Dna Manip
Dna Manip
Dna Manip
DNA
Manipulation
and Analysis
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Edited by
Garry Scarlett
Biophysics Laboratories, School of Biological Sciences, University of Portsmouth, Portsmouth, UK
Editor
Garry Scarlett
Biophysics Laboratories
School of Biological Sciences
University of Portsmouth
Portsmouth, UK
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
If there is such a thing as a single secret to the mystery of life, then that secret is probably
DNA. Since 1953 that has been a very badly kept secret, detailed descriptions and images of
its structure and function abound in everything from learned articles to pop culture.
However, we now not only know the structure of DNA but can decipher its code and
even modify that code. We have learned to read and write on a molecular and biological
level, the technologies that allow us to read and write DNA form the focus of this book. This
book is designed for scientists moving into recombinant DNA technologies, covering
techniques that are important for conducting experiments in fields as wide ranging as
developmental to structural biology. The chapters are laid out as readers of the long running
Methods in Molecular Biology series have come to expect, with an accessible theory section
followed by a detailed method and finally the ever-useful notes and troubleshooting pages.
No preface on the art of gene engineering could go without mentioning the discovery of
the structure of DNA and the subsequent cracking of the code by a series of elegant
experiments in the 1950s and 1960s. However, the key developments that are centrally
relevant to this book were the first recombinant DNA experiments of Berg and, shortly after,
Boyer-Cohen in 1971 and 1972; indeed, some of the principles in those early procedures
have recognisable descendants described in the following chapters. A landmark development
in the field in the mid-1970s was the arrival of Sanger dideoxy sequencing, replacing difficult
and dangerous chemical methods for determining the order of bases on a piece a DNA, with
a much easier enzymatic approach. The following decade of the 1980s was dominated by
two major breakthroughs, the polymerase chain reaction (PCR) and the less mentioned but
equally important phosphoramidite chemistry approaches to making short single-stranded
oligonucleotides. Without these developments, many of the techniques in this book would
simply not be possible, they have enabled not only de novo generation of DNA fragments but
also the rapid amplification, site selection and targeted mutation of sequences.
Initially, much of the early recombinant DNA work undertaken was on sequences of
DNA inserted into plasmids, replicating circular extra-chromosomal DNA found in bacteria
that encode useful attributes for the cell. Indeed, plasmids remain the work horses of the
DNA laboratory, and the first few chapters detail a variety of different methods of inserting
DNA sequences into them, each with specific strengths and weaknesses. Following on from
these, the next three chapters discuss how to manipulate and create DNA sequences, which
in conjunction with the molecular cloning methods discussed in the earlier part of the book
provide powerful options in the laboratory for the would-be molecular biologist. The last
decade of the twentieth century and the dawn of the new millennium saw increasingly rapid
strides in the complex field of genomic modification, not only bacterial genomes but also the
genomes of eukaryotic cells, including those in multicellular organisms. Early attempts at
eukaryotic genomic modification suffered from significant issues with targeting the correct
location within the genome, a problem that became worse the bigger the genome to be
manipulated. The first technology that really tackled this was based upon zinc finger
nucleases; this was rapidly replaced by TALENs before the extremely powerful and easy to
use CRISPR/Cas9 system rose to its current dominance. Chapters 9, 10, and 11 discuss
transgenics in the model system Xenopus, with Chaps. 10 and 11 describing methods for
CRISPR targeted gene knockouts and gene insertions, respectively. The book then has two
v
vi Preface
examples of the growing field of ‘cell-free’ DNA work, dealing with the raising of aptameric
sequences and high throughput array technologies. Chapters 14, 15 and 16 deal with the
key underpinning technologies of all DNA work, making oligonucleotides and reading the
sequence. Much of this is now available as outsourced services for starter laboratories, but
in-house provision allows for flexibility and increased speed. The last chapter deals with
ethical considerations. The growing importance of DNA and gene manipulation to wider
society has led to increasing public and governmental scrutiny, not only of ethics but also the
risks of recombinant DNA technologies. Many jurisdictions have now introduced guidelines
and laws governing the making and containment of genetically modified organisms, this is in
addition to legal implications of using animals in research.
Recombinant technologies have developed remarkably since those first experiments in
the early 1970s; the growth of manufactured kits and outsourced services have brought
nucleic acid–based experiments into the range of many laboratories that previously would
have struggled to set up the infrastructure. We hope this book helps provide the expertise for
scientists embarking on their first forays into these types of projects. Finally, I would like to
thank the many chapter authors to who have contributed and provided their time and
expertise to help support the scientific community with the resources held within this book.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
Contributors
ix
x Contributors
Abstract
Traditional molecular cloning involves a series of linked experimental steps performed with the overall goal
of isolating (“cloning”) a specific DNA sequence—often a gene. The main purpose of cloning is to study
either that DNA sequence or the RNA or protein product it encodes. Building on key enzymatic discoveries
in the late 1960s, gene cloning was pioneered in the early 1970s. Since then, DNA cloning and manipula-
tion have been used in every area of biological and biomedical research, from molecular genetics, structural
biology, and developmental biology to neurobiology, ancient DNA studies, and immunology. It is a
versatile technique that can be applied to a variety of starting DNA types and lengths, including cDNAs,
genes, gene fragments, chromosomal regions, or shorter fragments such as PCR products and functional
control regions such as enhancers or promoters. The starting DNA can originate from any cell, tissue, or
organism. In this chapter we will cover traditional (“classic”) molecular cloning strategy. This comprises six
linked stages in which (1) PCR is used to amplify a DNA region of interest that is then (2) digested with
restriction enzymes, alongside a selected vector, to produce complementary ends crucial for the two
molecules to be (3) ligated by an ATP-dependent DNA ligase, creating a recombinant DNA molecule.
The recombinant DNA is then (4) introduced into competent bacterial cells by transformation and
(5) grown on a selective agar media, followed by (6) colony-PCR for screening purposes. We provide a
worked example to demonstrate the cloning of an average-size gene (in this case the 2 kb DNA ligase A
gene) from E. coli into a common plasmid expression vector. We have included six color figures and two
tables to depict the key stages of a classical molecular cloning protocol. If you are cloning a segment of DNA
or a gene, remember that each DNA cloning experiment is unique in terms of sequence, length, and
experimental purpose. However, the principles of traditional cloning covered in this chapter are the same
for any DNA sequence; we have included a detailed notes section, so you should easily be able to transfer
them to your own work. Some of the following chapters in this volume will cover other, more recently
developed, cloning protocols.
Key words Molecular cloning, Gene cloning, Restriction endonucleases, Plasmid, PCR, Transforma-
tion, Bacteria
1 Introduction
1.1 Historical Before the 1970s, the largest challenge to the progression of chro-
Perspective mosomal and genetic research was lack of a reliable and reproduc-
ible method for isolating and analyzing DNA sequences of interest.
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_1,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
1
2 Ana Mikić et al.
This changed in the early 1970s when three key discoveries were
brought together to revolutionize genetics and initiate the field of
modern molecular biology.
The first discovery, in the late 1960s, occurred in the laboratory
of Werner Arber and Stuart Linn, whose group characterized the
first bacterial restriction endonucleases [1]. Restriction endonu-
cleases are a class of bacterial DNA-binding enzymes that evolved
as a defense against bacteriophage infection. They target and cleave
specific recognition sequences within bacteriophage genomes,
causing phage genome degradation. Although much attention
was given to the characterization of these enzymes, termed “restric-
tion factors,” their ability to cut specific DNA sequences was not
fully exploited until the laboratory of Daniel Nathans first used
them to map the Simian Virus 40 (SV40) genome [2]. Soon after,
many more restriction enzymes were isolated and their recognition
sequences determined, creating a way to selectively and specifically
cut DNA molecules. Today, thousands of restriction endonucleases
are known, with many hundreds available from commercial
suppliers.
The second key discovery that contributed to the development
of molecular cloning was made earlier in the 1960s by several
laboratories, including those of Martin Gellert, I. Robert Lehman,
Charles Richardson, and Jean Weigle. This was the discovery of a
class of enzymes able to perform the opposite function to restric-
tion endonucleases, namely, to join DNA ends. These are called the
DNA ligases [3, 4]. It is important to note that the formation of a
phosphodiester bond between two juxtaposed DNA ends (specifi-
cally, between a 5′-phosphate and 3′-hydroxyl group) by DNA
ligase is not a sequence-specific activity. Therefore, this discovery
provided a means to re-join any two DNA fragments cut by restric-
tion endonucleases. The first experiment to use restriction enzyme
digestion and subsequent ligation was performed in 1972 by Paul
Berg’s laboratory, successfully synthesizing the first recombinant
DNA molecule [5]. Although this confirmed the concept that the
DNA from any two species may be joined and a resulting recombi-
nant DNA molecule can be created, the process was long, labori-
ous, and had a low yield.
However, this changed with the third discovery—of bacterial
transformation by plasmids—that made it possible for the molecu-
lar cloning method to become a widely applicable technology.
Bacterial transformation was in fact discovered by Griffith in the
1930s, in studies on lethal and non-lethal bacterial strains
[6]. Transformation of bacterial cells enabled effective replication
of the created recombinant DNA molecule. However, its applica-
tion for the purpose of molecular cloning was not fully realized
until 1972, when Stanley Cohen and Herbert Boyer treated bacte-
rial cells with calcium chloride to induce the uptake of plasmids
carrying antibiotic-resistance genes [7]. This was a ground-
Classical Cloning 3
breaking study that set the stage for the first molecular cloning
experiment to be performed in 1973 by Boyer, Cohen, and Chang
[8]. Their method joined the individual digestion, ligation, and
transformation steps to pioneer gene cloning and lay the founda-
tion for the millions of molecular cloning experiments since.
1.3 Cloning Vectors A cloning vector is a small DNA molecule into which another DNA
molecule can be inserted without disrupting the stability of the
vector within a host organism. Cloning vectors can be of different
sizes and from different hosts. The most commonly used vectors
are bacterial plasmids, which are circular and typically several thou-
sand base pairs in size (2–10 kbp). Alternatively, bacteriophages,
cosmids, bacterial artificial chromosomes (BACs), yeast artificial
chromosomes (YACs), or mammalian artificial chromosomes
(MACs) have been developed (but are not covered further in this
chapter). Regardless of the type or size of a vector, all standard
cloning vectors must contain at least three structural features to
allow for the recombinant DNA to be made and replicated. A
vector must possess: (1) a multiple cloning site (MCS), also
known as a polylinker; (2) an origin of replication (ori), and (3) a
selectable marker gene.
A multiple cloning site is a region on a plasmid containing a
number of different restriction enzyme recognition sites, and into
which the exogenous DNA is incorporated during the cloning
experiment. An origin of replication allows a vector to replicate
independently of a host cell (such as an E. coli cell) to obtain
multiple copy numbers per cell cycle. A selectable marker gene
provides host cells with a selectable phenotype that can be used to
clearly distinguish between cells that contain the vector and the
ones that do not. This is frequently an antibiotic resistance gene
(such as for beta-lactamase), as only cells containing the vector will
be able to grow on a media containing the corresponding
antibiotic [9].
4 Ana Mikić et al.
Fig. 1 Recombinant DNA cloning. A pictorial overview of a typical molecular cloning experiment, in which a
chosen DNA sequence is amplified by PCR and ligated into a plasmid vector; the recombinant vector is then
transformed into bacterial cells. There are six distinct steps, and these are shown in Panels A and B. Panel A:
Molecular biology steps. Step 1: PCR amplification of a gene or region of interest to give a DNA insert for
cloning (light blue). Step 2: restriction enzyme digestion of both the DNA insert ends (red and purple) and the
chosen plasmid (green), to give compatible, cohesive (“sticky”) ends. Step 3: ligation reaction between the cut
DNA insert and cut plasmid; a new recombinant DNA molecule is born. Panel B: Microbiology steps. Step 4:
Transformation of recombinant plasmid (blue/green) into competent bacterial cells (orange) and growth of
transformed bacterial colonies. Step 5: Selection of successfully transformed cells via antibiotic resistance
(positive selection). Step 6: Screening of selected cells by colony-PCR, followed by DNA sequencing of the
plasmid and insert
Fig. 2 Types of plasmid vector: cloning vectors vs. expression vectors. A pictorial comparison between the
essential structural components of a standard cloning plasmid vector (such as pUC19) and an expression
plasmid vector (such as pET28b). Panel A: A standard cloning plasmid must contain an origin of replication
(blue), a selectable marker (ampicillin resistance in this example, purple), and a multiple cloning site, MCS
(red). The MCS contains a single recognition sequence for at least a dozen different restriction enzymes (to aid
flexibility when cloning) and is often found within the LacZ operon (green). Panel B: An expression plasmid
must contain all the structural parts of a standard cloning plasmid vector (origin of replication, a selectable
marker, and multiple cloning site) as well as additional sequences necessary for protein expression.
These are: a RNA polymerase promoter (yellow), ribosome binding site (dark pink), poly-A signal (orange),
and a terminator (brown) sequence. It may also contain N- or C- terminal tags, such as histidine- or
glutathione-S-transferase-tag (black lines) to aid recombinant protein purification
1.5 Recombinant After selecting the DNA sequence, the restriction endonuclease(s),
DNA Production and the cloning vector to be used in your experiment, the next step
of the molecular cloning method is to use the Polymerase Chain
Reaction (PCR) to amplify the gene of interest and combine it with
the plasmid, thus making a recombinant molecule (Fig. 1a). This
involves designing a PCR primer pair that specifically flanks the
region of DNA you wish to amplify. For good specificity and
stringency, the forward and reverse primers are typically ~20 bases
or more in length. Rarely will you find a restriction endonuclease
recognition site perfectly adjacent to the DNA sequence you wish
to clone. Therefore, each primer should also contain an extra
(non-binding) section with the desired restriction enzyme recogni-
tion sequence, as illustrated in Fig. 1a, Step 1. As an example, the
forward and reverse primers used for cloning the E. coli Ligase A
gene are shown in Fig. 3.
Following PCR amplification of the target DNA, the product
(often referred to as the “insert”) is purified and ready for cloning.
The next step is to enzymatically digest both the DNA insert and
the vector you wish to insert it into, using the restriction endonu-
clease enzymes you chose earlier (Fig. 1a, Step 2). This step creates
the complementary free ends on the two DNA molecules, which
are then combined in the final step, ligation. In this step, a recom-
binant DNA molecule is created by the action of DNA ligase
(Fig. 1a, Step 3). The most frequent choice of a DNA ligase for
this purpose is the T4 DNA ligase, which uses ATP as a cofactor and
is capable of ligating both sticky and blunt ends.
Although ligation protocol does not vary considerably between
cloning experiments, the possible resulting molecules do. A ligation
reaction can produce three different output molecules, as shown in
Classical Cloning 7
Fig. 3 Example of PCR primer pair for amplifying the E. coli Ligase A (LigA) gene. The target LigA gene is
depicted as a heteroduplex with the top strand in black and bottom strand in grey. The regions of the forward
primer (left) and reverse primer (right) that are complementary to the LigA gene are shown in blue. Hydrogen
bonding between gene and primer during PCR amplification is indicated by blue dots between the strands. The
recognition sequences for restriction endonucleases (NdeI, CATATG and BamHI, GGATCC) are shown in red.
Black bases on the primers’ 5′-ends are random-sequence bases added to ensure restriction enzyme binding
and cutting
Fig. 4: DNA insert can be joined with the cloning vector resulting
in a recombinant DNA molecule, a vector can self-ligate without
the inclusion of the target DNA sequence, or two target DNA
sequences can be joined in tandem [13]. The undesired vector
self-ligation and tandem insert DNA forms will be more likely to
form if blunt-ended restriction endonucleases are used. However,
the type of restriction endonuclease used is not the only contribut-
ing parameter. The output of the ligation reaction will also depend
on other properties of the DNA molecules utilized, such as length/
size of the insert or vector, length of the cohesive ends (1, 2, 3, or
4 nucleotide overhangs), and the concentration ratio of the DNA
insert to the vector in the reaction. The latter is an important
parameter, and often ligation reactions require oversaturation
with the DNA insert compared to the vector to ensure
incorporation.
8 Ana Mikić et al.
Fig. 4 Potential outcomes of a ligation reaction in molecular cloning. During any ligation reaction, the DNA
insert and cloning vector are first digested with the relevant restriction enzymes (top image). DNA strand
joining by a DNA ligase can then produce different outcomes: (1) the two ends of the linearized plasmid re-join
since the ends are close together in 3D space and more likely to meet each other and be resealed (undesired
outcome); (2) an insert and linearized vector meet and are joined successfully (desired outcome), or (3)
depending on the length of the inserts, free ends of the DNA insert join up to produce tandem (or more) repeats
(undesired outcome). The trick to cloning is to try a few different ratios of insert: linearized vector to favor
outcome (2)
1.6 Transformation The final steps of a typical cloning protocol aim to multiply the
synthesized recombinant DNA molecule in host cells (bacteria) and
to exploit various methods for the selection of the obtained colo-
nies. To multiply the recombinant DNA molecule, it must first be
introduced into the selected host cells in the fourth step of the
standard cloning protocol, transformation (Fig. 1b, Step 4). Simi-
lar to deciding on the appropriate restriction endonucleases and a
compatible vector, the choice of host cells in which you aim to
multiply your recombinant DNA molecules is also crucial for the
success of the experiment. This decision should be made early on
when designing the experiment and selecting the cloning vector, as
not all vectors can be taken up by all host cells.
In our case, using a plasmid vector, the most common choice is
to transform it into a bacterial cell. The available bacterial cells are
most often strains of Escherichia coli or Lactococcus lactis. These are
frequently engineered for protein expression, but some strains are
Classical Cloning 9
Fig. 5 Flowchart for selection of competent cells. This decision tree may help you select the best bacterial
strain for your cloning reaction. Work from left to right, and you can choose the bacterial strain best suited to
your downstream application. The cells used in our example method for cloning and expressing the LigA gene
(BL21 (DE3)) are underlined
not, so pay attention when selecting those you wish to use in your
experiment [14]. A useful flowchart to help you choose the right
cells is provided in Fig. 5. Other than the genetic constitution of
the bacterial cells used, the success of transformation will also
depend on the transformation method performed. The two distinct
methodological approaches are transformation by electroporation
or chemical transformation [15]. Transformation by electropora-
tion (also known as electropermeabilization) is achieved by apply-
ing a short, high-voltage pulse to bacterial cells. The electric shock
makes the cell membrane more permeable and susceptible to the
DNA uptake. This method is simple, fast, and gives high transfor-
mation efficiency for E. coli cells [16]. Alternatively, chemical trans-
formation is achieved by heat-shocking bacterial cells at 42 °C to
allow for the vector uptake. Prior to the heat shock, cells must be
treated with high salt concentrations to increase the cell membrane
permeability, and these are known as “competent” cells. Nowadays,
10 Ana Mikić et al.
1.7 Selection and Whether the host cells will take up the recombinant DNA molecule
Screening can never be guaranteed, therefore, numerous methods have been
developed to assess the success of the steps carried out so far.
However, even with these methods developed, testing each indi-
vidual colony would be a laborious task if not for a selection step.
Selection is the fifth step of the standard cloning protocol (Fig. 1b,
Step 5), in which successfully transformed bacterial cells are
selected for the phenotype they exhibit (positive selection) or lack
(negative selection). The principle of positive selection is that cells
are selected for the presence of a property encoded by the vector, as
only cells containing the vector will be able to form colonies. The
examples are auxotrophy and the aforementioned antibiotic selec-
tion test, where only bacterial cells that have obtained an antibiotic
resistance gene carried by the vector will grow on a media contain-
ing the same antibiotic [17]. In contrast, the principle of negative
selection is based on successfully transformed bacterial cells not
exhibiting a property encoded by the vector and, therefore, being
unable to grow and form colonies. The examples are SacB-counter
selection or toxin-antitoxin system. By performing the selection
step, the number of obtained colonies is limited only to the ones
that have been successfully transformed and contain the vector.
This enables you to focus on assessing the smaller number of
colonies for the presence of the target recombinant DNA
molecule [18].
The purpose of the sixth and last step of a standard cloning
protocol is screening (Fig. 1b, Step 6). Often, selection and screen-
ing tests can be performed simultaneously, as in the case of the well-
known blue-white test exploiting the lactose (X-gal) metabolism
pathway and β-galactosidase enzyme activity. This test offers a fast
and simple way of assessing bacterial colonies for the presence of
the target recombinant DNA molecule through observation of the
plates with the naked eye. Alternatively, colony PCR is a rapid
screening method in which primers used in the first step of the
cloning experiment are repurposed for the detection of recombi-
nant DNA molecules. However, these and many other methods
(reporter gene or lethal gene assays) can be erroneous and give a
false-positive result [19]. Therefore, the best practice is always to
send samples of your grown colonies to be sequenced using Sanger
dideoxy sequencing or other available sequencing methods. We
provide an example of a standard cloning experiment in Fig. 6 for
the cloning of the E. coli Ligase A and Ligase B genes.
Classical Cloning 11
A M 1 2 B
3000 bp
2000 bp
1500 bp
M 1 2 3 4 5
1000 bp
C
3000 bp
2000 bp
500 bp 1500 bp
1000 bp
500 bp
Fig. 6 Example cloning results. The genes for DNA Ligase A and B were amplified from E. coli strain K-12,
cloned into plasmid pET28b, transformed into BL21(DE3) cells, checked by colony PCR, and sequenced. Panel
A: 1% (w/v) agarose gel with 1 kb marker (lane M) and samples of amplified Ligase A gene (lane 1, green
arrow) and Ligase B gene (lane 2, blue arrow). Panel B: Agar plates showing transformants for Ligase A (left
plate) and Ligase B (right plate). Panel C: 1.5% (w/v agarose gel with 1 kb marker (lane M) and samples of
colony-PCR reactions for five separate colonies from the Ligase A plate in Panel B. A positive result is visible in
lane 1. Panel D: Sanger dideoxy sequencing showing that the Ligase A gene is successfully cloned, without
mutations; the green arrow indicates the methionine start codon for Ligase A
1.8 Current and Since its first application in 1973, molecular cloning has become an
Future Applications essential technique utilized in every area of biological studies
including genetics, molecular cell biology, developmental biology,
neurobiology, neuroscience, and immunology. Its importance is
highlighted by the fact that nowadays, for a simple molecular
cloning experiment, you may choose between 240 available proto-
cols exploiting more than 3000 restriction endonucleases that rec-
ognize 230 different sequences, at least 1000 different cloning
vectors, and hundreds of available host cells [20]. Moreover, new
advances of this technology are continuously being developed, such
as TA cloning, TOPO cloning, sequence and ligation independent
12 Ana Mikić et al.
2 Materials
2.1 Polymerase 1. DNA containing the target sequence, region, or gene that you
Chain Reaction (PCR) wish to clone. This can be in many different forms, including
and Agarose Gel intact cells, purified genomic DNA, plasmid preparations,
Electrophoresis DNA libraries, mitochondrial preparations, chloroplast pre-
parations, or previous PCR products (see Note 1). You will
need a ~20 μL stock containing 10 ng of the DNA in
nuclease-free water (0.5 ng/μL).
2. PCR primers that flank the DNA target sequence and that
contain the recognition sequence(s) of the chosen restriction
enzyme(s) (see Note 2). You will need a working stock of
10 μM in ~20 μL of nuclease-free water each.
3. PCR buffer (10× stock): 100 mM Tris–HCl, pH 8.6, 500 mM
KCl, 1.5 mM MgCl2, 50% (v/v) glycerol, 0.8% (w/v) IGEPAL
CA-630, 0.5% (w/v) Tween-20. Use 1× dilution.
4. Nucleotide mix: 10 mM dNTPs. Store at -20 °C.
5. Taq DNA Polymerase: 5 U/μL from a commercial supplier.
Store at -20 °C.
6. Thin-wall 0.2 mL polypropylene PCR tubes.
7. PCR thermocycler set with the following program: For the
selection of the annealing temperature (see Note 3).
8. Microcentrifuge tubes (1.5 mL).
9. DNA size ladder: a 500 bp—10 kb ladder provided with a 6×
DNA Gel Loading Dye. Bought from a commercial supplier
and stored at -20 °C.
10. A sterilized conical flask.
11. An agarose gel electrophoresis system. This includes a power-
pack, gel cassette with removable end seals, combs, agarose
powder (low endo-osmosis form), 10× TBE buffer stock,
10 mg/mL ethidium bromide (EtBr), and distilled water.
Classical Cloning 13
12. TBE buffer (10× stock). For 1 L add: 108 g Tris base, 55 g
boric acid, 900 mL distilled water, 40 mL 0.5 EDTA solution,
pH 8.0. To obtain the 1× TBE running buffer, dilute the 10×
TBE stock 10-fold in distilled water.
13. TBE running buffer (1×). For 1 L, add: 10 mL 10× TBE stock
to 900 mL distilled water. Add 50 μL of 10 mg/mL ethidium
bromide stock (0.5 μg/mL final concentration).
14. Small microwave oven.
15. An ultraviolet (UV) transilluminator capable of excitation at
302/312 nm (UV-B) or 365 nm (UV-A) for visualizing
DNA-ethidium bromide complexes in agarose gels. It should
be equipped with an LED camera for photo capture. Here we
use a G:BOX gel doc.
16. A commercial spin-column PCR purification kit. Use per man-
ufacturer’s instructions.
17. A small-volume UV spectrophotometer (e.g. a NanoDropTM
or PicoDropTM device).
2.2 Restriction 1. A cloning vector containing the same restriction enzyme rec-
Enzyme Digestion ognition sites that were introduced at the ends of the DNA
insert by the PCR primer pairs. Here we use the pET28b(+)
5368 bp expression plasmid vector with NdeI and BamHI
recognition sites with a kanamycin resistance gene. For the
guidance on vector selection (see Note 4).
2. NdeI restriction enzyme: 20 U/μL from a commercial sup-
plier. Store at -20 °C (see Note 5).
3. BamHI restriction enzyme: 20 U/μL from a commercial sup-
plier. Store at -20 °C (see Note 5).
4. Restriction enzyme buffer (10× stock), for example, rCutS-
martTM buffer: 200 mM Tris-acetate, 100 mM magnesium
acetate, 500 mM potassium acetate, 1 mg/mL recombinant
bovine serum albumin (BSA), pH 7.9 at 25 °C. Store at -20 °
C (see Note 6).
5. A water bath set to 37 °C.
2.4 Transformation Ensure your laboratory is prepared for the microbiology work.
While handling bacterial cells, you should be working using aseptic
techniques.
1. Competent bacterial cells. Selection depends on the down-
stream applications of the cloning experiment (for guidance
see Note 7). Here we used BL21-DE3 competent cells from
E. coli that can be used for protein expression. Cells are usually
obtained from a commercial supplier; refer to the manufac-
turer’s instructions for storing and avoid freeze/thawing until
being used. You may make your own competent cells, recipes
are available online for this.
2. Ice and ice bucket.
3. A water bath set to 42 °C.
4. Freshly prepared SOC broth: 0.5% (w/v) yeast extract, 2%
(w/v) tryptone, 10 mM NaCl, 2.5 mM KCl, 10 mM MgCl2,
10 mM MgSO4, 20 mM glucose, pH 7.0 (see Note 8). To
prepare 1 L of SOC media combine 20 g tryptone, 5 g yeast
extract, and 0.5 g NaCl. Stir the ingredients in 950 mL of
distilled water until they dissolve. Add 10 mL of 250 mM
KCl and adjust the pH to 7.0 with NaOH. Adjust the volume
to 1 L with distilled water. Transfer 250 mL to four clean Pyrex
bottles. Sterilize each by autoclaving at 121 °C (and 15 psi) for
20 min on liquid cycle with the cap loosely attached by auto-
clave indicator tape. Before using the media add 5 mL of sterile
2 M MgCl2 solution and 20 mL of sterile 1 M glucose solution,
gently mix.
2.5 Selection 1. Freshly prepared and pre-warmed (37 °C) SOC media selec-
tion plates (see Note 9). To prepare 1 L of SOC media combine
20 g tryptone, 5 g yeast extract, and 0.5 g NaCl. Stir the
ingredients in 950 mL of distilled water until they dissolve.
Add 10 mL of 250 mM KCl and adjust the pH to 7.0 with
NaOH. Adjust the volume to 1 L with distilled water. Add
1.5 g of bacto agar. Transfer 250 mL to four clean Pyrex
bottles. Sterilize each by autoclaving at 121 °C (and 15 psi)
for 20 min on liquid cycle with the cap loosely attached by
autoclave indicator tape. Once cooled to ~60 °C, add 5 mL of
sterile 2 M MgCl2 solution, 20 mL of sterile 1 M glucose
solution, and 50 mg/mL kanamycin (see Note 10), gently
mix. Pour approximately 15 mL into each labeled petri dish
(final concentration of 50 μg/mL kanamycin). Label, seal, and
store inverted at 4 °C until needed.
2. A water bath set to 37 °C.
3. Sterile 50-mL polypropylene tubes for liquid media growth.
4. Sterile, disposable plastic spreader.
5. Microbiology incubator-shaker set to 37 °C.
Classical Cloning 15
2.6 Screening by 1. Sterile 50-mL polypropylene tubes for liquid media growth.
PCR 2. The same number of sterile microcentrifuge tubes.
3. SOC liquid broth.
4. Nuclease-free water: high-quality molecular biology grade;
stored at 4 °C.
5. Benchtop cooled-centrifuge capable of 13,000 rpm (15,100 ×
g) and chamber temperature of 0 °C.
6. Taq PCR master-mix (1×): 10 mM Tris-HCl, pH 8.6, 50 mM
KCl, 1.5 mM MgCl2, 0.2 mM dNTPs (50 μM each), 5% (v/v)
glycerol, 0.08% (w/v) IGEPAL CA-630, 0.05% (w/v) Tween-
20, 2 μM forward primer, 2 μM reverse primer, 25 Units/mL
Taq DNA Polymerase. Prepare for the final reaction volume of
25 μL. For multiple PCR reactions (see Note 11). Keep on ice.
7. A laboratory vortex mixer.
8. PCR thermocycler set with the same program as used for PCR
amplification of the DNA insert. Increase the number of cycles
to 40.
9. Agarose gel electrophoresis equipment; same as in
Subheading 2.1.
10. A commercial spin-column plasmid purification kit. Use per
manufacturer’s instructions.
3 Methods
Order and prepare all the materials and reagents in advance. Con-
duct all steps at room temperature unless otherwise specified. Wear
lab coat, gloves, and appropriate PPE at all times to avoid contami-
nation and minimize the risk of nuclease degradation of samples.
Table 1
PCR cycle conditions
3.2 Restriction 1. Take a clean microcentrifuge tube and label it “Vector” or “V.”
Enzyme Digestion To the tube add 1 μg of the plasmid vector, 1 μL of the NdeI
enzyme, 1 μL of the BamHI enzyme, 3 μL of the rCutSmartTM
buffer, and bring up to 30 μL with nuclease-free water.
2. Take a clean microcentrifuge tube and label it “DNA” or “D.”
To the tube, add 1 μg of the purified DNA sample from the
PCR reaction, 1 μL of the NdeI enzyme, 1 μL of the BamHI
enzyme, 3 μL of the rCutSmartTM buffer, and bring up to
30 μL with nuclease-free water.
3. Incubate both tubes in a water bath at 37 °C for 1 h.
3.4 Transformation 1. Thaw the competent cells on ice for 10–30 min. Thaw as many
single-use competent cell tubes as transformation reactions you
aim to conduct. Label them “T1,” “T2,” etc.
2. Transform the cells by adding 5 μL of the “L” reaction mix to
each of the competent cell mixtures thawed (“T1,” “T2,”
etc.). Mix by gently flicking the tube 4–5 times.
3. Incubate the tubes labeled “T” on ice for 30 min (see Note 18).
4. Heat shock the cells by placing the “T” tubes in water bath at
exactly 42 °C for exactly 45 s (see Note 19). Do not mix or
shake.
5. Immediately place the “T” tubes back on ice for another 2 min.
18 Ana Mikić et al.
3.5 Selection 1. To each “T” tube add 250 μL of the pre-warmed SOC liquid
media and cap the tube.
2. Incubate the “T” tubes in water-bath at 37 °C for 1 h. Shake
vigorously (225 rpm) or rotate (see Note 20).
3. Mix the cells by flicking and inverting the “T” tubes. In the
clean microcentrifuge tubes, perform several 10-fold serial
dilutions of the “T” tube contents in the SOC liquid media.
We recommend three 10-fold dilutions.
4. Using a disposable plastic spreader, spread 50–100 μL of each
dilution onto a separate SOC selection plate, labeled with the
same number as the transformation reaction tube, by using a
spread plate technique. Work using aseptic techniques. You
should also include two control plates (see Note 21).
5. Incubate the inverted plates overnight at 37 °C. Carefully
examine the plates the next day for colonies.
3.6 Screening by 1. Depending on how many colonies have grown on your selec-
PCR (Colony PCR) tion plate(s) and how many you wish to screen, take the same
number of universal (20 mL) and microcentrifuge (1.5 mL)
tubes. Label them “A,” “B,” “C,” etc., so that each universal
tube matches one microcentrifuge tube.
2. To each universal tube add 5 mL of the liquid SOC media.
3. To each microcentrifuge tube add 10 μL of nuclease-free water.
4. Using a sterile pipette tip, pick a single colony from the selec-
tion plate. Dip the tip into the labeled microcentrifuge tube
containing 10 μL nuclease-free water and swirl for 10 s. Take
the pipette tip out of the microcentrifuge tube and drop it into
the universal tube containing liquid SOC media with the
matching label. Repeat this step for every colony you wish to
screen using a different set of labeled microcentrifuge/univer-
sal tubes (see Note 22). Set the universal tubes on the side of
your bench.
5. Collect all of the used microcentrifuge tubes and spin them in a
centrifuge for 2 min at maximum speed (15,000× g). You
should observe a pellet and supernatant.
6. Label the same number of PCR tubes to match one of the
microcentrifuge tubes as “A,” “B,” “C,” etc. To each PCR
tube add 2.5 μL of the supernatant from the corresponding
microcentrifuge tube.
7. To each PCR tube add 22.5 μL of the previously prepared PCR
master-mix.
8. Close the lid of PCR tubes and place them in the thermocycler.
Run the 3-step standard PCR cycling under the previously set
program outlined in Table 1 with the number of cycles
increased to 40.
Classical Cloning 19
4 Notes
1. Before any wet lab work decide on the purpose of your cloning
experiment. Selection of the cloning vector, restriction
enzymes, and competent bacterial cells will depend upon the
further applications of the cloned sequence of interest. Here we
aim to clone Escherichia coli DNA ligase A (ligA) gene
(2016 kb) using directional cloning (two restriction enzymes)
into an expression vector (pET-28b), which will be trans-
formed into competent cells capable of protein production
(BL21(DE3)) to obtain the 671aa monomer. For guidance
on choosing a vector and competent cells appropriate for
your experimental purpose (see Notes 4 and 7).
2. When designing your primers refer to the guidelines outlined
in Table 2. You can also use Fig. 1 as a visual aid.
3. Annealing temperature depends on the melting temperature
(Tm) of the primers used. You should aim for 5 °C below the
calculated Tm and for it to be in the range of 45–68 °C.
Annealing temperature can be optimized by performing an
annealing screen: a temperature gradient PCR starting 5 °C
below the calculated Tm.
4. When selecting a cloning vector consider the following
qualities:
(i) It must possess an origin of replication.
(ii) A high replication rate is desirable to obtain a high copy
number per cell cycle of a transformed cell.
(iii) It should possess a range of restriction enzyme recognition
sites to clone into. It must possess the restriction site for the
selected restriction enzymes to be used for target DNA
20 Ana Mikić et al.
Table 2
Primer design instructions
References
1. Linn S, Arber W (1968) Host specificity of 8. Cohen SN, Chang ACY, Boyer HW, Hellingt
DNA produced by Escherichia coli, in vitro RB (1973) Construction of biologically func-
restriction of phage fd replicative form. Proc tional bacterial plasmids in vitro. Proc Natl
Natl Acad Sci U S A 59(4):1300–1306 Acad Sci U S A 70(11):3240–3244
2. Danna K, Nathans D (1971) Specific cleavage 9. Preston A (2003) Choosing a cloning vector.
of Simian Virus 40 DNA by restriction endo- Methods Mol Biol 235:19–26
nuclease of hemophilus influenzae. Proc Natl 10. Carter M, Shieh J (2015) Chapter 10 – Molec-
Acad Sci U S A 68(12):2913–2917 ular cloning and recombinant DNA
3. Kellenberger G, Zichichi ML, Weigle JJ (1961) technology. In: Carter M, Shieh J (eds) Guide
Exchange of DNA in the recombination of to research techniques in neuroscience, 2nd
bacteriophage λ. Proc Natl Acad Sci U S A edn. Academic, New York, pp 219–237
47(6):869–878 11. Mertz JE, Davis RW (1972) Cleavage of DNA
4. Meselson M, Weigle JJ (1961) Chromosome by R1 restriction endonuclease generates cohe-
breakage accompanying genetic recombination sive ends. Proc Natl Acad Sci U S A 69(11):
in bacteriophage. Proc Natl Acad Sci U S A 3370–3374
47(6):857–868 12. Green MR, Sambrook J (2020) Cloning in
5. Jackson DA, Symons RH, Berg P (1972) Bio- plasmid vectors: directional cloning. Cold
chemical method for inserting new genetic Spring Harb Protoc 2020(11):485–488
information into DNA of Simian Virus 40: 13. Upadhyay A, Upadhyay K (2009) Recombi-
circular SV40 DNA molecules containing nant DNA technology. In: Basic molecular
lambda phage genes and the galactose operon biology. Himalaya Publishing House, Global
of Escherichia coli. Proc Natl Acad Sci U S A Media, pp 452–506
69(10):2904–2909 14. Fakruddin M, Mohammad Mazumdar R, Bin
6. Griffith F (1928) The significance of pneumo- Mannan KS, Chowdhury A, Hossain MN
coccal types. J Hyg 27(2):113–159 (2013) Critical factors affecting the success of
7. Cohen SN, Chang ACY, Hsu L (1972) Non- cloning, expression, and mass production of
chromosomal antibiotic resistance in bacteria: enzymes by recombinant E. coli. ISRN Bio-
genetic transformation of Escherichia coli by technol 2013:1–7
R-factor DNA. Proc Natl Acad Sci U S A 15. Hanahan D, Jessee J, Bloom FR (1991) Plas-
69(8):2110–2114 mid transformation of Escherichia coli and
24 Ana Mikić et al.
other bacteria. Methods Enzymol 204 and preventing false positives. In: Brown G
(C):63–113 (ed) Molecular cloning – selected applications
16. Dower WJ, Miller JF, Ragsdale CW (1988) in medicine and biology. In Tech, Rijeka
High efficiency transformation of E. coli by 20. Sambrook J, Russell DW (2001) Molecular
high voltage electroporation. Nucleic Acids cloning: a laboratory manual, vol 1, 3rd edn.
Res 16(13):6127–6145 Cold Spring Harbor Laboratory Press,
17. Manna S, Harman A, Accari J, Barth C (2013) New York
Altering the selection capabilities of common 21. Plasmids 101 A Desktop Resource Plasmids
cloning vectors via restriction enzyme 101: A Desktop Resource (3rd edn) (2017).
mediated gene disruption. BMC Res 6(1):1–9 Retrieved November 13, 2021, from www.
18. Nicholl DST (2002) Selection, screening and addgene.org
analysis of recombinants. An introduction to 22. Kibbe WA (2007) OligoCalc: an online oligo-
genetic engineering. Cambridge University nucleotide properties calculator. Nucleic Acids
Press, Cambridge, pp 132–150 Res 35. (Web Server issue)
19. Padmanabhan S, Banerjee S, Mandi N (2011)
Screening of bacterial recombinants: strategies
Chapter 2
Abstract
Molecular cloning is a routine technique for many laboratories with applications from genetic engineering
to recombinant protein expression. While restriction-ligation cloning can be slow and inefficient, ligation-
independent cloning uses long single-stranded overhangs generated by T4 DNA polymerase’s 3′ exonu-
clease activity to anneal the insert and plasmid vector prior to transformation. This chapter describes a fast,
high-efficiency protocol for inserting one or more genes into a vector using sequence- and ligation-
independent cloning (SLIC).
Key words SLIC, PCR, Cloning, T4 polymerase, Genetic engineering, Protein expression
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_2,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
25
26 Robert A. Holland
Fig. 1 General concept of SLIC. The gene of interest is amplified using primers with 5′ sequences (typically
15–40 bp) complementary to the insertion site within the vector. Incubation of the resulting PCR product with
T4 DNA polymerase generates complementary overhangs. Similarly, the vector is linearized either by
restriction digestion or PCR and overhangs generated by T4 DNA polymerase. When mixed, the insert and
vector anneal to form a nicked plasmid which can be transformed directly into competent cells where gaps in
the sequence are repaired
2 Materials
2.2 Gel Analysis 1. Tris-acetate EDTA (TAE) 50× stock: dissolve 242 g tris base,
57.1 mL glacial acetic acid, and 100 mL 0.5 M EDTA pH 8.0,
topped up to 1 L with deionized H2O.
2. 1% agarose gel: TAE, 10 g/L agarose.
3. Ethidium Bromide.
4. 6× DNA loading dye.
5. DNA ladder (1 kbp/100 bp).
6. Gel tank.
7. UV transilluminator suitable for agarose gel visualization.
3 Method
3.1 PCR See Note that unless specified, all steps should be carried out on ice.
Amplification of Insert
1. Dissolve the primers in nuclease-free H2O, aiming for a final
and Vector
concentration of 50–100 μM. Measure the exact concentration
using the NanoDrop 2000 spectrophotometer.
2. In 200 μL PCR tubes (one for each insert or vector), prepare
150 μL reaction mixture for the insert(s): 30 μL 5× Phusion
buffer, 0.5 μM forward primer, 0.5 μM reverse primer, 200 μM
dNTPs (each), 0.2–0.5 ng/μL DNA template, 0.02 U/μL
Phusion polymerase, topped up with nuclease-free H2O, and
mix thoroughly by pipetting. Split each reaction mixture
between 6× 25 μL reactions in separate PCR tubes. Reactions
can be overlaid with mineral oil (see Note 6).
3. Place each of the six reaction tubes (see Note 7) containing
insert or vector with equal spacing across the thermocycler and
program the thermocycler as follows:
1 cycle Initial denaturation, 98 °C, 10 s, 1 cycle
25 cycles Denature, 98 °C, 10 s
Anneal, 50–70 °C gradient, 30 s.
Extension, 72 °C, 15–30 s per kb (see Note 8).
1 cycle Final extension, 72 °C, 5 min.
4. While the PCR amplification is running, set up the gel tank
with a 1% agarose gel with TAE buffer. Load 5 μL from each
reaction tube, each mixed with 1 uL 6× DNA loading dye, into
separate wells of a 1% agarose gel, flanked by DNA ladder, and
run at 100 V, 30 min (for 100 mL gel). Visualize the gel using a
UV transilluminator and decide on the optimal annealing tem-
perature for vector and insert (see Note 9).
5. Scale up the amplification by repeating steps 2 and 3, except
with all 25 μL reactions at the optimal annealing temperature
decided on in step 4.
6. Pool reactions from each amplicon and remove 10 μL for
further analysis. Add DpnI and 10× fast digest buffer (5 μL
each per 50 μL) and incubate in the thermocycler at 37 °C for
10 min, followed by 4 °C (see Note 10).
7. Purify the amplicons using a PCR reaction cleanup kit, follow-
ing the manufacturer’s instructions, ensuring the product is
eluted in nuclease-free H2O. Measure DNA concentration
using a Nanodrop 2000 spectrophotometer.
8. Mix insert and vector in a molar ratio of 2.5:1 (0.0625 pmol
insert & 0.025 pmol vector), 1× T4 polymerase buffer and
0.5 U T4 polymerase, topped up 10 μL with nuclease-free
Sequence and Ligation-Independent Cloning 29
4 Notes
Fig. 2 Example gel visualizing cloning steps. This 8 kb vector had two bands
corresponding to circular and supercoiled plasmid. Vector linearization by PCR
removed a 500 bp sequence, resulting in a band of a lower molecular weight.
Recombination of linearized vector with a molar excess of 600 bp insert yielded
a high cloning efficiency, indicated by a single band at approximately 8 kb. The
low molecular weight contaminant was the same size as the insert, so it would
not affect transformation
References
1. Celie PH, Parret AH, Perrakis A (2016) academic.oup.com/nar/ar ticle-lookup/
Recombinant cloning strategies for protein doi/10.1093/nar/18.20.6069
expression. Curr Opin Struct Biol 38:145– 4. Tillett D, Neilan B (1999) Enzyme-free clon-
154. Available from: https://linkinghub. ing: a rapid method to clone PCR products
elsevier.com/retrieve/pii/S0959440X16300 independent of vector restriction enzyme
677 sites. Nucleic Acids Res 27(19):26e–26.
2. Shuldiner AR, Scott LA, Roth J (1990) Available from: https://academic.oup.com/
PCR-induced (ligase-free) subcloning: a rapid nar/article-lookup/doi/10.1093/nar/27.1
reliable method to subclone polymerase chain 9.e26
reaction (PCR) products. Nucleic Acids Res 5. Li MZ, Elledge SJ (2007) Harnessing homol-
18(7):1920–1920. Available from: https:// ogous recombination in vitro to generate
academic.oup.com/nar/ar ticle-lookup/ recombinant DNA via SLIC. Nat Methods
doi/10.1093/nar/18.7.1920 4(3):251–256. Available from: http://www.
3. Aslanidis C, de Jong PJ (1990) Ligation- nature.com/articles/nmeth1010
independent cloning of PCR products 6. Stevenson J, Krycer JR, Phan L, Brown AJ
(LIC-PCR). Nucleic Acids Res 18(20): (2013) A practical comparison of ligation-
6069–6074. Available from: https:// independent cloning techniques. PLoS One
32 Robert A. Holland
8(12):e83888. Available from: http://www. multiple DNA fragments with short end
pubmedcentral.nih.gov/articlerender.fcgi? homologies. PLoS One 10(9):e0137466
artid=3871625&tool=pmcentrez& 9. Jeong J-Y, Yim H-S, Ryu J-Y, Lee HS, Lee J-H,
rendertype=abstract Seen D-S et al (2012) One-step sequence- and
7. Scholz J, Besir H, Strasser C, Suppmann S ligation-independent cloning as a rapid and
(2013) A new method to customize protein versatile cloning method for functional geno-
expression vectors for fast, efficient and back- mics studies. Appl Environ Microbiol 78(15):
ground free parallel cloning. BMC Biotechnol 5440–5443. Available from: https://journals.
1 3 ( 1 ) : 1 2 . A v a i l a b l e f r o m : h t t p s : // asm.org/doi/10.1128/AEM.00844-12
bmcbiotechnol.biomedcentral.com/ar ti 10. Gibson DG, Young L, Chuang R-Y, Venter JC,
cles/10.1186/1472-6750-13-12 Hutchison CA, Smith HO (2009) Enzymatic
8. Kostylev M, Otwell AE, Richardson RE, assembly of DNA molecules up to several hun-
Suzuki Y (2015) Cloning should be simple: dred kilobases. Nat Methods 6(5):343–345.
Escherichia coli DH5á-mediated assembly of Available from: http://www.nature.com/arti
cles/nmeth.1318
Chapter 3
Abstract
Here we describe the in vivo DNA assembly approach, where molecular cloning procedures are performed
using an E. coli recA-independent recombination pathway, which assembles linear fragments of DNA with
short homologous termini. This pathway is present in all standard laboratory E. coli strains and, by
bypassing the need for in vitro DNA assembly, allows simplified molecular cloning to be performed without
the plasmid instability issues associated with specialized recombination-cloning bacterial strains. The
methodology requires specific primer design and can perform all standard plasmid modifications (inser-
tions, deletions, mutagenesis, and sub-cloning) in a rapid, simple, and cost-efficient manner, as it does not
require commercial kits or specialized bacterial strains. Additionally, this approach can be used to perform
complex procedures such as multiple modifications to a plasmid, as up to 6 linear fragments can be
assembled in vivo by this recombination pathway. Procedures generally require less than 3 h, involving
PCR amplification, DpnI digestion of template DNA, and transformation, upon which circular plasmids are
assembled. In this chapter we describe the requirements, procedure, and potential pitfalls when using this
technique, as well as protocol variations to overcome the most common issues.
Key words Molecular cloning, In vivo DNA assembly, recA-independent recombination, IVA clon-
ing, Sub-cloning, Site-directed mutagenesis
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_3,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
33
34 Sandra Arroyo-Urea et al.
7]. Using in vivo DNA assembly, all standard plasmid DNA mod-
ifications can be performed, including sequence insertions, dele-
tions, point-mutagenesis, and sub-cloning of large fragments
between vectors. When employed as a cloning tool, linear DNA
fragments are generated in vitro, primarily by PCR, with homolo-
gous termini of around 15–30 bp that direct plasmid assembly
in vivo. After transformation, endogenous single-stranded exonu-
cleases (ExoIII/V) [8] degrade the termini of these linear frag-
ments to single-stranded DNA, which allows annealing between
homologous fragments in vivo, before DNA repair (LigA) assem-
bles a circular plasmid [9, 10].
Cloning protocols generally comprise the following: (1) primer
design, (2) PCR amplification, to introduce modifications and
homologous sequences, (3) digestion of the parental DNA using
the methylase-dependent restriction enzyme DpnI, and (4) trans-
formation into standard laboratory E. coli before subsequent col-
ony screening and selection. This protocol can be used to perform
all types of plasmid modifications, from inserting and deleting
sequences to site-directed mutagenesis and sub-cloning, each dic-
tated by primer design. Furthermore, up to six DNA fragments can
be assembled simultaneously, allowing complex cloning strategies
to be achieved in a single step; however, method efficiency
decreases as procedure complexity increases. Given the principal
requirement for cloning using recombination is linear DNA frag-
ments, in vivo DNA assembly can also be combined with restriction
enzyme-linearized plasmids or synthesized linear double-stranded
genes, which can overcome PCR amplification issues or further
simplify procedures (Fig. 1). Given that there are no requirements
for commercial kits or specialized bacteria, this approach is accessi-
ble to any molecular biology laboratory. Additionally, since the
homology requirements are similar to other enzyme or
recombination-based commercial approaches (e.g., Gibson assem-
bly [11]), primers designed for in vivo assembly cloning can also be
combined/used with such in vitro assembly methods as an alterna-
tive backup route. Here, we describe reagents and protocols to
perform in vivo DNA assembly for plasmidic DNA cloning and
modification as performed in our laboratory.
2 Materials
3 Methods
3.1 Primer Design We recommend using software for visualizing both the original and
target DNA sequences for the design of oligos, as well as software
for the calculation of annealing temperatures (Tm). We use the
freely accessible Snapgene Viewer program for primer sequence
design and the OligoCalc webserver (http://biotools.nubic.
northwestern.edu/OligoCalc.html) for annealing temperature cal-
culation [12]. All Tm values reported in this chapter are calculated
using this webserver. Accurate primer design is critical to the suc-
cess of in vivo DNA assembly. Regardless of the modification to be
made (insertion, deletion, mutagenesis, or sub-cloning), primers
consist of two regions: a 3′ region that anneals to the template
DNA (template binding region) and a 5′ homologous region that
drives in vivo recombination. First, design the template binding
region, which has the same requirements of standard PCR oligo
design (at least 18–22 bp and Tm values of ~60 °C). The homolo-
gous region is included 5′ to this sequence, and it should be ≥15 bp
and have a Tm ≥ 50 °C (see Note 7). As a rule of thumb, a
homologous region of ~20–25 bp is sufficient to ensure efficient
recombination (usually providing a Tm ≥ 50 °C), and lengths of up
to 35 bp can been used to enhance efficiency when assembling
≥5 DNA fragments simultaneously. Specific primer design require-
ments for each DNA modification are as follows:
Fig. 2 Primer design to perform insertions. Primers must have template binding regions (grey) that bind either
side of the insertion site (red) and a homologous region at the 5′ end (orange box) that allows in vivo
recombination. Based on the length of the desired insertion (depicted in green), the homologous regions for
each of the primers could be: (a) the entire insert (when the insertion is ~20 bp), (b) a sub-region of the insert
(when the insertion is >25–30 bp), or (c) extended beyond the insert sequence alone (<18 bp).
3.1.2 Deletion A deletion occurs when a desired DNA sequence is removed from
the template plasmid. Primers for a deletion are designed such that
they have their template binding regions annealing either side of
the deletion site, amplifying outwards (i.e., amplifying the entire
plasmid aside from the fragment to remove). A 5′-DNA sequence is
added to one of the primers, which is homologous to the 5′ end of
the primer’s pair (Fig. 3a) (see Note 9).
Fig. 3 Primer design to perform deletions, mutagenesis, and sub-cloning. (a) Deletions. Primers bind astride
the sequence to remove, with homologous regions at termini encoded in one primer. (b) Mutagenesis. Primers
flank the mutation site, similarly to deletions, with the new codon encoded in primer sequences. (c) Sub
cloning. Four primers are needed to add a DNA fragment to a target vector. Insert amplification primers are
designed to have specific homology to target vector termini (specific homologous regions for Fw (purple box)
and Rv primer (blue box) match vector sequence)
3.2 Using PCR for In the vast majority of cloning approaches, PCR will be used to
DNA Modification and amplify and/or modify the DNA sequences involved. Employing
Amplification restriction enzyme-dependent DNA linearization or synthetic
genes are possible alternatives, which will be addressed in Subhead-
ing 3.5. PCR-based cloning proceeds as follows:
1. Prepare PCR Master Mix (see Materials) and freeze aliquots for
future use. Defrost on ice on the day of use.
2. If primers arrive desiccated, make up to 100 μM in deionized
water.
Molecular Cloning Using In Vivo DNA Assembly 39
3.4 Performing Here, we have detailed how specific primers can be used to create a
Complex Procedures variety of individual plasmid modifications; however, multiple
modifications are often required and are historically carried out
sequentially, increasing the length of a cloning procedure. Using
in vivo DNA assembly, up to six fragments have been assembled
simultaneously [2, 5], where final assembly from individual frag-
ments is driven by their unique homologous sequences. This means
40 Sandra Arroyo-Urea et al.
TE
V
3C V
TE
RFP
RFP
is G
H
FLAG
A
0X1 FL
Fig. 4 Example of in vivo DNA assembly for complex cloning procedures. By combining three pairs of primers,
a N-terminal HisTag (×10 HisTag, yellow) is exchanged for a FLAG tag (FLAG, pink) and a new coding
sequence (green) is added downstream of the Red Fluorescent Protein gene (RFP, red), while replacing 3C
cleavage site (3C, brown) for a TEV protease site (TEV, blue). These multiple modifications can be performed
simultaneously using standard in vivo assembly primers
3.5 Alternative In vivo DNA assembly can assemble any linear DNA fragment that
Routes for Linear contains homologous regions to another linear DNA. Although
Fragment Generation: the most common and practical route to obtaining such sequences
Restriction Enzymes is PCR, alternative routes are possible. Restriction enzymes can be
and Synthetic Genes used to linearize a vector at a desired site for modification, allowing
insertion of a fragment of interest without complete vector amplifi-
cation. The inserted fragment must be designed to contain termini
homologous to the vector sequence either side of the restriction
site, which typically involves PCR amplification of the insert.
Co-transformation of these fragments allows assembly of the
novel plasmid in vivo (see Note 19).
An increasingly useful and accessible route to obtain linear
DNA fragments is gene synthesis. Currently, linear double-
stranded synthetic DNAs (e.g., Gene Fragments or gBlocksTM)
can be designed and purchased to be readily used in transformation
for in vivo DNA assembly. Novel sequences must simply be
designed to include termini homologous to the vector insertion
location (as previously described). This approach can be used for a
multitude of purposes: new genes, synthetic proteins, DNA frag-
ments that are not amenable for PCR amplification or simply, a
DNA fragment with many modifications that would be trouble-
some to obtain from the parental DNA. For use in in vivo DNA
assembly cloning, synthetic genes are co-transformed with a linear-
ized vector (linearized through PCR or restriction enzymes) (see
Note 20).
4 Notes
References
1. Watson JF, Garcı́a-Nafrı́a J (2019) In vivo mutagenesis by placing homologous ends on
DNA assembly using common laboratory bac- DNA using polymerase chain reaction. Bio-
teria: a re-emerging tool to simplify molecular techniques 10:62–66
cloning. J Biol Chem 294:jbc. 8. Nozaki S, Niki H (2018) Exonuclease III
REV119.009109 (XthA) enforces in vivo DNA cloning of Escher-
2. Garcı́a-Nafrı́a J, Watson JF, Greger IH (2016) ichia coli to create cohesive ends. J Bacteriol
IVA cloning: a single-tube universal cloning 201:20181210
system exploiting bacterial In Vivo Assembly. 9. Conley EC, Saunders VA, Saunders JR (1986)
Sci Rep 6:27459 Deletion and rearrangement of plasmid DNA
3. Beyer HM, Gonschorek P, Samodelov SL et al during transformation of Escherichia coli with
(2015) AQUA cloning: a versatile and simple linear plasmid molecules. Nucleic Acids Res 14:
enzyme-free cloning approach. PLoS One 10: 8905–8917
1–20 10. Yang Y, Wang T, Yu Q et al (2021) The path-
4. Jacobus AP, Gross J (2015) Optimal cloning of way of recombining short homologous ends in
PCR fragments by homologous recombination Escherichia coli revealed by the genetic study.
in Escherichia coli. PLoS One 10:e0119221 Mol Microbiol:1–14
5. Kostylev M, Otwell AE, Richardson RE et al 11. Gibson DDG, Young L, Chuang RR-Y et al
(2015) Cloning should be simple: Escherichia (2009) Enzymatic assembly of DNA molecules
coli DH5a-mediated assembly of multiple up to several hundred kilobases. Nat Methods
DNA fragments with short end homologies. 6:343–345
PLoS One 10:1–15 12. Kibbe WA (2007) OligoCalc: an online oligo-
6. Huang F, Spangler JR, Huang AY (2017) In nucleotide properties calculator. Nucleic Acids
vivo cloning of up to 16 kb plasmids in E. coli is Res 35:43–46
as simple as PCR. PLoS One 12:1–21 13. Inoue H, Nojima H, Okayama H (1990) High
7. Jones DH, Howard BH (1991) A rapid efficiency transformation of Escherichia coli
method for recombination and site-specific with plasmids. Gene 96:23–28
Chapter 4
Abstract
The Gibson Assembly is a popular method for molecular cloning which has been developed specifically to
join several fragments together in a specific order, without the constraint of restriction enzyme sites. This
method is based on the assembly of overlapping fragments, generally produced by PCR, and then
combining them using three enzymes: a 5′ exonuclease, a DNA polymerase, and a DNA ligase, in an
isothermal reaction. Here, we describe this method, including the design of primers for the generation of
the overlapping fragments and the assembly; to this end, we provide an example involving joining two
fragments in a single plasmid.
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_4,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
45
46 Luisana Avilan
Fig. 1 Schema of the Gibson Assembly for two adjacent fragments. After including overlapping sequences by
PCR, the fragments are treated with a 5′ exonuclease, a thermophilic DNA polymerase, and a thermophilic
DNA ligase
2 Materials
3 Methods
3.1 Design of The design of primers for the PCR amplification needed to prepare
Overlapping Primers the fragment is a critical step in the Gibson method. The primers
should anneal well with the targeted DNA (i.e., the complementary
region between the target and the primer should have a Tm greater
than 55 °C) and meet the standard requirements for optimal PCR
amplification of a gene-specific sequence primer. In addition, it
should have an overlapping extension from the adjacent fragment
(overlapping 3′ primer extension). After amplification, these ampli-
cons will include the overlap region which should be between
15 and 40 bp and have a Tm greater than 48 °C (as summarized
in Fig. 2 and see Note 7).
1. Primers can be designed using either freely available or com-
mercial DNA sequence editors or viewers. Sequences can be
visualized in either single or double strand format (for example,
SnapGene Viewer). For the example shown in Fig. 3a, the
editor ApE was used to design the primers for the assemble of
fragments A and B into a vector. The primers have an overlap
sequence between 36 and 38 bases; a Tm of the gene-specific
sequence should be around an optimal value of 57 °C (see
Note 8).
2. For the forward primer for the fragment A (Fig. 3), start at the
5′ end of fragment A and advance in a 3′ direction until the
sequence has the expected Tm; a G or C must be the final base
at the 3′ termini. To add the overlapping extension at this
Gibson Assembly 49
Fig. 2 Schema of overlapping primers used for the Gibson Assembly method.
Each primer is an oligonucleotide composed a single of gene-specific region and
a tail that provides the overlap to the next fragment. The forward primer for the
first fragment (purple) and reverse primer for the last fragment (light blue) have
been omitted as their design will depend on the method of linearization of the
plasmid into which the fragments will be inserted. However, we recommend the
same strategy as assembly of the fragments, i.e., these primers will also require
a gene-specific region and a tail which provides the overlap to the relevant
fragment
Fig. 3 Overlapping primers for the cloning of fragment A and B on a vector. (a) Construction displayed on the
gene editor ApE showing the different fragments and the primers for the vector (dark grey), fragment A (red),
and fragment B (yellow). (b) The sequences of the primers, their Tm, and the sizes of the overlap sequences
are shown
50 Luisana Avilan
3.2 Obtaining the To prepare the fragments for the assembly, set up separate PCR
Fragments reactions containing the template DNA and the forward and
reverse primers.
1. Set up a standard PCR in a final volume of 50 μL. It is prefera-
ble to use a high-fidelity polymerase; we recommend Q5 high-
fidelity Taq from NEB. Use 5 ng of template, 0.5 μM of each
primer, and 0.2 μM of the dNTPs mix. For the buffer, follow
the instructions of the DNA polymerase manufacturer.
2. The cycling conditions are chosen according to the DNA poly-
merase manufacturer, In general, the annealing temperature is
chosen according to the Tm of the gene-specific sequence of
the primer. In the example shown in Fig. 2, the chosen anneal-
ing temperature for all the fragments was 57 °C; therefore, the
annealing temperature in the PCR profile should also be 57 °C.
However, this is only a guide and it may be necessary to
optimize the PCR by a trying several annealing temperatures
centered on 57. The extension time is chosen dependent on the
size of the fragment (see Note 10). In our example, we used:
1 min at 98 °C (initial denaturation), followed by 30 cycles of
98 °C for 45 s (cycle denaturation), 57 °C for 45 s (annealing),
1min at 72 °C (cycle extension), and finally a single tempera-
ture step of 10 min at 72 °C (final extension). The extension
time for the vector amplification was increased to 4 min due to
the length (see Notes 8 and 10).
3. If a vector is a template, add 1 μL of DpnI after the reaction and
incubate at 37 °C for 30 min.
Gibson Assembly 51
3.3 Assembly 1. Thaw an aliquot (15 μL) of 1.33X Master Mix and maintain
in ice.
2. Mix 5 μL of the DNA solution containing 0.05 pmol of each
fragment with the Master mix 1.33X in the final reaction. For
DNA fragments less than 1 kb, increase the amount of DNA to
0.25 pmol (see Note 12).
3. Incubate at 50 °C for 60 min. Vortex briefly the tube and spin
down the reaction.
4. Transform the competent cells with 2 μL of the reaction.
5. Analise transformants by picking individual colonies.
6. Screen by colony PCR or isolate the plasmid to analyze by
restriction enzymes or sequencing.
4 Notes
References
1. Jackson DA, Symons RH, Berg P (1972) Bio- 5. Hutchison CA 3rd, Chuang RY, Noskov VN,
chemical method for inserting new genetic Assad-Garcia N, Deerinck TJ, Ellisman MH,
information into DNA of Simian Virus 40: cir- Gill J, Kannan K, Karas BJ, Ma L, Pelletier JF,
cular SV40 DNA molecules containing lambda Qi ZQ, Richter RA, Strychalski EA, Sun L,
phage genes and the galactose operon of Escher- Suzuki Y, Tsvetanova B, Wise KS, Smith HO,
ichia coli. Proc Natl Acad Sci USA 69:2904– Glass JI, Merryman C, Gibson DG, Venter JC
2909 (2016) Design and synthesis of a minimal bacte-
2. Cohen SN (1973) Construction of biologically rial genome. Science 351:aad6253
functional bacterial plasmids in vitro. Proc Natl 6. Sayers JR, Eckstein F (1990) Properties of over-
Acad Sci USA 70:3240–3244 expressed phage T5 D15 exonuclease. Similari-
3. Gibson DG, Young L, Chuang RY, Venter JC, ties with Escherichia coli DNA polymerase I
Hutchison CA 3rd, Smith HO (2009) Enzy- 5′-3′ exonuclease. J Biol Chem 265:18311–
matic assembly of DNA molecules up to several 18317
hundred kilobases. Nat Methods 6:343–345 7. Ishino S, Ishino Y (2014) DNA polymerases as
4. Gibson DG, Glass JI, Lartigue C, Noskov VN, useful reagents for biotechnology–the history of
Chuang RY, Algire MA, Benders GA, Montague developmental research in the field. Front
MG, Ma L, Moodie MM, Merryman C, Microbiol 5:1–8. https://doi.org/10.3389/
Vashee S, Krishnakumar R, Assad-Garcia N, fmicb.2014.00465
Andrews-Pfannkoch C, Denisova EA, Young L, 8. Takahashi M, Yamaguchi E, Uchida T (1984)
Qi ZQ, Segall-Shapiro TH, Calvey CH, Parmar Thermophilic DNA ligase. Purification and
PP, Hutchison CA 3rd, Smith HO, Venter JC properties of the enzyme from Thermus ther-
(2010) Creation of a bacterial cell controlled by mophilus HB8. J Biol Chem 259:10041–10047
a chemically synthesized genome. Science 329:
52–56
Chapter 5
Abstract
DNA ends can become damaged for various reasons making them unsuitable for TA cloning techniques,
the easiest and most common of the DNA cloning technologies. Examples of end-damaged DNA include
ancient DNA and those produced by laboratory methods such as sonication. In this chapter, we discuss how
to deal with end-damaged DNA prior to cloning with either the popular pGEM®-T Easy Vector Systems
Kit and TOPO™ TA Cloning™ Kits.
Key words End repair, TA cloning, Damaged DNA ends, Ligation, Blunt ends
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_5,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
55
56 Charlotte Ayling
A
A
3’ adenine
overhang
T T
5’ thymine
overhang
1.2 Cloning of Non- Sequences other than those produced by PCR can also be cloned
PCR-Generated via the TA approach, as Taq is able to add a 3′ adenine to extant
Sequences and End sequences (for instance, fragmented genomic DNA) when incu-
Damage bated in the presence of dATP. As such, pools of heterogenous
unknown sequences can be easily cloned compared to other tech-
niques which often require an exact knowledge of a homogenous
insert sequence. However, as a caveat to the general flexibility of the
approach, the target sequence must contain blunt ends prior to the
TA and Damage 57
Table 1
Similarities and differences between the pGEM®-T Easy Vector Systems Kit and TOPO™ TA
Cloning™ Kit
TOPO™ TA Cloning™
Specifications pGEM®-T Easy Vector Systems Kit Kit
Antibiotic resistance Ampicillin (AmpR) Ampicillin (AmpR),
Kanamycin (KanR)
Recommended Top10, DH5α, JM109 Top10, DH5α
bacterial strain
Directionality No No
Replication of origin F1 Origin F1 Origin, pUC origin
Ligation time 1 h at room temperature, may be extended to 5 min at room
increase the number of colonies temperature
Blue white screening Yes Yes
1 2 3
kb
3.0
1.0
0.5
0.1
Fig. 2 Sonicated chromatin from stage 15/16 Xenopus laevis embryos on a 1% agarose gel. Lane 1, 1 kb plus
DNA ladder. Sizes are indicated. Lanes 2 and 3 show sonicated chromatin samples. The DNA is fragmented
and of various sizes, generating a smear down the gel; these fragments can be cloned but require an end
repair and the addition of A’s before TA cloning
For instance, ancient DNA (DNA greater than 50 years old) tends
to be degraded to fragments between 40 and 500 bp; this fragmen-
tation is due to hydrolytic depurination and subsequent single-
strand breaks is particularly prevalent at the ends of the DNA
[1]. Such material must first be end repaired to form blunt ends
prior to the addition of adenine and ligation into the vector.
2 Materials
2.1 End Repair 1. Thermo Scientific™ Fast DNA End Repair Kit.
2. 1.5 mL microcentrifuge tubes.
3. 0.5–10 μL pipette and sterile pipette tips.
4. Ice box and ice.
5. Heat block set to 20 °C or thermocycler.
6. Damaged DNA sample (0.5–5 μg).
6. Power pack.
7. Gel electrophoresis tank with lid comb, UV tray, and dams.
8. Digested sample.
9. Undigested sample.
10. 100 bp plus DNA ladder/1 kb plus DNA ladder.
11. Microwave.
3 Methods
3.1 End Repair The Thermo Scientific™ Fast DNA End Repair Kit is used per the
manufacturer’s instructions as follows:
1. Add the following components (keep on ice) to a sterile micro-
centrifuge tube to make up a total of 50 μL:
• 0.5–5 μg DNA fragments
• 5 μL 10X End Repair Reaction Mix
• 2.5 μL End Repair Enzyme Mix
• Up to 50 μL Nuclease-Free Water
2. Incubate the sample(s) in a heat block or thermocycler for
5 min (no longer than 20 min) at 20 °C (room temperature).
3. This can be stored at -20 °C indefinitely.
3.2 Adding the 1. Add the following to 7 μL of the end repaired sample:
Adenines • 0.7 μL 10 mM dATP
• 1 μL Dream Taq polymerase
• 1 μL Dream Taq buffer
2. Mix well and incubate at 72 °C for 8–10 min in a hot block or
thermocycler.
3. Place on ice.
3.3 Purification We generally use QIAquick PCR purification columns. All centri-
fuge steps are at ~16,000× g in a conventional benchtop microcen-
trifuge. The recommended size range for purification by these
columns is 100 bp to 10 kb (see Note 3).
1. Add 5 volumes of buffer PB to 1 volume of the PCR sample
and mix.
2. Place a QIAquick spin column in a provided 2 mL
collection tube.
3. To bind the DNA, apply the sample to the QIA quick column
and centrifuge for 60 s.
TA and Damage 61
3.4 Ligation The pGEM®-T Easy Vector Systems Kit (keep on ice) is used using
the following directions as specified by the manufacturer:
3.4.1 Using the pGEM®-T
Easy Vector Systems Kit 1. Briefly centrifuge the pGEM®-T or pGEM®-T Easy Vector and
Control Insert DNA tubes to collect contents at the bottom of
the tube.
2. In general, a 3:1 Molar ratio of insert to vector is recom-
mended; therefore, the mass of DNA to add varies with the
average insert length. To calculate the amount of PCR prod-
uct/DNA sample to add into the reaction, we use the Biomath
calculator available from the supplier (www.promega.com/
biomath/).
3. Set up the ligation reactions in 1.5 ml LoBind microfuge tubes
as shown in Table 2. Vortex the 2X Rapid Ligation Buffer
vigorously before each use.
4. Mix the reactions by pipetting. Incubate the reactions 1 h at
room temperature. Alternatively, incubate the reactions over-
night at 4 °C for the maximum number of transformants (see
Note 5).
5. Proceed to the transformation; the reaction can be stored at
-20 °C overnight.
Table 2
Reagent volumes for use with the pGEM®-T or pGEM®-T Easy kits
Table 3
Reagent volumes for use with the TOPO® kit
3.4.2 Using the TOPO™ Alternatively, the TOPO™ TA Cloning™ Kit can be used; follow
TA Cloning™ Kit the directions as specified by the manufacturer:
1. Set up the ligation by adding the reagents shown in Table 3.
2. Mix the reaction gently by pipetting up and down, and then
incubate the reaction at room temperature (22–23 °C) for
5 min.
3. Proceed to the transformation step; the reaction can be stored
at -20 °C overnight.
3.5 Transformation 1. Prepare LB agar plates with the suitable antibiotic for the
vector used (see Notes 6 and 7).
2. Thaw the DH5α cells on ice.
3. Add 1–100 ng of the DNA sample to the microcentrifuge tube
containing the cells and gently stir with the pipette tip.
4. Incubate for 30 min on ice.
5. Heat shock the cells by placing the tubes in a heat block set at
42 °C for 45 s.
6. Place the samples on ice for 2 min.
7. Add 250 μL of fresh LB broth/SOC medium to the micro-
centrifuge containing the cells and gently pipette up and down.
Make sure to use aseptic technique.
8. Incubate the cells in a shaking incubator set to 37 °C, 220 rpm
for 30–60 min.
9. While incubating the DH5α cells, place the LB plates (with the
suitable antibiotic) in the incubator set at 37 °C in order to dry
the plates prior to spreading.
10. Spin the cells in the centrifuge at 1000× g for 10 min to form a
visible pellet.
11. Discard the supernatant and resuspend in 100 μL of fresh LB
broth.
TA and Damage 63
12. Pipette 100 μL of the cells onto a LB agar plate (with the
suitable antibiotic) from the incubator and spread using aseptic
technique.
13. Incubate the plates overnight at 37 °C.
4 Notes
References
Abstract
The ability to enzymatically assemble DNA oligonucleotides into longer DNA duplexes in a process known
as gene synthesis has wide-ranging applications in the fields of genetic engineering and synthetic biology.
Thermodynamically balanced inside-out (TBIO) gene synthesis is one of several PCR-based primer exten-
sion gene synthesis protocols that have been developed. In TBIO gene synthesis, overlapping primers with
equivalent melting temperatures (Tms) are designed so that the 5′ half of the DNA is encoded by sense
primers and the 3′ half of the DNA molecule is encoded by antisense primers. Primer extension is initiated
at the center of the DNA and continues bidirectionally to progressively elongate the DNA molecule. Here
we provide the protocols necessary for performing TBIO gene synthesis to generate a DNA molecule of
interest.
Key words Bidirectional elongation, DNA, Genetic engineering, Gene synthesis, Overlapping pri-
mers, PCR, Primer extension, Synthetic biology
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_6,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
65
66 Timothy J. Ragan and Helen A. Vincent
a Primer design
Sense primers
: Matched Tm
Antisense primers
b PCR-based assembly
i Reaction mix
Primer Concentration
Step 2
Step 3
Step 4
Step 5
Step 6
2 Materials
3 Methods
The protocols provided below assume that the user is familiar with
standard molecular biology techniques, including measuring DNA
concentration, standard PCR primer design, PCR, PCR clean-up,
agarose gel electrophoresis, and gel-extraction of DNA.
TBIO Gene Synthesis 69
3.1 Primer Design The following protocol outlines how to manually design TBIO
and Synthesis gene synthesis primers (see Note 6).
1. Take the sense-strand sequence of the DNA to be generated by
TBIO gene synthesis (see Note 1) and identify a region of
approximately 20 nt (see Note 7) at the center of the sequence
(see Note 8) which is predicted to have a Tm of 64 °C ± 2 °C
when double-stranded (see Note 3) (Fig. 2a). Designate this
region O1–2 (see Note 9).
2. Identify the 60 nt (see Notes 10 and 11) sequence immediately
upstream of the 3′ end of O1–2 (Fig. 2b). Designate this
region 1 (see Note 12).
3. Identify the 60 nt (see Notes 10 and 11) sequence immediately
downstream of the 5′ end of O1–2 (Fig. 2c). Designate this
region 2 (see Note 13).
4. Identify a region of approximately 20 nt (see Note 7) extending
downstream from the 5′ end of region 1 which is predicted to
have a Tm of 64 °C ± 2 °C when double-stranded (see Note 3)
(Fig. 2d). Designate this region O3–1 (see Note 14).
5. Identify the 60 nt (see Notes 10 and 11) sequence immediately
upstream of the 3′ end of O3–1 (Fig. 2e). Designate this region
3 (see Note 15).
6. Identify a region of approximately 20 nt (see Note 7) extending
upstream from the 3′ end of region 2 which is predicted to have
a Tm of 64 °C ± 2 °C when double-stranded (see Note 3)
(Fig. 2f). Designate this region O2–4 (see Note 16).
7. Identify the 60 nt (see Notes 10 and 11) sequence immediately
downstream of the 5′ end of O2–4 (Fig. 2g). Designate this
region 4 (see Note 17).
8. Extend outwards incrementally, essentially repeating steps 4–7,
until the entire DNA sequence has been covered (see Note 18)
(Fig. 2h).
9. Synthesize (see Note 19) the odd-numbered sequences (see
Note 20) as sense primers (Fig. 2i).
10. Synthesize (see Note 19) the reverse complement of the even-
numbered sequences (see Note 21) as antisense primers
(Fig. 2i).
1
b
O1-2
60 nt
2
c
O1-2
60 nt
1
d
O3-1
Tm = 64º C
3
e 1
O3-1
2
f
O2-4
Tm = 64º C
4
g 2
O2-4
11 7 3 2 6 10
h 9 5 1 4 8 12
S6
i S5 S4 S3 S2 S1
AS1 AS2
AS3 AS4
AS5 AS6
Table 1
Optimal final primer concentrations
Number of primer pairs 2 3 4 5 6
Innermost primers 40 40 40 40 40
60 60 60 60
80 80
Final primer concentration (nM) Internal primers
100
Final total primer concentration (nM) 480 600 840 1000 1200
Table 2
Primer stock dilutions for single-round TBIO gene synthesis and round one of multi-round TBIO gene
synthesis
Number of primer pairs 2 3 4 5 6
Innermost primers 2 2 2 2 2
3 3 3 3
4 4
Primer concentration (µM) Internal primers
5
6 6 6
Outermost primers 10 10 10 10 10
Table 3
TBIO gene synthesis reaction mix
3.3 Multi-Round DNA molecules that require primer extension and assembly of
TBIO Gene Synthesis more than six primer pairs (see Note 22) can be generated using
multi-round TBIO gene synthesis (Fig. 3). The first round follows
the protocol for single-round TBIO gene synthesis (see Subheading
3.2), using up to six of the innermost primer pairs (see Note 22) to
generate an inside fragment (Fig. 3bi and ii). This inside fragment is
gel-purified (Fig. 3biii) and substituted for the innermost primer
pair in a second round of TBIO gene synthesis utilizing up to five of
the adjacent outer primer pairs (see Note 22) (Fig. 3c). The product
from the second round of TBIO gene synthesis can similarly be
gel-purified and substituted for the innermost primer in a third
round. This process can be repeated until the full-length DNA
molecule has been synthesized.
1. Perform an initial round of TBIO gene synthesis using the
innermost six primer pairs by following the protocol for
single-round TBIO gene synthesis (see Subheading 3.2)
(Fig. 3bi).
2. Gel-purify the DNA inside fragment (Fig. 3bii) (see Notes 29
and 30).
3. Prepare a 2μM stock dilution of the DNA inside fragment (see
Note 31).
4. Prepare stock dilutions of each primer in ddH2O according to
Table 4 (see Note 23).
TBIO Gene Synthesis 73
a Primer design
Primer concentration
Table 4
Primer stock dilutions for round two and subsequent rounds of multi-
round TBIO gene synthesis
Number of primer pairs 2 3 4 5 6
Inside fragment 2 2 2 2 2
3 3 3 3
4 4
Primer concentration (µM) Internal primers
5
6 6 6
Outermost primers 10 10 10 10 10
3.4 Recommended The following steps are optional, but are recommended to both
Steps maximize the yield of the full-length DNA molecule, and to verify
its sequence.
1. Amplify the full-length DNA molecule in a standard PCR
reaction using the reaction mixture following the final round
of TBIO gene synthesis as the template, and the outermost
primer pair as the primers.
2. Purify the full-length DNA molecule using a PCR clean-up kit
(e.g., QIAquick PCR Purification Kit, Qiagen).
3. Sequence the DNA molecule (see Notes 35–37).
TBIO Gene Synthesis 75
4 Notes
tool for the experienced user [8, 9]. Therefore, we advise users
to familiarize themselves with the TBIO gene synthesis primer
design process before relying on the output from an automated
design tool.
7. The length of this region should be adjusted to achieve a Tm of
64 °C ± 2 °C. 20 nt is a good starting point.
8. The exact position of this region with respect to the center of
the DNA sequence can be adjusted to aid the identification of a
region of approximately 20 nt with a Tm of 64 °C ± 2 °C, or to
avoid potentially problematic regions of the sequence such as
repeat sequences.
9. This region will be the overlap between the 3′ termini of the
innermost primer pair (S1 and AS1).
10. This will be the final length of each primer. The choice of a
length of 60 nt reflects a balance between primer purity, primer
cost, and the need for flexibility to adjust the length of overlap
regions. However, advances in solid phase DNA synthesis
mean that it is currently feasible to routinely use primers up
to 90 nt in length.
11. It can be helpful to position repeat sequences, or regions that
will be modified for structure-function studies, in the gaps that
occur between primer overlaps. This can be achieved by
increasing the final length of a primer.
12. This sequence will be synthesized as the innermost sense
primer (S1).
13. The reverse complement of this sequence will be synthesized as
the innermost antisense primer (AS1).
14. This region will be the overlap between the 3′ termini of the
antisense strand of the inside fragment generated by primer
extension of the innermost primer pair (S1 and AS1) and the
adjacent outer sense primer (S2).
15. This sequence will be synthesized as the adjacent outer sense
primer (S2).
16. This region will be the overlap between the 3′ termini of the
sense strand of the inside fragment generated by primer exten-
sion of the innermost primer pair (S1 and AS1) and the adja-
cent outer antisense primer (AS2).
17. The reverse complement of this sequence will be synthesized as
the adjacent outer antisense primer (AS2).
18. Approximately 500 nt can be covered with 12 60 nt overlap-
ping oligonucleotides or six primer pairs.
19. Custom DNA oligonucleotide synthesis services are widely
available (e.g., Eurofins, IDT, and Merck).
20. Those encoding the 5′ half of the DNA sequence.
TBIO Gene Synthesis 77
32. For example, the primer mix for two primer pairs would con-
tain 1μL of the 2μM stock dilution of the DNA inside frag-
ment, 1μL of each of two primers, and 12μL ddH2O. The
primer mix for six primer pairs would contain 1μL of the
2μM stock dilution of the DNA inside fragment, 1μL of each
of 10 primers, and 4μL ddH2O.
33. When using 60 nt oligonucleotides, each round of TBIO gene
synthesis elongates the DNA molecule by approximately
500 bp (see Note 18). To maintain a primer extension time
of 60 s/kbp, the primer extension time for the second round
should be increased from 30 s to 60 s. The primer extension
time should then be increased by 30 s for each subsequent
round of TBIO gene synthesis.
34. If subsequent rounds of TBIO gene synthesis are necessary to
generate the full-length DNA molecule.
35. Although the use of a high-fidelity proofreading DNA poly-
merase and thermocycling conditions that minimize the error
rate (see Note 26) should result in few errors, mutations could
still occur. Therefore, we advise checking the sequence of the
DNA molecule.
36. The DNA molecule may be sequenced directly using an appro-
priately designed sequence-specific primer. Alternatively, the
DNA molecule could be cloned into a suitable vector allowing
for amplification and sequencing using standard universal
primers.
37. If errors are detected, they should be corrected (e.g., using
standard site-directed mutagenesis protocols) before proceed-
ing to downstream applications.
Acknowledgements
References
1. Agarwal KL, Büchi H, Caruthers MH, Gupta N, and applications. FEMS Microbiol Rev 32:522–
Khorana HG, Kleppe K, Kumar A, Ohtsuka E, 540
Rajbhandary UL, Van de Sande JH, 4. Czar MJ, Anderson JC, Bader JS, Peccoud J
Sgaramella V, Weber H, Yamada T (1970) (2009) Gene synthesis demystified. Trends Bio-
Total synthesis of the gene for an alanine transfer technol 27:63–72
ribonucleic acid from yeast. Nature 227:27–34 5. Hughes RA, Miklos AE, Ellington AD (2011)
2. Carr PA, Church GM (2009) Genome Engi- Gene synthesis: methods and applications.
neering. Nat Biotechnol 27:1151–1162 Methods Enzymol 498:277–309
3. Xiong AS, Peng RH, Zhuang J, Gao F, Li Y, 6. Hughes RA, Ellington AD (2017) Synthetic
Cheng ZM, Yao QH (2008) Chemical gene DNA synthesis and assembly: putting the
synthesis: strategies, softwares, error corrections,
TBIO Gene Synthesis 79
synthetic in synthetic biology. Cold Spring Harb 8. Hoover DM, Lubkowski J (2002) DNAWorks:
Perspect Biol 9:a023812 an automated method for designing oligonu-
7. Gao X, Yo P, Keith A, Ragan TJ, Harris TK cleotides for PCR-based gene synthesis. Nucleic
(2003) Thermodynamically balanced inside-out Acids Res 30:43e
(TBIO) PCR-based gene synthesis: a novel 9. Hoover D (2012) Using DNAWorks in design-
method of primer design for high-fidelity assem- ing oligonucleotides for PCR-based gene syn-
bly of longer gene sequences. Nucleic Acids Res thesis. Methods Mol Biol 852:215–223
31:143e
Chapter 7
Abstract
Random mutagenesis of DNA sequences has the advantage of generating DNA sequences with novel
properties, either directly in the case of aptamers or through subsequent transcription/translation of the
mutated sequence in the case of proteins. In both cases no prior structural or mechanistic knowledge of the
molecule is required. For sequences greater than 100 bp, one of the easiest methods to introduce the
mutations is to use Error-prone PCR (EP-PCR) as discussed in this chapter. When coupled with an
appropriate selection or high throughput screening methodology, PCR-based random mutagenesis can
provide a powerful tool for modern molecular biologists.
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_7,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
81
82 F. A. Myers
2 Materials
2.2 Planning The first step once the sequence of DNA to be mutagenized is
chosen is to decide on the level of changes best suited to that
specific project. A mutation rate that is too low may result in
missing important variants of interest. Conversely, if the rate is
too high, this could generate a library which contains molecules
carrying many multiple mutations confusing downstream interpre-
tation. For instance, if an enzyme with a modified function is
required, a high mutation rate will result in many mutant proteins
but few that retain any function. If a very high mutation rate is
required (for example, in order to alter catalytic activity or generate
a novel binding site), a downstream screening method capable of
selection from a large number of variants would be required. In
general, we recommend a final mutation rate of 0.5–1% per base per
PCR.
2.3 Pilot EP-PCR Prior to running the ER-PCR, it is important to run a pilot reaction
Experiment to determine the amplification efficiency. The main key point is to
monitor the yield per amplification cycle; if too low (< 1.7 fold
increase in DNA product), this can lead to DNA fragments which
contain only one or both of the primer sites which can lead to
shorter amplicon fragments, and these will be selectively amplified,
skewing the end pool of products. The other key point is to
calculate the number of EP-PCR cycles necessary to achieve the
required mutation rate. The average number of mutations per
DNA template starting length is shown below in Table 1 as a
function of EP-PCR doublings versus template length. To deter-
mine this, the starting template should be diluted and then ampli-
fied by the basic method outlined below. After every few cycles, a
sample should be taken for analysis on agarose gel. This should
show a yield of >1.7 fold amplification per cycle, should this not be
the case (see Note 3).
Table 1
Average number of mutations per DNA template as function of length and number of doublings
Template length
EP-PCR doublings Mutations per nucleotide position 100 bp 200 bp 400 bp 800 bp 1600 bp
5 0.0033 0.33 0.66 1.3 2.6 5.3
10 0.0066 0.66 1.3 2.6 5.3 11
20 0.013 1.3 2.6 5.3 11 21
30 0.020 2.0 4.0 7.9 16 32
50 0.0033 3.3 6.6 13 26 53
84 F. A. Myers
3 Method
3.1 Mutagenesis of This protocol is based on the Caldwell & Joyce Method and uses a
Homogenous Starting 500 bp homogenous DNA template region which is run through
Sequences 15 PCR cycles.
1. Make up the following PCR reaction mixture in 100 μl PCR
tube on ice:
– 10 μl 100 mM Tris-HCl, pH 8.3 (10 mM final)
– 2.5 μl 2 M KCl (50 mM final)
– 3.5 μl 200 mM MgCl2 (7 mM final)
– 4 μl 25 mM dCTP, pH 7 (1 mM final)
– 4 μl 25 mM dTTP, pH 7 (1 mM final)
– 4 μl 5 mM dATP, pH 7 (0.2 mM final)
– 4 μl 5 mM dGTP, pH 7 (0.2 mM final)
– 2 μl 100 mM 5′ primer (2 mM final)
– 2 μl 100 mM 3′ primer (2 mM final)
– X μl DNA template (3 nM of target site, final) (see Notes 3
and 4).
– 2 μl 25 mM MnCl2 (0.5 mM final) (see Note 5)
– 1 μl 5 U/μl Taq DNA polymerase (0.05 U/μl final).
Make up to 100 μL with DNAse free water (Molecular
Biology Grade).
2. Place the tube into the thermocycler and perform ~15 PCR
cycles (see below). The cycling conditions will vary depending
on the template and the primers, but generic conditions are:
94 °C for 1 min (denaturation), 60 °C for 1 min (annealing),
and 72 °C for 3 min (extension) (see Note 6).
3. The final PCR product should be run on an ethidium bromide
agarose gel with a suitable size marker to check that the correct
sized amplicon has been achieved.
4. Clone and sequence a sample of the PCR DNA to determine
frequency of mutations in the amplicon products using the
TOPO T/A cloning kit and a commercial sequencing company
(see Notes 7, 8, and 9).
4 Notes
References
1. Leung DW, Chen E, Goeddel DV (1989) A 3. Cadwell RC, Joyce GF (1992) Randomization
method for random mutagenesis of a defined of genes by PCR mutagenesis. PCR Methods
DNA segment using a modified polymerase Appl 2:28–33
chain reaction. Technique 1:11–15 4. McCullum EO, Williams BAR, Zhang J, Chaput
2. Wilson DS, Keefe AD (2000) Random muta- JC (2010) Random mutagenesis by error-prone
genesis by PCR. Curr Protoc Mol Biol PCR. Methods Mol Biol 634:103–109
2000(8):3
Chapter 8
Abstract
Site-Directed Mutagenesis (SDM) allows for changes in the DNA sequence of plasmids using polymerase
chain reaction (PCR). It is a reliable, accessible, and rapid method which is the common initial step of many
biochemial or genetic experiments. Here we describe the various different forms of SDM before giving a
detailed method for the introduction of substitutions, insertions, or deletions using a fast, ligation-free
protocol, followed by colony PCR to screen for mutated sequences.
Key words Site-directed mutagenesis, Polymerase chain reaction, Mutation, Genetic alteration,
Plasmid manipulation, Colony PCR
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_8,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
87
88 Michael J. McClellan
2 Materials
2.1 SDM PCR 1. Q5 DNA polymerase and buffer (New England Biolabs) (see
Note 1).
2. SDM primers (see Subheading 3.2).
3. Plasmid purified from dam+ bacteria and eluted in water or
elution buffer.
4. 10 mM dNTP mix.
2.2 Digestion and 1. DpnI and rCutsmart buffer (New England Biolabs).
Transformation 2. XL-10 GOLD ultracompetant cells (Agilent) (see Note 1).
3. 14-mL snap cap Falcon polypropylene round-bottom tubes.
4. S.O.C. medium (ThermoFisher).
5. Water bath at 42 °C.
6. Agar plates with Antibiotic appropriate for plasmid.
2.3 Colony PCR and 1. Q5 DNA polymerase and buffer (New England Biolabs) (see
Sequencing Note 1).
2. Primers designed to amplify mutated region.
3. 10 mM dNTP mix.
4. LB broth with appropriate antibiotic.
SDM 89
3 Methods
3.1 Selection of 1. Select bases to mutate; as many as three can be easily changed in
Mutations and Design one reaction, above which efficiency may start to drop off.
of Primers 2. Bases to mutate should all be within 30 bp of each other. Short
deletions or insertions may also be introduced; however, any
more than three bases begins to reduce the yield of mutated
plasmid, as does deleted/inserted bases being
non-consecutive. If multiple changes of this type are required,
it may be best to do sequential rounds of mutagenesis.
3. Design primers, which are the direct complements of each
other, targeting the region of plasmid you wish to mutate.
4. Primers do not require 5′ phosphorylation, and standard
desalting purification is sufficient.
5. The melting temperature of primers should be high, >~75°, to
allow for mismatches and should possess G or C “clamps” at
either end, although this is not essential (see Note 2).
6. The primers should be complimentary to the plasmid sequence
except at the bases which are to be mutated (Fig. 1). No
mutated base should be less than 10 bp away from one end of
the primer.
7. The minimum size of the primers should be 30 bp to allow
sufficient distance between staggered nicks on the plasmid
(Fig. 2) to facilitate annealing. Both primers should contain
all mutated bases.
8. Larger primers can be used when targeting multiple sites,
although this increases the risk of secondary structure
formation.
Fig. 1 Schematic of primer design for the three basic forms of SDM, substitution, insertion, and deletion. In
each case, the primers are the exact complement of each other, and both primers contain the desired change
in sequence. Primers are also long, with a high Tm, and the mutations are central
Fig. 2 Introduction of mutation by PCR and the depletion of unmutated plasmid by DpnI digestion. Plasmid,
isolated from dam+ bacteria, will carry methylated adenine on both strands at all GATC sequences (blue DNA),
whereas newly synthesized DNA, extended from primers carrying the mutated sequence will not possess any
methylated bases (black DNA). Multiple rounds of PCR will enrich DNA carrying the mutation relative to the
methylated parental DNA. Four possible products exist after the final round of extension, all, excepting plasmid
with a staggered nick and carrying the desired mutation on both strands, will be digested by DpnI, which
selectively targets methylated and hemi-methylated GATC sequences
SDM 91
Table 1
Thermocycling conditions for SDM PCR
3.4 Colony PCR and 1. Select primers to amplify (and later sequence) the region con-
Sequencing taining the introduced mutation. These should not be more
than 500 bp apart, and neither primer should be within 20 bp
of the mutated bases to ensure high sequencing quality at the
critical point in the read (see Note 10).
92 Michael J. McClellan
Table 2
Thermocycling conditions for colony PCR
4 Notes
References
1. Gillam S, Smith M (1979) Site-specific muta- directed mutagenesis of large amplified mole-
genesis using synthetic oligodeoxyribonucleo- cules. Nucleic Acids Res 21(9):2277–2278
tide primers: II. In vitro selection of mutant 7. Liu H, Ye R, Wang YY (2015) Highly efficient
DNA. Gene 8(1):99–106 one-step PCR-based mutagenesis technique
2. Kunkel TA, Roberts JD, Zakour RA (1987) for large plasmids using high-fidelity DNA
Rapid and efficient site-specific mutagenesis polymerase. Genet Mol Res 14(2):3466–3473
without phenotypic selection. Methods Enzy- 8. Luna S, Mingo J, Aurtenetxe O, Blanco L,
mol 154:367–382 Amo L, Schepens J, Hendriks WJ, Pulido R
3. Jinek M, Chylinski K, Fonfara I, Hauer M, (2016) Tailor-made protein tyrosine phospha-
Doudna JA, Charpentier E (2012) A program- tases: in vitro site-directed mutagenesis of
mable dual-RNA-guided DNA endonuclease PTEN and PTPRZ-B. Methods Mol Biol
in adaptive bacterial immunity. Science 337: 1447:79–93. https://doi.org/10.1007/978-
816–821 1-4939-3746-2_5. PMID: 27514801
4. Cho SW, Kim S, Kim JM, Kim JS (2013) Tar- 9. Adereth Y, Champion KJ, Hsu T, Dammai V
geted genome engineering in human cells with (2005) Site-directed mutagenesis using Pfu
the Cas9 RNA-guided endonuclease. Nat Bio- DNA polymerase and T4 DNA ligase. Biotech-
technol 31:230–232 niques 38(6):864, 866, 868
5. Doudna JA, Charpentier E (2014) Genome 10. Wu D, Guo X, Lu J, Sun X, Li F, Chen Y, Xiao
editing. The new frontier of genome engineer- D (2013) A rapid and efficient one-step site-
ing with CRISPR-Cas9. Science 346(6213): directed deletion, insertion, and substitution
1258096 mutagenesis protocol. Anal Biochem 434(2):
6. Marini F 3rd, Naeem A, Lapeyre JN (1993) An 254–258
efficient 1-tube PCR method for internal site-
SDM 95
11. Liu H, Naismith JH (2008) An efficient 14. Emruzi Z, Aminzadeh S, Karkhane AA,
one-step site-directed deletion, insertion, sin- Alikhajeh J, Haghbeen K, Gholami D (2018)
gle and multiple-site plasmid mutagenesis pro- Improving the thermostability of Serratia mar-
tocol. BMC Biotechnol 4(8):91 cescens B4A chitinase via G191V site-directed
12. Monchietti P, López Rivero AS, Ceccarelli EA, mutagenesis. Int J Biol Macromol 116:64–70
Catalano-Dupuy DL (2021) A new catalytic 15. Liang Q, Chen L, Fulco AJ (1995) An efficient
mechanism of bacterial ferredoxin-NADP+re- and optimized PCR method with high fidelity
ductases due to a particular NADP+binding for site-directed mutagenesis. PCR Methods
mode. Protein Sci 30(10):2106–2120 Appl 4(5):269–274
13. Harrison JJEK, Tuske S, Das K, Ruiz FX, Bau- 16. Nishizawa-Yokoi A, Yamaguchi N (2018)
man JD, Boyer PL, DeStefano JJ, Hughes SH, Gene expression and transcription factor bind-
Arnold E (2021) Crystal structure of a retrovi- ing tests using mutated-promoter reporter
ral polyprotein: prototype foamy virus lines. Methods Mol Biol 1830:291–305
protease-reverse transcriptase (PR-RT).
Viruses 13(8):1495
Chapter 9
Abstract
Transgenic approaches using I-SceI are powerful genome modification methods for creating heritable
modifications in eukaryotic genomes. Such modifications are ideal for studying putative promoters and
their temporal and spatial expression patterns in real time, in vivo. Central to this process is the initial
engineering of a plasmid construct containing multiple DNA modules in a specific order prior to the
integration into the target genome. One popular way of doing this is based upon the pGateway system, the
modular form of which described in this chapter is known as pTransgenesis. We will initially describe the
protocol of obtaining the plasmid construct containing the required sequence modules, and then the
process of integrating the construct into the genome of a Xenopus embryo via co-injection with I-SceI and
subsequent screening for transgenics.
1 Introduction
1.1 Transgenesis as The term transgenic was first introduced in 1981 by John Gordon
a Tool for Reporter and Frank Ruddle when they successfully inserted exogenous
Gene Assays genetic material into the genome of fertilized mouse eggs
[1]. Since then, transgenesis has evolved into a widely used method,
across a number of species for both commercial and academic
purposes. Examples include the “spider goat” for spider silk pro-
duction in goat milk and golden rice, which is fortified for the
precursor to vitamin A [2, 3]. The first reported transgenic frog
was made in 1984 by Laurence Etkin when he introduced, via
microinjection, DNA constructs into one cell Xenopus laevis
embryos [4]. In these early experiments, various constructs were
injected as either linear or circular pieces of DNA into model
organisms, such as Xenopus, Drosophila, and Sea Urchins. Circular
plasmids were found to be integrated into the embryo genome less
often than the linearized genetic material, but the main problem
with the method was the highly mosaic expression pattern, where
the plasmid was integrated into the genomes of only patches of
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_9,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
97
98 Liliya Nazlamova
1.2 The I-SceI In order to produce a transgenic frog using the I-SceI technique,
Technique and the one-cell stage embryos are injected with a plasmid clone (Fig. 1),
pGateway System containing the insert of interest and a marker gene for indicating a
successful transgenesis event. I-SceI is an endonuclease isolated
from the yeast Saccharomyces cerevisiae that recognizes an 18-bp
non-palindromic sequence that does not occur within the genomes
of animal species but will linearize the plasmid within the cell,
preventing recircularization by endogenous ligases. In our hands
this method can give approximately 30% efficiency in generating F0
transgenic Xenopus.
Key to the I-SceI transgenesis protocol is the assembly of the
final injectable plasmid. In this chapter, we use a recombinase
approach based upon the commercially available pGateway system
to construct the initial plasmid and subsequently use this plasmid to
generate a transgenic frog for characterization of a previously
unknown promoter. The recombinase approach involves four vec-
tors, each of which carries one of the modules (such as a putative
promoter, GFP reporter) that will be combined to build the final
cassette (Fig. 2). These vectors are based upon the pGateway
plasmids which form the pGateway system [8, 9]. This is a modular
system and is transferable between different species such as Dro-
sophila, Zebrafish, Xenopus, and mammalian cells. An interesting
aspect of the pGateway plasmids is the inclusion of the ccdB gene
which encodes a gyrase inhibitor that is toxic to most E. coli strains.
However, after successful recombination, this gene is removed
from the plasmids as a by-product. Therefore, after bacterial trans-
formation, only the bacterial clones containing the successful
recombinants will survive, drastically reducing the background.
Although the pGateway system provides non-recombinant
plasmids, versions of the plasmids containing the required gene
modules can be obtained easily from a number of sources. The
ones in our example are available from European Xenopus Resource
Center (EXRC) (https://www.port.ac.uk/research/research-pro
jects/european-xenopus-resource-centre) and obviate the need for
insertion of genes into the base plasmid; the relationship between
the base plasmids and the ones obtained from the EXRC is shown
pGateway Transgenesis 99
+ I-SceI
n= 100
Fig. 1 Outline of the I-SceI transgenic procedure. A mix of I-SceI and an expression clone is injected into
one-cell Xenopus laevis embryos. The expression vector carries internal control modules. These include
flanking I-SceI restriction sites and 5′ and 3’ insulators (Tol2). A γ-crystalline promoter driving a GFP reporter
gene (indicated in green) is used as a marker for later identification of successful transgenics. A second set of
modules within the expression clone consists of the putative promoter to be tested (dark blue) and a second
reporter gene Katushka (red). Depending on the properties of the putative promoter, red fluorescence may be
observed at developmental stages
100 Liliya Nazlamova
Expression vector
Amp r
Fig. 2 MultiSite gateway 3-fragment recombination reaction. An overview of the pGateway cloning procedure
where the first step is PCR-amplification of a putative promoter construct to be recombined with the pDONR
vector. The resulting new plasmid designated entry clone p2 (putative promoter) is recombined with p1
(Y-crystalline/GFP), p3 (Katushka), and destination vector p4 in order to produce the expression vector. The
crossed lines represent recombination events between the designated att-sites and segments B1, B2, B3, and
B4 represent the positions of the resulting recombination sites. The selected recombination att-sites in all of
the plasmids allow for specific order of the cassette elements
Table 1
Plasmid nomenclature
2 Materials
3 Methods
Table 2
PCR cycle conditions
4 Notes
References
1. Gordon JW, Ruddle FH (1981) Integration 4. Etkin L et al (1984) Replication, integration,
and stable germ line transmission of genes and expression of exogenous DNA injected
injected into mouse pronuclei. Science into fertilized eggs of Xenopus laevis. Differen-
(New York, NY) 214(4526):1244–1246 tiation 26(3):194–202
2. Tang G et al (2009) Golden rice is an effective 5. Hartley KO, Nutt SL, Amaya E (2002) Tar-
source of vitamin A. Am J Clin Nutr 89(6): geted gene expression in transgenic Xenopus
1776–1783 using the binary Gal4-UAS system. Proc Natl
3. Williams D (2003) Sows’ ears, silk purses and Acad Sci 99(3):1377–1382
goats’ milk: new production methods and 6. Kroll KL, Amaya E (1996) Transgenic Xenopus
medical applications for silk. Med Device Tech- embryos from sperm nuclear transplantations
nol 14(5):9–11 reveal FGF signalling requirements during
pGateway Transgenesis 109
Abstract
CRISPR/Cas9 has become the favorite method for gene knockouts in a range of vertebrate model
organisms due to its ease of use and versatility. Gene-specific guide RNAs can be designed to a unique
genomic sequence and used to target the Cas9 endonuclease, which causes a double-stranded break at the
desired locus. Repair of the breaks through non-homologous end joining often results in the deletion or
insertion of several nucleotides, which frequently result in nonsense mutations. Xenopus frogs have long
been an excellent model organism in which to study gene function, and they have proven to be useful in
gene-editing experiments, especially the diploid species, X. tropicalis. In this chapter, we present our
protocols for gene disruption in Xenopus, which we regularly use to investigate developmental processes
and model human genetic disease.
Key words CRISPR/Cas9, Xenopus tropicalis, Xenopus laevis, Disease modeling, Gene knockout,
MicroCT
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_10,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
111
112 Anita Abu-Daya and Annie Godwin
2 Materials
2.2 Injection Delivery 1. Fertilised Xenopus eggs. The in vitro fertilization of eggs for
of CRISPR/Cas9 microinjection can be achieved using either fresh crushed testes
Constructs or frozen sperm.
2. A controlled temperature incubator (ideal range: 14–28 °C).
3. 2% L-Cysteine solution (Recommended: non-hydrochloride
(168149), Sigma-Aldrich), pH 7.8 for dejellying eggs. Store
the solution at 4 °C and use until precipitates are visible.
4. A 1× stock of Marc’s Modified Ringers (MMR) solution
(0.1 M NaCl, 2 mM KCl, 1 mM MgSO4, 2 mM CaCl2,
5 mM HEPES, and pH 7.4) is used to prepare the 0.05×
MMR (X. tropicalis) and 0.1× MMR (X. laevis) working
stocks. OPTIONAL: Commercially available penicillin
(10,000 U/mL) and streptomycin (10 mg/mL) solution can
be added to improve the survival of early staged embryos
(Nieuwkoop and Faber (NF) stages 1–41) [27]. Store at 4 °C.
5. Plastic petri-dishes lined with 1% agarose (in 0.05× MMR,
Store at 4 °C) or glass dishes should be used to house early-
staged (NF1 – NF10) X. tropicalis embryos. It is further
recommended at these stages that glass Pasteur pipettes are
used for embryo manipulation.
6. A picoinjector (for example, Medical Systems PLI-100 Picoin-
jector (Harvard Apparatus)), air compressor (for example, PT5
Bambi Air Compressor), a stereomicroscope (for example, 2×
magnification Nikon Stereomicroscope (SMZ800, C-W 10× B
Eyepieces)), with reticule micrometer and a cold light source
(for example, KL 2500 LED Lightsource (Schott)) are
required for microinjection.
7. Borosilicate glass capillaries (Harvard Apparatus), used to make
needles for microinjection.
8. Micropipet puller (for example, Sutter instrument, Model P-87
Flaming Brown Micropipette Puller) with an appropriate pro-
gram (for example, 801 ms (Heat), 250 ms (Pull), 190 ms
(Velocity), 160 ms (Time), adjust as required) to prepare
microinjection needles from capillaries.
9. Fine watchmaker’s forceps (for example, Dumont no.5) to cut
the end of needles.
CRISPR Knockout 115
3 Methods
Ensure all work complies with local ethical and animal scientific
requirements. Conduct all steps at room temperature and use
double-distilled water in solutions unless otherwise specified.
Wear lab coat, gloves and appropriate PPE at all times to avoid
nuclease degradation of samples and injury. The following protocol
is broken down into a series of steps to guide the creation and
analysis of mutant Xenopus tadpoles:
1. Designing guide RNAs.
2. Generating guide RNA from ssDNA templates.
3. Microinjection of guide RNA into Xenopus embryos.
4. Genotyping strategies to understand indel formation in cris-
pant embryo genomic DNA samples.
5. General phenotyping strategies for gross morphological
changes in crispant tadpoles.
CRISPR Knockout 117
3.1 Target The first and most important step in CRISPR/Cas9 gene disrup-
Identification and tion studies is designing guide RNAs which will target Cas9 to the
Oligonucleotide Design sequence of interest. There are many approaches to standardize the
design and improve the efficiency of prospective gene-editing con-
structs; we successfully use the freely available CRISPRscan and
inDelphi browser applications. CRISPRscan identifies appropriate
sgRNAs in a given sequence, estimates the efficiency of cutting
in vivo, and checks that the sequence is unique to minimize the
possibility of off-target effects [28, 29]. On the other hand, the
inDelphi mESC model has been shown to reliably predict frame-
shift frequency and precision events (range of indels) resulting from
CRISPR/Cas9 genome editing [30–32].
1. The gene of interest is first identified through the gene-pages
presented on Xenbase [33] using the most up to date genome
assembly (presently, X. tropicalis: v10 and X. laevis: v9.2). The
features of each gene’s structure can be identified in JBrowse
and modeled in a DNA analysis software facilitating annotation
(for example, SnapGene®5.2.4).
2. Any chromosomal or gene duplication should be considered in
the experimental design. See Note 2 for more details including
how to approach rare situations where the gene is poorly
annotated. When considering the CRISPR/Cas9 target
sequence it is best to choose a coding region not located within
20 bp of the end of the exon due to the potential for splicing
effects. If many sgRNAs are possible, we choose those pre-
dicted to have high mutagenic activity (CRISPRscan score
>30), with no viable off-target events (test in Xenbase blast
using the latest genome assembly version to make sure the
sequence is unique), and a high frameshift frequency (inDelphi
frameshift score >75% (mESC model)) [28, 30]. See Notes 3
and 4 for how to approach small exon genes, splice variants,
highly repetitive regions of the genome, large deletions, and
for recommended CRISPR/Cas9 experimental controls.
3. Species conservation (between Xenopus sp. and for example,
H. sapiens) and key protein domains can be mapped from
protein sequences stored in the National Centre for Biotech-
nology Information (NCBI) databases using a freely available
multiple sequence alignment tool (for example, Clustal Omega
(EMBL-EBI)). This information may guide targeting an
appropriate region in the protein (5′UTR, first exon, early
exon, key protein domain, patient variant site). See Notes 3
and 4 for more details.
4. It is recommended that three sgRNAs are designed from geno-
mic DNA sequences encompassing the target region
(c. 500 bp) to test in a preliminary experiment. This approach
allows the selection of the most effective sgRNA and minimizes
the likelihood the phenotype is due to off-target changes.
118 Anita Abu-Daya and Annie Godwin
3.2 Guide RNA For sgRNA synthesis we have had good success using the method
Synthesis described in Nakayama et al. [34] This relies on annealing two
single-stranded oligonucleotides, the gene-specific nucleotide sug-
gested by CRISPRscan, which contains a 5′ T7 promoter, and a
Universal CRISPR Oligonucleotide, which contains the Cas9 bind-
ing sequence. The two oligonucleotides have an overlapping seg-
ment and can be annealed and extended by Taq polymerase to
generate a DNA template from which the sgRNA can be tran-
scribed with T7 polymerase.
1. Set up the annealing and extension reaction to convert ssDNA
to dsDNA by mixing a 50 μL Taq PCR mix, 46 μL nuclease-
free water, 2 μL Universal CRISPR oligonucleotide (100 μM),
and 2 μL target-specific oligonucleotide (100 μM) in a thin-
walled PCR tube on ice.
2. Mix gently, remove 3 μL of the mixture and keep on ice as a
negative control. Run the annealing and extension reaction in a
thermal cycler as detailed in Table 1.
3. Following the annealing and extension reaction, take a 3 μL
sample from each tube and run each sample alongside the
negative control on a 1.2% agarose gel to assess the formation
of the dsDNA template (Fig. 1). The ssDNA and dsDNA
templates visualized using the intercalating reagent Ethidium
Bromide. dsDNA templates show no increase in size following
the annealing and extension reaction, however the dsDNA
bands are much brighter and tighter than ssDNA, indicating
that the procedure was successful.
4. sgRNA can now be transcribed from the dsDNA template
using the MEGAshortscriptTM T7 Transcription Kit
(Invitrogen – ThermoFisher Scientific) or a similar kit.
CRISPR Knockout 119
Table 1
CRISPR annealing and extension reaction
Fig. 1 ssDNA and dsDNA templates visualized using the intercalating reagent
Ethidium Bromide
3.3 Injection of The third step in CRISPR/Cas9 gene disruption studies is gener-
CRISPR/Cas9 ating the crispant animals. The protocol presented is an adaptation
Constructs into of Molecular Methods in Developmental Biology—Chapter 10:
Xenopus Embryos Microinjection into Xenopus Oocytes and Embryos [12].
1. Prepare fertilized eggs for injection by removing the outer jelly
coats in 2% L-Cysteine (non-hydrochloride solutions should be
used when handling X. tropicalis embryos) solution
(pH 7.8–8.0) with gentle rocking.
CRISPR Knockout 121
3.4 Genotyping The next step in CRISPR/Cas9 gene disruption studies is genotyp-
Embryos ing the crispant animals. The success of genome editing can be
judged using the T7 endonuclease I assay, or by Sanger sequencing
of amplicons containing the target site and tracking of indels by
decomposition (TIDE or Synthego ICE).
122 Anita Abu-Daya and Annie Godwin
Table 2
PCR amplification of genomic target region
Table 3
Thermocycler conditions for T7 endonuclease I assay
Fig. 3 Genotype analysis of F0 mosaic tadpoles. The target locus is amplified from genomic DNA preparations
of injected (crispant) and uninjected (control) tadpoles. Amplicons are digested with T7 Endonuclease I, which
reveals a second band unique to crispant tadpoles that correspond to the location of the CRISPR target site (A).
Sanger sequencing is used to confirm the presence of indels in these crispant samples. Analysis of the Sanger
sequencing trace files by Synthego ICE revealed the editing efficiency of the target domain is around 50% (B)
Fig. 4 Detailed structural differences are examined in Xenopus tadpoles using high-resolution Micro Computed
Tomography. The imaged volumes show a whole tadpole with a 3D reconstruction of the cardiovascular
system and gut visualized using TXM3DViewer (Carl Zeiss Microscopy) that can be exported as cross-section
high-resolution TIFF image files
3.5 Phenotyping The final step in CRISPR/Cas9 gene deletion studies is phenotyp-
Mutant Embryos ing the crispant animals. There are many strategies to explore the
effects of knockouts, here we report MicroCT as a broad and
extremely useful technique to investigate comprehensive or unex-
pected phenotypes (Fig. 4).
1. Gross morphological assessment can be performed under stan-
dard bright-field microscopy. For best results, image embryos/
tadpoles on agarose lined dishes. See Note 14 for considera-
tions where an unexpected phenotype presents in the F0 cris-
pant model.
2. Detailed structural differences can be examined using high-
resolution Micro Computed Tomography (MicroCT) in fixed
specimens contrast stained in Phosphotungstic acid (PTA,
Sigma-Aldrich) [38]. The recommended stage of analysis is
NF42-50. Resolution below NF30 is poor and penetrance of
the contrast reagent, using this methodology, above NF50 is
highly variable.
3. Fix terminally anaesthetized (0.2% MS222) tadpoles in glass
vials containing 5 mL MEMFA for 2 h and dehydrate in a series
of Methanol washes: 25%, 50%, 75%, and 100%. Replace the
100% Methanol once more, and store embryos at -20 °C.
4. Rehydrate fixed samples in a series of methanol washes: 100%,
75%, 50%, and 25% and replace the final methanol concentra-
tion with 1% PTA for 48 h (NB it is essential that no salts are
introduced after this step).
5. Remove the solution containing 1% PTA and wash all tadpoles
in nuclease-free water.
6. Embed specimens in 0.7% agarose. Dissolve the agarose in
nuclease-free water and maintain it at 60 °C on a heat block
until use. Cool the agarose solution to 30 °C, immerse the
tadpole in agarose and quickly transfer the tadpole into the
CRISPR Knockout 125
4 Notes
References
1. Naert T, Vleminckx K (2018) CRISPR/Cas9 human disease genes in Xenopus. Dev Biol
disease models in zebrafish and Xenopus: the 408:196–204
genetic renaissance of fish and frogs. Drug Dis- 14. Hassnain Waqas SF et al (2017) Adipose tissue
cov Today Technol 28:41–52 macrophages develop from bone marrow–
2. Doudna JA, Charpentier E (2014) The new independent progenitors in Xenopus laevis
frontier of genome engineering with and mouse. J Leukoc Biol 102:845–855
CRISPR-Cas9. Science (80-) 346:1258096 15. McQueen C, Pownall ME (2017) An analysis
3. Shalem O et al (2014) Genome-scale CRISPR- of MyoD-dependent transcription using
Cas9 knockout screening in human cells. Sci- CRISPR/Cas9 gene targeting in Xenopus tro-
ence 343:84 picalis embryos. Mech Dev 146:1–9
4. Carroll D (2011) Genome engineering with 16. Blum M, Ott T (2018) Xenopus: an under-
zinc-finger nucleases. Genetics 188:773–782 valued model organism to study and model
5. Bibikova M, Golic M, Golic KG, Carroll D human genetic disease. Cells Tissues Organs
(2002) Targeted chromosomal cleavage and 205:303–313
mutagenesis in Drosophila using zinc-finger 17. Hwang WY, Marquez J, Khokha MK (2019)
nucleases. Genetics 161:1169–1175 Xenopus: driving the discovery of novel genes
6. Nakajima K, Nakajima T, Takase M, Yaoita Y in patient disease and their underlying patho-
(2012) Generation of albino Xenopus tropicalis logical mechanisms relevant for organogenesis.
using zinc-finger nucleases. Dev Growth Differ Front Physiol 10:953
54:777–784 18. Sater AK, Moody SA (2017) Using Xenopus to
7. Miller JC et al (2011) A TALE nuclease archi- understand human disease and developmental
tecture for efficient genome editing. Nat Bio- disorders. Genesis 55:e22997
technol 29:143–148 19. Grainger RM (2012) Xenopus tropicalis as a
8. Ishibashi S, Cliffe R, Amaya E (2012) Highly model organism for genetics and genomics:
efficient bi-allelic mutation rates using past, present, and future. In: Methods in
TALENs in Xenopus tropicalis. Biol Open 1: molecular biology (Clifton, NJ), vol 917.
1273–1276 Humana Press, pp 3–15
9. Jinek M et al (2012) A programmable dual- 20. Abu-Daya A, Sater AK, Wells DE, Mohun TJ,
RNA-guided DNA endonuclease in adaptive Zimmerman LB (2009) Absence of heartbeat
bacterial immunity. Science (80-) 337:816– in the Xenopus tropicalis mutation muzak is
821 caused by a nonsense mutation in cardiac myo-
10. Nakayama T et al (2013) Simple and efficient sin myh6. Dev Biol 336:20–29
CRISPR/Cas9-mediated targeted mutagenesis 21. Abu-Daya A et al (2011) The secreted integrin
in Xenopus tropicalis. Genesis 51:835–843 ligand nephronectin is necessary for forelimb
11. Sive HL, Grainger RM, Harland RM (2010) formation in Xenopus tropicalis. Dev Biol 349:
Microinjection of Xenopus embryos. Cold 204–212
Spring Harb Protoc 5:pdb.ip81 22. Goda T et al (2006) Genetic screens for muta-
12. Guille M (1999) Microinjection into Xenopus tions affecting development of Xenopus tropi-
oocytes and embryos. Methods Mol Biol (Clif- calis. PLoS Genet 2:e91
ton, NJ) 127:111–123 23. Abu-Daya A, Khokha MK, Zimmerman LB
13. Bhattacharya D, Marfo CA, Li D, Lane M, (2012) The hitchhiker’s guide to Xenopus
Khokha MK (2015) CRISPR/Cas9: an inex- genetics. Genesis 50:164–175
pensive, efficient loss of function tool to screen
130 Anita Abu-Daya and Annie Godwin
24. Macken WL et al (2021) Biallelic variants in modeling of editing outcomes in Xenopus and
COPB1 cause a novel, severe intellectual dis- zebrafish embryos. Sci Rep 10:14662
ability syndrome with cataracts and variable 32. Shen MW et al (2018) Predictable and precise
microcephaly. Genome Med 13:1–19 template-free CRISPR editing of pathogenic
25. Session AM et al (2016) Genome evolution in variants. Nature 5637733(563):646–651
the allotetraploid frog Xenopus laevis. Nature 33. Xenbase. http://www.xenbase.org/entry/
538:336–343 34. Nakayama T et al (2014) Cas9-based genome
26. Hellsten U et al (2010) The genome of the editing in xenopus tropicalis. In: Methods in
western clawed frog xenopus tropicalis. Science enzymology, vol 546. Academic, pp 355–375
(80-) 328:633–636 35. T7 Endonuclease I assay – CRISPR fly design.
27. Nieuwkoop PD, Faber J (1994) Normal table https://www.crisprflydesign.org/t7-endo-i-
of Xenopus Laevis (Daudin): a systematical & assay/.
chronological survey of the development from 36. Brinkman EK, Chen T, Amendola M, van
the fertilized egg till the end of metamorphosis. Steensel B (2014) Easy quantitative assessment
Garland Pub of genome editing by sequence trace decompo-
28. CRISPRscan: Welcome. https://www. sition. Nucleic Acids Res 42:e168–e168
crisprscan.org/ 37. Industrialized CRISPR Platforms for Acceler-
29. Moreno-Mateos MA et al (2015) CRISPRs- ated Discovery to Clinic. https://www.
can: designing highly efficient sgRNAs for synthego.com/platforms.
CRISPR-Cas9 targeting in vivo. Nat Methods 38. Metscher BD (2009) Micro CT for compara-
12:982–988 tive morphology: simple staining methods
30. Indelphi. https://indelphi.giffordlab.mit.edu/ allow high-contrast 3D imaging of diverse
31. Naert T et al (2020) Maximizing CRISPR/ non-mineralized animal tissues. BMC Physiol
Cas9 phenotype penetrance applying predictive 9:11
Chapter 11
Abstract
Xenopus has long had a reputation for being a powerful model organism for use in developmental cell and
biochemistry research. With the advent of gene-editing technologies, and the full genome sequencing of
Xenopus genomes revealing the extent of the genetic conservation between Xenopus and humans, Xenopus
has the potential to become an ideal model for human genetic disease. However, the inability to produce
non-mosaic, precise DNA insertions through homology directed repair has limited the strength of Xenopus
this field. Furthermore, it has prevented researchers from taking full advantage of fusion tagging, a method
for directly tagging genes with either epitope or fluorescent tags, allowing the visualization, quantification,
and tracking of proteins without the use of protein-specific antibodies. Here, we describe a method for
precise DNA insertion into oocytes using CRISPR/Cas9, followed by in vitro maturation and fertilization
by intracytoplasmic sperm injection (ICSI), culminating in the production of embryos carrying a
non-mosaic, heterozygous insertion.
Key words CRISPR, Xenopus, Precise insertion, Oocytes, ICSI, Sperm nuclei, Disease model, Epi-
tope tag
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_11,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
131
132 Sian Angela Martin
2 Materials
2.2.2 Long Single 1. Q5 high-fidelity polymerase and master mix (New England
Stranded DNA Synthesis Biolabs).
2. dNTP mix.
3. Molecular grade water.
4. Guide-it long ssDNA Production System v2 kit (Takara Bio).
2.6 Intracytoplasmic 1. Injection dishes made with 2% agarose in 0.4× MMR (see
Sperm Injection (ICSI) Note 1).
2. Glass micro-injection needles pre-cut to 40 μm.
3. Tygon tubing (see Note 2).
4. Ficoll 400.
5. 0.4× MMR with penicillin-streptomycin (1 mL/L).
6. Syringe pump.
3 Methods
3.1 Designing sgRNA When designing sgRNA for an insertion, first you must identify
and lssDNA Inserts where in the gene it should be inserted, for instance, when tagging
a gene with an epitope tag or fluorescence tag, the insertion should
be directly downstream of the 3′ stop codon or directly upstream of
the 5′ ATG. Whereas, if you want to make a specific mutation
within the open reading frame by insertion, the location of the
cut site is dictated by the location of the desired mutation. How-
ever, the location of the cut site (sgRNA site) can be up to 30 bp
away from the desired insertion site, so there is some leeway when
finding an efficient sgRNA site. Once a region (30 bp either side of
your desired insertion point) has been identified the region
sequence can be inputted into scoring algorithms designed to
predict cutting efficiency and find the best sgRNA sites in your
sequence. Many sgRNA tools are available on the Internet, our tool
of choice is CRISPRscan (https://www.crisprscan.org/). Once a
suitable sgRNA is found, it can be tested for cutting efficiency by
embryo injection followed by T7 endonuclease assay. The insertion
design includes the sequence to be inserted with 500 bp sequences
flanking upstream and downstream that are homologous to the
genomic sequence to be targeted for insertion.
3.2 BAC Cloning BAC libraries are used to gain access to gene sequences which will
be used to make the homology portion of your long single stranded
DNA insert. These BAC libraries are available at the European
Xenopus resource center (EXRC).
1. Streak BAC culture onto LB agar plates containing 50 μg/mL
ampicillin and grow over night in a 37 °C incubator.
136 Sian Angela Martin
3.3 Long Single Using Gibson assembly cloning, we can amplify multiple fragments
Stranded DNA of our insert DNA and fuse them together whilst also inserting the
(lssDNA) Insert complete insert DNA sequence into a pUC19 vector ready for
Synthesis cloning. When inserting a short DNA sequence such as an epitope
tag, this can be achieved by designing two sets of primers to make
3.3.1 Gibson Assembly two fragments. The first fragment will include a 5′ 20 bp overlap
Cloning with the 5′ end of the linearized pUC19 vector and the 500 bp
homology arm upstream from the insert sequence (epitope tag or
specific mutation). The second fragment will include the 500 bp
homology arm downstream from the insert sequence with a 20 bp
overlap with the 3′ end of the linearized pUC19. Both fragments
will include a 20 bp overlap at the insert sequence where they will
be fused during Gibson assembly. Included in the primers compris-
ing of the cut site, three or more silent mutations should be
included in the primers over the cut site to prevent secondary
cutting of the insert by Cas9.
1. Amplify your fragments by PCR using high-fidelity polymer-
ase. The PCR protocol will need to be optimized for each set of
primers to produce a clean product.
2. Linearize the pUC19 vector using EcoRI and HindIII restric-
tion enzymes.
A CRISPR/Cas-Based Method for Precise DNA Integration in Xenopus laevis. . . 137
3.3.2 Single Stranded 1. Amplify the whole insert including the homology arms
DNA Synthesis through PCR using primers with a phosphate modification at
the 5′ end of one of the primers and a phosphothioate modifi-
cation at the 5′ end of the other (see Note 3).
2. Denature one of the DNA strands using Guide-it long ssDNA
Production System v2 kit (Takara Bio) following the manufac-
turer’s instructions.
3. Check the single stranded DNA on a 1% agarose gel against the
double stranded template.
4. Aliquot and store the single stranded DNA at -30 °C.
3.4 Oocyte All oocyte work, apart from Liberase defolliculation should be
Preparation carried out in a dedicated 18 °C room.
1. On the day of the experiment, clean the work area thoroughly
with ethanol (see Note 4).
2. On the day of the experiment, collect the ovary of one or two
female frogs and wash thoroughly but gently in 16 °C MMR
(see Note 5).
3. Snip off a small piece of ovary and check the quality and stage of
the oocytes under a microscope (see Note 6).
4. If the quality is satisfactory, fill a 90 mm dish with 1×
MMR + gentamycin and tear the ovary into small ~2 cm pieces
using clean forceps.
5. Transfer 5 mL of ovary pieces into a 50 mL Falcon tube and
add 1× MMR + gentamycin to 12 mL.
6. Add 7 units of Liberase TM to the tube and incubate at room
temperature on a rocker set to 12 RPM for 1 h.
7. Gently wash the defolliculated oocytes 7 times in 1X MMR +
gentamycin, then once with OCM. After the last wash, pour
the oocytes into a 90 mm agarose dish and let them rest for 1 h
in a 16 °C incubator.
138 Sian Angela Martin
3.6 Oocyte Culture 1. Culture the oocytes for 72 h in 90 mm agarose dishes (100 per
and Maturation dish) filled with OCM at 16 °C. Once every 24 h clean out any
dying oocytes and transfer the remainder to a fresh dish
of OCM.
2. Incubate a 30 mM stock solution of progesterone at room
temperature for 1 h with occasional vortexing.
3. Vortex the stock solution then add to OCM at a final concen-
tration of 3 μM and mix thoroughly.
4. Pour the maturation solution into 90 mm dishes and transfer
the oocytes to the dishes (see Note 8).
5. Incubate the oocytes at room temperature for 3–4 h or until a
white maturation spot appears on the top of the oocyte, signal-
ing germinal vesicle breakdown (GVBD).
6. Incubate the oocytes at 18 °C for a further 3–4 h or until a
small white dot can be seen in the middle of the white spot (see
Note 9).
7. Check for prick activation by puncturing the matured oocyte
with a microinjection needle. Activation is characterized by the
contraction and subsequent relaxation of the animal pole. We
check for activation in 5 oocytes every half an hour after the
appearance of the white dot until at least 4 out of 5 oocytes
activate.
3.7 Sperm Nuclei 1. On the day of the experiment, collect testes from a male frog.
Preparation 2. Wash testes in ice cold 1× MMR and clean of any blood vessels
by rolling them gently on tissue. If there are any blood vessels
which cannot be removed by this method, use watchmaker’s
A CRISPR/Cas-Based Method for Precise DNA Integration in Xenopus laevis. . . 139
23. Work out the dilution needed for 1–1.5 nuclei per 10 nL
injection using the following calculation:
– n × 10 = undiluted count per injection (UC)
– Dilution = 1 μL in UC μL of SDB
3.8 ICSI Fertilization 1. Set the syringe pump to 0.6 μL/min and run it for 1 min to
in Oocytes ensure that it is pumping mineral oil through the Tygon
tubing.
2. Cut glass needles (1 for every 100 oocytes) to 40 μm, measur-
ing with a stage graticule, and cutting with watchmaker’s
forceps.
3. Once the oocytes are prick activating, start loading the oocytes
on to injection dishes filled with 6% Ficoll in 0.4× MMR and let
them sit in Ficoll for 30 min to 1 h before the first injection.
4. Back fill one of the pre-cut needles with diluted sperm nuclei
suspension until it is completely full and push the back end of
the needle into the Tygon tubing running from the
syringe pump.
5. Attach the needle with Tygon tubing to the needle holder next
to the microscope and lower the tip of the needle into your first
injection dish.
6. Check that sperm nuclei suspension is flowing through the
needle by turning the syringe pump on and looking for the
trail of suspension exiting the needle. This can be seen due to
the difference in viscosity between the suspension and the
ficoll. Try moving the needle from side to side and up and
down to see the flow-through more clearly.
7. Inject each oocyte for 1 second, inserting the needle at the edge
of the maturation spot on the animal pole. Change the needle
and sperm nuclei dilution every 100 oocytes.
8. After sperm nuclei injection leave the injected oocytes in their
injection dishes and incubate at 18 °C. Do not disturb them
until they reach blastula stage (see Note 11).
9. At stage 8–9, select all developing embryos and transfer them
into a fresh dish of 0.1× MMR and incubate at 18 °C.
10. Check on the embryos periodically throughout gastrulation,
removing any dying embryos and freezing for genotyping.
Embryos fertilized by ICSI are particularly sensitive during
this period.
11. Grow the embryos on as normal.
A CRISPR/Cas-Based Method for Precise DNA Integration in Xenopus laevis. . . 141
4 Notes
pipette tips. We use clean razor blades to cut the ends off our
tips for sperm work. This prevents mechanical damage to the
sperm or nuclei.
11. Comparison experiments performed in our lab have shown
that embryos fertilized by ICSI have a 100% higher survival
rate past gastrulation when they are not moved or disturbed
during pre-gastrula development. Removal of dead unfertil-
ized eggs may be beneficial; however, in our experience, we
have found that, in this setting, neighboring dead cells do not
appear to affect developing embryos.
References
16. Heasman J, Holwill S, Wylie CC (1991) 18. Kroll KL, Amaya E (1996) Transgenic Xenopus
Chapter 11: Fertilization of cultured Xenopus embryos from sperm nuclear transplantations
oocytes and use in studies of maternally inher- reveal FGF signaling requirements during gas-
ited molecules. Methods Cell Biol 36 trulation. Development
(C):213–218. https://doi.org/10.1016/ 19. Miyamoto K, Simpson D, Gurdon JB (2015)
S0091-679X(08)60279-4 Manipulation and in vitro maturation of xeno-
17. Amaya E, Kroll KL (1996) A method for gen- pus laevis oocytes, followed by intracytoplasmic
erating transgenic frog embryos. In: Molecular sperm injection, to study embryonic develop-
embryology: methods and protocols, 97th ment. J Vis Exp 96(96):52496. https://doi.
edn, pp 393–414 org/10.3791/52496
Chapter 12
Abstract
Nucleic acid aptamers are short sequences of single-stranded (ss) DNA or RNA that fold into a three-
dimensional shape with useful binding properties. Traditionally, these properties have included specific
recognition and binding of ions, small-molecules, proteins, and enzyme targets. Increasingly though,
aptamers are being raised against complex subcellular or cellular targets. These broader-affinity aptamers
can be usefully employed for detection, labeling, or therapeutic targeting of intact/living cells, whether
prokaryotic or eukaryotic. Aptamers are usually developed from a random-sequence oligonucleotide library
by repeated rounds of selection and amplification, a process named “systematic evolution of ligands by
exponential enrichment” (SELEX). We describe here a widely applicable cell-SELEX method for raising
aptamers against bacteria, using Escherichia coli strain HB101 as an example. Our cell-SELEX method uses
a cycle of four stages: (1) incubation of a fluorescently labeled random-sequence ssDNA library with
bacterial cells; (2) separation of cell-associated ssDNA from free ssDNA; (3) amplification of bound
ssDNA by PCR, and (4) use of lambda-exonuclease to selectively regenerate ssDNA for further rounds
of selection.
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_12,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
145
146 Robert Gowland and Darren M. Gowers
2 Materials
2.4 SELEX Round 1. PCR primers for SELEX: these are called SELEX-Top and
One: Amplification and SELEX-Bot, and for each, you will need a stock of ~100 μL
Regeneration of ssDNA at 50 μM in nuclease-free water. Then make 100 μL of a
working stock of each at 20 μM in nuclease-free water. See
Fig. 1b above for sequences (see Note 3).
2. Taq PCR mix (2×): 10 mM Tris-HCl, pH 8.6, 50 mM KCl,
1.5 mM MgCl2, 0.2 mM dNTPs, 5% (v/v) glycerol, 0.08%
150 Robert Gowland and Darren M. Gowers
Cell-SELEX Oligonucleotides
5’-(HEX)GCGACCCAAGGAATTCTGCTGAAGG-3’ 5’-(PHOS)GGCTGGTGACGGATCCGGTACGATG-3’
C TOP cloning primer (25 nt) BOT cloning primer (25 nt)
5’-GCGACCCAAGGAATTCTGCTGAAGG-3’ 5’-GGCTGGTGACGGATCCGGTACGATG-3’
EcoRI BamHI
D pJET1.2 forward seq primer (23 nt) pJET1.2 reverse seq primer (24 nt)
5’-CGACTCACTATAGGGAGAGCGGC-3’ 5’-AAGAACATCGATTTTCCATGGCAG-3’
Fig. 1 Cell-SELEX oligonucleotides. The nucleic acid sequences of oligos used in this cell-SELEX method are
shown. (a) The 100 nt starting library sequence (top) contains a 5′-hexachlorofluorescein group (pink) for
visualization and steric inhibition of lambda exonuclease digestion, along with two 25 nt flanking sequences
and a central 50 nt random-sequence cassette (N = A/T/C/G). (b) The two SELEX primers for use in amplifying
the purified cell-associated sequences. The TOP SELEX primer contains a 5′-HEX group (pink) and the BOT
SELEX primer contains a 5′-phosphate group, essential for lambda exonuclease specificity (orange). (c) The
two cloning primers are identical to the SELEX primers above, but lack any 5′-modification. The TOP cloning
primer contains a single EcoRI site (green) and the BOT cloning primer contains a single BamHI site (blue). (d)
The forward and reverse sequencing primers for the pJET series of cloning vectors are shown
Fig. 2 Aptamer selection by Cell-SELEX. The starting pool of random-sequence single-stranded DNA (colored
strands, top left) is mixed with bacterial cells (top right). Three rounds of cell resuspension and centrifugation
remove weakly-bound ssDNA sequences. The remaining steps round the Cell-SELEX clock involve purifying
the bound ssDNA sequences, PCR-amplifying them to dsDNA, returning the dsDNA to ss form (using lambda
exonuclease digestion), and then reincubating the enriched pool for another round of cell binding and selection
Table 1
PCR cycle conditions
2.5 Further SELEX 1. Sufficient microfuge boxes: one per round. Label clearly and
Rounds keep all samples at 4 °C.
2.6 Cloning and 1. Cloning materials and methods as per your laboratory’s pre-
Sequencing of ferred method. See Fig. 1c for cloning primers (see Notes 4
Aptamer Candidates and 19).
2. Sequencing materials and methods as per your laboratory’s
preferred method. Most labs send batches of plasmids for
sequencing to a commercial company. See Fig. 1d for sequenc-
ing primers for pJET1.2 vectors (see Notes 5 and 20).
3 Methods
Order and prepare all the materials and reagents in advance. Con-
duct all steps at room temperature unless otherwise specified. Wear
lab coat, gloves, and appropriate PPE at all times to avoid contami-
nation and minimize the risk of nuclease degradation of samples.
3.1 Preparation of 1. Use a sterile loop to streak a fresh LB-amp plate with your
Bacterial Target target bacteria (here, HB101 freshly transformed with
pAT153) and grow inverted overnight at 37 °C (see Note 7).
2. Pick a single well-spaced colony to inoculate 10 mL LB-amp
broth and start growing aerobically at 37 °C, shaking at
200 rpm; note the time.
3. Measure the OD600 in a spectrophotometer or cell-density
reader every 40 min by taking a 2 mL sample to a sterile plastic
cuvette; blank against a separate 2 mL sample of sterile LB-amp
media. It can be useful to plot the growth curve as you go.
4. Once the optical density reaches ~0.4 (about 4 h), take a 1 mL
sample to a fresh microcentrifuge tube and, with a balance
tube, centrifuge at 8000 × g for 3 min to pellet the cells (see
Note 8).
5. Remove the liquid media from each by first decanting it away
and then using a pipette to remove any residual LB media.
Label this tube “EC1” (E. coli pellet 1) and store in a box at
room temperature until required (see Note 9).
3.2 SELEX Round 1. Using a hot-block, heat the tube containing 100 μL of 30 μM
One: Binding SELEX library oligonucleotide (in your selected buffer, here
1×PBS) to 95 °C for 5 min, then allow it to cool slowly to room
temperature. This allows any ssDNA secondary structures
to form.
2. Add 1 mL of 1×PBS to tube EC1 and gently resuspend the
E. coli pellet by pipetting slowly; avoid introducing air bubbles.
3. To the resuspended cells, add 50 μL of the cooled 30 μM
SELEX library oligonucleotide dissolved in 1×PBS (giving
~1.4 μM final), mix gently, and leave to incubate at room
temperature for 30 min (see Note 10).
4. Centrifuge at 8000 × g for 3 min to collect the cells.
5. Take a 5 μL sample of the supernatant, add to 10 μL D-PAGE
loading buffer and label this “S1-1” (Sample 1, round 1) (see
Note 11).
6. Remove and dispose of all the remaining supernatant (~1 mL)
by pipette.
154 Robert Gowland and Darren M. Gowers
3.3 SELEX Round 1. Heat-kill the resuspended cells at 95 °C for 5 min (with the cap
One: Purifying Bound on and a weight on top) then cool on ice for 5 min. Briefly
DNA centrifuge to collect all liquid at the bottom of the tube.
2. Transfer the contents (~100 μL) to a “Phase-lock gel” tube,
add 100 μL of PCI, and mix (do not vortex).
3. Centrifuge at 16,000 × g for 3 min to separate the organic and
aqueous phases.
4. Pipette the top (aqueous) layer into a fresh tube and add 25 μL
of 10 M ammonium acetate (to give a final concentration of
2 M).
5. Add two-volumes (250 μL) of freezer-cold 100% (v/v) etha-
nol, mix and store at -20 °C for 30 min before centrifuging at
16,000 × g for 20 min at 0 °C (with the microfuge hinge facing
“up” in the centrifuge rotor).
6. Remove all the ethanolic supernatant by pipette, taking care
not to disturb the precipitated DNA on the side under the
hinge.
7. Wash the precipitated DNA by adding 500 μL of freezer-cold
70% (v/v) ethanol down the side of the tube opposite the pellet
and centrifuge at 16,000 × g for 5 min at 0 °C.
8. Remove all the ethanolic supernatant by pipette and air dry
with the cap open for 5 min at room temperature.
9. Resuspend the DNA pellet in 50 μL nuclease-free water, taking
care to resuspend the DNA under the hinge as well. Label this
tube “Bound DNA” (see Note 14).
10. Take a 5 μL sample of Bound DNA and add to 10 μL D-PAGE
loading buffer and label “S5-1.”
3.4 SELEX Round 1. Prepare a PCR master-mix. Into a single tube (on ice) add
One: Amplification and 330 μL nuclease-free water, 15 μL of SELEX-Top primer,
Regeneration of ssDNA 15 μL of SELEX-Bot primer, and 375 μL of 2×Taq PCR mix.
Mix gently and keep on ice until ready; the total volume is
735 μL (see Note 15).
Aptamers and SELEX 155
Table 2
SELEX samples per round and PAGE type to run them on
3.5 Further SELEX 1. To refine the evolving pool of aptamers, you may proceed with
Rounds as many further rounds of SELEX as you like by repeating
Subheadings 3.1, 3.2, 3.3 and 3.4. Use a fresh bacterial pellet
for each round and remember to update the names of each
sample (“EC2,” “S1-1,” “S2-2,” etc.).
2. To start Round Two, repeat Subheadings 3.1, 3.2, 3.3 and 3.4,
but in place of using 50 μL of the starting oligonucleotide pool
(as you did in Round One), use 50 μL of the ssDNA R2I
(Round Two Input) from Subheading 3.4, step 13. Remember
to heat the R2I sample to 95 °C for 5 min, then allow to cool
slowly to room temperature. This allows any ssDNA secondary
structures to form.
3. Continue as required for Round Three, Round Four, and so on
(see Note 18).
3.6 Cloning and 1. For each round of SELEX, check that you have a 10 μL sample
Sequencing of from Subheading 3.4, step 7 (labeled “Round One Cloning,”
Aptamer Candidates “Round Two Cloning,” and so on).
2. Clone a sample (10 to 20) of the aptamer candidates from each
round. Use your laboratory’s preferred cloning, transformation
and recombinant plasmid purification protocols. Figure 1c
shows the cloning primers we use, and these contain restriction
sites for EcoRI and BamHI, respectively (see Note 19).
Aptamers and SELEX 157
3.7 Analyzing and 1. Align the sample of nucleotide sequences within each round
Ordering Aptamer using a multiple sequence alignment (MSA) tool. We recom-
Candidates mend using the EBI Clustal Omega, MUSCLE, or MAFFT
online tools (www.ebi.ac.uk/tools/msa/).
2. Use Weblogo (www.weblogo.berkeley.edu) to create a nucleo-
tide frequency histogram of the central 50 nt cassette for each
SELEX round.
3. Compare the predicted secondary structure for the individual
sequences in each round using either the University of Vienna
RNAfold webserver (http://rna.tbi.univie.ac.at/) or mFold
tool on the UNAfold server (www.unafold.org).
4. Construct a list of ssDNA aptamer sequences that you would
like to test. We recommend screening several from each round,
though those from later rounds are likely to show greater
affinity. Order your chosen sequences from an oligonucleotide
supplier, ensuring they are HPLC- or PAGE-purified (see Note
21).
5. Conduct further biochemical, biophysical and microscopy
experiments as required. The 5’-HEX group is very useful to
have for visualizing and quantitating binding. An important
control experiment is to test the binding of a scrambled version
of you best-binding aptamer sequence. This scrambled control
has the same number of A, G, C and T nucleotides as your
candidate aptamer, but in a random order. Another control is
to test your best putative aptamer sequences against a sample of
the starting random pool. Finally, you can explore how chang-
ing the backbone to nuclease-resistant linkages (such as PNA,
LNA) affects binding and so on.
4 Notes
6. You (obviously) will not know the sequence for these until you
have completed Stage 3.7, so there is inevitably a short hiatus
between finishing SELEX and ordering aptamer candidates.
We recommend these oligos are HPLC- or PAGE-purified; it
is always best to order the full 100 nt length (i.e. including the
primer sites) and not just the 50 base central cassette.
7. Use a freshly grown culture from a single colony every round.
As mentioned earlier, you may choose any bacterial species to
study; transformation with a plasmid for antibiotic selection
makes plating and growth easier.
8. In LB-amp, we find that an E. coli culture with an OD600 of
0.45 has approximately 107 cells/mL.
9. It is important to remove as much of the LB growth media as
possible. In the next round, label the pellet “EC2” and so on.
10. This is the SELEX binding step. You can increase or decrease
the binding duration and/or temperature. In fact, the more
stringent the binding conditions, the greater the likelihood of
obtaining aptamers with an improved Kd. Note though that in
the early few rounds, you may like to have less-stringent con-
ditions and increase binding stringency in later rounds.
11. This 15 μL sample, along with other samples taken throughout
the round, is run on a gel later, at the end of Subheading 3.4
and corresponds to unbound oligo sequences.
12. This is the first of three SELEX wash steps to remove any
ssDNA that is not tightly bound to the bacteria.
13. At this stage, only ssDNA that is able to bind to the bacteria
will remain. In the next stages, this is purified from the bacteria
(Subheading 3.3), amplified by PCR (Subheading 3.4), cloned
(Subheading 3.5), and input for another round of SELEX
(Subheading 3.6).
14. The tube Bound DNA contains purified ssDNA molecules
from the starting aptamer pool that were able to bind to the
bacteria.
15. The PCR master-mix contains every PCR component except
template. Enough is made for fifteen 50 μL PCR reactions,
though only fourteen PCR reactions are run; the extra volume
is spare, to allow for pipetting error, though can be run as a
further no-template control if required.
16. This is the step at which the dsDNA pool is converted back to
ssDNA, due to the lambda exonuclease preferentially hydro-
lysing the “lower” 5′-phosphate-labelled strand; the “upper”
strand remains intact due to chemical protection by the
5′-HEX group. You may use 10 or 20 units of lambda exonu-
clease, though we have found that 5 units is easily sufficient to
accomplish bottom strand digestion.
160 Robert Gowland and Darren M. Gowers
17. Running the samples is very useful, and you will need one
denaturing (D-PAGE) gel and one native (N-PAGE) gel per
round. This is to check for the presence or absence of DNA at
each stage, and especially for correct amplicon sizes and PCR
controls. This completes one full round of SELEX.
18. We have found that 4–8 rounds of binding and selection is
sufficient to evolve a pool of cell-binding aptamers. For each
subsequent SELEX round, remember to modify all the sample
labels to match the correct round number; for example, “S1-
2,” “S2-2,” “Round Two Cloning,” and so on.
19. For example, cutting the PCRs with EcoRI and BamHI and
ligating into your cloning vector of choice (such as pUC19,
2686 bp) before transforming and sequencing a sample of the
recombinant plasmids. In our lab, we use the CloneJET PCR
cloning kit and the pJET1.2/Blunt vector (2974 bp) for rapid
cloning. For this method, follow the supplied kit protocol. In
brief, this involves cutting 1 μL of the PCR sample with EcoRI
and BamHI, blunting the product and blunt-end cloning into
pJET1.2/Blunt, followed by transformation into competent
DH5α cells. Transformants are grown on LB-amp plates and
subject to colony PCR with the SELEX PCR primers; clones
showing a 100 bp fluorescent product are grown on in 5 mL
LB-amp broth, a plasmid minipreparation performed and a
sample sent for sequencing. It is possible to sequence.
20. We recommend sequencing at least 10–20 (or more) different
clones per round, to give a statistically useful picture of the
evolving aptamer pool. Some labs routinely use next-genera-
tion sequencing (NGS) rather than cloning and transformation
and this allows a greater number of sequencing reads per
round. Interestingly, it is occasionally observed through
sequence analysis that after extended rounds of selection
(5+), amplicons with additional Bot primer sequences at the
3′ terminus can be generated. These additional sequences can
be removed using the appropriate endonuclease (BamH1)
digestion. It may also be that these additional primer sequences
are accumulated during PCR or molecular cloning.
21. It is best practice to ensure they have the same format as the
100 nt ssDNA library pool, with a 5′-HEX group and the two
flanking 25 nt primer sequences, but with the 50 nt random
central cassette replaced with the defined sequence to be
tested. For economy, you can omit the 25 nt flanking regions.
Acknowledgments
References
1. Joyce GF (1989) Amplification, mutation and 8. Kim YS, Song MY, Jurng J, Kim BC (2013)
selection of catalytic RNA. Gene 82:83–87 Isolation and characterization of DNA apta-
2. Ellington AD, Szostak JW (1990) In vitro mers against Escherichia coli using a bacterial
selection of RNA molecules that bind specific cell-systematic evolution of ligands by expo-
ligands. Nature 346:818–822 nential enrichment approach. Anal Biochem
3. Tuerk C, Gold L (1990) Systematic evolution 436:22–28
of ligands by exponential enrichment: RNA 9. Stoltenburg R, Reinemann C, Strehlitz B
ligands to bacteriophage T4 DNA polymerase. (2007) SELEX – a (r)evolutionary method to
Science 249:505–510 generate high-affinity nucleic acid ligands. Bio-
4. Chai C, Xie Z, Grotewold E (2011) SELEX mol Eng 24:381–403
(Systematic Evolution of Ligands by EXponen- 10. Gooch J, Tungsirisurp S, Costanzo H,
tial Enrichment) as a powerful tool for deci- Napier R, Frascione N (2021) Generating apta-
phering the protein-DNA interaction space. mers towards human sperm cells using mas-
Methods Mol Biol 754:249–258 sively parallel sequencing. Anal Bioanal Chem.
5. Lakhin AV, Tarantul VZ, Gening LV (2013) (Epub ahead of print)
Aptamers: problems, solutions and prospects. 11. Kolm C, Cervenka I, Aschl UJ, Baumann N,
Acta Naturae 5:34–43 Jakwerth S, Krska R, Mach RL, Sommer R,
6. Davydova A, Vorobjeva M, Pyshnyi D, DeRosa MC, Kirschner AKT, Farnleitner AH,
Altman S, Vlassov V, Venyaminova A (2016) Reischer GH (2020) DNA aptamers against
Aptamers against pathogenic microorganisms. bacterial cells can be efficiently selected by a
Crit Rev Microbiol 42:847–865 SELEX process using state-of-the art qPCR
and ultra-deep sequencing. Sci Rep 10:20917
7. Darmostuk M, Rimpelova S, Gbelcova H,
Ruml T (2015) Current approaches in 12. Keefe AD, Pai S, Ellington AD (2010) Apta-
SELEX: an update to aptamer selection tech- mers as therapeutics. Nat Rev Drug Discov 9:
nology. Biotechnol Adv 33:1141–1161 537–550
Chapter 13
Abstract
RNA performs a wide variety of vital cellular functions. These functions typically require interactions with
other biological macromolecules, often as part of an intricate communication network. High-throughput
techniques capable of analyzing RNA-based interactions are therefore essential. Functional-RNA arrays
address this need, providing the capability of performing hundreds of miniature assays in parallel. Here we
describe a method to generate functional-RNA arrays using in vitro transcription of a DNA template array
and in situ RNA capture. We also suggest how functional-RNA arrays could be applied to investigating
RNA-RNA interactions.
Key words Binding partner, Functional-RNA, In situ capture, In vitro transcription, Microarray,
RNA array, RNA-RNA interactions, Surface immobilization
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_13,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
163
164 Helen A. Vincent et al.
Fig. 1 Generation of functional-RNA arrays by in vitro transcription and in situ RNA capture. Custom DNA
in vitro transcription templates are designed and synthesized. These are spotted onto a microarray slide to
produce a DNA in vitro transcription template array. A DNA in vitro transcription template array—in vitro
transcription reagent mix (IVT)—RNA capture surface “sandwich” is assembled. As in vitro transcription
proceeds, RNA is captured in situ to produce a corresponding functional-RNA array
2 Materials
2.1 In Vitro 1. RNA secondary structure prediction software (see Note 1).
Transcription
Template Design and
Synthesis
2.2 Generation of the 1. Streptavidin-coated glass microarray slide (see Notes 2–4).
DNA In Vitro 2. 20–500 nM 5′-biotinylated DNA in vitro transcription tem-
Transcription plate(s) in phosphate-buffered saline (PBS), pH 7.4 (see Notes
Template Array 5–7).
3. Automated arrayer (see Note 8) or a micropipette.
4. PBST: PBS, pH 7.4 supplemented with 0.05% (v/v) Tween
20.
5. PBS.
6. H2O.
7. Microarray slide scanner (see Notes 9 and 10).
2.3 Generation of the 1. DNA in vitro transcription template array (see Subheading 3.2).
Functional-RNA Array 2. Streptavidin-coated glass microarray slide (see Notes 2–4).
3. Parafilm.
4. T7 in vitro transcription reagent mix (see Note 11): 1X MEGA-
script T7 Transcription Reaction Buffer, 1X MEGAscript T7
Transcription Enzyme Mix, 0.4 mM ATP, 0.4 mM CTP,
0.4 mM GTP, 0.4 mM UTP.
5. PBST.
6. PBS.
7. H2O.
8. 50–100 nM linker probe (a fluorescently labeled DNA oligo-
nucleotide with a sequence complementary to the unstruc-
tured linker between the RNA of interest and the
immobilization aptamer (see Subheading 3.1 and Fig. 2) in
2X saline-sodium citrate (SSC) supplemented with 0.1% w/v
sodium dodecyl sulphate (SDS) (see Notes 12–14).
9. LifterSlip (Thermo Fisher Scientific) (see Note 15).
10. Microarray slide scanner (see Notes 9 and 10).
Generation of Functional-RNA Arrays 167
3 Methods
The protocols provided below assume that the user has experience
working in a molecular biology laboratory and is familiar with
standard molecular biology techniques. The protocols are intended
to provide a starting point when generating functional-RNA arrays
for the first time. It is anticipated that the user will bring their
system-specific knowledge to these protocols and will adapt them
where appropriate.
Subheading 3.1 involves the custom design and synthesis of
in vitro transcription templates. This will require forward planning.
Once this step is complete, Subheadings 3.2–3.4 can be completed
in one day or they can be split over two days with Subheading 3.2
completed on the first day and Subheadings 3.3 and 3.4 completed
on the second day.
Fig. 2 Design of the in vitro transcription template. A schematic of the in vitro transcription template which
consists of a short immobilization linker (see Note 20), a promoter (see Note 21), transcription system-specific
sequence (see Note 22), sequence encoding the RNA of interest, an unstructured linker (see Note 23),
sequence encoding an immobilization aptamer (see Notes 24 and 25) and an optional fluorophore (see Note
26). A suggested starting DNA sequence for each component of the in vitro transcription template is indicated,
except for the user-specified RNA of interest sequence which is represented by . . .(N)n. . .
Fig. 3 Example RNA secondary structures predictions. (a) The sequence of the full-length RNA transcript (RNA)
which consists of GGG (T7 transcription system-specific sequence; grey), MicA (a small regulatory RNA of
interest; blue), an unstructured linker (magenta) and the streptavidin-binding RNA aptamer (SAapt; immobili-
zation aptamer; gold). The RNA secondary structures, predicted by RNAfold (rna.tbi.univie.ac.at) [12], for the
full-length RNA transcript (FL), MicA and the streptavidin-binding RNA aptamer (SAapt) are shown below the
RNA sequence in dot-bracket notation. (b–d) The RNA secondary structures, predicted by RNAfold, visualized
in forna [16]. The predicted secondary structures for MicA and the streptavidin-binding aptamer are the same
in the context of the full-length transcript and in isolation
170 Helen A. Vincent et al.
Fig. 4 Spotting and visualization of the DNA in vitro transcription template array. The DNA in vitro transcription
template array is prepared by spotting 5′-biotinylated DNA in vitro transcription templates onto a streptavidin-
coated microarray slide. Spotting can be performed manually using a micropipette, or it can be automated
using an arrayer (see Note 31). Both methods result in discrete spots containing multiple molecules of the
immobilized DNA in vitro transcription template. Following spotting, the DNA in vitro transcription template
array is washed. If the DNA in vitro transcription templates are also fluorescently labeled (see Note 26), the
DNA in vitro transcription template array can be visualized using a microarray scanner and quantified (see
Note 37)
Fig. 5 In vitro transcription and in situ RNA capture. (a) Assembly of the DNA in vitro transcription template
array—in vitro transcription reagent mix (IVT mix)—RNA capture surface “sandwich.” The RNA capture
surface (a streptavidin-coated microarray slide) is positioned surface-side up. A small piece of parafilm is
positioned at each of the short edges of the RNA capture surface to act as spacers (see Note 40). In vitro
transcription reagent mix is pipetted onto the RNA capture surface and the DNA in vitro transcription template
array, array-side down, is carefully lowered onto the RNA capture surface to complete the “sandwich”
assembly. (b) The “sandwich” assembly is incubated at 37 °C for 30–90 min during which time RNA is
synthesized by in vitro transcription and captured in situ by the RNA capture surface. (c) The DNA in vitro
transcription template array and the newly generated corresponding functional-RNA array are carefully
separated
Fig. 6 Probing the functional-RNA array with linker probe. The functional-RNA array can be probed with a
fluorescently labeled DNA oligonucleotide with a sequence complementary to the unstructured linker region
between the RNA of interest and the streptavidin-binding RNA aptamer (see Note 12). This allows visualization
and quantification of the RNA levels on the functional-RNA array (see Note 46). A solution of fluorescently
labeled linker probe is pipetted over the functional-RNA array (see Note 13). This is covered with a LifterSlip
and incubated at room temperature for 30 min in the dark (see Note 47). During incubation, the linker probe
binds to the unstructured linker on the immobilized functional-RNAs. Following incubation, the LifterSlip is
removed, the functional-RNA array is washed, and linker probe fluorescence is detected using a microarray
scanner and quantified (see Note 48)
Generation of Functional-RNA Arrays 173
3.4 Application of 1. Design the functional-RNA array layout (see Notes 32, 49 and
Functional-RNA Arrays Fig. 7).
to the Evaluation of 2. Design each DNA in vitro transcription template that will be
RNA-RNA Binding needed to generate the functional-RNA array (see
Specificity Subheading 3.1).
3. Generate the DNA in vitro transcription template array (see
Subheading 3.2).
4. Generate the functional-RNA array (see Subheading 3.3) and
use immediately.
5. Probe the functional-RNA array with the RNA binding partner
probe (see Notes 16–18, 50–55). (1) Place the functional-
RNA array with the array surface-side up (see Note 44) and
pipette 90 μL RNA binding partner probe (see Notes 16–18,
50 and 52) onto the functional-RNA array and cover with a
24 × 60 mm LifterSlip (see Note 53). (2) Incubate for 30 min
at room temperature in the dark (see Note 54). (3) Remove the
LifterSlip, transfer the functional-RNA array slide to a 50 mL
Falcon tube containing 45 mL hybridization buffer (see Note
44) and rotate on a rolling platform for 5 min at room temper-
ature. (4) Repeat step 3 two more times (see Note 56). (5) Dry
the functional-RNA array slide by centrifugation at 500 × g for
5 min at room temperature (see Note 36).
6. Visualize the functional-RNA array using a microarray slide
scanner (see Note 55).
7. Quantify and analyze the data (see Fig. 7).
174 Helen A. Vincent et al.
Fig. 7 Application of functional-RNA arrays to the evaluation of RNA-RNA binding specificity. Functional-RNA
arrays can be used to detect RNA-RNA interactions [5, 10]. (a) A functional-RNA array is designed with
appropriate positive (+) and negative (-) control RNAs and a variety of test RNAs (T1-T7) (see Note 49). (b) A
DNA in vitro transcription template array is generated (see Notes 5–7). (c) A functional-RNA array is generated
by in vitro transcription and in situ RNA capture and probed with linker probe (see Notes 12–14, 32 and 46).
(d) The functional-RNA array is probed with RNA binding partner probe (see Notes 16–18, 32 and 51).
Fluorescence should be clearly detected for the positive control and not for the negative control. The presence
or absence of fluorescence for the test RNAs can give a yes-no output for binding or the relative intensity of the
RNA detected may be able to give an indication of the relative binding affinity
4 Notes
Unstructured linker RNA sequence Streptavidin-binding RNA aptamer (SAapt) RNA sequence Reference
ACACACACACACACACACAC GCAUGCAUACCGACCAGAAUCAUGCAAGUGCGUAAGAUA [5, 6, 10]
GUCGCGGGCCGGGAUGCAUGC
UUUUUUUUUUUUUUUUUUUU UGUGUGACCGACCAGAAUCAUGCAAGUGCGUAAGAUAGU [5]
CGCGGGCCGGGCACACA
UUUUUUUUUUUUUUUUUU UAGAGACCGACCAGAAUCAUGCAAGUGCGUAAGAUAGUCG [5]
CGGGCCGGGCUCUA
GUGUGACCGACCAGAAUCAUGCAAGUGCGUAAGAUAGUC [5]
GCGGGCCGGGCACAC
AUGCAUGCACCGACCAGAAUCAUGCAAGUGCGUAAGAUA [5]
GUCGCGGGCCGGGGCAUGCAU
CGAUCGAUACCGACCAGAAUCAUGCAAGUGCGUAAGAUAG [6]
UCGCGGGCCGGGAUCGAUCG
AAUAAUAAUAAUAAUAAUAAU AUGCAUGCACCGACCAGAAUCAUGCAAGUGCGUAAGAUA [5]
GUCGCGGGCCGGGGCAUGCAU
The SAapt RNA sequences consist of a core sequence (regular text) [17] and a variable sequence at the 5′ and 3′ ends (bold and underlined) that extends a duplex and stabilizes the
aptamer [17]
Generation of Functional-RNA Arrays
179
180 Helen A. Vincent et al.
Acknowledgements
References
1. Cech TR, Steitz JA (2014) The noncoding the fabrication of RNA microarrays. Comput
RNA revolution – trashing old rules to forge Struct Biotechnol J 17:862–868
new ones. Cell 157:77–94 5. Phillips JO, Butt LE, Henderson CA,
2. Marz M, Stadler PF (2011) RNA Devonshire M, Healy J, Conway SJ,
interactions. In: Collins LJ (ed) RNA Infra- Locker N, Pickford AR, Vincent HA, Calla-
structure and Networks, Advances in Experi- ghan AJ (2018) High-density functional-RNA
mental Medicine and Biology, vol 722. arrays as a versatile platform for studying
Springer, New York RNA-based interactions. Nucleic Acids Res
3. Li PCH, Sedighi A, Wang L (eds) (2016) 46:e86
Microarray technology, Methods in Molecular 6. Henderson CA, Rail CA, Butt LE, Vincent
Biology, vol 1368. Humana Press, New York HA, Callaghan AJ (2019) Generation of small
4. Lietard J, Somoza MM (2019) Spotting, tran- molecule-binding RNA arrays and their
scription and in situ synthesis: three routes for
184 Helen A. Vincent et al.
Abstract
Oligonucleotides are used in a variety of molecular biology techniques, from next-generation sequencing to
genetic testing. Maintaining the sequence fidelity of synthetic oligonucleotides is critical to their use. This
chapter describes the steps of solid phase oligonucleotide synthesis and purification, which enables the
synthesis of oligonucleotides with specific sequences and high purity.
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_14,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
185
186 John Brazier
2 Materials
2.2 Oligonucleotide 1. C8 or C18 reverse phase HPLC column suitable for oligonu-
Purification Using cleotide purification and compatible with your HPLC
Reverse-Phase HPLC instrument—For a 1 μmole scale synthesis a 250 × 4.60 mm
(RP-HPLC) column would be an appropriate size.
2. Buffer A: 0.1 M aqueous triethyl ammonium acetate pH 6.5.
Add 14 mL of triethylamine (HPLC grade) to 900 mL of water
(HPLC grade). Adjust to pH 6.5 by addition of acetic acid
(HPLC grade) and make up total volume to 1 L with water
(HPLC grade).
3. Buffer B: 0.1 M triethylammonium acetate in 65% (v/v) Ace-
tonitrile/ water at pH 6.5. Add 14 mL of triethylamine (HPLC
grade) to 300 mL of water (HPLC grade) and 650 mL of
acetonitrile (HPLC grade). Adjust to pH 6.5 by addition of
acetic acid (HPLC grade) and make up total volume to 1 L with
water (HPLC grade) (see Note 5).
4. 5′-DMT deprotection: 20% v/v acetic acid in water.
188 John Brazier
2.3 Final Desalting of 1. C8 or C18 reverse phase HPLC column suitable for oligonu-
the Oligonucleotide cleotide purification and compatible with your HPLC
Solution Using RP- instrument—for a 1 μmole scale synthesis a 250 × 4.60 mm
HPLC column would be an appropriate size.
2. 1 M aqueous triethyl ammonium bicarbonate at pH 7.8. Add
140 mL of triethylamine (HPLC grade) to 860 mL of water
(HPLC grade). Using a sintered filter, bubble CO2 gas (usually
produced from sublimation of solid CO2) through the aqueous
triethyl ammonium hydroxide solution until the solution
reaches pH 7.8.
3. Buffer C: 0.1 M aqueous triethyl ammonium bicarbonate. Add
100 mL of 1 M aqueous triethyl ammonium bicarbonate,
pH 7.8 to 900 mL of water (HPLC grade).
4. Buffer D: 0.1 M triethyl ammonium bicarbonate 65:35 v/v
acetonitrile/ water. Add 100 mL of 1 M aqueous triethyl
ammonium bicarbonate, pH 7.8 to 250 mL of water (HPLC
grade), and 650 mL of acetonitrile (HPLC grade).
3 Methods
3.1 Automated 1. Install each of the synthesis reagents onto the instrument:
Oligonucleotide activator, oxidizer, de-block, capping mix, and anhydrous
Synthesis wash (see Note 6).
2. Install each of the nucleotide phosphoramidite solutions (see
Note 6).
3. Prime the instrument with each reagent (see Note 7).
4. Select the synthesis cycle and the synthesis scale on your
instrument.
5. Input your desired sequence into the instrument (see Note 8).
6. Choose whether to remove the 5′-DMT group from your final
oligonucleotide (see Note 9).
7. Install the correct solid phase synthesis column (see Note 10).
8. If your instrument allows, check that the reagent quantities are
sufficient for the sequence, scale, and synthesis cycle that you
have chosen.
9. Start the synthesis.
10. Once the synthesis is complete, and if present on your instru-
ment, you should check the trityl monitor to make sure that
each coupling step was successful (see Note 11).
Phosphoramidite Chemistry 189
3.2 Oligonucleotide 1. Install the HPLC column, buffer A: 0.1 M aqueous triethyl
Purification Using ammonium acetate pH 6.5, and buffer B: 0.1 M triethylam-
Reverse Phase-HPLC monium acetate in 65% (v/v) Acetonitrile/ water at pH 6.5,
(RP-HPLC) (See using the instructions supplied with your HPLC instrument
Note 18) (see Note 19).
2. Run an analytical trace of your oligonucleotide using a gradient
of 0–100% buffer B over 25 min (see Note 20), using UV
detection at 260 nm.
3. Use your analytical trace to determine the elution time of your
oligonucleotide (see Notes 21 and 22).
4. Run several purification traces of your oligonucleotide using a
gradient of 0–100% buffer B over 25 min (see Notes 22 and
23), collecting the solution that is eluted at the elution time of
your oligonucleotide.
5. Combine the eluted solution you have collected into one sam-
ple and dry your oligonucleotide by gentle evaporation of the
buffer until approximately 50 μL remain.
Phosphoramidite Chemistry 191
3.3 Final Desalting of 1. Install the HPLC column, buffer C: 0.1 M aqueous triethyl
the Oligonucleotide ammonium bicarbonate, and buffer D: 0.1 M triethyl ammo-
Solution Using RP- nium bicarbonate 65:35 v/v acetonitrile/ water, using the
HPLC instructions supplied with your HPLC instrument (see
Note 19).
2. Run an analytical trace of your oligonucleotide using a gradient
of 0–100% buffer D over 30 min (see Note 25), using UV
detection at 260 nm.
3. Use your analytical trace to determine the elution time of your
oligonucleotide.
4. Run several purification traces of your oligonucleotide using a
gradient of 0–100% buffer D over 30 min (see Note 23),
collecting the solution that is eluted at the elution time of
your oligonucleotide.
5. Combine the eluted solution you have collected into one sam-
ple and dry your oligonucleotide by gentle evaporation of the
buffer.
6. Resuspend your dried oligonucleotide in 500 mL of water and
evaporate to dryness again. Repeat this process twice more.
7. Your dried oligonucleotide is now purified and ready for use in
any subsequent experiments.
4 Notes
Abstract
Polymerase Chain Reaction (PCR) products have been traditionally characterized by cloning and cycle
sequencing. However, when quick sequencing data are required, the cloning step may be omitted and PCR
products can be sequenced directly. We describe here a sequencing protocol that involves the gold standard
Big Dye chemistry in a low throughput format using one of the latest sequencing platforms the ABI
Seqstudio. Our cycle sequencing protocol follows the following steps: (1) purification of the PCR product
with a spin column-based kit; (2) quality & quantity assessment of the PCR product with the use of
spectrophotometry & gel electrophoresis; (3) setup and amplification of the cycle sequencing reaction;
(4) Capillary Electrophoresis; (5) Sequence Data Analysis.
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_15,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
195
196 George D. Zouganelis and Nikolaos Tairis
Table 1
Capillary electrophoresis systems (Life Technologies) [5]
2 Materials
Ensure that the work takes place in a dedicated laboratory and that
PPE is worn at all times in order to avoid contamination. Use
deionized water for buffer dilution and PCR grade molecular biol-
ogy water (12 Ω) for cycling reactions. When planning your work,
primers should be ordered first as they may take longer time to
arrive as they are customary synthesized.
3 Methods
3.1 PCR Product Prior to cycle sequencing, PCR products are purified from the PCR
Purification and Master mix and primer dimers with the use of Qiaquick PCR
Clean up Purification Kit (Qiagen) [7]. In case that PCR results in nonspe-
cific products, a special protocol that involves gel excision is
required.
3.2 Specific PCR 1. To 1 volume of each PCR sample, add 5 volumes of PB buffer
Product and mix well.
2. Apply the mixture to each spin column which is placed in a
collection tube.
3. Spin the columns in a microcentrifuge at 13000 rpm at room
temperature for 30–60 s.
4. Discard the flow through. Place the spin column in the collec-
tion tube and add 750 μL of PE Buffer to the spin column (see
Note 9).
5. Spin at 13000 rpm for 30–60 s. Discard flowthrough, place
back to collection tube, and repeat spin to dry the spin column.
6. Remove spin column and add it to a 1.5 mL microcentrifuge
tube. Add 30–50 μL EB buffer or water.
7. Spin at 13000 rpm for 30–60 s to elute DNA.
200 George D. Zouganelis and Nikolaos Tairis
3.3 Excision Gel 1. Excise the gel band with a new scalpel and place in a micro-
Protocol centrifuge tube. Minimize the amount gel at a maximum of
400 mg.
2. Add 300 μL of buffer QG per 100 mg of gel for 1% agarose.
For >2% agarose gels, add 600 μL QG per 100 mg of gel.
3. Incubate the microcentrifuge tube with the QC buffer and the
gel slice at 50 °C for 10 min with periodical vortexing until
melting.
4. Add 100 μL of isopropanol per 100 mg of gel slice if the
fragment in question is ≤500 bp and 4Kb > (see Note 10).
5. Mix the resulting liquid well and place it to the spin column,
centrifuge for 1 min at 13000 rpm and discard the flow-
through. If the liquid is more than 700 μL repeat step until
all liquid flows through.
6. Follow steps 4–7 as described in Subheading 2.1.
3.4 PCR Product Purified PCR products can be readily tested for concentration using
Quality and Quantity the Nanodrop™ 2000 instrument (Thermo Fisher) [8], which is an
Assessment accurate and versatile instrument that requires minimum amount of
sample (1–2 μL) and lower detection limit of 2 ng/μL. Template
3.4.1 Measurement of concentration is critical for the downstream sequencing reactions.
Concentration with
Nanodrop™ 2000 1. Double click on the desktop NanoDrop™ 2000 software icon
and select the “Nucleic Acids” icon.
2. Select “Add to report” so the measurements will be saved in
the report.
3. Use appropriate buffer to establish a blank. An appropriate
blank would be the solution that the PCR product is eluted
or dissolved in.
4. Pipette 1–2 μL of the blank solution onto the bottom pedestal,
lower the arm, and click the “Blank” button. Ensure that the
arm is always down for all measurements.
5. Clean the bottom pedestal with a Kimwipe, enter the sample
ID, and then click the “Measure” button.
3.4.2 Agarose Gel Prior to downstream sequencing applications, the PCR products
Electrophoresis should be checked with agarose gel electrophoresis to ensure:
(a) the integrity of the template and (b) the absence of primer
dimers that may interfere with downstream sequencing reactions.
For gel electrophoresis, the materials used are listed in Subheading
2.1, and an established methodology is followed [9].
Sanger Sequencing 201
3.5 Cycle In cases where the amplification product is less than 800 bp, one
Sequencing direction sequencing reaction may be sufficient to obtain the nec-
essary data. However, to obtain maximum data, two reactions in
3.5.1 Considerations for
forward and reverse fashions are advised. Forward and reverse
Sequencing Primer
sequencing is invaluable not only for improved coverage but also
Selection and Design
as a means of cross-referencing and validating sequencing results.
Very often the primers used in either direction could be the original
primers used for fragment amplification. Nevertheless, in cases
where resequencing is required or the fragments are too large (>
1500), nested primers must be employed in order to maximize
coverage. The criteria for primer selection and design are as follows
[10, 11]:
1. Primers should be 20–30 bp in length.
2. Close to 50% GC content.
3. The primers should include a C or G at the 3′ of their sequence.
4. Thymidine at 3′ and 5′ and four or more repeated bases should
be avoided.
5. Primers should not ideally form secondary structures or
hybridize with each other forming hairpins and loops. This
issue can be checked with the use of appropriate applications
available from primer synthesis vendors.
6. Melting temperature range is 55–65 °C.
7. Primers should be checked for specificity to the target with the
use of nBlast program and or annealing with the use of an
alignment program such as MEGA7.
8. As cycle sequencing reaction is often unreadable for the first
30–40 bp, the primer should be designed upstream of the area
of interest.
Finally, there is the option of including tag sequences to the
amplification primers. These tag sequences have to complements
corresponding to universal primer sequences listed in Table 2 that
are known to be reliable for DNA sequencing.
Table 2
Recommended universal primers for cycle sequencing [12]
3.5.2 Cycle Sequencing The cycle sequencing reaction is setup with the use of Big Dye™
Reaction Terminator v 3.1 Cycle Sequencing (Applied Biosystems) [13]. It is
recommended that the reaction is setup in a dedicated PCR cabinet
to avoid cross contamination. In addition, frequently calibrated
pipettes dedicated for sequencing should be used for the setup
cycle sequencing and other procedures.
1. Thaw the contents of the Big Dye™ Terminator v 3.1 kit and
keep in ice.
2. Prepare 3.2 μM solutions for sequencing primer(s) and keep
in ice.
3. Label 1.5 mL microcentrifuge tubes accordingly noting sample
and primer used, and add the following components as
described in Tables 3 and 4. The total volume of the reaction
is 20 μL. Mix well, centrifuge and keep on ice.
4. There is an option of diluting the sequencing reaction. For that
purpose, the Big Dye™ Terminator v 1.1 & v 3.1 5X Sequencing
Buffer should employed as shown in the example in Table 5.
Diluted reactions are not recommended to run without optimiza-
tion as the resulting sequence may be compromised. The volume
of Buffer is given by the formula: Buffer Volume = 0.5 * [(Total
Reaction Volume)/2.5 – Reaction Mix].
Table 3
Components required per cycle sequencing reaction
Table 4
Recommended DNA quantities per reaction
Table 5
Example of a diluted sequencing reaction
Example
Component Quantity volume
Big dye™ terminator 3.1 ready reaction 4 μL 4 μL
mix
Big dye™ terminator v 1.1 & v 3.1 5X 2 μL 2 μL
sequencing buffer
Sequencing primer 3.2 pmoles 1 μL
Water (molecular biology grade) Variable 11 μL
Template Variable depending on concentration of 2 μL
template (see Table 2)
Total 20 μL
Table 6
Cycle sequencing reaction parameters
3.6 Cycle After completion of the cycle sequencing, the excess dye termina-
Sequencing Product tors are required to be removed. For this purpose, we employ the
Purification DyeEx 2.0 Spin kit (Qiagen) [14], which uses gel filtration
technology.
1. The column is gently vortexed in order to resuspend the resin.
2. The cap on the column must be slightly loosened (about one
quarter turn) so vacuum within the tube is avoided.
204 George D. Zouganelis and Nikolaos Tairis
3.7 Sample 1. Thaw a fresh aliquot of Hi-Fi™ Formamide. Make sure that it
Preparation has not been through more than two freeze-thaw cycles.
2. Resuspend the samples in 10–20 μL Hi-Fi™ Formamide. Do
not resuspend in water as the sample stability will be decreased.
3. Transfer the samples in MicroAmp™ Optical 96-Well Reaction
Plates or MicroAmp™ Reaction Tubes. The tubes are provided
in strips of 8 and should be kept in place by the MicroAmpTM
96-well tray and tray retainer.
3. Cover the plates or tubes with the appropriate septa by aligning
the holes of the septa with the wells or the tubes. Ensure that
the septa are in position by appropriate pressing.
4. Centrifuge briefly the plates or reaction tube assemblies to
ensure that the contents are at the bottoms of their tubes.
5. Run the samples as soon as possible after resuspension and
setup.
3.8 Running The instrument electrophoresis run can be setup from a connected
Sequencing on the PC through a mobile device or from the instrument touch screen
Capillary [15]. Here, we describe the setup from the touch screen. Prior to
Electrophoresis starting the run, it is important to check the storage history of the
System (Seqstudio™) cartridge (see Note 11).
1. From the start up menu, touch the Eject button and carefully
open the instrument door according to the screen prompt.
2. Open the lid by pressing the release button on the autosampler.
3. Ensure that the Cathode Buffer Container is full above the fill
line. Replace it accordingly (see Notes).
4. Place the plate or the tube assembly firmly in the autosampler
and close the lid until it clicks shut.
5. Select “Retract Plate” from the “Start Menu” and close the
instrument door.
6. To run the system go to the home screen and setup run and
follow the instructions in the previous section or select.
Sanger Sequencing 205
Table 7
Run modules, read lengths, and appropriate run times
3.9 DNA Sequence Sequence can be viewed on the SeqStudio instrument. However,
Viewing and Analysis we find that the viewing and manipulation of sequences is more
convenient on a PC with the aid of freeware. For routine use, the
most cost efficient approach is to use the light version of Chro-
mas™ (Technelysium Pty LTd) that allows visualization of the
sequence and export to FASTA format which is useful when further
bioinformatics processing is necessary. Access to sequence traces
with Chromas™ is very simple as the user simply have to down load
and install the program in the PC. Simply clicking on AB1 files will
open the traces in Chromas. To analyze and process the sequence,
take the following steps:
1. Without scrolling inspect the first 50 bp. You should see peaks
of large intensity that are not distinctly separated. This is typical
as primer binding affects the reading. Click on the sequence
letters in the region of the first 50 bp. At the top right of your
screen a quality value will appear. An acceptable quality value
should be Q > 20. The majority of the first 40–50 bp usually
are below 20. Based on this the region should be excluded.
2. Scroll the sequence the to the left. In a good quality sequence
(Fig. 4b) the nucleotide peaks are very well separated from each
other and the Q > 20 for all the nucleotides. On the contrary, a
problematic sequence appears with very high noise or a large
number of nucleotides with Q < 20 (see Troubleshooting). In
that case, the sequence is rejected from further processing.
3. If you identify spurious nucleotides (i.e., 5 in a sequence of
500 bp) in the middle region with Q < 20, this may be due to
heterozygosity in particular loci (see Subheading 3.9
Troubleshooting).
4. While scrolling to still to the left there is a point where the
signal drops. This is the end of the sequence (see Fig. 4d).
5. When you are satisfied with the quality of sequence, select from
the “Edit” menu the “Trim Low-quality” function and then
“Export” the sequence as a “FASTA” file for further
processing.
3.10 Trouble- In this section we list some examples of problematic cycle sequenc-
shooting ing of PCR products with the Seqstudio system. Troubleshooting
advice is listed below every listed example.
Fig. 4 Typical images of a sequence as shown in Chromas™ lite. (a) The beginning of the sequence is affected
by binding primer. (b) The middle part of a good quality sequence exhibits high Q values (Q > 20), and the
peaks are well separated from each other. (c) The middle part of a poor quality sequence where there are
stretches of multiple nucleotides (yellow boxes) with low Q values (Q < 20). (d) The end of the sequence
where there is no coherent laser signal
208 George D. Zouganelis and Nikolaos Tairis
3.10.2 Poor Start When there is a poor start followed by a weak signal, this may
Followed by a Weak Signal attributed either to primer binding to itself or there is other primer
present (Fig. 6). This can be addressed either by redesigning a new
sequencing primer or by checking the efficiency of PCR purification
via gel electrophoresis.
3.10.3 Overlapping In the case of overlapping peaks in sequencing data, the sequence
Peaks might exhibit multiple priming sites or the sequencing primer
might not be sufficiently purified by the manufacturers and the
impure mixture may be giving shadowing sequences (Fig. 7). In
both cases, the use of newly synthesized primers is advised. There is
a possibility that there is heterozygosity in the form of single
nucleotide polymorphisms (SNPs) in which case cross-referencing
with the sequence of the complementary strand is strongly advised
for confirmation.
3.10.4 Failed Sequence When the run fails to produce sequencing data, the probable causes
might be the suitability of the sequencing primer (Fig. 8), the
freshness of the primers stock, the quantity and quality of DNA,
and the presence inhibitors within template DNA. It is advisable to
repeat reaction first with a fresh aliquot of sequencing prime,
measure the concentration of template DNA, and ensure that a
Sanger Sequencing 209
3.10.5 Secondary In some cases, sequence may start well but the signal may weaken
Structure rapidly (Fig. 9). This is due to repeat regions such as GT or CT
repeats that may cause the sequencing signal to be depleted due to
formation of DNA loops or secondary structures. It is advisable to
add 1 μL of DMSO in the reaction or design nested sequencing
primers close to the secondary structure.
4 Notes
3. Entry level gel documentation systems that allow for the use of
safe gel staining dyes are sufficient for this protocol.
4. It is recommended to use an Applied Biosystems™ thermal
cyclers as Big Dye™ chemistry kits are optimized with the use
of these instruments. Alternatively, cycle sequencing reaction
optimization might be necessary.
5. The PC used for data visualization and storage in this protocol
should be run on Windows™ 8 or 10 operation systems.
6. A wide range ladder should be used (100–10,000 bp) as the
PCR products may vary in size.
7. Premade loading dye (6X Loading Dye) in order to ensure
consistency during loading is recommended.
8. For staining of DNA in agarose we prefer to use dyes that are
safer than ethidium bromide such as SYBRSafe™ and
Noveljuice™.
9. Before the use of PE Buffer, add the appropriate
(as recommended on the bottle) amount of ethanol
(96–100%).
10. Fragments between 500b and 4 Kb are not affected by the
addition of isopropanol.
11. The cartridge can be stored up to 4 months on or off the
instrument. If the cartridge is stored off the instrument, it
should be kept at 2–8 °C and in an up-right position with an
integrated capillary protector and an optical cover installed.
References
1. Saiki RK, Scharf S, Faloona FA, Mullis KB, 2. Bevan SI, Rapley R, Walker MR (1992)
Horn CT, Erlich HA, Amheim N (1985) Enzy- Sequencing of PCR amplified DNA. PCR
matic amplification of β-globin genomic Methods Appl 1:222–228
sequences and restriction site analysis for diag-
nosis of sickle cell anemia. Science 230:1350–
1354
Sanger Sequencing 211
Abstract
Long read Nanopore sequencing can be utilised to determine the quality and accuracy of genetically
engineered changes in animals, which often produce heterogenous samples. The protocol presented in
this chapter can be used for a range of both low and high throughput sequencing applications. DNA must
be repaired, barcoded and ligated to sequencing adapters prior to sequencing. Quality of sequencing data
produced is dependent on stringent adherence to the protocol. However, nanopore sequencing is a fast
moving field, therefore it is worth considering using the most up to date chemistry available.
1 Introduction
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_16,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
213
214 Angela H. Beckett and Samuel C. Robson
example, if the smallest fragment on the gel is from Pool 1(A), and
the second smallest fragment is from Pool 2 (C), the first nucleotide
in the reverse complement of the sequence is G, the second is T and
so on. In recent years, this process has been automated through the
use of capillary electrophoresis and base-specific fluorescent tags.
Second- and third-generation sequencing technologies have
since been developed, such as short- and long-read high-through-
put sequencing methods developed by Illumina and Oxford Nano-
pore Technologies (ONT), respectively [5]. The increased
throughput, speed and quality of these technologies have led
them to become widely adopted. Illumina utilises a bridge amplifi-
cation technique; a sequencing adapter is bound to the end of DNA
fragments, which are complementary to a lawn of oligos bound to
the glass surface of the Flow Cell. Polymerase creates a complement
of the hybridised strands, after which the double strand is dena-
tured and the original template is washed away. The remaining
strand folds over and binds to a second type of oligo bound to
the Flow Cell surface forming a bridge, where it is replicated by
polymerase and denatured into single strands (Bridge Amplifica-
tion). The replication is repeated through multiple cycles, creating
clusters of identical DNA oligos. Fluorescently tagged nucleotides
are added one by one, with the signal released after each addition
indicating which base has been incorporated. This occurs across all
fragments on the Flow Cell simultaneously. Emitted fluorescence is
detected, and the sequence for forward strands is calculated, after
which the reverse strands are transcribed and the process is
repeated. This massively parallel system greatly sped up sequencing
efforts. However, this method is limited as it only generates short
sequencing reads, thus resulting in potential gaps in the aligned
sequence or difficulty resolving highly repetitive regions. This limi-
tation has been addressed in third-generation long read sequenc-
ing, including nanopore-based sequencing from Oxford nanopore
Technology (ONT) and Single Molecule Real-Time (SMRT)
approaches from Pacific Biosciences (PacBio), capable of sequenc-
ing long fragments of DNA.
In this chapter, the methodology for library preparation
and sequencing using nanopore sequencing is described. ONT
have developed a small sequencing machine (MinION), roughly
the size of a standard stapler, which is portable and inexpensive
(<£1000). Researchers have taken the MinION to the Arctic to
perform field experiments [6] as well as using it on the international
space station [7, 8]. Despite the low initial cost of this instrument,
it is important to consider the cost of the sequencing consumables
and reagents. Each experiment is performed on a disposable Flow
Cell, depending on the number of samples sequenced and the
duration of sequencing these can be used for a maximum of 72 h.
The Flow Cell contains over 1000 nanopores, which are
1 nanometre-wide transmembrane proteins perforating an electro-
conductive membrane. DNA is bound to the pore by adapters and
NGS 215
dsDNA
Helicase
cis (-) enzyme
Protein pore
Lipid bilayer
trans (+)
ssDNA
Electrical current
Base-called data
T G A C
Fig. 1 A schematic diagram of the mechanism of Oxford Nanopore Technologies (ONT) sequencing. Double-
stranded DNA (dsDNA) is unwound by a helicase enzyme, and a single strand of DNA (ssDNA) is translocated
through the nanopore. Each nucleotide base causes a characteristic change in the ionic flow through the
nanopore, which can be computationally decoded to determine the underlying DNA sequence. (Figure created
using BioRender.com)
Fig. 2 Process Flow Chart and Timeline. Approximate timings of library preparation and sequencing. The
process can be expedited by increasing the number of lab workers during the End Repair and Barcoding steps.
(Figure created using BioRender.com)
2 Materials
2.1 Consumables 1. 2.5 μL, 10 μL, 20 μL, 200 μL and 1000 μL Filter tips, Sterile.
2. 96-Well Reaction Plate, half-skirt and adhesive PCR plate seals.
3. Flat 8-cap strips.
4. 0.5 mL Screw Cap Tubes.
5. Invitrogen™ Qubit™ Assay Tubes.
6. 1.5 mL DNA Lo-Bind Tube.
6. Microcentrifuge.
7. Vortexer.
8. Flow Cell MIN 109D (ONT).
9. 96-well cooling block.
10. Centrifuge tube Mini-cooler.
11. ONT Sequencer (see Note 2).
12. Magnetic Rack (see Note 3).
13. Timer.
14. P2, P10, P20/P100, P200, P1000 Single Channel Pipette.
15. 8x P10 Multichannel Pipette.
16. Repeat Pipettor (see Note 4).
3 Method
3.2 Post-PCR Quality 1. Quantify DNA from all samples, the positive control and the
Control NFW negative control using the Qubit Fluorometer (ng/μL).
2. Label the required number of Qubit, 0.5 mL tubes to test the
samples, including two additional tubes for the calibration
standards (see Note 7).
NGS 219
Table 1
Mastermix 1 reagent volume per sample
3.5 Barcode Clean- 1. In a new 1.5 mL DNA Lo-bind Eppendorf tube, pool all 10 μL
Up barcoding reactions together (per library, e.g., 960 μL for
96-barcoded samples).
NGS 221
Diluted
sample MM1 MM2
01 09 01 09 01 09
02 0 02 10 02 10
03 11 03 11 03 11
04 12 04 12 04 12
05 13 05 13 05 13
06 14 06 14 06 14
07 15 07 15 07 15
08 16 08 16 08 16
12.0 µl 0.75 µl
Fig. 3 Example 96-well plate layout for 16 samples. As indicated by arrows, 12 μL of diluted sample DNA
(containing 400 ng or 130 ng) with a total volume of 15 μl from samples 01–08 can be transferred to
Mastermix 1 (diluted sample from Column 1: A1-H1 transported to Column 3: A3-H3) simultaneously using an
8x P10 Multichannel pipette. After Mastermix 1 has finished incubation and Mastermix 2 has been added to
the plate, 0.75 μl of Mastermix 1 can be transferred by multichannel to Mastermix 2 (Column 3: A3-H3
transported to Column 5: A5:H5). (Figure created using BioRender.com)
Table 2
Mastermix 2 reagent volumes. ×24-×96 sample volumes have overage included
Table 3
Adapter ligation mix
Table 4
Sequencing reagents required to prepare library for loading onto flow cell
Kit Reagent
Ligation Sequencing Kit Sequencing Buffer
Ligation Sequencing Kit Loading Beads
Flow Cell Priming Kit Flush Buffer (FB)
Flow Cell Priming Kit Flush Tether (FLT)
NGS 225
5. Load Flow Cell into the instrument by sliding it under the clip
and pushing down gently.
6. If using Flow Cell containing storage buffer (first use Flow
Cells and Flow Cells which have been stored after the second
use), go to the “Start” menu and run a “Flow Cell Check.” A
Flow Cell check takes ~20 min, so continue with the next step
while waiting for it to complete. The number of pores available
will display on the “sequencing overview” screen. Any value
over 800 pores passes the ONT warranty threshold. If the
number of available pores is below 800 on a new Flow Cell,
and it is less than 3 months old; set it aside, inform ONT and
request a replacement (see Note 36).
7. Meanwhile, add 30 μL FLT to the FB tube and mix well by
vortex to create Flush buffer-mix.
8. Once the Flow Cell check is complete, open the priming port
and take a P1000 pipette with the volume to 800 μL. Holding
the pipette completely upright, place the empty pipette tip into
the priming port and remove any air from the inlet port by
turning the volume dial anti-clockwise (Fig. 5). Remove
~30 μL of the buffer from the Flow Cell (see note 37).
9. Load 800 μL of Flush buffer-mix into the Flow Cell via the
priming port (Fig. 5), dispense slowly and smoothly to mini-
mise shear forces on the library and avoid the introduction of
any air bubbles (Fig. 5) (see note 38).
10. Wait for 5 min, in the meantime move to the next step.
1. Slide open the priming port cover by 2. Place tip vertically into priming port and 3. Load 800µl of Flush Buffer-mix slowly
90˚ to reveal the priming port dial anticlockwise to remove air bubble through the priming port
4. After 5 minutes, open the SpotON 5. Load 200µl of Flush Buffer-mix slowly 6. Load the library dropwise into the
port through the priming port SpotON port
Fig. 5 Priming and loading a Flow Cell. Flow Cell is primed with flush buffer-mix prior to the library being
loaded via the SpotON port. (Image adapted from © 2022 Oxford Nanopore Technologies plc)
226 Angela H. Beckett and Samuel C. Robson
Table 5
Sequencing reagent quantities required to prepare library for loading onto
flow cell
Fig. 6 Flow Cell health and duty time plot. X2 shows a low number of sequencing pores despite there being a
high number of available pores, likely caused by insufficient adapter ligation. X1 shows a healthy Flow Cell
run, with a high proportion of available pores being utilised for sequencing. (Figure created using BioRender.
com)
4 Notes
use sequential barcodes for the next experiment so that they are
used evenly and do not result in cross-contamination
between runs.
2. MinION, GridION or PromethION. GridION and Pro-
methION have an inbuilt computer, whilst the MinION
requires a computer.
3. It is recommended to use the Invitrogen™ DynaMag™-2
Magnet. This magnet makes it easier to perform high-quality
washes of DNA.
4. These pipettes are optional and are worth purchasing if high-
throughput testing is to be performed.
5. ONT reagents are packaged as 6 libraries per tube, do not
sub-aliquot these. NEB reagents come packaged in larger
volumes, sub-aliquot into smaller volumes. If possible, aliquot
into single-use volumes. This prevents cross-contamination
events. NFW should be dispensed into a single-use aliquot,
and not reused. Whenever an aliquot has been used and put
back for future use, mark the lid with a pen so that if a contam-
ination event occurs, pre-used tubes can be destroyed. Give
each set of reagent aliquots a unique ID so that if a contamina-
tion event occurs, the causative reagent can be identified and
destroyed. Use seperate PCR hoods for aliquoting reagents and
performing library preparation.
6. Wearing a separate lab coat for library prep will reduce the
likelihood of contamination when the PCR for sequential
experiments is performed.
7. Only use the specific Qubit tubes which are of desired thick-
ness. Do not label the side of the tube as this could interfere
with the sample read.
8. The final volume in each tube should be 200 μL. Use a P2
pipette for adding 1 μL for high accuracy, when pipetting such
small volumes, it is best practice to break the surface tension in
the tip before use. Pipette the volume of liquid required and
expel to the second stop to break the surface tension, prior to
pipetting the desired volume for use in the process. Holding
the pipette upright at a 90° angle, whilst drawing up liquid into
the pipette tip, will produce more accurate results than holding
at a 45° angle.
9. Do not centrifuge the tubes, they are thin-walled and will break
under centrifugal force.
10. Calibrate the instrument with the standards at the start of the
week or if it has been used for another type of assay (such as
RNA). It does not need to be calibrated before each use.
11. Value is usually ~30–50.
12. Value is usually ~1600–1900.
NGS 229
13. If the PCR positive control value contains >20 ng/μl and
NFW is <20 ng/μl proceed with experiment, otherwise halt
experiment. If the sample contains <20 ng/μl but the PCR
positive control is >20 ng/μl, test another sample. It may be
that one sample had a low starting concentration. If many of
the samples contain <20 ng/μl and the positive control con-
tains >20 ng/μl, this suggests samples prior to PCR were low
quality and may require clean-up. If the positive control is also
low, this suggests an issue with the PCR itself. When a PCR has
been performed, the negative control often looks higher than it
would without PCR, as primer-dimers can cause a signal. This
can be checked by performing gel electrophoresis or TapeSta-
tion analysis on the negative control. Primer-dimers appear as
smears on the gel but contamination will show as a band the
size of the PCR amplicon. In some instances, low levels of
contamination are masked and cannot be detected until
sequenced. However, if there has been a catastrophic failure,
such as a sample mix-up in which DNA was added to the NFW,
this will be detectable. For a 25 μL PCR reaction pool, the
Qubit quantification should be <20 ng/μL.
14. When 230/260 and 260/280 values are outside of the pre-
scribed range this indicates that there are inhibiting substances
carried over from the PCR protocol. Clean-up DNA by: bind-
ing to AMPure beads at a 2× vol ratio, washing with 80%
ethanol and resuspending in TE buffer/NFW using the same
methodology as the bead washing steps throughout the proto-
col. Retest concentration and purity after clean-up. It is also
possible to use spin-column kits such as Qiagen QIAquick PCR
Purification Kit.
15. A 130 ng input is based on 200 fmol of 1 Kb fragment length.
This quantity can be adjusted based on the fragment length of
the amplicon. If 130 ng of DNA contains larger 5Kb frag-
ments, there will be less fragments which need end-repair
than if 130 ng contains many smaller 1Kb fragments. Thus,
to ensure that there is sufficient enzyme to perform end-repair
on all DNA fragments, the fmol concentration must be taken
into account. The fmol input can be calculated as such:
First: moles dsDNA (mol) = mass of dsDNA (g)/((length
of dsDNA (bp) × 617.96 g/mol/bp) + 36.04 g/mol).
Second: moles of dsDNA ends = moles dsDNA (mol) × 2.
Third: DNA copy number = moles of dsDNA × 6.022e23
molecules/mol.
This can be more easily calculated using online biocalcula-
tors. The amplicon length can be confirmed using gel electro-
phoresis, or for a more sensitive result a Tapestation or
Bioanalyser. Furthermore, 150 ng of DNA is diluted to a
total volume of 15 μl, so that in the next step, when 12 μl is
230 Angela H. Beckett and Samuel C. Robson
26. Once opened ethanol will absorb water and evaporate, reduc-
ing the ethanol content over time. When the ethanol concen-
tration reduces, the water concentration increases and removes
DNA from the beads during the wash step. Therefore, it is
essential that Ethanol dilutions are prepared fresh daily.
27. Smaller experiments (<15 samples per tube) often need longer
than 2 min to form the bead pellet. Wait until the liquid in tube
is completely clear before proceeding to the next step. This can
take up to 5 min.
28. A lot of care must be taken at this step. If the supernatant is
removed too quickly, it can pull DNA off the beads. Use a
P200 to remove the supernatant, set the volume to half the
total volume in the tube. That is, if there is 300 μL in the tube,
set the pipette to 150 μL so that supernatant can be removed in
a slow and controlled fashion. Keep the pipette tip angled away
from the beads and just below the meniscus of the supernatant;
pipette as slowly as possible; move pipette tip down the tube as
more volume is removed. Before discarding the supernatant,
check the liquid in the pipette tip for beads. If there is any
discoloration indicating the presence of beads, return the
supernatant back to the tube, wait for beads to pellet onto
magnet, and repeat. If it is not possible to remove the last
50 μL without accidentally removing beads, use a P20 or P10
pipette. Holding the magnetic rack at an angle can make it
easier to visualise the beads and the pipetting (Fig. 4).
29. Use 0.5–20 μL pipette tips with a P10 pipette. The longer tip
length allows for greater dexterity within the tube, making it
easier to avoid the bead pellet. For larger experiments with
higher sample numbers and thus more beads, the incubation
time will need to be extended up to 3 min. Do not allow the
pellet to dry completely; it will crack and become difficult to
re-suspend.
30. Use NFW if immediately continuing protocol. If leaving over-
night before proceeding, use Elution Buffer or Tris-EDTA
Buffer pH 8.0. Tris-EDTA and Elution buffer stabilise the
DNA for storage. This is not required if immediately using
the eluate for the next steps of the protocol. This step is a
good place to stop if there is not enough time to continue
the rest of the library preparation. Throw away the tube con-
taining 70% ethanol so that it is not accidentally used in later
steps.
31. For greater precision, when transferring eluate, transfer
3 × 10 μL with a P10 pipette to avoid accidental transfer of
beads.
232 Angela H. Beckett and Samuel C. Robson
40. Once mixed with loading beads and sequencing buffer, the
library must be used immediately and cannot be stored for
use at another time. Always ensure the Flow Cell check has
been successfully completed before performing this step.
41. Steady pipette by resting the shaft of the pipette against the
back of hand. When dispensing, the buffer may bubble out of
the Spot-on port. If this occurs, pause or slow down dispensing
to prevent buffer overflow. Do not raise thumb on the pipette,
thus drawing liquid back up into the pipette. If the buffer is
removed from the sensor array and air is introduced, damage to
Flow Cell pores will be irreparable. Pores in contact with air
bubbles will perish and become inactive; this will be indicated
in the low number of available pores reported in the sequenc-
ing overview.
42. Do not pipette directly into the SpotON port as this will
introduce air and damage the sensor array. Sometimes when
the Flow Cell has been used before, the SpotON port can
become blocked with dried precipitate from the last library.
Try carefully re-opening and closing the port, or if this fails
carefully chip away the precipate from around the port using a
small sterile pipette tip, being careful not to touch the sensor
array. If this does not resolve the issue then a new Flow Cell will
need to be used.
43. Label experiment as desired with an easily identifiable naming
scheme to track sequencing runs. We recommend prepending
IDs with the run date for simple chronological ordering (e.g.,
20201120_Experiment001_Run001).
44. Ensure there is enough disk space on the computer for the run
to complete. Delete previous runs as required. If there is not
enough disk space, sequencing will automatically stop. If this
occurs, delete previous files and then restart sequencing with a
new experiment ID. Data from both runs can be combined
during analysis by the bioinformatician. If the Flow Cell has
been sat inactive for a long time, restarting the run on the same
Flow Cell may not produce any data. If there is any library
remaining, prepare a new library for sequencing and run on a
fresh Flow Cell.
45. Wait 5 min before checking the run (at the start of the run the
Flow Cell health is loading and is not accurate). A common
issue seen at this stage is that there is a low number of pores
being utilised for sequencing despite there being a large num-
ber available (Fig. 6). This is usually caused by an error during
the adapter ligation steps. If the adapter has been insufficiently
ligated or missed in error, despite there being a high concen-
tration of DNA in the second quantification, the DNA will not
be transported through the pores. If the DNA is not being
transported through the pores, a low quantity of data will be
234 Angela H. Beckett and Samuel C. Robson
produced and the Flow Cell Health will rapidly decline as pores
perish when they are not being utilised. Library preparation
will have to be repeated and loaded onto a new Flow Cell. If the
Flow Cell Health rapidly drops to show mostly recovering and
inactive pores (indicated in blue – Fig. 6), this suggests insuffi-
cient quantity of DNA being added.
46. After the run has finished, it is normal to see small condensa-
tion bubbles above the sensor array.
Acknowledgments
References
1. Piccinni M, Sharpe C, Guille M (2019) Using 7. Castro-Wallace SL, Chiu CY, John KK et al
HDR and a template to introduce an in-frame (2017) Nanopore DNA sequencing and
HA tag on the 3′ end of the Xenopus laevis genome assembly on the international Space
gata2.L open reading frame. microPubl Biol Station. Sci Rep 7. https://doi.org/10.1038/
2019. https://doi.org/10.17912/MICRO S41598-017-18364-0
PUB.BIOLOGY.000170 8. Stahl-Rommel S, Jain M, Nguyen HN et al
2. Ran F, Hsu P, Wright J et al (2013) Genome (2021) Real-time culture-independent micro-
engineering using the CRISPR-Cas9 system. bial profiling onboard the international Space
Nat Protoc 8:2281–2308. https://doi.org/ Station using Nanopore sequencing. Genes 12:
10.1038/NPROT.2013.143 1 – 1 8 . h t t p s : // d o i . o r g / 1 0 . 3 3 9 0 /
3. Koch B, Nijmeijer B, Kueblbeck M et al (2018) GENES12010106
Generation and validation of homozygous 9. Korber B, Fischer WM, Gnanakaran S et al
fluorescent knock-in cells using CRISPR/ (2020) Tracking changes in SARS-CoV-
Cas9 genome editing. Nat Protoc 13:1465. 2 spike: evidence that D614G increases infec-
https://doi.org/10.1038/NPROT.2018.042 tivity of the COVID-19 virus. Cell 182:812.
4. Furtmann N, Schneider M, Spindler N et al https://doi.org/10.1016/J.CELL.2020.
(2021) An end-to-end automated platform 06.043
process for high-throughput engineering of 10. du Plessis L, McCrone JT, Zarebski AE et al
next-generation multi-specific antibody thera- (2021) Establishment and lineage dynamics of
peutics. MAbs 13:1955433. https://doi.org/ the SARS-CoV-2 epidemic in the UK. Science
10.1080/19420862.2021.1955433 (New York, NY) 371:708–712. https://doi.
5. Goodwin S, McPherson JD, McCombie WR org/10.1126/SCIENCE.ABF2946
(2016) Coming of age: ten years of next- 11. nCoV-2019 sequencing protocol v3 (LoCost).
generation sequencing technologies. Nat Rev https://www.protocols.io/view/ncov-2019-
Genet 17(6):333–351. https://doi.org/10. sequencing-protocol-v3-locost-bh42j8ye.
1038/nrg.2016.49 Accessed 15 Aug 2021
6. Goordial J, Altshuler I, Hindson K et al (2017) 12. Community – Protocol – Native barcoding
In situ field sequencing and life detection in genomic DNA (with EXP-NBD196 and
remote (79°26′N) Canadian high Arctic per- S Q K - L S K 1 0 9 ) . h t t p s : // c o m m u n i t y.
mafrost ice wedge microbial communities. nanoporetech.com/protocols/native-
Front Microbiol 8. https://doi.org/10. barcoding-96-gdna/v/nbe_9121_v109_
3389/FMICB.2017.02594 revb_19jan2021. Accessed 15 Aug 2021
Chapter 17
Abstract
Never before have we had the methods to edit or manipulate genes and genomes in the way we can today.
This volume has described technologies that, a generation ago, were unthinkable. But such power raises
broader issues on how we wield it. These concerns are relevant at a number of levels, from basic safety,
through the importance of scientific reproducibility and transparency, to Ethical, Philosophical, or even
Political considerations. But why should this matter to a laboratory scientist – surely results are the main aim
of experiments? However, such a view misses the important point that alongside our research generating
new information, experience gained conducting research can also be used for influence far beyond the lab
bench. Indeed, how research is conducted can be as important as the results themselves because our actions
also reflect and inform the values that society and us as individuals view as important. Rather than focusing
on the details of policy or legislation, this chapter therefore seeks to reflect on how Ethics, Legality, and
Safety impact the molecular biologist, and why researchers need to view these not as bureaucracy, but rather
as a critical part of their wider scientific identity and task.
1 Introduction
The words “Health & Safety” and “Ethics” often raise feelings of
frustration and consternation among laboratory scientists. Science
is hard enough without having to contend with bureaucracy,
administration, and often the perception that while having roles
that supposedly support research, others actively seem not to want
experiments to be conducted. This razor sharp focus on getting the
experiment done is entirely necessary for researchers to remain
motivated through frustrations and set-backs, but can obscure the
broader picture of why experiments are conducted in the first place.
The intricacies and complexity of biochemical systems mean that
although the relevance of a specific result is often limited, the
experience gained by the researcher, and the attitude and behavior
they demonstrate while conducting the work, can sometimes be as
important as the result itself. Society needs people who are able to
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4_17,
© The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature 2023
235
236 Simon E. Kolstoe
2 Research Integrity
get to this are simply not available to anyone outside the specific
research groups with many years of prior experience or preparatory
work. Add to this the complexities caused by variations in the purity
and availability of even simple consumables, and the pressures
created by limited funding and the constant push for novel results,
and it is really no surprise that many experiments or results cannot
legitimately be reproduced.
This situation is clearly regrettable, but there are only really
three courses of action that concerned scientists (and others within
the research system) can take. The first is to devote considerable
(and potentially up to 50% of available science funding) to repeat-
ing experiments. While such a course of action will definitely
improve the quality of the literature and thus the reliability of
existing knowledge, it is unlikely to produce the rapid results
needed (and expected) by the wider community that commissions
and funds research. The second response might be to acknowledge
the problem, but then just accept that this is a hazard of the
profession, and thus something any good researcher needs to take
into account when they rely on previous work to plan their new
experiments. Indeed many, perhaps more experienced, researchers
might be heard to comment anecdotally that they have always
known that there is a lot of “nonsense” in the literature. The
problem with this approach is that it is extremely wasteful, with
one famous study, commenting on medical research in particular,
reporting that “85% of research is wasted, usually because it asks the
wrong questions, is badly designed, not published or poorly reported”
[7]—such a figure is not helpful when trying to argue for increased
funding! As a consequence, the third, and only really practical
response to the reproducibility problem, is to pursue a middle
path of acknowledging the problem, acknowledging the reason
for the problem, and then seeking to conduct research in such a
way that minimizes waste wherever possible. Scientists should also
engage more broadly within both their research areas and also
wider academic communities supporting efforts to improve
research culture through education, and initiatives aimed at helping
improve the research process as a whole. Far from being a distrac-
tion, these activities are fundamental to the process of doing good
science and thus being a researcher.
Based on this argument it should be clear that the attitude of
researchers is vitally important for the production of good science.
A 2014 survey by the UK’s Nuffield Council on Bioethics (among
mostly life scientists) highlighted specific virtues (or character
traits) viewed as essential for the production of high-quality
research. These were being rigorous, accurate, original, honest,
and transparent, followed by supporting collaboration, multidisci-
plinarity, openness, and creativity [8]. Such traits can only really be
learned “on the job” so to speak, emphasizing the importance of
strong mentoring and good academic role models. Often only lip
Ethics and Legality 239
3 Research Governance
3.1 Health and Safety Health and Safety procedures should be familiar to anyone working
(Physical, Chemical, within a laboratory. Biosafety levels dictating containment arrange-
Biological) ments and are broadly consistent internationally. Biosafety Level
1 (BSL1) covers the handling of well-characterized agents that do
not cause disease in humans; Biosafety Level 2 (BSL2) covers the
handling of mildly infectious agents (to both humans or the envi-
ronment); Biosafety Level 3 (BSL3) covers the handling of highly
infectious and lethal agents, and Biosafety Level 4 (BSL4) is the
highest level for handling aerosol transmitted lethal agents. The
majority of genetic modification experiments can be routinely han-
dled at BSL1; however, if organisms are being created that can
potentially infect humans (most commonly using viral vectors),
BSL2 or above may be needed.
Every laboratory with a BSL categorization will confirm to
certain standards but should also have in place specific local risk
assessments that researchers need to be made aware of prior to
working in the laboratory. Local risk assessments will often also
cover other physical, chemical, and radiological risks that, while not
directly part of Biosafety, are often dictated by parallel standards. As
a consequence, local risk assessments and arrangements will often
refer to Biosafety Standards but then extend to also include the
various other (commonly nationally defined) standards that are
being addressed. Importantly, in order to meet such standards,
laboratories also often have to register with a regulator and allow
themselves to be open to both internal and external inspection so as
Ethics and Legality 241
3.2 Genetically Closely linked to the more general health and safety arrangements
Modified Organism described above, countries may also have specific legislation or
Legislation/Policy policies governing the laboratory handling of genetically modified
organisms in particular. Often containment arrangements for such
organisms closely parallel Biosafety Level arrangements wherein the
level of BSL is dictated by the type of genetic modification. Such an
alignment makes pragmatic sense as it allows for multiple proce-
dures, both genetic and biochemical, to be carried out within the
same laboratories and under safety arrangements that are suitable
for all such work. However, such legislation also often includes the
requirement for registration of experiments as countries or regions
seek to keep a record of the types and details of genetic modification
work that is being carried out. Such reporting requirements do vary
considerably between settings, but whereas general chemical/
biological/physical standards are ensured through regular inspec-
tions, genetic modification work may trigger additional reporting
or registration requirements. Here, it should be noted that experi-
ment using human tissue, especially human embryos, is often
highly regulated and subject to numerous laws and standards that
are legally enforced by various national regulators. Indeed the
majority of court cases and criminal convictions handed out to
scientists due to their research activities are within this area. As a
consequence all researchers wishing to work in this area should be
encouraged and ideally funded to attend specific courses relevant to
their regulatory context.
3.3 Ethics Policies The above two sections focus mainly on the practical elements of
laboratory work with the aim being to protect researchers and the
environment. Coupled to this there may be broader limitations on
the type of genetic work that can be carried out. Such limitations
are often defined nationally by statute and reflect each nation’s
political and social acceptance of genetic modification. Here inter-
national statements (such as UNESCO’s Universal Declaration on
Bioethics and Human Rights) are clearly influential, but how the
principles are translated into national law is mostly based on local
dialogue. Regulators may well be established to oversee legislation,
authorize experiments or laboratories, and conduct inspections
alongside (or as part of) Health and Safety regulation. This formal
process is often coupled with less formal arrangements dictated by
funders who may, for instance, prohibit certain types of working
being conducted using money from their funding schemes.
242 Simon E. Kolstoe
Similarly there are some funders who will not release funds to
institutions or organizations that conduct any work—even if
funded by others—that go against their principles. Such arrange-
ments are generally outlined clearly in guidance from regulators or
funders and then transferred into local policy in Universities or
other organizations seeking to conduct relevant work.
The above three legal and governance areas reflect processes
that can broadly be handled using administrative processes, be it
through inspection, standard operating procedures, or the audit of
policy requirements. Compliance with such processes is often not
negotiable from the individual scientist perspective. However, sci-
ence moves quickly and such processes may not always be sufficient,
or relevant, to newer discoveries or methodologies. In such cases
mechanisms need to be in place to discuss new situations and
arrange suitable oversite. This may be through updating regula-
tions or engaging with the wider research community to decide
what work is acceptable or not. Here independent review can
be key.
4 Independent Review
All scientists understand the value, but perhaps also weakness and
frustrations, of peer review. This is traditionally encountered at the
beginning and end of research through the review of grants sub-
mitted to funders, and then of manuscripts submitted to journals.
Independent review is also used during promotion and recruitment
processes. But such peer reviews can be frustrating as they are open
to abuse. However, if reviewers take seriously the virtues described
above in the researcher integrity section, they can add significant
value to the research process. This is because it is not easy to design
research, choose methodologies, analyze data, and then interpret-
ing results (let alone communicate them clearly). While increasingly
sophisticated guidance and even computational methods are avail-
able, sometimes there is no substitute for another scientist, with
sufficient experience and ideally a level of neutrality, reviewing
the work.
But within fields that conduct research with human participants
independent review also serves a second function beyond reviewing
scientific validity. Following the Second World War, the develop-
ment of the Nuremberg code and the subsequent 1964 Declaration
of Helsinki by the World Medical Association research ethics com-
mittees were established as central to any research involving human
participants:
The research protocol must be submitted for consideration, comment,
guidance and approval to the concerned research ethics committee before
the study begins. [9]
Ethics and Legality 243
5 Conclusion
References
1. Berg P (2008) Asilomar 1975: DNA modifica- 3. Cyranoski D (2020) What CRISPR-baby
tion secured. Nature 455:290–291. https:// prison sentences mean for research. Nature
www.nature.com/articles/455290a 577:154–155. https://www.nature.com/arti
2. Institute of Medicine (US) Committee to cles/d41586-020-00001-y
Study Decision Making (1991) Biomedical 4. Lander, E. S., Baylis F (2019) Feng Zhang et al
politics. In: Hanna KE (ed) Asilomar and Adopt a moratorium on heritable genome edit-
recombinant DNA: The end of the beginning. ing. Nature 567:165–168. Available from:
National Academies Press (US), Washington, https://doi.org/10.1038/d41586-019-
DC. Available from: https://www.ncbi.nlm. 00726-5. https://www.nature.com/articles/
nih.gov/books/NBK234217/ d41586-019-00726-5
Ethics and Legality 245
Garry Scarlett (ed.), DNA Manipulation and Analysis, Methods in Molecular Biology, vol. 2633,
https://doi.org/10.1007/978-1-0716-3004-4,
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer
Nature 2023
247
DNA MANIPULATION AND ANALYSIS
248 Index
In vitro ........................................................ 34, 68, 87–94, Plasmid
114, 132, 164–172, 174–178, 180–182 CloneJET................................................................. 160
In vivo .......................... 33–37, 39–43, 99, 111, 117, 128 pBlueScript ................................................................ 22
I-SceI .............................98, 99, 101, 102, 105, 107, 108 pBR322...................................................................... 22
pET-28b(+) .........................................................19, 21
K pGEM®-T Easy ..................................................56, 64
Katushka ................................................ 99–101, 104–106 pJET1.2 ................................................................... 160
pUC19.......................................................... 5, 22, 160
L Polyacrylamide gel electrophoresis (PAGE) ...............148,
152, 156, 213
Lambda-exonuclease............................................ 145–160 Polymerase chain reaction (PCR) ........................... v, 4, 6,
Legality ................................................................. 235–244 7, 9, 11–13, 15–19, 21–23, 25–30, 34–36, 38–43,
Ligase .......................................2, 6–9, 11, 13, 16, 19, 22, 46–53, 55–57, 60–65, 68, 71–73, 77, 81–94, 99,
45–47, 51, 56, 61, 98, 158, 217, 220, 221, 223 103, 107, 108, 113, 115, 118, 122, 128, 136,
Long single stranded DNA (lssDNA) ................ 135–137 137, 147–151, 154, 155, 158–160, 180,
195–210, 213, 217, 218, 228–230
M
PPE ............................................12, 15, 57, 99, 113, 116,
Melting temperature (Tm) ...............................19, 29, 42, 133, 148, 153, 198, 199
66, 67, 89, 93, 134, 137, 158, 201 Primer
Microarray ........................................... 164–177, 180–183 design.............................................. 20, 29, 34, 36–38,
Micro computed tomography (MicroCT) .................112, 42, 46, 48–50, 68–70, 75, 85, 90, 93, 178, 201
122, 124, 125 overlapping ............................. 48, 49, 66, 75, 76, 208
Microinjection .............................. 97, 114, 116, 119, 138 Promoter ........................................... 4, 5, 20, 31, 43, 75,
Multiple cloning site (MCS) ........................................ 1, 5 88, 98–101, 103, 104, 107, 118, 125, 167, 168,
Mutagenesis 177, 178
deletions...............................................................52, 87 Protein expression .........5–7, 14, 25, 125, 131, 164, 195
insertions .............................................................52, 87 pTransgenesis ..........................................................97–108
random ...................................................................... 81
site-directed (SDM) ......................34, 52, 78, 87, 195 R
substitutions ................................................. 52, 82, 87 Recombinant ........................................ v, vi, 1–23, 25, 98,
101, 104, 155, 158, 160, 195, 236
N Red fluorescent protein (RFP) ....................................... 40
Nonsense mutations ................................... 111, 125, 238 Restriction enzyme
blunt ends ..............................................................6, 20
O sticky ends...................................................4, 6, 20, 25
Reverse-phase HPLC (RP-HPLC) ....187, 188, 190–193
Oligonucleotide ....................................... v, vi, 49, 50, 52,
Ribosomal binding site (RBS)....................................4, 20
65, 66, 76, 78, 112, 113, 118, 126, 146–150,
Riboswitches................................................ 145, 146, 163
153, 155, 157, 158, 164, 166, 172, 176–178,
Ribozymes ................................................... 145, 146, 163
180, 185–193, 196
RNA ................................................. 5, 31, 113, 114, 116,
Oocytes ................................................................ 118, 119,
118, 120, 121, 128, 145–147, 163–183, 185, 228
132–134, 137–139, 141
RNAfold ...................................................... 157, 169, 174
Origin of replication (ori)...................................... 1, 5, 19
S
P
Safety.............................................................209, 235–244
PCR SAR-CH4 insulator................................................ 99, 101
colony ................................................. 4, 9, 18, 19, 23, SARS-CoV-2 ........................................................ 216, 226
34, 51, 64, 88, 91–93, 158, 160, 213 Secondary structure .......................... 20, 50, 89, 93, 120,
error-prone .......................................................... 81–86 147, 153, 155, 157, 166–169, 178, 201, 209, 210
Peer review .................................................................... 242 Selectable marker
Phenotyping ........................................112, 116, 122, 125 ampicillin ..........................5, 27, 48, 59, 64, 104, 107
Phosphoramidite chemistry...................................... v, 185 chloramphenicol..................................................27, 48
Phosphotungstic acid (PTA) ...................... 112, 116, 122 kanamycin .......................................13, 14, 21, 27, 30,
Phusion DNA polymerase ........................................47, 51 48, 59, 64, 103, 107
DNA MANIPULATION AND ANALYSIS
Index 249
Sequencing T
long read.................................................................. 214
low throughput ....................................................... 197 Taq DNA Polymerase .......................................12, 15, 51,
nanopores ....................................................... 214, 216 55, 56, 81, 82, 84, 85, 113, 115, 150
next generation .............................................. 160, 216 T4 DNA polymerase .................................................25–27
Sanger ............................................... 9, 11, 23, 31, 99, Thermodynamically balanced inside-out
121, 123, 127, 128, 158, 195–197, 213 (TBIO)...........................................................65–78
Single guide RNA (sgRNA) ............. 112, 113, 117, 118, Transformation..............................................2–4, 7, 9, 14,
120–122, 125–129, 135, 138 16, 18, 20–22, 29, 31, 34–36, 39–43, 47, 59,
Site-specific recombination........................................... 100 61–64, 88, 91, 93, 94, 98, 148, 155, 158–160
Snapgene Viewer .......................................................36, 48 Transgenesis ............................................................97–108
SOC broth ....................................................................... 14
U
Software
ABI Seqstudio ...............................197, 198, 204, 206 Untranslated region (UTR) ......................................... 117
ApE editor ................................................................. 50
OligoCalc...................................................... 20, 36, 42 V
SnapGene....................................................36, 48, 113
Vectors .................................................. 1, 4–9, 11, 13, 16,
Spectrophotometer ...........................................13, 17, 22,
19–23, 25–31, 34, 36–38, 41–43, 46, 48–50, 52,
27–29, 99, 102, 113, 120, 149, 150, 153, 155
55–59, 61, 62, 64, 68, 78, 87, 98–101, 103, 104,
Sperm nuclei ................................................ 132, 139, 141
106, 133, 136, 150, 158, 160, 240
ssDNA................................................ 116, 118, 119, 134,
137, 138, 146, 147, 149, 151–157, 159, 160, 215 X
Sticky ends .......................................................... 25, 43, 46
Streptavidin.................................................. 174, 175, 178 Xenopus
Synthetic biology ...................................... 45, 46, 66, 164 laevis.................................................... 57, 97, 99, 112,
Systematic evolution of ligands by exponential 115, 125, 127, 131–142
enrichment (SELEX)................................ 145–160 tropicalis................................................. 112, 115, 125
X-gal................................................................................... 9