A comprehensive review on CRISPR/Cas9
Replying to the comments of peer review:
1. Combinations of keywords have been used to avoid going into application based
studies and clinical trials. Data regarding one word keywords vs combined keywords
had been given in a previous assignment. The combinatorial keywords allow for more
speci c hits, keep the review focused on the technique and its major applications
rather than going awry. Hence, such complex keywords were used out of necessity
rather than choice given than the amount of literature that exists under the banner of
CRISPR/Cas9. Combination keywords have also been used because it was observed
that searches for a complex keyword retrieved hits which had partial identity to the
keyword. An example of this would be using the keyword Cas9 Gene editing. If a
speci c search for Cas9 gene editing is made, the review will pop up early due to
complete identity with the keyword. However, if a non-speci c search using the query
‘gene editing’ is made, the review will still pop up, albeit a bit later. This would not
happen in the non-speci c case if the keyword had just been Cas9 or gene editing.
Thus, using complex keywords would allow a speci c search to nd the review early,
yet will not completely disqualify the review from popping up due to partial identity
between keywords and the search query. The keywords were designed keeping that
cyber trick in mind, thus, they have not been changed.
2. No changes to the abstract have been made as the author feels it has enough
information already and the word limit must be strictly adhered to. The abstract was
kept playful intentionally. The technique is talked about as a genetic editing technique,
and the fact that it has variants is mentioned. Further, the criticism that the system of
CRISPR must be de ned seems a bit misplaced. CRISPR has no given system. It
works in many systems and stems from multiple bacteria. Even its discovery included
multiple systems. Hence de ning a basic CRISPR ‘system’ is not possible.
3. All full forms have already been mentioned in the text. Cpf1 has no full form. KRAB full
form has been added.
4. Speci c grammatical and syntax errors have been corrected.
5. More schematics have been added where appropriate. Explanations have been
provided in the gure legends as well.
6. APOBEC1 is APOBEC1. A search for APOBAC1 yields APOBEC1 as a suggestion by
google. It is thus a suggestion to the reviewer to please make such comments
carefully and after conducting proper research.
7. The manuscript has been kept as brief as possible. Nuances have been avoided
multiple times in order to not go completely away from the main point of the review.
fi
fi
fi
fi
fi
fi
fi
fi
fi
fi
1
Seeing as how the purpose of the review is to serve as a hub of all things related to
CRISPR, such length is unavoidable as the scope of the review is extremely wide.
Such a criticism is understandable given that in the basics of CRISPR section, a lot of
details about the functioning of the enzyme have been given, but given the fact that
the review wants the reader to understand CRISPR at a satisfactory, it is necessary
that such details are provided, otherwise, the reader might get a false sense of the
true exibility CRISPR/Cas9 application. Also, the details mentioned in this section
have been used in other sections to explain follow-up concepts, hence no details
have been removed.
8. A lot of subheadings have been used to cover the vast area under the banner of
CRISPR/Cas9 application. Each subheading covers a major application of the
technique. Thus, a reduction in their number would mean blatantly ignoring the
existence of a major part of the technique and its future, hence taking away from the
reason behind the review, which is to provide a ‘COMPREHENSIVE’ look at the
technique in its entirety. Thus, no sub-headings have been removed.
9. Finally, colourful fonts have been chosen in order to di erentiate between the various
kinds of headings. Major headings, section headings, section sub-headings, subheadings under section sub-headings, all have been provided a di erent colour so
that the reader is not lost when they read a heading. It is therefore a request to the
reviewer then to please understand the reason behind the colours and not just
criticise their use in general. Such colours also introduce visual variety in a text which
is long and contains many details. Colours have been accepted as a part of
schematics, it is time we adapt them into the main text as well, as a visual treat to the
eyes. Thus, keeping all that in mind, the colour scheme has been kept unchanged.
10. References were checked and were deemed appropriate. Citations in the main text
have been xed.
11. An author’s note: Most of this review has been written at the level of an
undergraduate student or a student who is currently beginning their graduation. It is
for this very reason that casual and playful language is used and nuances have been
avoided to keep the review from getting too technical and complicated. The review
expressly mentions where nuances have been avoided so that readers are not
completely blindsided. Thus, it aims to be more of a gentle read rather than a
technical scienti c text. It is therefore requested that the reader read through the
review in that eye rather than judging this text on the same merits as that of a formal
scienti c text. To achieve such a review has been a painstaking endeavour. A
balancing act of what to include and what not to include. Almost all major uses of the
technique have been covered and all major new developments have been covered as
well, thus the review ends up becoming a long read. To avoid having the reader go
through all of it at once, the review has been cleanly divided into three major sections.
The introduction lists what all each section includes so that the reader can jump
directly to that section and hence have quick access to the information that they
need. This is another reason that the colour scheme is being kept, so that the reader
knows when the jump to another major section is occurring. A mini conclusion has
been added as a cool down after the read.
ff
ff
fi
fi
fi
fl
2
CRISPR/Cas9 system: Origins, the present
scenario and beyond
By Mittu Udaat
In 2002, Ruud Jansen and colleagues coined the term CRISPR (Clustered Regularly
Interspaced Short Palindromic Repeats) for a sequence of direct repeats interspaced with
non-repetitive spacers. What followed in the next 2 decades has turned the CRISPR/Cas
system into one of the most exible and widely used techniques in Biological research
and has brought about a revolution in the eld of genetic editing. It has expanded far
beyond that with variants coming out every year which makes keeping track of the latest
developments of this tool di cult. The aim of this review thus, is to serve as a hub of
everything related to the CRISPR/Cas9 system, be it historical perspective, major variants
in use today or newer versions and improvements to the technique popping up every day.
Hence, anyone looking to familiarise themselves with the technique has quick access to
all things CRISPR via this review.
Keywords: Clustered regularly interspaced short palindromic repeats, CRISPRassociated proteins, CRISPR-Cas systems, Cas9 Gene editing, CRISPR-associated
protein 9, CRISPR
Introduction:
fi
fi
fi
fi
fi
fl
ffi
fl
fi
fi
CRISPR/Cas9 system is a technology that was developed by Jennifer Doudna and
Emmanuelle Charpentier in 2012 as a method of targeted gene editing. (18) Since its
development, this technology has created waves of scienti c applications and has
generated myriads of variants. (36) Such has been its impact that almost every Biological
science lab in the world seems to have adopted it in some manner. It is thus important for
any person who wants to conduct research in this eld to be aware of the existence of
this technique, have a general idea of its mechanism of function and possess some
knowledge of the many ways in which it is being applied in modern scienti c research. To
aid in that, this review tells the story of how CRISPR/Cas9 system was found and how it
was turned into a tool, how it functions (the molecular mechanism) and this is followed by
an exploration of the major applications of the technique today. As a treat for those who
are interested, a section discussing the future of CRISPR/Cas9 has been included which
discusses the latest phenomenons in the evolution of CRISPR/Cas9 technology. All of this
is underlined by a nal section which talks about the human side of science, the ethics of
using a gene editing technology. Thus, this review consists of three separate sections:
1. The Origins: A section which talks about the history of development of CRISPR/Cas9
2. The Present Scenario: A section which explains the mechanism of action of the
method and covers the major applications of CRISPR/Cas9 system.
3. Beyond: The nal section which talks about the interesting experiments that have
been conducted in order to push this technique even further. Articles have been
discussed which describe how major obstacles limiting the universal application of
this technology are being tackled. This section also discusses the ethics of use of
such a powerful and exible technology.
It is requested of the reader to pick a section of their choice from the three described
above based on what information they would like to absorb rst and what their purpose is
3
for reading this review. For that very reason, the sections have been kept somewhat stand
alone. With that said, let's dive into the rst section.
The Origins:
In this section, we take a look at the journey the scienti c community took in order to
create the CRISPR/Cas9 system. Step by step, we shall take a look at the landmark
discoveries that happened in the unbelievable ride that scientists experienced on the road
to developing the most exible and applicable biotechnological method ever.
History of CRISPR:
Finding the elusive repeats:
In 1987, as a part of an article completely unrelated to the repeats now known as
CRISPR, the rst instance of a sequence of repeats interspersed with spacers was
mentioned, that too as an afterthought in the ‘Discussion’ section. The article was about a
product of the iap gene in Escherichia coli and by accident, regions surrounding the iap
gene were sequenced. The discussion mentioned that the locus had regions of homology
spaced by 32 nucleotide spacers and that the biological function of such a construct was
‘not known’. (1) It was an obscure beginning for a tool that in a matter of a few decades
would go on to dominate the world of biotechnology. After the discovery of the repeats,
no major waves were made. The research article became part of an ocean of scienti c
literature, with a little pearl hidden in its written word. Fast forward now to 1993, in an
early article about spoligotyping for di erentiation of Mycobacterium tuberculosis strains,
a mention was made about a sequence of direct repeats with a length of 36 nucleotides.
(2) Again, no speci c follow up was made for these repeats.
As happens often in science, in the same year, in a completely unrelated scienti c study
about the alterations of genetic loci of a microorganism in conditions of varying salt
concentrations, Mojica, a graduate student rediscovered the same repeats in Haloferax
mediterranei and for the rst time, someone’s interest was sparked by these unknown
repeats. In 1993, he published an article describing these palindromic repeats of 30
nucleotides interspersed with short sequences and noted that these repeats were unlike
any other. Little did he know at that time, the gravity of the discovery he had made and
what a wild re his spark of interest would generate. In follow up works, he noted that
these repeats were present in multiple organisms, and by the year 2000, he had
characterised these repeats in almost 20 di erent organisms. (3)
fi
fi
fi
fi
ff
fi
ff
fi
fl
fi
fi
fi
By this time, thanks to the work of Mojica, a good number of scientists had taken note of
this sequence that was then called ‘Short regularly spaced repeats’ (SRSRs) and by the
year 2002, it had been discovered that these repeats were present in a large number of
organisms and there were dedicated genes (cas genes) present in their vicinity. (4) It was
in 2002, that on the suggestion of Mojica, Ruud Jansen and colleagues coined the term
‘Clustered regularly interspaced short palindromic repeats’ for this family of repeats. (4)
The article published by Jansen et al. was also one of the rst articles to capture the
structure of the entire CRISPR construct. (3) The structure of the CRISPR locus was
4
elucidated. It was time then to gure out, what do these repeats do and why are they
dispersed across so many organisms.
From repeats to the tool:
Spacers are of extrachromosomal origin We arrive at a point now where the various parts of the CRISPR loci have been found, yet
their function remains unknown. In 2002, it was still not known what the spacers between
the repeats were responsible for, or what they were homologous to. This mystery was
solved independently by three players, an old one, Mojica and the new ones Gilles
Vergnaud and Alexander Bolotin.
Mojica, in order to decipher the mystery behind the spacers, chose the most obvious
method. He picked up sequences of these spacers and used the bioinformatic tool
BLAST to search for similar sequences. It was in 2005 that he discovered that one of his
spacer sequences was similar to a P1 bacteriophage. The curious thing about the strain
of the organism with this spacer was that this very organism was also resistant to P1
bacteriophage. Thus, Mojica hypothesised that the loci might have functions similar to the
adaptive immune system. (5)
Independently of Mojica, Vergnaud studied the spacers present in Yersinia pestis and he
discovered that the spacers present in the CRISPR locus of the microbe were derived
from a bacteriophage and that these spacers were added in a directional manner such
that a new spacer was always added at the front end. (6) Thus, in this manner, two
individual scientists came to the conclusion that the CRISPR locus must be responsible
for immunity against incoming infections. It was aptly described as ‘memory of past
genetic aggressions’. The third player, Alexander Bolotin also arrived at the same
conclusion as the formers. What is notable about his work is that Bolotin was the rst
person to suggest a possible mechanism of the functioning of CRISPR locus. He
suggested that the spacers when transcribed, must work in an antisense inhibition
manner, by binding directly to bacteriophage RNA and preventing its proper functioning,
in a manner similar to RNAi (RNA interference). (7)
CRISPR is responsible for ‘adaptive’ immunity The fact that spacers in the CRISPR loci were of extrachromosomal origin and that they
were derived from bacteriophages immediately gave birth to the speculation that the
CRISPR locus might be involved in conferring adaptive immunity to a bacterial system. In
2007, a landmark article by Phillipe Horvath and colleagues proved beyond any doubt
that CRISPR in-fact led to development of adaptive immunity. Horvath and colleagues
used a bacteriophage sensitive Streptococcus thermophilus and two di erent
bacteriophages isolated from di erent samples of yogurt. In this article, they showed that
resistance to infection was acquired only when a corresponding spacer which was
identical to the bacteriophage was present inside the CRISPR locus. Another curious
discovery was that an increase in the number of the same spacers would lead to
increased resistance. The crux of the article however was that 100% identity between the
spacer and the bacteriophage genome was required for proper functioning.
Bacteriophages which displayed a single nucleotide polymorphism bypassed the CRISPR
adaptive immune system and thus killed the microbes. Similar results were observed for
fi
ff
ff
fi
5
spacers which displayed any genetic alterations. The same group also showed that a
protein of the Cascade region Csn1 (now Cas9) acted as a nuclease. (8)
Thus, by 2007, the exact function of the CRISPR locus was elucidated. The scienti c
community then aimed its e orts towards understanding the mechanism of CRISPR
adaptive immunity in its entirety by looking at it on the molecular level.
The bits and bobs of CRISPR based adaptive immunity In 2008, Van der Oost and colleagues looked to understand how various parts of the
CRISPR locus functioned. To do that, they took two Escherichia coli strains, one with a
CRISPR locus and one without and transferred the CRISPR locus from the one containing
the locus into the strain without one. Then, they knocked out each component of the
CRISPR locus one at a time and that led to the discovery of 5 proteins which are now
termed ‘Cascade’ proteins and which were named ‘cas’ for CRISPR associated. Cascade
proteins were found to be necessary for cleaving a precursor RNA transcribed from the
CRISPR locus to form crRNAs (CRISPR RNAs). The crRNAs consisted some of the last
nucleotides of the repeat region, the complete spacer and the bases at the beginning of
the repeat region which immediately followed the spacer. Such a construction led to
secondary structure formation due to complementarity between the repeat elements
since they are palindromic. The same group also went on to prove that crRNAs were
required for CRISPR based immunity by programming the crRNAs to target the lambda
phage. This was the rst time ever that anyone had arti cially programmed a CRISPR
locus to speci cally target a gene of interest. (9)
Even after all this information was available, it was still not clear whether CRISPR RNA
targeted RNA or DNA? According to Bolotin, RNA was the target of interest, but this was
refuted by a student by the name of Mara ni. (3) In 2008, Mara ni and Sonheimer used a
modi ed plasmid which had a self splicing region in a reporter gene in the plasmid. The
experiment was simple. If crRNAs targeted only RNA, no changes would be noticeable
since the given region of the reporter plasmid would be spliced out and the rest of the
mRNA would be targeted as is normal. But if crRNA attacked DNA, the extra region
inserted inside the reporter would be incompatible with the crRNA and thus, CRISPR
based immunity would not be observed. Using this method, they proved that crRNAs did
target DNA and that CRISPR system was basically a targeted DNA endonuclease. (10)
After all this, focus was shifted towards the main mechanism of CRISPR function. By
now, it was known that Cas9 functioned as a nuclease (8) and that crRNA targeted DNA.
(10) Garneau et al. followed up these studies and went on to then show that CRISPR/Cas
in Streptococcus thermophilus produced blunt ended double stranded cuts. The cuts
were quickly followed by a conserved sequence of 3 nucleotides called the protospacer
adjacent motif. (PAM) (11,12,13,14) This showed that the cuts were made precisely at the
regions of complementarity.
Another piece to the puzzle of CRISPR function was found by Charpentier and Vogel in
2012. In an independent experiment, they found that the third most expressed RNA in
Streptococcus pyogenes was a region from the CRISPR locus which would be called the
tracrRNA (Trans activating crRNA). This RNA had perfect complementarity to a region of
the crRNA. Thus, Vogel and Charpentier believed that this RNA might have something to
do with the functioning of the CRISPR system. They proved via knockout experiments,
that this region was necessary for the proper functioning of the system. (3)
fi
ffi
fi
ffi
ff
fi
fi
fi
6
By this point, the necessary components of the CRISPR system - crRNA, tracrRNA and
Cas9 - were known. It was time to see then how far this system could go.
Making of the tool The rst step in application of the CRISPR system was to see whether a CRISPR locus
from one organism could function normally in another organism. Siksnys and his group
proved this by transferring the CRISPR locus from Streptococcus thermophilus to
Escherichia coli and observing that the system functioned normally despite being in a
di erent microbe. (16) Siksnys then studied whether the same function would be possible
in vitro. He puri ed a streptavidin bound Cas9 and then showed that this Cas9 functioned
the same in vitro. In the same experiment, it was also shown that spacers in the CRISPR
locus can be manipulated in whichever way desired to target di erent sequences and the
system would still function the same. (17) This occurred in 2012.
For any of those who are familiar with the recent Nobel Prize (the year 2020) awarded for
designing the CRISPR/Cas9 technology, I am sure they would know that it was in this
very year that Jennifer Doudna and Emmanuelle Charpentier published the revolutionary
article which launched CRISPR/Cas9 as a tool of genetic editing. What Doudna and
Charpentier did di erently from the group of Siksnys was that they combined the crRNA
and tracrRNA into a singular construct and called it the single guide RNA (sgRNA). They
showed in vitro that this sgRNA alone was enough to allow Cas9 to perform its nuclease
activity. In the very same article, Doudna and Charpentier commented that this tool had
the potential to be exploited as a system for ‘RNA-programmable gene editing’. The
article therefore was aptly named,
“A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial
immunity” (18)
Figure-1 contains the timeline of the events covered in this section in brief along with
some major discoveries in recent years. At the end of 2012, the journey from the initial
discovery of the repeats to the construction of a functional tool was completed. It was
time then to utilise this tool and see how far it could be pushed.
Figure-1: A Timeline of CRISPR/Cas related discoveries
ff
ff
fi
fi
ff
Reprinted from Nidhi et al, Novel CRISPR–Cas Systems: An Updated Review of the Current Achievements, Applications, and
Future Research Perspectives (103)
7
The Present Scenario:
Basics of CRISPR/Cas9 system:
Before we dive into the myriads of applications of the CRISPR/Cas9 system, it is
important to possess a basic understanding of how it functions. It is better to understand
the adaptive immune system as a whole rather than just the technique because the more
knowledge one possesses, the more creative one can be. The functioning of the CRISPR/
Cas based adaptive immune system consists of the following steps1. Spacer acquisition
2. Biogenesis and maturation of CRISPR RNAs
3. Interference
Spacer Acquisition:
The CRISPR locus The rst step in the process is the capture of invading DNA and then adding it into the
CRISPR locus so that a crRNA can be produced to allow the machinery to target the
invader's genome. (19,20) The CRISPR locus is structured as follows - an AT-rich leader
sequence is present right before the CRISPR repeats. As the leader sequence ends, you
encounter the rst repeat which can range in size from 21-55bp, though usually, the size
is somewhere between 21-47bp. (21,22,23) This repeat is followed by a spacer of almost
the same length which can bind to the target, this is then immediately followed by another
repeat and in this manner, the repeat-spacer-repeat pattern continues. (22) This region
containing the leader sequences and repeats is anked by the CRISPR associated genes
(position varies from organism to organism) which code for the expression of Cas
proteins. (4) It is in this structure that a part of the invader’s genome must be added. The
invader need not be a pathogen. (10)
The process The process of addition of a new spacer is also called adaptation and the new spacer
which is added is called the protospacer. (21) It begins with the recognition of foreign
DNA. The di erence between host and foreign DNA (bacteriophage or plasmid) is chi
sites. Spacer acquisition requires AddAB (gram positive bacteria) or RecBCD system
(gram negative bacteria) to cleave regions of the invaders genome in order to insert it into
the CRISPR locus. These chi sites reduce the activity of RecBCD and AddAB proteins. It
must be intuitive then that these chi sites are highly enriched in the host genome as
compared to the invader’s. There is a bias then of the machinery towards cutting DNA
which has a lower amount of these sites. Another bias is produced by the presence of
free DNA ends. The bacterial chromosome is circular in nature and thus contains no free
ends of DNA, however, often, the genome of an invading bacteriophage is linear in nature,
which allows the cutting machinery to home in on these sites. (24,25,26)
The next step is to pick out a region of the invader’s genome which can serve as a
protospacer. This selection is made on the basis of the presence of a 3 nucleotide
fl
fi
ff
fi
8
sequence called the protospacer adjacent motif (PAM). Genomic regions which are
present immediately upstream of the PAM are picked up for use as a protospacer. The
PAM is an absolute necessity for the system to perform both spacer acquisition and
interference. The CRISPR locus of the host lacks this PAM and thus, there is no selftargeting. (19,21) The PAM recognising domain in Cas9 protein is responsible for
detecting it and thus, it aids in protospacer acquisition by cutting the region present
directly upstream of the PAM. The cleaved protospacer is then added into the CRISPR
locus at the front end, directly downstream of the leader sequence and used for
production of CRISPR RNAs. (19) This process can be di erent for di erent types of
CRISPR/Cas systems, for example, type-I systems use Cas1-Cas2 complex to perform
this function, as opposed to Cas9. (27,28) In type-I systems, spacer acquisition occurs at
a lower rate when there are no pre-existing spacers present in the CRISPR locus which
have either complete or partial homology to the invader’s DNA. In case a pre-existing
spacer exists, the rate of spacer acquisition is much faster. This is called ‘Primed spacer
acquisition’. (29,30)
Biogenesis of CRISPR RNAs:
The newly acquired spacer or pre-existent spacers are now transcribed to form the precrRNA. This long piece of RNA must be processed in order to convert it into a mature
crRNA which can be used to guide Cas proteins to perform interference. For the purpose
of maturation, exist proteins which di er from system to system. Cas6 protein is
responsible for maturation in type-I and type-III CRISPR/Cas systems. The Cas6 protein
cleaves the pre-crRNA in such a manner that a 5’ tag is present at the end of the process.
Subsequently, an unknown protein causes further shortening of the RNA and leads to the
production of a 3’ end which forms a secondary structure in the form of a loop. This
completes the maturation process. (31,32)
For type-II systems, the mechanism is entirely di erent which is what we shall cover here.
In type-II Cas systems (Cas9 is the interference protein), a tracrRNA is required. This
tracrRNA has a nucleotide sequence which is complementary to the repeat containing
region of the pre-crRNA. TracrRNA binds to pre-crRNA, an interaction which is stabilised
by Cas9. This tracrRNA:crRNA duplex is recognised by RNaseIII which cleaves this
duplex and forms an intermediate crRNA. The nal step in the conversion of intermediate
crRNA to mature crRNA remains unknown. (15)
After a mature crRNA is produced, the last step of the process is interference. In order to
keep the review brief, the interference mechanism of only CRISPR/Cas9 will be covered
as anything beyond it does not qualify under the scope of this review.
Interference:
In this stage, the mature crRNA acts as a guide for the Cas proteins to speci cally target
invader DNA in order to introduce double stranded breaks (DSBs) in it. (21)
In CRISPR/Cas9 based interference, the tracrRNA:crRNA duplex is responsible for
guiding the Cas9 protein to the target and introduce a DSB. Cas9 rst binds to the guide
RNA via direct interactions with the secondary structures formed by the RNA. (33) This
forms the structure necessary for search of target DNA. The detection of target DNA
occurs in a stepwise manner where the Cas9 rst searches via assessing for presence of
fi
ff
fi
ff
ff
fi
fi
ff
9
a PAM. Upon nding it, base complementarity between the crRNA and the target DNA
must occur for the recognition to be completed. In case the PAM sequence is not correct
or if the crRNA and the region anking the PAM are not complementary, Cas9 rapidly
dissociates away and no nicking takes place. However, if everything checks out, Cas9
dwells in that region for a long time and can conduct its endonuclease activity. (34)
When an interaction between a PAM and Cas9 occurs, local DNA structure is altered via
the introduction of a kink and turn, directly upstream of the PAM sequence. This kink and
turn of the target DNA ips a nucleotide of the non-target strand thus making the target
strand available for crRNA, hence facilitating interaction between the crRNA and the
target DNA. Base complementarity then takes over and in this manner, a duplex is formed
between the crRNA and a single strand of the target. (34) The displaced DNA strand of
the target (also known as R loop) is stabilised by a large number of van der Waals and
hydrophobic interactions. (35) The Cas9 is now ready to cleave the target DNA.
Cleaving of target DNA is performed by two domains of the Cas9 protein, the RuvC and
HNH nuclease domains. Each domain is responsible for cleaving one strand of the target
DNA 3 base pairs upstream of the PAM. Such a nicking process creates a blunt ended
DSB. (34) During the process of cleaving, the HNH and RuvC domains of Cas9
communicate with each other. After the R loop is formed, Cas9 undergoes a
conformational change which brings the HNH domain closer to the target DNA. The
conformational change in Cas9 causes crRNA strand invasion to move further. More base
pairing causes greater conformational changes till an active conformation of the HNH
domain is achieved. This active conformation in turn allosterically controls the RuvC
domain. HNH is connected to RuvC through two linkers (namely L1 and L2) which allow
for communication between them. These two linkers undergo large conformational
changes themselves and direct the non-target strand towards the catalytic region of the
RuvC domain. It is at this point that the Cas9 protein is completely active. The target
strand is cleaved by the HNH domain and the non-target strand, after being inserted into
the RuvC domain, is cleaved. Subsequent to the cleavage, Cas9 remains attached to its
target until some other factors remove the enzyme in order to recycle it. (34,35)
Figure-2: A schematic of CRISPR/Cas9 based interference
The process of CRISPR/Cas9 based activity has 3 major steps. First, the crRNA is transcribed from the CRISPR locus (called
biogenesis). Second, this newly transcribed RNA (called pre-crRNA) is cleaved in order to form a mature crRNA construct that
can interact with tracrRNA. This process of cleaving of pre-crRNA is called maturation. The mature crRNA and a tracrRNA
present inside a Cas9 protein interact to form the crRNA:tracrRNA duplex and thus the CRISPR/Cas9 construct is ready for
target recognition. The nal step of the process is recognition of the target genomic region by the guide RNA. The target is
recognised by nucleotide base complementarity between the crRNA and the target. After the target region is recognised in
such a manner, it is then cleaved by the Cas9 enzyme. This process is called interference. (Nuances of the process have been
mentioned in the main text of the review.)
Schematic created with BioRender.com
fl
fl
fi
fi
10
Major variants of CRISPR/Cas9 system in use
today:
In this section of the review, we shall go through the creative ways in which this technique
has been applied. CRISPR/Cas9 is exible enough that one is limited only by their own
imagination. Variants of CRISPR/Cas9 come under two major categories: Variants in gene
editing and variants which are used in functions other than gene editing.
Cas9 enzyme variants in gene editing:
As mentioned earlier, Cas9 enzymes assess a PAM in order to recognise their target and
start the base complementarity and interference process. Then, do all Cas9 enzymes
have the same PAM preference? The answer would be no. The fun of biology lies in the
inherent diversity and variations in systems across a wide range of organisms. As such,
there are Cas9 enzymes which have di erent PAM preferences. Not just that, Cas9
enzymes vary in their sizes and sgRNA requirements as well. (36,37) This creates for us a
large pool to sift through in order to nd a Cas9 enzyme which suits our requirements the
most. Let us look at these Cas9 enzymes rst. The various Cas9 enzymes in use today
have been mentioned in table-1. Such a large variety of Cas9 enzymes is in use today due
to various reasons ranging from compatibility to target organism (mostly PAM
compatibility), the size of the Cas9 enzyme, size of the guide RNA etc. (36) Factoring
these variables, the Cas9 currently in rst place as far as usage goes is SpCas9
(Streptococcus pyogenes). The obvious reason behind it is the elementary PAM
requirement - NGG - where N could be any nucleotide. This allows for more target
exibility as compared to other enzymes. fnCas9 does possess the same PAM
requirement, but the fact that it is larger in size as compared to spCas9 limits its usage.
Size matters because the CRISPR assembly needs to be delivered into the target
organism and a larger size means more problems in e cient transfer of the CRISPR/Cas
system into the target organisms. Although there are Cas9 enzymes which are smaller
than spCas9, it turns out that natural Cas9 enzymes which are smaller in size have more
complex PAM requirements, thus the game becomes a balancing act. A larger size means
more di culty in delivering the assembly, a smaller size means more PAM complexity.
One thus has to balance these trade-o s in order to pick the enzyme most suited for their
use. It is due to the aforementioned reasons then that spCas9 dominates the eld (36)
Cpf1 enzymes have been mentioned in the table as well since they allow for much more
e cient target cutting, are smaller in size as compared to most Cas9 proteins, require
only one guide RNA as compared to a tracrRNA:crRNA duplex, are responsible for
producing mature crRNA on their own and do not rely on any other enzyme for this
function. Beyond all that, they cut downstream of the PAM sequence and introduce
staggered cuts as compared to upstream and blunt ended cuts, which is what Cas9
fi
ffi
fi
fi
ff
ff
fi
fi
fl
ffi
11
ffi
fl
Figure-2 consists of a schematic which shows the major steps of the entire process.
Many nuances have been skipped in order to make the mechanism easier to understand
and to stay within the scope of the review. To understand more about the mechanism, it is
recommended that the reader follow up on the speci c references mentioned in this
section. With a basic understanding of how CRISPR/Cas9 works, we shall now turn the
discussion towards the applications of this revolutionary technology.
enzymes do. This makes them much more reliable for genetic editing as compared to
Cas9, yet these enzymes still have not overtaken Cas9 enzymes as far as mainstream use
is concerned. Some reasons for this are that the Cpf1 enzymes require tighter
temperature control in order to function properly and because the size of the guide RNA is
smaller, there are greater chances for secondary structure formation which may be
di erent from what is desired. Besides that, Cpf1 enzymes can be used for any function
that Cas9 enzymes can perform. Please keep that in mind as we explore the various
applications of the CRISPR/Cas9 system. (37,38,39,40)
This table lists the details of all the major Cas9 enzymes currently in use today. Two Cpf1 (now Cas12a)
enzymes have been mentioned as well since their impact cannot be entirely ignored. (36,37)
Terminology used N = Any nucleotide; R = Pyrimidines; W = Adenine or Thymine; V = Guanine or Cytosine or Adenine.
The idea behind Cas9 based gene editing -
ff
Cas9 via the use of a sgRNA can be used to target any region of the DNA. By now, it must
be clear that this statement is not completely accurate. Using Cas9 and a sgRNA, you
can target regions of the DNA which have the appropriate PAM following it or preceding it
(depending on whether you are using Cas9 or Cpf1). The manner in which Cas9 based
genome editing works is quite simple. The scientist relies on the ability of the tool to
create a DSB. Once this double strand break is created, it is detected by an organism’s
12
repair machinery. At this point, NHEJ (non homologous end joining) or HDR (Homologydirected repair) takes over. (20)
Each of these mechanisms has di erent usages. In case a scientist wants to produce a
knockout, NHEJ is preferred. NHEJ despite being a method of DSB repair for DNA, is
prone to either cause some deletions or random insertions. Introduction of either of these
leads to an alteration in the genetic code of DNA (based on codon triplets). Due to the
introduction of or removal of nucleotide bases, a frameshift occurs. This shift causes
alterations in the way the mRNA produced by this DNA fragment is read for translation. In
such a manner, a non-functional protein is produced. Thus, a gene is e ectively knocked
out. HDR is employed when you want to perform a targeted gene deletion, or to insert a
gene or a gene segment into the genome. This has been described in Figure-3. (20,41)
How to use the tool The following considerations must be made in order to use the tool:
1. Choice of target DNA- It is obvious that your target of choice should always be next
to a PAM, otherwise your experiment will not work. A useful tip here is to target a
region as early on in the coding region of the gene as possible. Doing this will
accentuate the e ect of deletions or insertions caused by the system. There will still
be a lot of trial and error as the design of your sgRNA will depend on not only the
target, but also the organism in which you are performing the experiment. (42,43)
2. sgRNA design - An sgRNA must contain the following 3 regions, a guide sequence
which is complementary to the target DNA, an intermediate region which resembles
the formation of a duplex between crRNA and tracrRNA and nally, it must contain a
tracrRNA region which folds to form secondary structures in a manner such that a
Cas9 enzyme can bind to the sgRNA. sgRNAs must be designed to target regions
close to the preferred PAM for your Cas9 enzyme. The promoter to be used to
express your sgRNA must be appropriate for your target organism (as in, whether
your system is a prokaryote or a eukaryote). It is also pertinent to mention here that
the GC content of your sgRNA should not be too low or too high. Further, repeated
nucleotides at a stretch have been shown to reduce the e ectiveness of the sgRNA.
In order to support you, there are multiple sgRNA design tools available online,
including one from Zhang lab. You can employ these to design a guide RNA as per
your speci cations. (20,44)
3. Delivery method - The nal step in the process is picking out what you will use to
deliver the CRISPR assembly inside your organism. Methods of delivery include
simple methods such as plasmid vectors which have the CRISPR assembly under the
action of inducible promoters in order to control the function of CRISPR/Cas system.
For experiments which go on for a while, it is better to knock in the CRISPR assembly
into the chromosome of the organism and create a stable cell line such that your
CRISPR locus is faithfully replicated in each generation. Such methods are
employable for gene knockout or knock in experiments. But, in case of gene therapy,
Adenovirus based systems are employed but this comes with a lot of complications
(for example immune responses generated by the adenovirus) and are beyond the
scope of this review. (45,46,47)
Now that we know how to apply the tool (at a basic level), it is time to start exploring its
applications in the scienti c world.
ff
fi
ff
ff
fi
fi
ff
fi
13
Applications of CRISPR/Cas9 in genome editing Because of its ease of use, this tool has been used extensively in reverse genetic studies
to understand the functions of genes (gene knockout experiments), understanding
diseases and in gene therapy as well. By using targeting via sgRNAs properly, genes can
be knocked out and corresponding changes in phenotypes can be studied. (36) Using
sgRNAs for targeted knock in for genes, one can produce transgenic organisms in order
to develop model organisms for study of diseases. This has been achieved in fungi (48),
plants (49) and a large number of animals as well. (50) CRISPR/Cas9 excels more than
other tools at such feats simply because of its ease of use. Applying this concept at a
wider range, a large number of sgRNAs have been used in a system at the same time to
achieve large scale alterations. An instance would introducing 2 DSBs in the same
segment of DNA so as to cause an inversion of the gene segment. The same method can
be used to cause translocations by producing DSBs in multiple segments. All this may
sound speculative, but these methods have been used in experiments already. Such
chromosomal alterations have been employed to study oncogenesis (51), disease
progression (52) and e ects of chromosomal translocations. (53) The same method can
then be applied to perform large scale screens to understand every aspect behind any
given biological event. This technique has also been used for gene therapy of genetic
disorders such as cystic brosis, and the infamous case of He Jiankui, a scientist who
edited genomes of human babies in order to make them less susceptible to HIV. These
applications are beyond the scope of this review, but are interesting reads. It is
encouraged therefore, that you go and read about these events. (20,54)
Figure-3: A schematic depicting CRISPR/Cas9 based gene editing
CRISPR/Cas9 based gene editing relies on the repair mechanisms inside organisms to
produce its effects. NHEJ is used to mainly disrupt or knock out your gene of interest as
this method of repair is error prone. HDR is used when you want to knock in a gene,
perform a targeted deletion or correct a gene of interest.
fi
ff
Schematic created with BioRender.com
14
CRISPR/Cas9 beyond gene editing:
It is beyond gene editing where CRISPR/Cas9 has left all other genome editing
techniques in the dust. We shall cover these applications one by one now.
Most of these techniques employ a catalytically dead cas9 enzyme (dCas9) which has
been arti cially engineered to remove its nuclease activity. This is achieved by introducing
targeted mutations in the HNH and RuvC domains of Cas9 enzyme in order to prevent
cleaving of target DNA. (36) Section by section, we will now explore the application of the
Cas9 enzyme beyond prototypical gene editing.
Control of gene expression dCas9 has been used beyond gene editing in creative ways. A dCas9 enzyme possesses
the ability to bind to target DNA but does not cleave it. This property has been employed
to cause interference in the genome to cause gene repression. This technique is named
CRISPRi for CRISPR interference. The concept behind it is that upon target recognition,
dCas9 binds to the target region. This binding prevents other enzymes from accessing
this region of the genome which leads to its silencing. The advantage of this technique is
that we can mix the inducible promoter trick mentioned earlier along with CRISPRi to
temporally control the silencing of a gene and when required, we can revert it to make the
gene functional. Such an application is important when studying genes which are
essential for cell development or growth. Thus, CRISPRi knocks down a gene instead of
knocking it out. Besides this, one can attach a repressor to a dCas9 and then target this
repressor to the gene of interest to silence it. An example of such a repressor are KRAB
domain containing proteins (Krüppel associated box containing protein). The addition of a
repressor enhances the silencing ability of dCas9 and allows for more accurate targeting.
(45,55)
Opposite to this, you can attach an activator to the dCas9 in order to enhance expression
of a protein. This methodology is called CRISPRa (CRISPR activation). To achieve this,
you can introduce a dedicated activator of a gene and couple it to dCas9. This will recruit
proteins to the target site thus inducing your gene of interest. Nowadays, this technique
has evolved to an unprecedented level. Instead of a single activator, scientists have used
activator complexes in order to achieve a higher level of induction. To provide an
example, Chavez et al. used a tripartite complex. In their article, VP64 was used as an
initial activator and a corresponding increase in transcriptional activity was noted.
Following that, they created bipartite fusions with the C-terminal domain of the dCas9VP64 complex, using either p65 or Rta. These double activator complexes were more
e cient in inducing the gene of interest as compared to the individual activator. The nal
step was to create a tripartite complex where both p65 and Rta were fused with the
dCas9-VP64 complex. This complex achieved the highest level of activation! (56)
In this manner, one can manipulate gene expression tightly using dCas9 according to
their will. It is possible to simultaneously introduce orthogonal activator and repressor
constructs attached to dCas9s under di erent inducible promoters to control expression
of a gene of interest and study its function.
fi
ff
fi
ffi
15
Shining a spotlight on chromosomes, live cell imaging with
Cas9 In this variation, instead of attaching an activator or repressor to the dCas9, one can
simply attach a uorescent protein to it and target any genetic locus of interest. By doing
this, you can view genomic structures live and understand their organisation. dCas9 has
not exactly revolutionised this methodology. TALEs (transcription activator-like e ectors),
that are another type of DNA targeting proteins, have been used before to perform similar
experiments, however, dCas9 being more exible, has overtaken other methods of live
cell imaging. Such a technique is a far cry from the days of uorescence in situ
hybridisation where a cell would have to be heated and xed before making observations.
Thus, a considerable jump has been achieved in live cell imaging using CRISPR/Cas9.
(57,58,59)
Alter chromatin architecture at your whim Large scale chromatin structure alterations can also be achieved using dCas9 tethered
with appropriate proteins. A study by Morgan et al. used dCas9 enzymes tethered to ABI1
or PYL1. They introduced plasmids with the dCas9-ABI1/PYL1 constructs in human bone
marrow cells and achieved large scale chromatin structure manipulation. The idea behind
the technique is that 2 di erent dCas9 enzymes are tethered to two di erent proteins,
ABI1 and PYL1. The trick is that these two proteins, which are bound to independent
dCas9s have the ability to bind to each other. So, when two of these constructs bind to
two di erent target DNA regions, the a nity between the proteins tethered to dCas9s will
cause them to bind, thus bringing target regions close to each other and altering the
structure of chromatin. In the same article, they also showed that simply altering the
chromatin structure is enough to induce expression of genes. They named the technique
‘reversible chromatin loop reorganisation using nuclease-de cient Cas9’ or CLOuD9 in
short. (60)
Controlling the epigenome Human DNA is packed tightly in order to t a large amount of genetic information into
minuscule spaces. This packing is performed by the presence of proteins called Histones.
DNA winds around histones and thus gets packed in tightly wound structures called
nucleosomes. Consequently, genes which are packed tightly are silenced since proteins
cannot reach them. In contrast, genes which are active are loosely packed to allow
various enzymes access to those regions in order to transcribe and express them. The
wondrous thing about nucleosomes is that by manipulating the modi cations of histones,
the tightness of packing of a certain region of the genome can be controlled. Dedicated
enzymes exist for these functions. Beyond that, DNA itself can be directly subject to
modi cations (such as CpG island methylations). (61)
You must get it by now. Dedicated enzymes for a given function? Existence of dCas9?
Flexible targeting using sgRNAs? This combination of factors thus allows us to tether
various epigenetic modi er enzymes such as Histone methylases, Histone acetylases,
Histone deacetylases etc. to dCas9 in order to manipulate the targeted epigenetic
landscape and thus, study the e ects of di erent epigenetic modi cations in a genetic
region. Today, many such epigenetic proteins have been tethered to dCas9 to study
di erent functions. (62,63,64)
ff
ff
fi
fi
fl
fi
fi
ff
fl
fi
ffi
ff
ff
fi
fl
ff
fi
ff
16
Split Cas9 A split Cas9 is an enzyme which has been literally split into two pieces. When split, the
Cas9 enzyme will be inactive. In order to activate it, a substrate is added which allows the
split Cas9 parts to interact and thus form a functional Cas9. At a deeper level, what needs
to be done is to attach dimerisation domains to pieces of the Cas9 enzyme. These
dimerisation domains can be substrate inducible like the one produced by Zetsche et al.
(65) who used rapamycin binding domains tethered to split Cas9s. Introduction of
rapamycin in the system activates the Cas9 by bringing the pieces of Cas9 together.
Nihongaki et al. instead of using substrate based domains, used photo inducible domains
called magnets which interact with each other only upon irradiation by blue light. (66) In
this manner, by keeping the Cas9 separated and bringing it together with a special
stimulus, greater spatiotemporal control of the system can be achieved.
It is important to mention here, for any of the techniques mentioned above, inducible
promoters which control the expression of Cas9 itself can be applied. By doing that, we
can introduce layers of control in our experiment. As a result, accuracy of the system can
be increased and o -target e ects of the same can be reduced dramatically!
All of the strategies mentioned above have been summarised in gure-4. With this, we
conclude our foray into the major variations of CRISPR/Cas9 techniques presently in use.
In the next section, we shall explore the future of this technique.
Figure-4: Various strategies of CRISPR/Cas9 application in use today
The gure has been divided into two parts. The two strategies on the left are DNA editing strategies. (Direct targeted
gene editing has been covered in the Basics of CRISPR section and Base editing, which has been explained in the future
of CRISPR section.) On the right are strategies which employ the use of a dCas9 enzyme and thus are the applications
which make use of the fact that the dCas9 enzyme can be targeted to a speci c genomic region using a sgRNA. Various
effector enzymes or molecules are attached to such a dCas9 enzyme and thus, they receive a ride via dCas9 to the
genomic region of interest. All of these strategies have been discussed in the CRISPR/Cas9 beyond gene editing
section. Please refer to each section to learn more about these strategies.
17
fi
fi
ff
ff
fi
Reprinted from Adli, M. (2018). The CRISPR tool kit for genome editing and beyond. In Nature Communications (Vol. 9, Issue 1). Nature Publishing
Group
Beyond:
The major variations and techniques under the domain of CRISPR/Cas9 have been
discussed. In this section, we move beyond the present and try to understand what lies in
the future for CRISPR/Cas9. We shall look into the latest developments in the eld and
understand the obstacles that are being tackled in order to evolve the technique further.
Finally, we shall cover the human side of science, the ethics of the application of such a
technique.
Future of CRISPR/Cas9:
Second generation CRISPR tools:
Second generation CRISPR tools signify a leap from random base alterations to targeted
base alterations. These tools tether a di erent type of enzyme to the dCas9 protein so
that bases can be edited. The dCas9 tethering to an enzyme trick is not exactly a new
idea, but by using an entirely di erent class of enzymes, one can alter bases in a genome
without introducing DSBs. Imagine the exibility this would provide! The routine Cas9
simply causes DSBs and it is the NHEJ repair machinery which then causes deletions or
insertions. Of course, this would be non-speci c. HDR could be an option then? But, it
turns out that even when HDR is supposed to happen, the cell can choose NHEJ based
repair because it is more e cient, leading to undesired results. But in the case of Second
generation CRISPR tools, without breaking the DNA at all, one possesses the ability to
edit DNA by making speci c and targeted base alterations. (36,67,68) This shall be
explained further.
Liu et al. create the rst CRISPR/Cas9 base editor In 2016, from the laboratory of Liu came out an article which described a paradigm shift
in the world of CRISPR/Cas9. What prompted their research was the information that
upon binding of Cas9 to the protospacer in the target DNA, an R loop is formed i.e.
ssDNA (single stranded DNA) is present in that location. Cytosine bases of a segment of
ssDNA which is exposed are prone to deamination by enzymes called cytidine
deaminases. Removal of an amino group by these enzymes converts a Cytosine base into
Uracil. This Uracil during the course of replication is then read as a Thymine. Thus, a
deamination event converts a C-G pair into a T-A pair. In their study, di erent cytidine
deaminases were tethered with dCas9. The highest e ciency was achieved by the rat
APOBEC1. The enzyme in vitro and in vivo (albeit at a lower e ciency) converted
Cytosines to Uracils which were replicated in progenies as a T-A pair. This was the rst
generation of Base editors (BE1) with lower in vivo e ciency rates because of the cellular
repair machinery which could detect the U-G mismatch pair formed after the deamination
and correct it. To avoid the cellular repair, an enzyme called Uracil DNA glycosylase
inhibitor was fused to the C terminal end of the BE1 complex. This construct was called
BE2. What this BE2 could accomplish was that it could inhibit the enzyme responsible for
the repair of the U-G mismatch. A higher e ciency was achieved in vivo with this base
editor. Liu and colleagues were still not satis ed however. The nal step in the evolution
fi
fi
ff
fi
ffi
ffi
ffi
fi
fi
ffi
ff
fl
ff
fi
ffi
fi
18
process was to restore some of the cleaving activity of dCas9 by reverting the mutations
in the HNH domain of dCas9 to form an enzyme called the nickase Cas9, which would
cleave only one strand of DNA instead of both. What this nickase Cas9 could do was to
bias the cellular repair machinery to use U as the template for repair of the U-G mismatch
pair instead of G. Thus, prompting the repair machinery itself to create the edit by nicking
the strand which contains the G. So, the outcome of this kind of repair would be a U-A
pair which would then become a T-A pair during replication. This was the rst instance of
targeted base editing using CRISPR/Cas9 without the introduction of DSBs. (69)
In the years since targeted base editing was achieved, a lot of modi cations have been
made to the base editors. An example would be Koblan et al. attaching nuclear
localisation signals to the base editor complex in order to improve nuclear localisation of
BEs and thus increase their e ciency. This led to enzymes called BE4max and
ancBE4max. (70) More cytidine deaminases have been found since then, with di erent
sequence preferences in order to widen the pool of targets which can be altered. (68)
Many developments and improvements have been made since then, a good amount
coming from Liu’s group. It is encouraged then that the reader follow his work if they want
to explore base editing further.
C-G to T-A isn’t all of it Cytidine deaminase developed by Liu and colleagues allowed for C-G to T-A base pair
transitions. After they achieved that in 2016, they did not rest on their laurels. In 2017,
they published another article, describing an Adenine deaminase which caused an
Adenine to Inosine conversion, the inosine then being read as a G, converted an A-T pair
into a G-C pair. The process seems simple, but developing an enzyme which converts AT to G-C was not easy. It turns out, Adenine deaminases don’t occur naturally. Even if
they do, they only perform deamination of Adenosine bases present in RNA, not DNA.
The di culty then was to engineer an enzyme which causes deamination of Adenosine in
DNA. To achieve that, they took an adenine deaminase from Escherichia coli (ecTadA) and
tried to evolve it into an enzyme which would be able to process Adenine using DNA as a
substrate. The trick they used was ingenious. They tethered this ecTadA to a dCas9 and
in the genome of the organism, introduced faulty antimicrobial resistance genes which
would become e ective only if there were to be a base pair conversion from A-T to G-C.
They grew these organisms with the ecTadA-dCas9 fusion in the presence of the
antimicrobial and selected for a mutant which could survive. In this manner, they got an
adenine deaminase which could target DNA. (71)
Since then, much more e cient and e ective adenine deaminases have been developed.
Scientists have used techniques of evolution such as Phage assisted continuous
evolution (PACE) or Phage assisted non-continuous evolution (PANCE) to enhance these
enzymes. These enzymes show greater catalytic activity and greater compatibility to a
larger variety of dCas9s, hence improving their exibility of use. (72)
So, with these classes of enzymes, you can achieve four DNA base transitions, making
precise DNA base editing applicable in a large amount of instances.
Going beyond four transition mutations -
ff
fi
fi
fl
ff
ffi
ffi
ff
ffi
Anzalone et al. recently described a method of editing bases precisely using a reverse
transcriptase attached to a nickase Cas9 and an important piece, the prime-editing guide
19
RNA (pegRNA). The prime editing guide RNA has a twofold function. It guides the nickase
Cas9-reverse transcriptase assembly to the target site and it also serves as a template for
the reverse transcriptase to generate DNA from the pegRNA. The newly synthesised DNA
can then bind to the target via base complementarity and then get faithfully reproduced
during replication. A caveat in this process is that two complimentary strands exist at the
end of this process, one strand that was bound to the target originally and another strand
which is synthesised by the reverse transcriptase. It turns out that the original strand that
is selectively degraded by cellular endonuclease as compared to the newly synthesised
strand which causes the equilibrium to shift towards the newly synthesised DNA strand.
In this manner, using the guide RNA itself as a template, base edits were made. Third
generation prime editors exist today which are much more e cient and thermostable as
compared to their ancestors. (73,74)
This method of editing bases has been termed ‘Prime editing’ and allows one complete
freedom to edit bases in whichever way they seem t. Of course, the normal limitations
exist for this system as well, some of which are PAM speci city of Cas9, problems with
the delivery of the system and a ected e ciency of the conversions due to cellular repair
machinery. Greater e ciencies have been achieved however for Cytosine BEs, Adenosine
BEs and Prime BEs by using secondary sgRNAs to nick the non-template strand such
that the cellular repair machinery uses the edited DNA strand as a template rather than
the non-edited one. This would shift the equilibrium towards the desired outcome. (68)
Precise base editing has allowed applications such as generating premature stop codons
(for example converting a TGA into a TAA) in order to knockout genes (CRISPR STOP).
(75) One could also tackle diseases which are caused due to a SNP (single nucleotide
polymorphism) and use editing to permanently cure the disease. Despite their recent
advent, precise base editors have been able to work their way into mainstream CRISPR/
Cas9 applications. (68) Much work still needs to be done to avoid o target e ects of
these editors and to increase their e ciency. It is only a matter of time then that these
methodologies start dominating the eld of therapeutic genetics.
Pushing CRISPR/Cas9 even further:
In this section, it is basically a free for all. It will contain descriptions of the latest
techniques and instances where some of the major obstacles to achieving the perfect
CRISPR/Cas9 construct have been tackled. Thus, this section is basically a treat for any
fans of CRISPR/Cas9 (courtesy of the author).
Controlling CRISPR/Cas9 Throughout this review, mentions have been made regarding o target e ects of the
technique. It is important to be able to control the assembly tightly so as to avoid these
e ects. We have gone through inducible promoters for Cas9 and split Cas9s which
themselves can be under the control of di erent inducible substrates, but even then, after
the machinery is activated, it stays in the cell for some amount of time which could lead
to o target e ects. So, to counter that, we shall discuss 2 solutions, sgRNA based
control and Anti-CRISPRs.
ff
ff
ff
ff
ffi
fi
fi
ff
ffi
fi
ffi
ff
ffi
ff
ff
ff
20
sgRNA based control In 2020, Hao et al. described a method of controlling the CRISPR/Cas9 system by
focusing on the sgRNA rather than the Cas9 itself. They described oligonucleotide based
constructs with a toehold at the end which would allow precise control of the CRISPR/
Cas9 assembly using oligonucleotide inputs. (76)
Cas9 needs to bind to the tracrRNA region of the sgRNA in order to be guided to the
target. This binding occurs due to the presence of secondary structures in the tracrRNA
region. What Hao and colleagues did was that they designed oligonucleotides that were
complimentary to the secondary structures of the sgRNA such that base complementarity
between the oligonucleotides (blockers) and the sgRNA regions would cause these
blockers to bind to the sgRNA, a ecting the secondary structure of the sgRNA which is
necessary for its interaction with Cas9. As a result, these blockers strip the ability of the
sgRNA to bind to Cas9. Another ingenious trick they used was at the end of these
oligonucleotides, they added bases which were not complimentary to the sgRNA. These
bases would then hang away from the oligonucleotide-sgRNA complex and allow for
another oligonucleotide (input) to bind to these bases. The technique works as follows.
Initially, the sgRNA and blocker are co-expressed. The sgRNA and blocker bind and thus
Cas9 is unable to bind to the sgRNA (OFF-state). Now, when the assembly needs to be
activated, you arti cially insert inputs which would be complimentary to the hanging
region of the blocker. This would cause the blocker and input to bind to each other thus
freeing the sgRNA to form its secondary structures. The free sgRNA would then interact
with Cas9 and guide it to the target (ON-state). Now, for a little while the assembly will be
active, but after a certain amount of time, the equilibrium between the blockers and inputs
will shift since blockers are generated internally as compared to the arti cially inserted
inputs. In this manner, you would block the sgRNA again and the assembly would again
be in the OFF-state. The same process has been described in Figure-5. (76)
Figure-5: Modifying the sgRNA to control the CRISPR/Cas9 system
a. A description of how base complementarity is applied to affect the secondary
structure of the sgRNA and hence prevent it from binding to a Cas9 enzyme. b.
Insertion of an input which is complementary to the toehold switch removes the
blocking strand and thus, turns on the sgRNA. c. A schematic of the technique. A
sgRNA is in the OFF-state when blocked using a blocking strand and upon insertion of
the input, the blocking strand is removed and thus, Cas9 can interact with the sgRNA
in its ON-state and produce a downstream effect
fi
ff
fi
Reprinted from Hao, Y., Li, J., Li, Q., Zhang, L., Shi, J., Zhang, X., Aldalbahi, A., Wang, L., Fan, C., & Wang, F.
(2020). Programmable Live-Cell CRISPR Imaging with Toehold-Switch-Mediated Strand Displacement.
Angewandte Chemie - International Edition
21
Thus, using strand displacement reactions, you can dictate when the assembly is on or
o . Further, you can use di erent combinations of blockers and inputs such that only a
speci c combination of inputs gives you the desired result. Hao et al. also go on to
mention that using this methodology, you can construct logical gates for CRISPR/Cas9
function inside the cell. (76)
Anti-CRISPRs Anti-CRISPRs (Acr) are another method of controlling CRISPR/Cas9. CRISPR exists as a
defense against mobile genetic elements (MGEs), but since this is biology we are talking
about, the recipients of the attacks by CRISPR/Cas will not just stay silent and take it.
They will have to evolve in some manner. The answer various MGEs came up with was a
group of proteins called Acr proteins. Acrs are a diverse set of proteins coded by MGEs
which directly interact with CRISPR proteins and inhibit their functions. The stage at
which inhibition occurs can vary from DNA binding by crRNA to cleavage of the DNA by
Cas proteins. (77)
Acr proteins were discovered by Joseph Bondy-Denomy by analysing phages that were
escaping the wrath of the CRISPR/Cas system. After such a discovery, a search began,
looking for such genes that could protect a phage from the CRISPR/Cas system. During
that search, it was found that Acr proteins cluster together in the form of a loci, so despite
having minimal similarities to each other or to Cas proteins, multiple Acr proteins were
discovered and analysed. Since then, these proteins have found applications in regulating
the CRISPR/Cas system. (78,79,80)
In a system with a CRISPR/Cas construct, Acr proteins can either be introduced
exogenously or can be introduced as a genetic construct under the control of an inducible
promoter to be expressed whenever required so as to stop the function of the CRISPR/
Cas system. Thus, Acr proteins can be used to gain another layer of control over the
CRISPR/Cas system in order to avoid the o -target e ects. (77)
The most exciting possible application of this discovery would be in creating CRISPR/Cas
based gene drives. Such gene drives would basically consist of releasing CRISPR/Cas
engineered organisms into the environment at a population level such that the engineered
locus spreads in a population via natural methods of reproduction. The engineered locus
would then, after a certain amount of time, be present in all members of a population as
they move through generations. An example of such a gene drive would be one designed
by Gantz et al. who created a transgenic Anopheles stephensi (vector for malaria) so that
it became resistant to the malarial parasite Plasmodium, making it impossible for the
vector to carry the pathogen, hence stopping the spread of malaria entirely. (81) This
would be the ideal scenario, however, questions have been raised against releasing
organisms with modi ed genomes in the environment as it may have unpredictable
deleterious e ects. However, imagine a scenario where you have an Acr system present
in the gene drive alongside the CRISPR/Cas system. So, you always have a bona de
‘KILL SWITCH’ to stop the gene drive in case something goes awry. Thus, Acr proteins
give rise to exciting new possibilities in the world of CRISPR/Cas. Beyond this, Acr
proteins could also be used to reduce the pathogenicity of virulent organisms which
depend on their CRISPR/Cas system for their defense. (77,81)
fi
ff
ff
ff
fi
ff
fi
ff
22
A bonanza of interesting leaps, techniques and applications Cas9 as a highly speci c biosensor Cas9 until now has been applied in genetic applications such as editing, silencing, even
activating gene segments and other interesting applications such as targeted live cell
imaging. Extending the live cell imaging technique a step further, what Guk et al. did was
they used targeted CRISPR/Cas9 to detect a Methicillin-resistant Staphylococcus aureus
(MRSA) using a fusion of dCas9 with SYBR green dye to target the mega region of the
MRSA genome. Not only that, they were able to discern between a MRSA and Methicillin
sensitive Staphylococcus aureus (MSSA). What they achieved was a a combination of
Fluorescence in situ hybridisation and CRISPR to create a diagnostic construct, hence
creating a CRISPR/Cas9 based biosensor for detecting MRSA. CRISPR/Cas9 in similar
manners has been used to detect multiple bacterial resistance genes and to discern hostpathogen interactions as well. Since then, not only Cas9, but also other Cas proteins such
as Cas12a and Cas13 have been used to design highly speci c diagnostic tools. (82,83)
Looking for a PAM free Cas9 Many a time in this review, it has been mentioned that Cas9 requires a PAM in order to
perform its function. This limits the diversity of gene sequences that are truly editable. In
order to increase the pool of sequences that can be edited, scientists have been looking
for naturally occurring Cas9s with di ering PAM preferences, hoping to nd that one
hidden gem, which would be a Cas9 enzyme that does not require a PAM at all. Though
that approach is ne, some scientists have decided to take a di erent route, arti cially
engineering enzymes to produce di ering PAM speci cities. To that e ort, a large amount
of engineered Cas9s have been developed. (84)
An engineered Cas9 which is of great interest to the scienti c world was developed by
Nishimashu et al. They designed a variant of SpCas9 called spCas9-NG. A wild type
SpCas9 has a NGG PAM preference but after the introduction of mutation in the Arginine
residue present at amino acid position 1335 of the SpCas9 and a follow up evolution
experiment, a Cas9 was generated which did not need the second G of the PAM for
target recognition. Thus, they developed a Cas9 which only required NG as its PAM. This
signi ed a quantum leap in the kind of sequences you could edit. (85)
Following this experiment, Walton et al. looked to truly generate a Cas9 without any PAM
preference at all. They performed targeted mutagenesis (which they called structure
guided mutagenesis) and generated two Cas9 variants. One was a SpG variant which
recognised an NG sequence as PAM similar to Nishimashu’s group, but the second
variant was SpRY. SpRY was a Cas9 enzyme which recognised the sequence NR as a
PAM (where R = A or G). It also recognised a RY sequence (where Y = C or T) as a PAM,
although to a lesser extent. As you can appreciate, having an enzyme which can
recognise an NA or an NG sequence as a PAM would dramatically increase the variety of
genetic sequences you can target. Thus, this experiment was a great leap in the world of
engineered Cas9 enzymes. To this day, SpRY is the most exible Cas9 enzyme. (86)
Since then, many scienti c groups have been looking to create a PAM free Cas9. In the
pool of such literature, an article has been gaining some steam recently. This article was
written by Qi et al., the contents of which describe creating a SpRY toolbox which allows
fi
fi
ff
ff
fi
fi
fl
fi
ff
ff
fi
fi
fi
fi
23
them to nearly edit the entire plant genome at their will. The word PAM-less was
mentioned in the title of their article and in a Scitech interview, they mentioned that this
toolbox removes the biggest bottleneck as far as CRISPR/Cas9 genome editing is
concerned, which is the PAM. (87,88) What this article signi es is a paradigm shift in the
search for a PAM free Cas9. It introduces a new approach. The approach of combining
di erent Cas9 enzymes so as to have a repertoire that together can target all possible
PAM combinations allowing us to e ectively edit any DNA sequence, rather than
searching for that one magical enzyme which can edit all sequences. This approach is
being adopted by the scienti c community as there is increasing literature which talks
about lling the holes in the current PAM repertoire as compared to just searching for one
PAM free nuclease. Other reasons also can be attributed to this shift away from a PAM
free nuclease. A truly PAM free Cas9 would have greater o target e ects and would
demand more stringent control as compared to a Cas9 which has a preferred PAM. Even
beyond that, a PAM free Cas9 would have to assess the entire genome for its target and
as a result, the time taken for such experiments would increase by a large amount. These
trade-o s will have to be considered if one day we truly achieve a PAM free Cas9. But,
based on the current trend, a complete Cas9 repertoire is the direction which seems more
appropriate. (84)
Remote controlled gene editing? We have read about light inducible split Cas9s, however, what those Cas9s require are
tedious systems such as a laser or an optical bre in order to activate those Cas9
enzymes which limits the exibility of the technique. Yu et al. designed a system to
remove that barrier. They developed a system called FAST which stands for Far-red light
(FRL) activated Split Cas9 system. This system allowed them to use LED lights to shed far
red light on mice and achieve photo inducible editing in the internal organs. (89)
The FAST system was constructed using two FRL sensors - BphS and p65-VP64-BldD;
and two cohesion proteins - DocS and Coh2. They fused these to a split dCas9 enzyme
and thus produced a system which upon induction by Far-red light would fuse the split
Cas9s together and allow them to perform their activity. This system pushes the optical
repertoire for Cas9 which usually would require phototoxic wavelengths of light in order to
be activated. Using just an LED for the same function would allow photoinducible Cas9
control in vivo without damaging the organism. (89)
Knock in anything! CRISPR and Transposons join hands Knocking out using CRISPR is fairly easy. Knocking in is much more di cult. Especially
with large genomic segments, the e ciency of the CRISPR/Cas9 system plummets
because it depends on HDR. In contrast to Cas9, transposons have the ability to insert
large genomic segments, however, they lag behind CRISPR/Cas9 in its ability to target
speci c targets. So, it was easy for someone to imagine a scenario where Cas9 could
take care of the guiding function and transposons could take care of the knocking in
function. Came forth a fusion of the two, CasTn system. Cas-Tn means Cas-Transposon
where a transposase (Himar1) is fused to a dCas9 enzyme to create a targeted
transposase construct. Using this assembly, Sway Chen and Harris Wang were able to
achieve a targeted knock in at a rate which was greater than 300 times than that of an
isolated transposase on its own. In the study, by using knockdown of uorescent proteins
such as mCherry and GFP, they showed that their construct could achieve targeted
knockdowns. Further, using a promoter less puromycin resistance gene as a payload for
ffi
fl
ff
fi
ff
fi
ff
ffi
fi
fl
ff
fi
fi
ff
24
delivery, they displayed that the CasTn could be used for targeted insertion of genes.
They therefore demonstrated that Transposition and CRISPR/Cas9 can be combined to
create a novel and much more accessible platform for gene knockins as compared to
what these two systems could achieve by themselves. The process has been explained in
Figure-6. (90)
Figure-6: A schematic depicting the process of targeted knock in of a transposon via tethering
to a dCas9 enzyme
A Himar transposase is tethered to a dCas9 enzyme along with the transposon which is supposed to be
knocked in. This assembly is guided to the region of interest using the sgRNA which nds the target genomic
regions and binds to the dCas9. Thus, the entire assembly is driven to the region of interest where via the action
of the transposase, the transposon is knocked in.
Reprinted from Chen, S. P., & Wang, H. H. (2019). An Engineered Cas-Transposon System for Programmable and Site-Directed DNA
Transpositions. The CRISPR Journal, 2(6), 376–394.
With this ends the fan service. All of these techniques and applications serve as a
possible direction of CRISPR/Cas9 development. Thus, the future itself is the future
perspective. As it is impossible to entirely predict human behaviour, it is also impossible
to predict the extent of human creativity. Thus, these techniques here have been
mentioned to serve as some ideas that might allow you to maybe one day create a
CRISPR/Cas9 variant of your own. As a follow up, there are many more interesting
articles related to creative applications of CRISPR/Cas9 coming out everyday. The go to
journal for accessing or keeping track of these articles would be the CRISPR journal
which is a dedicated journal that keeps track of CRISPR based technologies. It can be
accessed using this link: https://home.liebertpub.com/publications/the-crispr-journal/642
Now, we move onto a much more human discussion. We have all these powers in our
reach, but should we use them? If yes, then how should we use them.
Ethics of CRISPR/Cas9 application:
'With great power, comes great responsibility’ said Ben Parker to a young Peter as they
drove to a library one evening. To those who are uninitiated, this is a direct quote from
Spider-Man and is one of the most iconic lines in superhero literature. Uncle Ben said that
to Peter Parker in order to teach him that any sort of power comes with a certain amount
of responsibility and it is upto us to not abuse it or not to harm others, but to help them
and make the world better a better place. This quote precisely explains the dilemma
scientists face with this revolutionary technique. We can edit almost all gene segments,
fi
25
Use of CRISPR/Cas beyond biological research:
CRISPR/Cas9 has been applied extensively in biological research in order to understand
the depths of the workings of biological systems. However, given that this technique is so
exible, it has been applied in multifarious real world applications.
To this day, many feats have been achieved using CRISPR/Cas9. Some of them include
genetically engineered plants which boast greater resistance to stress (91,92) and
increased carbon xation e ciency. (93) Variants of Cas9 have also been used to
arti cially generate resistance against RNA based plant viruses. (103)
Not just plants, CRISPR/Cas9, despite its aws, has been able to achieve incredible feats
in the world of animal therapeutics. Duchenne Muscular Dystrophy (94), ⍺-1 antitrypsin
de ciency (95), haemophilia (96) and even the battle against the dreaded HIV have all
received contributions because of CRISPR/Cas9. (97) Recently, this system has also been
used as a diagnostic tool to detect SARS-CoV-2. (98) Beyond these, it is expected that
CRISPR/Cas9 will serve as an e ective weapon in the ght against some viruses which
can lead to latent infections e.g. Epstein-Barr virus, John Cunningham virus etc. (103)
Besides all these, cancer diagnostics have been revolutionised by the advent of CRISPR
variant based techniques such as SHERLOCK. (99) Also, in the battle against cancer,
positive results have been reported in mouse models where prevention of development of
cancer was achieved by preventing gain of function mutations, (100) inhibiting loss of
function mutations, (101) along with immunotherapy using genetically modi ed Chimeric
antigen receptor T cells, all experiments which used CRISPR/Cas9. (102)
In 2018, CRISPR was used by He Jiankui to edit human embryos and the rst CRISRP/
Cas based clinical trial to treat HIV was also set in motion. In 2019, in vivo clinical trials
using CRISPR/Cas9 in order to treat blindness were authorised, signifying the giant leaps
the technique has taken since its inception in 2012. (103) In coming years, the trend might
shift towards more invasive germline editing procedures. But as we attempt to perform
more and more invasive procedures, questions arise regarding natural laws we might be
breaking. Is it really okay for us to mess with nature like this? What possible dangers
might we be dealing with by editing embryos? And if we are editing human embryos and
various other organisms, where do we draw the line?
Before we deal with these questions, it is important to look at unauthorised uses of
CRISPR, especially He Jiankui’s attempt at editing human embryos.
Major misuse of the tool:
On 27 November 2018, an entire sect of the scienti c community sat back in shock and
awe. Their smartphones bombarded with one hashtag. The infamous hashtag that
signi ed a line being crossed in the world of genetic editing which was supposedly the
called the red line, as in, must never be approached, let alone crossed. #CRISPRbabies
trended on twitter. For the rst time, someone had used CRISPR/Cas9 to directly edit the
fi
fi
fi
fi
fl
ff
ffi
fi
fi
fi
fi
26
fi
fl
we can alter the course of evolution and we can manipulate not just DNA, but even RNA
at our will. But should we do all that? Who should have access to a technology which in
the wrong hands could lead to a catastrophe? Let us explore these questions together.
What He Jiankui had just done was that he used a edgling and o target e ect prone
technology to edit human embryos. Although he performed such an act to protect two
children from the wrath of HIV, still, the scienti c community almost lashed out. People
understood that to want to help someone in need is a noble quality, however, using an
unruly tool whose e ects one cannot fully predict is not only irresponsible, but also
verging on criminal. The Chinese judicial system agreed with the world's opinion on this
one as Jiankui was sentenced to 3 years in prison and was slapped with a US $430,000
ne. His cohorts were also punished with prison sentences, albeit shorter ones. (105) No
one questioned the fact that Jiankui intended only to help two kids so that they could live
a normal life, but in doing so he endangered two human lives. If things had gone sour and
some truly harmful o target snips had occurred, maybe two lives would have been lost.
Fortunately, the two babies were born healthy, but this still does not exempt Jiankui of the
act he committed. What redoubled his egregious act was the fact that Jiankui in his study
started with 8 HIV positive couples. This experiment was largely regarded as a failure of
‘self-regulation’ in the world of biological science. (104,106)
More problematic was the fact that this was not an isolated incident. Whispers had been
swirling around gene editing in humans being performed in China since as early as 2015,
but nothing became of them since the projects were kept highly classi ed. Some of these
were revealed later on, one example being Huang’s attempt at editing embryos in 2015 in
order to combat thalassemia. These projects were functioning even though the progenitor
of the technique, Jennifer Doudna, strictly preached against such an application. She was
right in doing so, since these projects reported o target e ects in the genomes of the
embryos. So, any application of CRISPR/Cas9 to edit anything in the human genome
could be potentially catastrophic. It could become as serious of a crime as manslaughter,
since it is argued that any embryo beyond 14 days in age is essentially a human life. (54)
Although the actions of He Jiankui were ill advised, his experiment did have a positive
e ect. This experiment sparked a wave of debates which discussed on a global scale the
ethics of application of CRISPR/Cas9 and at some level, the ethics of its distribution. He
Jiankui himself contributed to these debates by mentioning 5 guidelines for CRISPR/Cas9
usage in human embryo editing 1. Empathy for patients - for some families, early gene surgery may be the only way to
cure disease.
2. Only for serious disease, never for vanity.
3. No one can control a child’s life. A gene-edited child retains the same rights as a
“normal” child.
4. Genes do not de ne us. DNA does not predispose us.
5. Everyone deserves freedom from genetic disease, regardless of wealth.
The above guidelines are a direct quote from the book Editing humanity by Kevin Davies,
which covers this entire asco in detail. (54) This gives us a lot of talking points to tackle.
ff
fi
ff
ff
fl
ff
fi
fi
ff
ff
fi
27
ff
fi
genomes of twin babies, Lula and Nuna, in order to immunise them to HIV (CCR5 gene
was targeted which is what HIV strains use to infect human cells). But, no one
commended this action. In fact, the very next day, when the perpetrator He Jiankui
walked on stage during the gene editing summit in Hong Kong, he was greeted with
almost a disdain from his peers. Headlines poured from various news outlets, CRISPRbaby scientist fails to satisfy. (54,104)
The nal conversation we shall have then will be discussing these guidelines and ethical
conundrums. A large section of this debate shall be my personal opinion and the reader is
free to agree or disagree with them.
The ethical dilemma of genetic engineering in
humans:
Imagine a scenario. A couple walks to you and tells you that their child will be born with a
debilitating disease. You possess a tool to solve their problems, but it is risky. It will not
work as intended every time. What will you do? Will you argue that the unborn child has
its own rights and you cannot perform a permanent procedure and change that life
forever? What another argument would be is that, will you risk loss of life in order to
improve the quality of life for an embryo? At this point you enter a whole other realm of
philosophical thinking - is it better to be disabled and be alive or to not be alive at all?
These arguments seem to have no end, even then, I shall humbly present my take on
them.
Should He Jiankui have done what he did? The simple answer is no. You simply do not
take a risk this big, no matter what the situation may be because the risk does not lie with
one generation. This is a permanent genetic change which will be inherited by
subsequent generations, hence, one misstep may have doomed an entire line of
descendants. So, it was a highly irresponsible decision that was spurred on by emotions
rather than logic. Further, one might go so far as to say that Jiankui might have wanted to
capitalise on the desperation of those couples, but with regards to this, I shall give him
the bene t of the doubt. Sometimes you have to let something bad happen in order to
avoid an eventuality which is much worse. A chess player would empathise with this
analogy, and so would anyone who has lived long enough in this world which is not just
roses and rainbows. So, even though what Jiankui did was bad, it did serve as a wake up
call to all of us.
Should humans be allowed to edit the genomes of other humans? If it is for genetic
therapy in adults, an authorised clinical trial is where this such editing should happen. Of
course, all those who are participating in the trial must be made aware of the risks they
are taking. This is how medicine has worked for more than a century now. But where this
should not happen for now is in germline cell gene editing. Despite one’s consent,
germline editing is something which could adversely a ect the coming generations and
thus the decision does not really lie with the person consenting to the CRISPR based
therapy.
Should this happen in embryos? It could be done for embryos but not fait accompli like
what Jiankui did, who informed the world of his act after the fact. Again, such an act
should be performed as part of a clinical trial, under strict observation and such embryos
should not be implanted into a surrogate mother, so as to avoid risks to both the mother
and the unborn child. Thus, such a clinical trial must be only for research purposes until a
satisfactorily accurate method of gene editing is developed.
Who should have access to CRISPR/Cas9 technology? This one is a touchy subject.
As a scientist, one wants knowledge to be disseminated far and wide into the world, so
that anyone in need of it can access it. However, nefarious individuals exist on this planet.
ff
fi
fi
28
Many a times there have been instances where bio-weapons were used to terrorise
innocent people. Powdered Bacillus anthracis comes to mind rst of all as I was warned
multiple times as a child, not to open just any letter and in case there was some sort of
powder in it, to contact the doctor at once. So, in a world where bio-weapons are already
a reality, what could CRISPR/Cas9 editing do? It is not too far fetched to say that CRISPR
based editing and knock-ins could create pathogenic strains with increased virulence and
deleterious e ects, di cult though it may be. So, should anyone still have access to
CRISPR/Cas9? I would still say yes. However, in an ideal world, CRISPR/Cas9
engineering would be taught to only trained scientists who would allow a higher level of
transparency in their labs’ proceedings and it is via these scientists that both the
knowledge regarding CRISPR/Cas9 and its application would be accessed and
distributed. This does not mean that all the open access literature must be done away
with. This only means that any lab working with CRISPR/Cas9 must open its door to
investigations and become even more transparent as compared to a lab which does not. I
understand that this could hinder a lab’s functioning and would make CRISPR a di cult
subject to conduct research on, however, such measures must be enforced in order to
avoid a nearly criminal act as what He Jiankui did. A lot of conversations and debates
need to be had before we take any steps in the direction of human gene editing. I would
like to point you to one such conversation with Jennifer Doudna, the creator of this
technique.
ffi
fi
fi
ffi
fl
ff
The conversation would be Jennifer Doudna’s interview at the World Science Festival in
October 2019. (108) Interesting questions were addressed to her during her time on
stage. How could CRISPR base editing improve one’s quality of life? Could it maybe
increase a person’s life span? She answered by basically saying that CRISPR based
therapy could not achieve an increase in a person’s life span because we do not
understand ageing completely, but it could be used to prevent the shortening of lifespan
by helping in therapy of illnesses such as cancer or by allowing humans to ght against
infections. In another part of the interview, she addressed the question of designer
babies. She again went on to say that achieving traits such as increased intelligence or
increased athleticism could be under the combined control of thousands of genes, so
realistically, achieving that still would not be possible. Same answer for super soldiers. So
sadly, no Captain Americas for us in the near future. She also addressed notions such as
bringing back species which had gone extinct in recent times. Or even organising gene
drives, which she admitted were possible to some extent with the current state of
CRISPR technology. She also took the time to discuss one of her articles in nature titled ‘Genome-editing revolution: My whirlwind year with CRISPR’ (107) an article which was
prompted by the fear that CRISPR/Cas9 would be used to edit human embryos. As the
interview continued, she discussed many a topic, but what she stressed most of all was
the fact that it is important for us to have ethical debates regarding CRISPR and that
these debates should go global so that we can educate people. I would urge the reader to
go on YouTube and watch this video and thus introspect and have a conversation with
themselves regarding CRISPR/Cas9. As for my introspection, I agree with Jennifer on all
of the points. What I would like to add to all of this is that CRISPR is an important
technology. It is not too far fetched to say that it could have the same implications as
Alfred Nobel’s invention of dynamite, which I would like to remind the reader, was at one
point intended to make roads on mountains by blowing away portions of it, but as we
know now, the invention of dynamite instead led to the weaponisation of the world, an
eventuality which has exacted devastating damages. In the same tune, CRISPR/Cas
system has amazing therapeutic potential, however it could be dangerous because it can
be learnt quickly, it is exible and it can also be applied quickly and thus, in nefarious
hands, it could endanger the world. What can be taken away from all of this is that
29
managing CRISPR is not a scienti c responsibility alone, it is a global responsibility. In the
future, there may come a time where CRISPR touches all of us intimately. It may become
a part of our lives. So, we must develop international and national level regulatory
infrastructures so that knowledge regarding CRISPR can be distributed in a safe manner
in order to ensure that this gorgeous gift that biological sciences has provided us, does
not get abused like many other scienti c gifts that came before it, because this time, we
really cannot a ord any such mistakes. The bottomline therefore is this. CRISPR can be
used in humans but only for therapeutic purposes under strict regulation. CRISPR for
vanity is completely out of the question. The myriads of uses of this technology such as
genetic engineering of crops, biological research, ghting against antimicrobial resistance
and all other applications in the same vein must be encouraged. The world must start
thinking about how to use CRISPR/Cas properly and how to prevent its misuse. Finally,
decisions involving CRISPR/Cas must be made devoid of emotion, based on pure
scienti c logic. Thus, a lot of responsibility rides on our shoulders. A few missteps here
and there could doom an entire race. So, instead of shying away from it, we must face it
with vigour. We must own up to the challenge. Humanity birthed this technology and thus
all of humanity combined must raise this wonder child properly.
Conclusion:
Beyond all the heavy talk, to end this review on a positive note, I would like to add that a
world where humans will have access to their own genome via CRISPR/Cas9 may not be
too far away. A world where cancer is not a terminal malady but a mild disease which can
be xed with a trip to the doctor’s o ce for a little hit of CRISPR/Cas9 may not be
completely imaginary. A world where resistant bacteria do not pose major threats
because we have a knight in shining armour, armed and ready to ght for us, may soon
be upon us. Dreams of a scienti c utopia pop up whenever the future of CRISPR/Cas9 is
mentioned. Which dreams shall we attain and which shall be lost in the annals of human
endeavour, only time will tell. But what CRISPR/Cas9 most de nitely has done is
reinvigorated the world of science by attracting a new generation of budding scienti c
minds to the eld.
In this review, we have explored the entire journey of CRISPR/Cas9 system. A simple
sequencing mistake led to the discovery of CRISPR repeats and in the following years,
the very repeats generated a technology which will de ne biological scienti c e orts in
the coming century. To be a part of such an e ort, we understood the basics of the
mechanism by which this system functions, followed by where it stands in the current
world. Then we took a foray into the recent exciting variations that have been coming up
in the past 5 years and discussed how these very developments shall lead us into the
future. All of this scienti c text was underlined by a human discussion of the morality of
CRISPR/Cas9 usage, a conversation which I would say is just as important as the
scienti c aspect of this review, if not more. Hence, we arrive at the end of this beautiful
ride. The sole purpose behind this review was to introduce the uninitiated to the world of
CRISPR and to give them enough knowledge and ideas to set them on their way. Thus,
such a large text was generated in pursuit of that goal. To conclude this text then what I
would like to say is that millennia of scienti c thoughts, studies and experiments led us to
this unbelievable discovery. It must therefore be treasured, applauded, disseminated and
applied, albeit responsibly.
fi
ff
fi
fi
fi
fi
fi
ff
fi
fi
ffi
fi
fi
fi
ff
fi
fi
fi
fi
30
I would like to thank Professor Saikrishnan Kayarat for providing me with this opportunity
to review a technique which motivated to become a scientist. His advice and teachings
have been invaluable as I wrote this review. Further, I want to thank IISER Pune for having
such a course as Literature review which trains budding scientists in the art of writing
scienti c literature. This review, although will not be published, shall always remain my
rst piece of scienti c literature.
References:
History of CRISPR:
1.
2.
-.
13.
14.
Ishino, Y., Shinagawa, H., Makino, K., Amemura, M., & Nakata, A. (1987). Nucleotide Sequence of the
iap Gene, Responsible for Alkaline Phosphatase Isozyme Conversion in Escherichia coli, and
Identi cation of the Gene Product. In JOURNAL OF BACTERIOLOGY (Vol. 169, Issue 12).
Van Soolingen,’, D., De Haas,’, P. E. W., Hermans, P. W. M., Groenen,’ And, P. M. A., & Van Embdenl, J.
D. A. (1993). Comparison of Various Repetitive DNA Elements as Genetic Markers for Strain
Di erentiation and Epidemiology of Mycobacterium tuberculosis. In JOURNAL OF CLINICAL
MICROBIOLOGY.
Lander, E. S. (2016). The Heroes of CRISPR. In Cell (Vol. 164, Issues 1–2, pp. 18–28). Cell Press.
https://doi.org/10.1016/j.cell-
Jansen, R., Van Embden, J. D. A., Gaastra, W., & Schouls, L. M. (2002). Identi cation of genes that are
associated with DNA repeats in prokaryotes. Molecular Microbiology, 43(6),-. https://
doi.org/10.1046/j-.x
Mojica, F. J. M., Díez-Villaseñor, C., García-Martínez, J., & Soria, E. (2005). Intervening sequences of
regularly spaced prokaryotic repeats derive from foreign genetic elements. Journal of Molecular
Evolution, 60(2), 174–182. https://doi.org/10.1007/s-
Pourcel, C., Salvignol, G., & Vergnaud, G. (2005). CRISPR elements in Yersinia pestis acquire new
repeats by preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary
studies. Microbiology, 151(3), 653–663. https://doi.org/10.1099/mic-
Bolotin, A., Quinquis, B., Sorokin, A., & Dusko Ehrlich, S. (2005). Clustered regularly interspaced short
palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology, 151(8),-. https://doi.org/10.1099/mic-
Barrangou, R., Fremaux, C., Deveau, H., Richards, M., Boyaval, P., Moineau, S., Romero, D. A., &
Horvath, P. (2007). CRISPR provides acquired resistance against viruses in prokaryotes. Science,-),-. https://doi.org/10.1126/science-
Sorek, R., Kunin, V., & Hugenholtz, P. (2008). CRISPR - A widespread system that provides acquired
resistance against phages in bacteria and archaea. Nature Reviews Microbiology, 6(3), 181–186.
https://doi.org/10.1038/nrmicro1793
Sontheimer, E., and Marra ni, L. (2008). Target DNA interference with crRNA. U.S. Provisional Patent
Application 61/009,317, led September 23, 2008; later published as US2010/- (abandoned).
Garneau, J. E., Dupuis, M. È., Villion, M., Romero, D. A., Barrangou, R., Boyaval, P., Fremaux, C.,
Horvath, P., Magadán, A. H., & Moineau, S. (2010). The CRISPR/cas bacterial immune system cleaves
bacteriophage and plasmid DNA. Nature,-), 67–71. https://doi.org/10.1038/nature09523
Horvath, P., Romero, D. A., Coûté-Monvoisin, A. C., Richards, M., Deveau, H., Moineau, S., Boyaval,
P., Fremaux, C., & Barrangou, R. (2008). Diversity, activity, and evolution of CRISPR loci in
Streptococcus thermophilus. Journal of Bacteriology, 190(4),-. https://doi.org/10.1128/
JB-
Deveau, H., Barrangou, R., Garneau, J. E., Labonté, J., Fremaux, C., Boyaval, P., Romero, D. A.,
Horvath, P., & Moineau, S. (2008). Phage response to CRISPR-encoded resistance in Streptococcus
thermophilus. Journal of Bacteriology, 190(4),-. https://doi.org/10.1128/JB-
Mojica, F. J. M., Díez-Villaseñor, C., García-Martínez, J., & Almendros, C. (2009). Short motif
sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology, 155(3),
733–740. https://doi.org/10.1099/mic-
fi
ffi
fi
fi
fi
fi
31
ff
fi
Acknowledgements:
15. Deltcheva, E., Chylinski, K., Sharma, C. M., Gonzales, K., Chao, Y., Pirzada, Z. A., Eckert, M. R., Vogel,
J., & Charpentier, E. (2011). CRISPR RNA maturation by trans-encoded small RNA and host factor
RNase III. Nature,-), 602–607. https://doi.org/10.1038/nature-. Sapranauskas, R., Gasiunas, G., Fremaux, C., Barrangou, R., Horvath, P., & Siksnys, V. (2011). The
Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli. Nucleic Acids
Research, 39(21),-. https://doi.org/10.1093/nar/gkr606
17. Gasiunas, G., Barrangou, R., Horvath, P., & Siksnys, V. (2012). Cas9-crRNA ribonucleoprotein complex
mediates speci c DNA cleavage for adaptive immunity in bacteria. Proceedings of the National
Academy of Sciences of the United States of America, 109(39). https://doi.org/10.1073/
pnas-. Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J. A., & Charpentier, E. (2012). A programmable
dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science,-), 816–821.
https://doi.org/10.1126/science-
Basics of CRISPR/Cas9 system:
19. McGinn, J., & Marra ni, L. A. (2019). Molecular mechanisms of CRISPR–Cas spacer acquisition. In
Nature Reviews Microbiology (Vol. 17, Issue 1, pp. 7–12). Nature Publishing Group. https://doi.org/
10.1038/s-. Wang, H., La Russa, M., & Qi, L. S. (2016). CRISPR/Cas9 in Genome Editing and beyond. Annual
Review of Biochemistry, 85, 227–264. https://doi.org/10.1146/annurev-biochem-. Hille, F., & Charpentier, E. (2016). CRISPR-cas: Biology, mechanisms and relevance. In Philosophical
Transactions of the Royal Society B: Biological Sciences (Vol. 371, Issue 1707). Royal Society of
London. https://doi.org/10.1098/rstb-. Barrangou, R., & Marra ni, L. A. (2014). CRISPR-cas systems: Prokaryotes upgrade to adaptive
immunity. In Molecular Cell (Vol. 54, Issue 2, pp. 234–244). Cell Press. https://doi.org/10.1016/
j.molcel-. Tyson, G. W., & Ban eld, J. F. (2008). Rapidly evolving CRISPRs implicated in acquired resistance of
microorganisms to viruses. Environmental Microbiology, 10(1), 200–207. https://doi.org/10.1111/
j-.x
24. Ivančić-Bace, I., Cass, S. D., Wearne, S. J., & Bolt, E. L. (2015). Di erent genome stability proteins
underpin primed and Naïve adaptation in E. Coli CRISPR-Cas immunity. Nucleic Acids Research,
43(22),-. https://doi.org/10.1093/nar/gkv1213
25. Levy, A., Goren, M. G., Yosef, I., Auster, O., Manor, M., Amitai, G., Edgar, R., Qimron, U., & Sorek, R.
(2015). CRISPR adaptation biases explain preference for acquisition of foreign DNA. Nature,-),
505–510. https://doi.org/10.1038/nature-. Modell, J. W., Jiang, W., & Marra ni, L. A. (2017). CRISPR-Cas systems exploit viral DNA injection to
establish and maintain adaptive immunity. Nature,-), 101–104. https://doi.org/10.1038/
nature-. Wang, J., Li, J., Zhao, H., Sheng, G., Wang, M., Yin, M., & Wang, Y. (2015). Structural and Mechanistic
Basis of PAM-Dependent Spacer Acquisition in CRISPR-Cas Systems. Cell, 163(4), 840–853. https://
doi.org/10.1016/j.cell-. Heler, R., Samai, P., Modell, J. W., Weiner, C., Goldberg, G. W., Bikard, D., & Marra ni, L. A. (2015).
Cas9 speci es functional viral targets during CRISPR-Cas adaptation. Nature,-), 199–202.
https://doi.org/10.1038/nature-. Richter, C., Dy, R. L., McKenzie, R. E., Watson, B. N. J., Taylor, C., Chang, J. T., McNeil, M. B., Staals,
R. H. J., & Fineran, P. C. (2014). Priming in the Type I-F CRISPR-Cas system triggers strandindependent spacer acquisition, bi-directionally from the primed protospacer. Nucleic Acids Research,
42(13),-. https://doi.org/10.1093/nar/gku527
30. Fineran, P. C., Gerritzen, M. J. H., Suárez-Diez, M., Kun̈ ne, T., Boekhorst, J., Van Hijum, S. A. F. T.,
Staals, R. H. J., & Brouns, S. J. J. (2014). Degenerate target sites mediate rapid primed CRISPR
adaptation. Proceedings of the National Academy of Sciences of the United States of America,
111(16), E1629–E1638. https://doi.org/10.1073/pnas-. Carte, J., Wang, R., Li, H., Terns, R. M., & Terns, M. P. (2008). Cas6 is an endoribonuclease that
generates guide RNAs for invader defense in prokaryotes. Genes and Development, 22(24),-. https://doi.org/10.1101/gad-. Charpentier, E., Richter, H., van der Oost, J., & White, M. F. (2015). Biogenesis pathways of RNA
guides in archaeal and bacterial CRISPR-Cas adaptive immunity. In FEMS Microbiology Reviews (Vol.
39, Issue 3, pp. 428–441). Oxford University Press. https://doi.org/10.1093/femsre/fuv023
ffi
ff
ffi
ffi
ffi
fi
fi
fi
32
33. Jiang, F., & Doudna, J. A. (2017). CRISPR-Cas9 Structures and Mechanisms. In Annual Review of
Biophysics (Vol. 46, pp. 505–529). Annual Reviews Inc. https://doi.org/10.1146/annurevbiophys-. Sternberg, S. H., Redding, S., Jinek, M., Greene, E. C., & Doudna, J. A. (2014). DNA interrogation by
the CRISPR RNA-guided endonuclease Cas9. Nature,-), 62–67. https://doi.org/10.1038/
nature-. Jiang, F., Taylor, D. W., Chen, J. S., Kornfeld, J. E., Zhou, K., Thompson, A. J., Nogales, E., & Doudna,
J. A. (2016). Structures of a CRISPR-Cas9 R-loop complex primed for DNA cleavage. Science,-), 867–871. https://doi.org/10.1126/science.aad8282
Major variants of CRISPR/Cas9 system in use today:
36. Adli, M. (2018). The CRISPR tool kit for genome editing and beyond. In Nature Communications (Vol. 9,
Issue 1). Nature Publishing Group. https://doi.org/10.1038/s-. Alok, A., Sandhya, D., Jogam, P., Rodrigues, V., Bhati, K. K., Sharma, H., & Kumar, J. (2020). The Rise
of the CRISPR/Cpf1 System for E cient Genome Editing in Plants. Frontiers in Plant Science, 11, 264.
https://doi.org/10.3389/fpls-. Zetsche, B., Gootenberg, J. S., Abudayyeh, O. O., Slaymaker, I. M., Makarova, K. S., Essletzbichler, P.,
Volz, S. E., Joung, J., Van Der Oost, J., Regev, A., Koonin, E. V., & Zhang, F. (2015). Cpf1 Is a Single
RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System. Cell, 163(3), 759–771. https://doi.org/
10.1016/j.cell-. Fonfara, I., Richter, H., BratoviÄ, M., Le Rhun, A., & Charpentier, E. (2016). The CRISPR-associated
DNA-cleaving enzyme Cpf1 also processes precursor CRISPR RNA. Nature,-), 517–521.
https://doi.org/10.1038/nature-. Yamano, T., Nishimasu, H., Zetsche, B., Hirano, H., Slaymaker, I. M., Li, Y., Fedorova, I., Nakane, T.,
Makarova, K. S., Koonin, E. V., Ishitani, R., Zhang, F., & Nureki, O. (2016). Crystal Structure of Cpf1 in
Complex with Guide RNA and Target DNA. Cell, 165(4), 949–962. https://doi.org/10.1016/
j.cell-. Lieber, M. R. (2010). The mechanism of double-strand DNA break repair by the nonhomologous DNA
end-joining pathway. In Annual Review of Biochemistry (Vol. 79, pp. 181–211). https://doi.org/10.1146/
annurev.biochem-. Wang, T., Wei, J. J., Sabatini, D. M., & Lander, E. S. (2014). Genetic screens in human cells using the
CRISPR-Cas9 system. Science,-), 80–84. https://doi.org/10.1126/science-. Shalem, O., Sanjana, N. E., Hartenian, E., Shi, X., Scott, D. A., Mikkelsen, T. S., Heckl, D., Ebert, B. L.,
Root, D. E., Doench, J. G., & Zhang, F. (2014). Genome-scale CRISPR-Cas9 knockout screening in
human cells. Science,-), 84–87. https://doi.org/10.1126/science-. Gilbert, L. A., Horlbeck, M. A., Adamson, B., Villalta, J. E., Chen, Y., Whitehead, E. H., Guimaraes, C.,
Panning, B., Ploegh, H. L., Bassik, M. C., Qi, L. S., Kampmann, M., & Weissman, J. S. (2014).
Genome-Scale CRISPR-Mediated Control of Gene Repression and Activation. Cell, 159(3), 647–661.
https://doi.org/10.1016/j.cell-. Gilbert, L. A., Larson, M. H., Morsut, L., Liu, Z., Brar, G. A., Torres, S. E., Stern-Ginossar, N.,
Brandman, O., Whitehead, E. H., Doudna, J. A., Lim, W. A., Weissman, J. S., & Qi, L. S. (2013).
XCRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell, 154(2), 442.
https://doi.org/10.1016/j.cell-. Dykxhoorn, D. M., & Lieberman, J. (2005). The silent revolution: RNA interference as basic biology,
research tool, and therapeutic. In Annual Review of Medicine (Vol. 56, pp. 401–423). Annu Rev Med.
https://doi.org/10.1146/annurev.med-. Gaj, T., Epstein, B. E., & Scha er, D. V. (2016). Genome engineering using Adeno-associated virus:
Basic and clinical research applications. In Molecular Therapy (Vol. 24, Issue 3, pp. 458–464). Nature
Publishing Group. https://doi.org/10.1038/mt-. Dicarlo, J. E., Norville, J. E., Mali, P., Rios, X., Aach, J., & Church, G. M. (2013). Genome engineering in
Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Research, 41(7),-.
https://doi.org/10.1093/nar/gkt135
49. Li, J. F., Norville, J. E., Aach, J., McCormack, M., Zhang, D., Bush, J., Church, G. M., & Sheen, J.
(2013). Multiplex and homologous recombination-mediated genome editing in Arabidopsis and
Nicotiana benthamiana using guide RNA and Cas9. In Nature Biotechnology (Vol. 31, Issue 8, pp. 688–
691). Nature Publishing Group. https://doi.org/10.1038/nbt.2654
ffi
ff
33
50. Hsu, P. D., Lander, E. S., & Zhang, F. (2014). Development and applications of CRISPR-Cas9 for
genome engineering. In Cell (Vol. 157, Issue 6, pp-). Elsevier B.V. https://doi.org/10.1016/
j.cell-. Blasco, R. B., Karaca, E., Ambrogio, C., Cheong, T. C., Karayol, E., Minero, V. G., Voena, C., & Chiarle,
R. (2014). Simple and Rapid InVivo Generation of Chromosomal Rearrangements using CRISPR/Cas9
Technology. Cell Reports, 9(4),-. https://doi.org/10.1016/j.celrep-. Choi, P. S., & Meyerson, M. (2014). Targeted genomic rearrangements using CRISPR/Cas technology.
Nature Communications, 5(1), 1–6. https://doi.org/10.1038/ncomms4728
53. Maddalo, D., Manchado, E., Concepcion, C. P., Bonetti, C., Vidigal, J. A., Han, Y. C., Ogrodowski, P.,
Crippa, A., Rekhtman, N., Stanchina, E. De, Lowe, S. W., & Ventura, A. (2014). In vivo engineering of
oncogenic chromosomal rearrangements with the CRISPR/Cas9 system. Nature,-), 423–428.
https://doi.org/10.1038/nature-. Editing Humanity | Book by Kevin Davies | O cial Publisher Page | Simon & Schuster. (n.d.). April 2020,
from https://www.simonandschuster.com/books/Editing-Humanity/Kevin-Davies/-. Qi, L. S., Larson, M. H., Gilbert, L. A., Doudna, J. A., Weissman, J. S., Arkin, A. P., & Lim, W. A. (2013).
Repurposing CRISPR as an RNA-γuided platform for sequence-speci c control of gene expression.
Cell, 152(5),-. https://doi.org/10.1016/j.cell-. Chavez, A., Scheiman, J., Vora, S., Pruitt, B. W., Tuttle, M., P R Iyer, E., Lin, S., Kiani, S., Guzman, C.
D., Wiegand, D. J., Ter-Ovanesyan, D., Bra , J. L., Davidsohn, N., Housden, B. E., Perrimon, N., Weiss,
R., Aach, J., Collins, J. J., & Church, G. M. (2015). Highly e cient Cas9-mediated transcriptional
programming. Nature Methods, 12(4), 326–328. https://doi.org/10.1038/nmeth.3312
57. Shachar, S., Voss, T. C., Pegoraro, G., Sciascia, N., & Misteli, T. (2015). Identi cation of Gene
Positioning Factors Using High-Throughput Imaging Mapping. Cell, 162(4), 911–923. https://doi.org/
10.1016/j.cell-. Miyanari, Y., Ziegler-Birling, C., & Torres-Padilla, M. E. (2013). Live visualization of chromatin dynamics
with uorescent TALEs. Nature Structural and Molecular Biology, 20(11),-. https://doi.org/
10.1038/nsmb.2680
59. Chen, B., Gilbert, L. A., Cimini, B. A., Schnitzbauer, J., Zhang, W., Li, G. W., Park, J., Blackburn, E. H.,
Weissman, J. S., Qi, L. S., & Huang, B. (2013). Dynamic imaging of genomic loci in living human cells
by an optimized CRISPR/Cas system. Cell, 155(7),-. https://doi.org/10.1016/
j.cell-. Morgan, S. L., Mariano, N. C., Bermudez, A., Arruda, N. L., Wu, F., Luo, Y., Shankar, G., Jia, L., Chen,
H., Hu, J. F., Ho man, A. R., Huang, C. C., Pitteri, S. J., & Wang, K. C. (2017). Manipulation of nuclear
architecture through CRISPR-mediated chromosomal looping. Nature Communications, 8(1), 1–9.
https://doi.org/10.1038/ncomms-. Bannister, A. J., & Kouzarides, T. (2011). Regulation of chromatin by histone modi cations. In Cell
Research (Vol. 21, Issue 3, pp. 381–395). Cell Res. https://doi.org/10.1038/cr-. Liu, X. S., Wu, H., Ji, X., Stelzer, Y., Wu, X., Czauderna, S., Shu, J., Dadon, D., Young, R. A., &
Jaenisch, R. (2016). Editing DNA Methylation in the Mammalian Genome. Cell, 167(1), 233-247.e17.
https://doi.org/10.1016/j.cell-. Xu, X., Tao, Y., Gao, X., Zhang, L., Li, X., Zou, W., Ruan, K., Wang, F., Xu, G. L., & Hu, R. (2016). A
CRISPR-based approach for targeted DNA demethylation. Cell Discovery, 2(1), 1–12. https://doi.org/
10.1038/celldisc-. Shi, Y., Lan, F., Matson, C., Mulligan, P., Whetstine, J. R., Cole, P. A., Casero, R. A., & Shi, Y. (2004).
Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell, 119(7), 941–953.
https://doi.org/10.1016/j.cell-. Zetsche, B., Volz, S. E., & Zhang, F. (2015). A split-Cas9 architecture for inducible genome editing and
transcription modulation. In Nature Biotechnology (Vol. 33, Issue 2, pp. 139–142). Nature Publishing
Group. https://doi.org/10.1038/nbt.3149
66. Nihongaki, Y., Kawano, F., Nakajima, T., & Sato, M. (2015). Photoactivatable CRISPR-Cas9 for
optogenetic genome editing. Nature Biotechnology, 33(7), 755–760. https://doi.org/10.1038/nbt.3245
Future of CRISPR/Cas9:
67. Eid, A., Alshareef, S., & Mahfouz, M. M. (2018). CRISPR base editors: Genome editing without doublestranded breaks. In Biochemical Journal (Vol. 475, Issue 11, pp-). Portland Press Ltd.
https://doi.org/10.1042/BCJ-
fi
fi
fi
ffi
ffi
ff
ff
fl
34
68. Kantor, A., McClements, M. E., & Maclaren, R. E. (2020). Crispr-cas9 dna base-editing and primeediting. In International Journal of Molecular Sciences (Vol. 21, Issue 17, pp. 1–22). MDPI AG. https://
doi.org/10.3390/ijms-. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A., & Liu, D. R. (2016). Programmable editing of a
target base in genomic DNA without double-stranded DNA cleavage. Nature,-), 420–424.
https://doi.org/10.1038/nature-. Koblan, L. W., Doman, J. L., Wilson, C., Levy, J. M., Tay, T., Newby, G. A., Maianti, J. P., Raguram, A., &
Liu, D. R. (2018). Improving cytidine and adenine base editors by expression optimization and
ancestral reconstruction. Nature Biotechnology, 36(9), 843–848. https://doi.org/10.1038/nbt.4172
71. Gaudelli, N. M., Komor, A. C., Rees, H. A., Packer, M. S., Badran, A. H., Bryson, D. I., & Liu, D. R.
(2017). Programmable base editing of T to G C in genomic DNA without DNA cleavage. Nature,-), 464–471. https://doi.org/10.1038/nature-. Richter, M. F., Zhao, K. T., Eton, E., Lapinaite, A., Newby, G. A., Thuronyi, B. W., Wilson, C., Koblan, L.
W., Zeng, J., Bauer, D. E., Doudna, J. A., & Liu, D. R. (2020). Phage-assisted evolution of an adenine
base editor with improved Cas domain compatibility and activity. Nature Biotechnology, 38(7), 883–
891. https://doi.org/10.1038/s--z
73. Anzalone, A. V., Randolph, P. B., Davis, J. R., Sousa, A. A., Koblan, L. W., Levy, J. M., Chen, P. J.,
Wilson, C., Newby, G. A., Raguram, A., & Liu, D. R. (2019). Search-and-replace genome editing without
double-strand breaks or donor DNA. Nature,-), 149–157. https://doi.org/10.1038/
s-. Hos eld, D. J., Mol, C. D., Shen, B., & Tainer, J. A. (1998). Structure of the DNA repair and replication
endonuclease and exonuclease FEN-1: Coupling DNA and PCNA binding to FEN-1 activity. Cell, 95(1),
135–146. https://doi.org/10.1016/S-. Kuscu, C., Parlak, M., Tufan, T., Yang, J., Szlachta, K., Wei, X., Mammadov, R., & Adli, M. (2017).
CRISPR-STOP: Gene silencing through base-editing-induced nonsense mutations. Nature Methods,
14(7), 710–712. https://doi.org/10.1038/nmeth.4327
76. Hao, Y., Li, J., Li, Q., Zhang, L., Shi, J., Zhang, X., Aldalbahi, A., Wang, L., Fan, C., & Wang, F. (2020).
Programmable Live-Cell CRISPR Imaging with Toehold-Switch-Mediated Strand Displacement.
Angewandte Chemie - International Edition, 59(46),-. https://doi.org/10.1002/
anie-. Marino, N. D., Pinilla-Redondo, R., Csörgő, B., & Bondy-Denomy, J. (2020). Anti-CRISPR protein
applications: natural brakes for CRISPR-Cas technologies. In Nature Methods (Vol. 17, Issue 5, pp.
471–479). Nature Research. https://doi.org/10.1038/s-. Choi, C. Q. (2021). CRISPR Meets Its Match. ACS Central Science, acscentsci.1c00427. https://
doi.org/10.1021/acscentsci.1c-. Pawluk, A., Davidson, A. R., & Maxwell, K. L. (2018). Anti-CRISPR: Discovery, mechanism and
function. Nature Reviews Microbiology, 16(1), 12–17. https://doi.org/10.1038/nrmicro-. Hynes, A. P., Rousseau, G. M., Agudelo, D., Goulet, A., Amigues, B., Loehr, J., Romero, D. A.,
Fremaux, C., Horvath, P., Doyon, Y., Cambillau, C., & Moineau, S. (2018). Widespread anti-CRISPR
proteins in virulent bacteriophages inhibit a range of Cas9 proteins. Nature Communications, 9(1), 1–
10. https://doi.org/10.1038/s--w
81. Gantz, V. M., Jasinskiene, N., Tatarenkova, O., Fazekas, A., Macias, V. M., Bier, E., & James, A. A.
(2015). Highly e cient Cas9-mediated gene drive for population modi cation of the malaria vector
mosquito Anopheles stephensi. Proceedings of the National Academy of Sciences of the United States
of America, 112(49), E6736–E6743. https://doi.org/10.1073/pnas-. Yadav, N., Narang, J., Chhillar, A. K., & Rana, J. S. (2021). CRISPR: A new paradigm of theranostics. In
Nanomedicine: Nanotechnology, Biology, and Medicine (Vol. 33). Elsevier Inc. https://doi.org/10.1016/
j.nano-. Guk, K., Keem, J. O., Hwang, S. G., Kim, H., Kang, T., Lim, E. K., & Jung, J. (2017). A facile, rapid and
sensitive detection of MRSA using a CRISPR-mediated DNA FISH method, antibody-like dCas9/
sgRNA complex. Biosensors and Bioelectronics, 95, 67–71. https://doi.org/10.1016/j.bios-. Collias, D., & Beisel, C. L. (2021). CRISPR technologies and the search for the PAM-free nuclease. In
Nature Communications (Vol. 12, Issue 1). Nature Research. https://doi.org/10.1038/
s--y
85. Nishimasu, H., Shi, X., Ishiguro, S., Gao, L., Hirano, S., Okazaki, S., Noda, T., Abudayyeh, O. O.,
Gootenberg, J. S., Mori, H., Oura, S., Holmes, B., Tanaka, M., Seki, M., Hirano, H., Aburatani, H.,
Ishitani, R., Ikawa, M., Yachie, N., … Nureki, O. (2018). Engineered CRISPR-Cas9 nuclease with
expanded targeting space. Science,-),-. https://doi.org/10.1126/science.aas9129
fi
ffi
fi
35
86. Walton, R. T., Christie, K. A., Whittaker, M. N., & Kleinstiver, B. P. (2020). Unconstrained genome
targeting with near-PAMless engineered CRISPR-Cas9 variants. Science,-), 290–296. https://
doi.org/10.1126/science.aba8853
87. https://www.sciencedaily.com/releases/2021/01/-.htm
88. Ren, Q., Sretenovic, S., Liu, S., Tang, X., Huang, L., He, Y., Liu, L., Guo, Y., Zhong, Z., Liu, G., Cheng,
Y., Zheng, X., Pan, C., Yin, D., Zhang, Y., Li, W., Qi, L., Li, C., Qi, Y., & Zhang, Y. (2021). PAM-less plant
genome editing using a CRISPR–SpRY toolbox. Nature Plants, 7(1), 25–33. https://doi.org/10.1038/
s-. Yu, Y., Wu, X., Guan, N., Shao, J., Li, H., Chen, Y., Ping, Y., Li, D., & Ye, H. (2020). Engineering a far-red
light-activated split-Cas9 system for remote-controlled genome editing of internal organs and tumors.
Science Advances, 6(28). https://doi.org/10.1126/sciadv.abb1777
90. Chen, S. P., & Wang, H. H. (2019). An Engineered Cas-Transposon System for Programmable and SiteDirected DNA Transpositions. The CRISPR Journal, 2(6), 376–394. https://doi.org/10.1089/
crispr-
Ethics of CRISPR/Cas9 Application:
91. Klap, C., Yeshayahou, E., Bolger, A. M., Arazi, T., Gupta, S. K., Shabtai, S., Usadel, B., Salts, Y., &
Barg, R. (2017). Tomato facultative parthenocarpy results from Sl AGAMOUS-LIKE 6 loss of function.
Plant Biotechnology Journal, 15(5), 634–647. https://doi.org/10.1111/pbi-. Lu, H. P., Luo, T., Fu, H. W., Wang, L., Tan, Y. Y., Huang, J. Z., Wang, Q., Ye, G. Y., Gatehouse, A. M. R.,
Lou, Y. G., & Shu, Q. Y. (2018). Resistance of rice to insect pests mediated by suppression of serotonin
biosynthesis. Nature Plants, 4(6), 338–344. https://doi.org/10.1038/s-. Salesse-Smith, C. E., Sharwood, R. E., Busch, F. A., Kromdijk, J., Bardal, V., & Stern, D. B. (2018).
Overexpression of Rubisco subunits with RAF1 increases Rubisco content in maize. Nature Plants,
4(10), 802–810. https://doi.org/10.1038/s-. Min, Y. L., Li, H., Rodriguez-Caycedo, C., Mireault, A. A., Huang, J., Shelton, J. M., McAnally, J. R.,
Amoasii, L., Mammen, P. P. A., Bassel-Duby, R., & Olson, E. N. (2019). CRISPR-Cas9 corrects
Duchenne muscular dystrophy exon 44 deletion mutations in mice and human cells. Science
Advances, 5(3). https://doi.org/10.1126/sciadv.aav4324
95. Bjursell, M., Porritt, M. J., Ericson, E., Taheri-Ghahfarokhi, A., Clausen, M., Magnusson, L., Admyre, T.,
Nitsch, R., Mayr, L., Aasehaug, L., Seeliger, F., Maresca, M., Bohlooly-Y, M., & Wiseman, J. (2018).
Therapeutic Genome Editing With CRISPR/Cas9 in a Humanized Mouse Model Ameliorates α1antitrypsin De ciency Phenotype. EBioMedicine, 29, 104–111. https://doi.org/10.1016/
j.ebiom-. Ohmori, T., Mizukami, H., Ozawa, K., Sakata, Y., & Nishimura, S. (2015). New approaches to gene and
cell therapy for hemophilia. Journal of Thrombosis and Haemostasis, 13(S1), S133–S142. https://
doi.org/10.1111/jth-. Traylen, C. M., Patel, H. R., Fondaw, W., Mahatme, S., Williams, J. F., Walker, L. R., Dyson, O. F., Arce,
S., & Akula, S. M. (2011). Virus reactivation: A panoramic view in human infections. In Future Virology
(Vol. 6, Issue 4, pp. 451–463). Future Virol. https://doi.org/10.2217/fvl.11.21
98. Ding, X., Yin, K., Li, Z., & Liu, C. (2020). All-in-One Dual CRISPR-Cas12a (AIOD-CRISPR) Assay: A
Case for Rapid, Ultrasensitive and Visual Detection of Novel Coronavirus SARS-CoV-2 and HIV virus.
In bioRxiv (p-). bioRxiv. https://doi.org/10.1101/-. Huang, C. H., Lee, K. C., & Doudna, J. A. (2018). Applications of CRISPR-Cas Enzymes in Cancer
Therapeutics and Detection. In Trends in Cancer (Vol. 4, Issue 7, pp. 499–512). Cell Press. https://
doi.org/10.1016/j.trecan-. Bu, X., Kato, J., Hong, J. A., Merino, M. J., Schrump, D. S., Lund, F. E., & Moss, J. (2018). CD38
knockout suppresses tumorigenesis in mice and clonogenic growth of human lung cancer cells.
Carcinogenesis, 39(2), 242–251. https://doi.org/10.1093/carcin/bgx137
101. Takeda, H., Kataoka, S., Nakayama, M., Ali, M. A. E., Oshima, H., Yamamoto, D., Park, J. W.,
Takegami, Y., An, T., Jenkins, N. A., Copeland, N. G., & Oshima, M. (2019). CRISPR-Cas9-mediated
gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver
genes. Proceedings of the National Academy of Sciences of the United States of America, 116(31),-. https://doi.org/10.1073/pnas-. Eyquem, J., Mansilla-Soto, J., Giavridis, T., Van Der Stegen, S. J. C., Hamieh, M., Cunanan, K. M.,
Odak, A., Gönen, M., & Sadelain, M. (2017). Targeting a CAR to the TRAC locus with CRISPR/Cas9
enhances tumour rejection. Nature,-), 113–117. https://doi.org/10.1038/nature-. Nidhi, S., Anand, U., Oleksak, P., Tripathi, P., Lal, J. A., Thomas, G., Kuca, K., & Tripathi, V. (2021).
Novel crispr–cas systems: An updated review of the current achievements, applications, and future
fi
36
-.
research perspectives. In International Journal of Molecular Sciences (Vol. 22, Issue 7). MDPI AG.
https://doi.org/10.3390/ijms-
Cyranoski, D. (2018). CRISPR-baby scientist fails to satisfy critics. In Nature (Vol. 564, Issue 7734, pp.
13–14). Nature Research. https://doi.org/10.1038/d--w
Cyranoski, D. (2020). What CRISPR-baby prison sentences mean for research. In Nature (Vol. 577,
Issue 7789, pp. 154–155). NLM (Medline). https://doi.org/10.1038/d--y
Sand, M., Bredenoord, A. L., & Jongsma, K. R. (2019). After the fact—the case of CRISPR babies. In
European Journal of Human Genetics (Vol. 27, Issue 11, pp-). Nature Publishing Group.
https://doi.org/10.1038/s-
Doudna, J. (2015). Genome-editing revolution: My whirlwind year with CRISPR. In Nature (Vol. 528,
Issue 7583, pp. 469–471). Nature Publishing Group. https://doi.org/10.1038/528469a
CRISPR in Context: The new world of Human genetic engineering, World Science festival, https://
www.youtube.com/watch?v=RNRZchHaKgw
37