Rep:1–9. https://doi.org/ efficient one-step site-directed and site-
10.1007/S11033-022-07664-Y/TABLES/2 saturation mutagenesis protocol. Nucleic
14. Forner J, Kleinschmidt D, Meyer EH, Acids Res 32:e115. https://doi.org/10.
Fischer A, Morbitzer R, Lahaye T, Schöttler 1093/nar/gnh110
MA, Bock R (2022) Targeted introduction of 22. Liu H, Naismith JH (2008) An efficient
heritable point mutations into the plant mito- one-step site-directed deletion, insertion, sin-
chondrial genome. Nat Plants 8:245–256. gle and multiple-site plasmid mutagenesis pro-
https://doi.org/10.1038/s41477-022- tocol. BMC Biotechnol 8:91. https://doi.org/
01108-y 10.1186/1472-6750-8-91
15. Guo J, Zeng L, Chen H, Ma C, Tu J, Shen J, 23. Xia Y, Chu W, Qi Q, Xun L (2015) New
Wen J, Fu T, Yi B (2022) CRISPR/Cas9- insights into the QuikChange process guide
mediated targeted mutagenesis of BnaCOL9 the use of Phusion DNA polymerase for site-
advances the flowering time of Brassica napus directed mutagenesis. Nucleic Acids Res 43:
L. Int J Mol Sci 23:14944. https://doi.org/ e12. https://doi.org/10.1093/nar/gku1189
10.3390/ijms232314944 24. Silva D, Santos G, Barroca M, Collins T (2017)
16. Higuchi R, Krummel B, Saiki RK (1988) A Inverse PCR for point mutation
general method of in vitro preparation and introduction. In: Domingues L (ed) PCR.
specific mutagenesis of DNA fragments: study Methods and protocols, Springer protocols.
of protein and DNA interactions. Nucleic Methods in molecular biology, 1st edn.
Acids Res 16:7351–7367 Springer New York, New York, pp 87–100.
17. Triglia T, Peterson MG, Kemp DJ (1988) A https://doi.org/10.1007/978-1-4939-
procedure for in vitro amplification of DNA 7060-5_5
segments that lie outside the boundaries of
known sequences. Nucleic Acids Res 16:8186
Chapter 19
Abstract
PCR with degenerate primers can be used to identify the coding sequence of an unknown protein or to
detect a genetic variant within a gene family. These primers, which are complex mixtures of slightly different
oligonucleotide sequences, can be optimized to increase the efficiency and/or specificity of PCR in the
amplification of a sequence of interest by the introduction of mismatches with the target sequence and
balancing their position toward the primers 5′- or 3′-ends. In this work, we explain in detail examples of
rational design of primers in three different applications, including the use of specific determinants at the
3′-end, to (i) improve PCR efficiency with related sequences for members of a protein family by complete
degeneration at a core box of conserved genetic information at the 3′-end with the reduction of degenera-
tion at the 5′-end, (ii) optimize specificity of allelic discrimination of closely related DNA sequences of
orthologous by 5′-end fully degenerate primers, and (iii) increase the PCR efficiency of primers by targeting
DNA sequences belonging to specific phylogenetic groups, within a large and diverse gene family, allowing
the use of multiplex/degenerate PCR.
Key words PCR, Degenerate primers, 5′-end, 3′-end, PCR specificity, PCR efficiency, Sequence
alignment
1 Introduction
Lucı́lia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8_19,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
239
240 Maria Jorge Campos et al.
2 Materials
2.1 Sequence 1. Text processor, e.g., Microsoft Word, OpenOffice Text Docu-
Manipulation and ment or Pages for Mac.
Primer Design 2. Multiple alignment programs, e.g., http://www.ebi.ac.uk/
Tools/msa/ or https://www.expasy.org/resources/uniprot-
clustalo.
3. Primer design algorithms, e.g., http://www.ncbi.nlm.nih.
gov/tools/primer-blast/, https://www.thermofisher.com/
es/es/home/life-science/oligonucleotides-primers-probes-
genes/custom-dna-oligos/oligo-design-tools/oligoperfect.
html, and https://eurofinsgenomics.eu/en/ecom/tools/pcr-
primer-design/, or commercial informatics program as
OLIGO Primer Analysis Software (Molecular Biology Insight).
Design of Degenerate Primers 241
Table 1
Genetic code and compressed notation code according to IUPAC [5]
Compressed
Amino acid Codons notation codon Nucleotide Base
A Alanine GCT, GCC, GCA, GCG GCN A Adenine
R Arginine CGT, CGC, CGA, CGG, AGA, AGG CGN, MGR C Cytosine
N Asparagine AAT, AAC AAY G Guanine
D Aspartic acid GAT, GAC GAY T Thymine
C Cysteine TGT, TGC TGY R A or G
Q Glutamine CAA, CAG CAR Y C or T
E Glutamic acid GAA, GAG GAR S G or C
G Glycine GGT, GGC, GGA, GGG GGN W A or T
H Histidine CAT, CAC CAY K G or T
I Isoleucine ATT, ATC, ATA ATH M A or C
L Leucine TTA, TTG, CTT, CTC, CTA, CTG YTR, CTN B C or G or T
K Lysine AAA, AAG AAR D A or G or T
M Methionine ATG H A or C or T
F Phenylalanine TTT, TTC TTY V A or C or G
P Proline CCT, CCC, CCA, CCG CCN N Any base
S Serine TCT, TCC, TCA, TCG, AGT, AGC TCN, AGY
T Threonine ACT, ACC, ACA, ACG ACN
W Tryptophan TGG
Y Tyrosine TAT, TAC TAY
V Valine GTT, GTC, GTA, GTG GTN
Start ATG
Stop TAA, TGA, TAG TAR, TRA
2.2 PCR, Gel 1. 200 μL and 1.5 mL DNase-free plastic microtubes (see
Electrophoresis, and Note 1).
Image Acquisition 2. Thermocycler.
3. Gel electrophoresis system.
4. UV transilluminator or gel acquisition system.
3 Methods
3.1 Primer Design Fully conserved motifs of 4–6 consecutive amino acids, the core
from Protein Family box, are required to design the oligonucleotide 3′-end with, at
Alignment: Reduction least, 11 nucleotide positions. This is considering that the first
of 5′-End Complexity position, in the reverse primer, or the last position, in the forward
primer, leading to ambiguity, is omitted from the oligonucleotide
sequence to start reducing its complexity. In general terms, a good
principle for designing highly efficient degenerate primers would be
to establish a limit of degeneracy up to 64 (the number of different
sequences within the mixture primer), meaning that the core box
should not contain at all amino acid residues encoded by six codons
(L, R, or S) and very few encoded by three (I) or four codons (A, G,
P, V, or T). In contrast, amino acids encoded by two (C, D, E, F,
H, K, N, T, or Y) or particularly only one codon (M or W) are
strongly recommended (Table 1). The remaining sequence of pri-
mers, typically 7–12 nucleotides toward the 5′-end, is less critical
for DNA synthesis and, thus, is the region where efforts to reduce
degeneration should be focused. An example of core box selection
for primer design is shown in Fig. 1, where relationships between
CepA1 CepA2
Bfra 110…PQGGIEMSIADLLKYTLQQSDNNACDI……92……VTMGHKTGTGDRNAKG…55
Bun1 109…PDQDFTITLRELMQYSISQSDNNACDI……92……TVVGHKTGSSDRNADG…53
Bthe 105…PQGGFNIDIADLLNYTLQQSDNNACDI……92……VTIGHKTGTGDRNAKG…53
Bun2 127…SGPVISLTVRDLLRYTLTQSDNNASNL……94……VVIAHKTGSGYVNENG…57
Pden 127…SGPVISLTVRDLLRYTLTQSDNNASNL……94……VVIAHKTGSGDVNENG…57
Coch 127…SGPVISLTVRDLLRYTLTQSDNNASNL……94……VVIAHKTGSGDVNENG…57
. : : : :*:.*:: ******.:: ..:.****:. * .*
Fig. 1 Protein alignment and core box selection for primer design. Sequences shown are: Bfra, AAA22905.1;
Bun1, AAA66962; Bthe, NP_813418.1; Bun2, AAB17891; Pden, AAM48119.1; Coch, AAL79549.2. Multiple
alignement was performed by Clustal Omega. Bold characters represent strongly conserved positions. Grey
boxes indicate identical residues. Black boxes represent CepA characteristic residues used to design primers
with 3’-end specific determinants for discriminatory PCR. The expected size of cepA fragments amplified by
PCR with cepA1/A2 primers is 329 bp [8]
Design of Degenerate Primers 243
Table 2
Primer design for detection of cepA orthologous from Bacteroides and
related anaerobic Gram-negative bacteria
Forward Primer
Amino acids S D N N A C D
Coding sequence TCN GAC AAC AAC GCN TGC GAC
AGC T T T T T
T
Abbreviated WSN GAY AAY AAY GCN TGY GAY
Reverse Primer
Amino acids G H K T G T/S G/S
Coding sequence GGN CAC AAA ACN GGN ACN GGN
T G TCN TCN
AGC AGC
T T
Abbreviated GGN CAY AAR ACN GGN WSN BSN
3.2 Primer Design In primer design for allelic discrimination, the number of mis-
from DNA Alignment matches at the last positions from the 3′-end of the primer is crucial
(I): Reduction of for annealing and specific elongation by the DNA polymerase,
Specificity by which supports the mismatch amplification mutation assay or
Degeneration of 5′-End MAMA-PCR [7]. The discriminatory capability of this technique
is based on the fact that each particular allele of a polymorphic
position is complementary to the 3′-end of its corresponding pri-
mers, where annealing is further weakened by an additional mis-
match with its target sequences, allowing amplification with one
but not two mismatches. The technique is so sensitive that addi-
tional mispairing of primers produced by genetic variability of
sequences would give rise to false negatives. To solve that, in a
particular case that requires the use of DNA from closely related
species as target sequences, we designed degenerate primers with
allele-specific determinant at their 3′-ends that allow discrimination
of the C-257-T polymorphism of gyrA sequence, responsible for
quinolone resistance of Campylobacter isolates [12]. This primer
design, shown in Fig. 2, was performed by including degeneration
toward the primer 5′-ends, increasing the target recognition to
both major species of thermophilic Campylobacter isolated from
humans, C. jejuni and C. coli, without compromising the allele-
specific amplification of gyrA for both species.
3.3 Primer Design Many protein families are too large and/or diverse to support
from DNA Alignment specific primer design, knowing that the efficiency limit for product
(II): Reduction of detection by agarose gel electrophoresis should be 128–256 primer
Degeneration by sequences in the PCR mix (see Subheading 3.1). This situation has
Sequence Clustering been addressed to detect the occurrence, in enterobacteria, of mcr
genes [13], which are plasmid determinants, conferring colistin
Design of Degenerate Primers 245
3.4 Protocol for 1. Gather all the DNA or protein sequences of interest from
Protein or DNA online databases as the National Center for Biotechnology
Sequence Analysis Information (NCBI, http://www.ncbi.nlm.nih.gov/nuccore
for nucleotides or http://www.ncbi.nlm.nih.gov/protein/ for
proteins) in FASTA format (see Note 6).
2. Paste the sequences in a text processor document.
246 Maria Jorge Campos et al.
A B
> ATGATGCAG----------CATAC 13… …GGA--TTA---TCCGACTTGGGGCAAGG 457
> ---ATGACA----------TCACA 10… …CAA--TTA---TCCAACATGGGGCAAAG 451
> ---ATGACA----------CAGCA 10… …GGA--TTA---TCCGACATTGGGTAAGA 451
> ATGCGGTTGTCTGCATTTATCACT 24… …ACG--------AACGGGCTGGAAACAAG 466
> ATGTTCAAG--------------T 9 … …AGACAATAAGCGGGGAGCTTTTAGAATT 459
> ------GTG--------------A 4 … …TCA--TTA---TCAGCCCTTTTTTAAGG 445
> -----------------------A 1 … …TGA—-ATATGAGGAAAAATGGTTCAAAG 445
> ------------------------ -6… …GCG—-CTATCCGGCAAACTGGTACAAGG 439
> -----------------------A 1 … …TCA-—ATATCCTGAAAAATGGTATAAAG 445
> -----------------------A 1 … …TGA—-ATATGCAGACAAATGGTACAAAG 445
> -----------------------A 1 … …CCG-—TTATCCTGAGACATGGATAAAGG 445
> ATGTGGT----------------T 8 … …CAT-—TCGCACCGGAAAATGGTGGTATG 445
> ATGTCGTTATT-----------GC 13… …CCG-—TAAGCCCGCCACACGGTGGCGCG 457
> ATGTTGAAGCG-----------CC 13… …CAA-—ACCCACCACCTCACGCCTGCGCA 457
> ATGTTAAAGCG-----------CT 13… …CCG—-CCCGGCGACGCCGCGCTTACGTA 457
B (cont.)
TGCGGCACATCGACGGCGTATTCTGTGCCGTGTATGTTCAGCTA 884… …AACGAATGCCGCGATGTCGGTAT 1106… …AGCAATGCCTATGATGTCTCAATGCTGTATGTCAGCGATCAT 1398
TGTGGCACATCGACGGCGTATTCTGTGCCGTGTATGTTCAGCTA 878… …AACGAATGCCGTGATGTCGGTAT 1100… …GAAGCGAACTACGATGTCGCCATGCTCTATGTCAGTGACCAC 1392
TGTGGCACATCGACGGCGTACTCTGTGCCGTGTATGTTCAGTTA 878… …AACGAATGTCGTGATGTCGGTAT 1100… …CAGGCCAACTATGATGTTGCCATGCTCTATGTCAGCGACCAC 1392
TGCGGGACGGATACGGCTACATCCCTTCCCTGCATGTTTTCCCT 887… …GAGCGCTGCCTGGATGAAATTCT 1091… …-TCCGCTCACACGACACGGCGCTGCTGTACGTTTCCGATCAT 1377
TGCGGAACGGCTACCGCAATATCACTACCCTGCATGTTCTCGCG 875… …GGTACATGCTTTGATGAGGTGTT 1088… …TCCGGGATGCGTGACGTTGCTATGATATATCTTTCTGATCAT 1377
TGCGGCACGGCCACGGCGGTGTCTCTACCCTGTATGTTTTCACG 863… …CAATATTGTTTTGACCAAGTATT 1067… …CAGGATATGTTCGATACTGCAATGCTGTATCTCTCTGACCAT 1359
TGTGGGACTGCAACCGCTGTATCCGTCCCCTGCATGTTCTCCAA 860… …AACACATGCTATGACGAGGTTGT 1064… …GAAGATAAGTACAACACCGCGTTGCTCTACGTCTCCGATCAT 1353
TGCGGCACGGCCACAGCGGTGTCGGTGCCCTGCATGTTCTCCAA 854… …AACACCTGCTATGACGAGGTTGT 1058… …GAAGATAAGTACAACACGGCGTTGATCTACCTCTCTGATCAC 1347
TGCGGTACCGCTACCGCAATATCCGTTCCGTGCATGTTCTCGAA 860… …AAAACGTGCCATGACGAGGTGAT 1064… …AGCGATCAGTACAACACCGTGCTGCTTTATGTGTCCGATCAT 1353
TGTGGCACGGCGACCGCAGTCTCGGTGCCCTGTATGTTCTCGGA 860… …AAAACCTGCTATGACGATGTTAT 1064… …AGCGAACAGTACAACACCGTACTGCTGTATGTGTCCGATCAC 1353
TGTGGCACATCCACAGCCATATCTGTTCCATGCATGTTTTCAGA 860… …AACTCATGTTATGATGAGGTTAT 1067… …AAAGATGAGTATGATACTGTTTTATTATATGTCTCTGACCAT 1356
TGTGGCACGGAAACCGCTGTTTCCGTCCCCTGCATGTTCTCCGG 866… …AAATCCTGTATCGACGACGTTAA 1070… …CAGGCCAACATGAACACGGCGCTCATTTACCTCTCCGATCAC 1359
TGCGGCACGGCAACTGCGGTTTCCGTCCCCTGTATGTTCTCTAA 875… …GGCGAGTGCTACGATGAGGTCCT 1079… …CAGGATAAATTTACCACTAGCCTGGTTTATTTGTCCGACCAC 1368
TGCGGCACAGCAACAGCCGTCTCAGTGCCGTGCATGTTCTCGGA 875… …GGCGAATGCTATGACGAAGTGCT 1079… …CAGGATAAATTTACCACCAGCCTGGTTTATCTTTCTGACCAC 1368
TGCGGTACGGCGACCGCGATCTCCGTTCCCTGCATGTTTTCTGA 875… …GGCGAGTGCTACGATGAGGTGTT 1079… …CAGGATAAATTCACAACCAGCCTGGTCTATCTTTCCGATCAC 1368
** ** ** ** ** ** * ** ** ***** ** ** * * ** * ** **
Fig. 3 Phylogenetic analysis of colistin resistance genes and their multiple alignment of sequences to design
cluster-specific primers. (a) Neighbor joining tree of the mcr-gene family, with multiple sequence alignment
performed by Clustal X 2 and the phylogeny emulated by NJPlot 2.3 (bootstrap values indicated close to the
corresponding nodes). eptA sequences representative of enterobacterial groups presenting acquired colistin
resistance are indicated: E, Enterobacter spp.; K, Klebsiella spp.; E, Escherichia coli; S, Salmonella enterica.
hPET corresponds to a chromosomal gene encoding a hypothetical phosphoethanolamine transferase located
in the chromosome of some enterobacterial strains, which genome have been made available but their colistin
resistance phenotype remains unknown. (b) Selection of primer sequences for cluster-specific mcr genes
from the multiple sequence alignment. For every pair of forward and reverse primer (right and left sequences,
respectively), the same color code shown in the phylogenetic tree is utilized, and variants included in
degeneration are indicated in green (final sequences of primers in Table 3). Sequences shown are: mcr-1,
AP026796.1; mcr-2, LT598652.1; mcr-3, CP053734.1; mcr-4, NG_057470.1; mcr-5, MK731977.1; mcr-6,
NG_055781.1; mcr-7, NG_056413.1; mcr-8, CP049891.1; mcr-9, CP049986.1; mcr-10, LC548754.1; hPET,
CP044315.2; eptA-E, CP073771.1; eptA-K, CP044050.1; eptA-C, CP054230.1; eptA-S, CP049308.1
Table 3
Primer design for specific detection of mobilizable colistin resistance (mcr) determinants from
enterobacteria
Product
Cluster Forward primer Reverse primer (pb)
Cluster 1 (mcr- TGYGGCACATCGACGGCG ATACCGACATCRCGRCATTCG 266
1/-2) TA TT
Cluster 2 (mcr- GTSCCCTGCATGTTC AACGCSGTGTTGTACTTATC 494
3/-7) TCCAA TTC
Cluster 3 (mcr- ACGGCYACSGCRRTRTCWC ATGRTCAGARAGATAYAKCA 534
4/-8) TAC TWGCA
Singleton (mcr-5) GCGGTTGTCTGCATTTA CTTGTTTCCAGCCCGTTCGT 464
TCACT
Design of Degenerate Primers 247
3. Copy all the sequences and use the algorithm Clustal on online
platforms to align the multiple DNA or protein sequences.
Clustal can be found in the EBI multi Sequence Alignment
(http://www.ebi.ac.uk/Tools/msa/), the Swiss Institute of
Bioinformatics (https://www.expasy.org/resources/uniprot-
clustalo), the Kyoto University Bioinformatics Center
(http://www.genome.jp/tools/clustalw/), or other
platforms.
4. Look for a core box of successive and conserved amino acid
residues in the protein alignment (Fig. 1) or an 18 to 24 con-
served DNA sequence around the target polymorphism (see
Note 7) (Fig. 2). Protein sequences must be reverse translated
to their genetic code (Table 2). This can be done on platforms
as the Sequence Manipulation Suit (http://www.bioinformat
ics.org/sms2/) or BioPHP (http://www.biophp.org/
minitools/protein_to_dna/demo.php). It is possible to obtain
the codon usage of several organisms on online platforms like
the Codon Usage Database (http://www.kazusa.or.jp/
codon/ ) or EMBL-EBI (http://www.ebi.ac.uk/Tools/st/
emboss_backtranseq/).
5. Consider a conserved region that has at the penultimate or
ultimate position of the 3′-end of the possible primer a con-
served position that can be used to distinguish a desirable trait
and design the primer considering degenerate positions
between target sequences, preferably at the primer 5′-ends
(Table 1). An extra and obligated mismatch of the 3′-end
should be included to weak primer annealing and increase
specificity toward allele-specific (Fig. 2).
6. Primer design quality can be assessed by checking the forma-
tion of hairpins and primer dimerization. This information
might be obtained by using the PCR Primer Stats option at
the Sequence Manipulation Suite (https://www.bioinformat
ics.org/sms2/).
7. Once primers are designed, they can be ordered using an online
service from an oligonucleotide synthesis company.
3.5 PCR A successful PCR relies on a number of factors some related to the
quality of the primer design. Primer length should be between
16 and 28 nucleotides, which depending on the GC content should
produce primers melting temperatures (Tm) between 50 and 62 °
C. It is not judicious to have Tm difference, between the two
primers, greater than 5 °C [16], and the annealing temperature
can be empirically determined as being 5 °C lower than the primer
with the lowest Tm. Since for degenerate primers the working
sequence(s) of primers is(are) unknown, a good approach is to set
annealing temperature between 50 and 55 °C, although specific
primers corresponding to one known homologue could be used to
estimate more accurately the PCR conditions.
248 Maria Jorge Campos et al.
3.6 Thermocycling Incubate the 200 μL PCR microtubes containing the DNA and the
master mix in a thermocycler with the following program: initial
denaturation 95 °C for 2 min, 35 cycles of denaturation at 95 °C for
30 s, primer annealing between 50 °C and 60 °C for 1 min, exten-
sion at 72 °C for 1 min, and a final extension at 72 °C for 10 min.
The PCR can be maintained at room temperature indefinitely.
3.7 Gel Analysis 1. Prepare a 0.75 to 1.0% agarose gel (see Note 10).
2. Load 10 μL of each PCR product in each gel well.
3. Connect the power supply to 80 V and wait approximately 1 h
or until the fastest moving band in the PCR buffer is close to
the gels hedge.
4. Observe the gel in the UV transilluminator or gel acquisition
system, and look for the presence of the expected size
amplified band.
Design of Degenerate Primers 249
4 Notes
Acknowledgments
Bibliography
1. Saiki RK, Scharf S, Faloona F, Mullis KB, Horn 7. Cha RS, Zarbl H, Keohavong P, Thilly WG
GT, Erlich HA, Arnheim N (1985) Enzymatic (1992) Mismatch amplification mutation assay
amplification of beta-globin genomic (MAMA): application to the c-H-ras gene.
sequences and restriction site analysis for diag- PCR Methods Appl 2(1):14–20
nosis of sickle cell anemia. Science 4732:1350– 8. Garcı́a N, Gutiérrez G, Lorenzo M, Garcı́a JE,
1354 Pı́riz S, Quesada A (2008) Genetic determi-
2. Saiki RK, Gelfand DH, Stoffel S, Scharf SJ, nants for cfxA expression in Bacteroides strains
Higuchi R, Horn GT, Mullis KB, Erlich HA isolated from human infections. J Antimicrob
(1988) Primer-directed enzymatic amplifica- Chemother 5:942–947
tion of DNA with a thermostable DNA poly- 9. Lorenzo M, Garcı́a N, Ayala JA, Vadillo S,
merase. Science 4839:487–491 Pı́riz S, Quesada A (2012) Antimicrobial resis-
3. Chothia C, Lesk AM (1986) The relation tance determinants among anaerobic bacteria
between the divergence of sequence and struc- isolated from footrot. Vet Microbiol 157(1–2):
ture in proteins. EMBO J 4:823–826 112–118
4. Iserte JA, Stephan BI, Goñi SE, Borio CS, 10. Ikemura T (1985) Codon usage and tRNA
Ghiringhelli PD, Lozano ME (2013) Family- content in unicellular and multicellular organ-
specific degenerate primer design: a tool to isms. Mol Biol Evol 1:13–34
design consensus degenerated oligonucleo- 11. Ben-Dov E, Shapiro OH, Siboni N, Kushmaro
tides. Biotechnol Res Int 2013:383646 A (2006) Advantage of using inosine at the 3′
5. Nomenclature Committee of the International termini of 16S rRNA gene universal primers for
Union of Biochemistry (NC-IUB) (1985) the study of microbial diversity. Appl Environ
Nomenclature for incompletely specified bases Microbiol 11:6902–6906
in nucleic acid sequences. Recommendations. 12. Hormeño L, Palomo G, Ugarte-Ruiz M, Por-
Biochem J 229(2):281–286 rero MC, Borge C, Vadillo S, Pı́riz S,
6. Haras D, Amoros JP (1994) Polymerase chain Domı́nguez L, Campos MJ, Quesada A
reaction, cold probes and clinical diagnosis. (2016) Identification of the main quinolone
Sante 4(1):43–52 resistance determinant in campylobacter jejuni
Design of Degenerate Primers 251
Lucilia Domingues (ed.), PCR: Methods and Protocols, Methods in Molecular Biology, vol. 2967,
https://doi.org/10.1007/978-1-0716-3358-8,
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer
Nature 2023
253
PCR: METHODS AND PROTOCOLS
254 Index
I Q
Internal amplification control (IAC) ..........45, 64, 66–72 Quantification .................................................2, 5, 11, 19,
Intramolecular secondary structure ................... 160–162, 21, 26–28, 57, 62, 66, 86, 87, 89, 92, 96, 101,
164, 165, 169, 170 105–114, 118, 121–123, 128, 133–147,
Inverse PCR (iPCR) ............................................ 223–237 151–157, 197
Inverted terminal repeat (ITR) ...................................161, Quantitative PCR (qPCR).........................................2, 18,
162, 164, 170 28, 29, 53–66, 68–72, 105–114, 118–121, 123,
126, 127, 133–147, 151–157, 167, 168
L
R
Lactococcus lactis................................................... 151–157
Long amplicons............................................................. 190 Reaction efficiency ..............................106, 109, 113, 147
Long-range PCR.................................................. 181–191 Real-time PCR ..................................................18, 41, 42,
44–49, 54, 63–72, 75–83, 86–91, 94–96, 98, 100,
M 101, 118, 120, 121, 123, 124, 126, 129
Megaprimers...............................194, 196, 200, 201, 205 Real-time qPCR ..................................151–157, 163, 168
Melting .................................... 26, 32, 34, 41–51, 53–62, Recombination efficiency..................................... 117–129
Restriction enzymes ..........................................20, 22, 23,
99, 111, 119, 121, 123, 126, 127, 147, 154, 156,
157, 183, 185, 189, 194, 195, 205, 216, 235, 247 27, 129, 195, 198, 203, 204, 227, 233
Microbial diversity studies .............................................. 32 RNA isolation.........................5, 133, 135–137, 144, 145
RNA quality......................................... 133–135, 137–140
Milk adulteration.................................................. 173–179
Minicircles (MCs) ................................................ 117–129
S
Mitochondrial D-loop DNA ............................... 173–179
Mixed bacterial cultures................................................ 106 Saccharomyces cerevisiae.............. 198, 210, 212, 214–218
Multiplex real-time PCR ..........................................63–72 Sample collection ................................3–5, 133, 135, 136
SARS-CoV-2 ............................................................... 1–11
N Sequence alignment ..................................... 94, 100, 177,
Nonoverlapping primers ...................................... 224–226 244, 247, 249
Nuclear DNA .................................................................. 76 Shiga toxin-producing Escherichia coli
(STEC).................................................... 63–67, 70
P Single nucleotide polymorphism (SNP) ................ 42, 43,
46, 48, 50, 51, 164
ParA resolvase....................................................... 124, 125 Site-directed mutagenesis (SDM) ............. 124, 165, 194,
PCR additives ............................................................44, 46 223–237
PCR efficiency ............................................ 62, 86, 87, 96, Species differentiation .......................................v, 173–179
101, 127, 178, 205, 210, 240 Species-specific PCR assay .............................................. 17
PCR enhancers ...........................182, 183, 188, 212, 213
PCR specificity .............................................................. 213 T
Pentaplex qPCR assay ...............................................64, 66
Theobroma cacao........................................................75, 76
Plasmid copy number determination..........151–153, 155
Polyacrylamide gel ....................................................33–35 Thermal cycling.......................................... 2, 4, 9–10, 22,
Polymerase chain reaction (PCR) .............................2, 17, 28, 29, 164, 185, 186, 189, 212
Third-generation PCR.................................................... 18
31, 41, 63, 76, 118, 159, 173, 181, 193, 209,
224, 239
W
Primer design .......................................44, 48, 88, 98, 99,
119, 127, 152, 153, 156, 169, 183–185, 189, Wastewater based epidemiology (WBE) ...................... 1, 2
194, 198–201, 204, 224–226, 229, 234–236,
240, 242–247 Y
Proofreading enzyme .................................................... 182
Yeast ..................................... 70, 194, 198, 209–219, 228
Protein engineering ............................................. 194, 223