CRISPR/Cas9 technology for targeted genome editing
CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) are the segments of prokaryotic DNA containing short repeats in its nucleotide sequence. Today we know that this is a bacterial protection system against viral DNA. The molecular components of CRISPR/Cas9 system have been used for a...
Gespeichert in:
Datum: | 2015 |
---|---|
Hauptverfasser: | , , |
Format: | Artikel |
Sprache: | English |
Veröffentlicht: |
Інститут молекулярної біології і генетики НАН України
2015
|
Schriftenreihe: | Вiopolymers and Cell |
Schlagworte: | |
Online Zugang: | http://dspace.nbuv.gov.ua/handle/123456789/152569 |
Tags: |
Tag hinzufügen
Keine Tags, Fügen Sie den ersten Tag hinzu!
|
Назва журналу: | Digital Library of Periodicals of National Academy of Sciences of Ukraine |
Zitieren: | CRISPR/Cas9 technology for targeted genome editing / N.A. Lomov, V.V. Borunova, M.A. Rubtsov // Вiopolymers and Cell. — 2015. — Т. 31, № 4. — С. 243-248. — Бібліогр.: 32 назв. — англ. |
Institution
Digital Library of Periodicals of National Academy of Sciences of Ukraineid |
irk-123456789-152569 |
---|---|
record_format |
dspace |
spelling |
irk-123456789-1525692019-06-13T01:26:25Z CRISPR/Cas9 technology for targeted genome editing Lomov, N.A. Borunova, V.V. Rubtsov, M.A. Reviews CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) are the segments of prokaryotic DNA containing short repeats in its nucleotide sequence. Today we know that this is a bacterial protection system against viral DNA. The molecular components of CRISPR/Cas9 system have been used for a gene editing in eukaryotes since 2013. But as any other method it also has the limitations and drawbacks. Here we are going to review the history of CRISPR biology and to discuss the possibilities that this new technology provides to researchers as well as the prospects for its use in the medical research and treatment. CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) – послідовності в геномі прокариот, які складаються з коротких повторів, що перемежовуються унікальними послідовностями. Це система бактеріальної захисту від вірусної ДНК. Молекулярні компоненти даної системи з 2013 року використовуються як інструмент редагування еукаріотічесого геному, хоча дана технологія і має деякі обмеження і недоліки. У даному огляді ми торкнемося історію застосування системи CRISPR / Cas9 і обговоримо можливості, які дана технологія надає для дослідження і лікування різних захворювань. CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) – последовательности в геноме прокариот, которые состоят из коротких повторов, перемежающихся уникальными последовательностями. Это система бактериальной защиты от вирусной ДНК. Молекулярные компоненты данной системы с 2013 года используются как инструмент редактирования эукариотического генома, хотя данная технология и имеет некоторые ограничения и недостатки. В данном обзоре мы затронем историю применения системы CRISPR/Cas9 и обсудим возможности, которые данная технология предоставляет для исследования и лечения различных заболеваний. 2015 Article CRISPR/Cas9 technology for targeted genome editing / N.A. Lomov, V.V. Borunova, M.A. Rubtsov // Вiopolymers and Cell. — 2015. — Т. 31, № 4. — С. 243-248. — Бібліогр.: 32 назв. — англ. 0233-7657 DOI: http://dx.doi.org/10.7124/bc.0008E7 http://dspace.nbuv.gov.ua/handle/123456789/152569 577.21 en Вiopolymers and Cell Інститут молекулярної біології і генетики НАН України |
institution |
Digital Library of Periodicals of National Academy of Sciences of Ukraine |
collection |
DSpace DC |
language |
English |
topic |
Reviews Reviews |
spellingShingle |
Reviews Reviews Lomov, N.A. Borunova, V.V. Rubtsov, M.A. CRISPR/Cas9 technology for targeted genome editing Вiopolymers and Cell |
description |
CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) are the segments of prokaryotic DNA containing short repeats in its nucleotide sequence. Today we know that this is a bacterial protection system against viral DNA. The molecular components of CRISPR/Cas9 system have been used for a gene editing in eukaryotes since 2013. But as any other method it also has the limitations and drawbacks. Here we are going to review the history of CRISPR biology and to discuss the possibilities that this new technology provides to researchers as well as the prospects for its use in the medical research and treatment. |
format |
Article |
author |
Lomov, N.A. Borunova, V.V. Rubtsov, M.A. |
author_facet |
Lomov, N.A. Borunova, V.V. Rubtsov, M.A. |
author_sort |
Lomov, N.A. |
title |
CRISPR/Cas9 technology for targeted genome editing |
title_short |
CRISPR/Cas9 technology for targeted genome editing |
title_full |
CRISPR/Cas9 technology for targeted genome editing |
title_fullStr |
CRISPR/Cas9 technology for targeted genome editing |
title_full_unstemmed |
CRISPR/Cas9 technology for targeted genome editing |
title_sort |
crispr/cas9 technology for targeted genome editing |
publisher |
Інститут молекулярної біології і генетики НАН України |
publishDate |
2015 |
topic_facet |
Reviews |
url |
http://dspace.nbuv.gov.ua/handle/123456789/152569 |
citation_txt |
CRISPR/Cas9 technology for targeted genome editing / N.A. Lomov, V.V. Borunova, M.A. Rubtsov // Вiopolymers and Cell. — 2015. — Т. 31, № 4. — С. 243-248. — Бібліогр.: 32 назв. — англ. |
series |
Вiopolymers and Cell |
work_keys_str_mv |
AT lomovna crisprcas9technologyfortargetedgenomeediting AT borunovavv crisprcas9technologyfortargetedgenomeediting AT rubtsovma crisprcas9technologyfortargetedgenomeediting |
first_indexed |
2025-07-14T04:02:16Z |
last_indexed |
2025-07-14T04:02:16Z |
_version_ |
1837593512682455040 |
fulltext |
243
ISSN 0233-7657
Biopolymers and Cell. 2015. Vol. 31. N 4. P. 243–248
doi: http://dx.doi.org/10.7124/bc.0008E7
Reviews
UDC 577.21
CRISPR/Cas9 technology for targeted genome editing
N. A. Lomov1, 3, V. V. Borunova1, 3, M. A. Rubtsov1, 2, 3
1 M. V. Lomonosov Moscow State University,
Leninskie Gory, 1/12, Moscow, Russian Federation, 119991
2 I. M. Sechenov First Moscow State Medical University,
8, Trubetskaya Str. Moscow, Russian Federation, 119991
3 LIA 1066 French-Russian Joint Cancer Research Laboratory
Villejuif, France–Moscow, Russian Federation
ma_rubtsov@mail.ru
CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) are the segments of prokary-
otic DNA containing short repeats in its nucleotide sequence. Today we know that this is a bacterial
protection system against viral DNA. The molecular components of CRISPR/Cas9 system have been
used for a gene editing in eukaryotes since 2013. But as any other method it also has the limitations
and drawbacks. Here we are going to review the history of CRISPR biology and to discuss the possi-
bilities that this new technology provides to researchers as well as the prospects for its use in the
medical research and treatment.
K e y w o r d s: CRISPR/Cas9, genome targeting, genome editing, personalized therapy, chromosomal
translocations, DNA repair.
© 2015 N. A. Lomov et al.; Published by the Institute of Molecular Biology and Genetics, NAS of Ukraine on behalf of Biopolymers and Cell.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/),
which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited
History
The specifi c sequences in bacterial genomes, called
CRISPR, were discovered in 1987. They consist of
repeats, alternating by unique sequences or «spac-
ers» [1] (Fig. 1, A). Its function has been unsolved
until 2005 when it was found that these «spacers»
have a sequence equal to a viral DNA. It was also
observed that there are the CRISPR-associated
genes and these genes encode nucleases. After-
wards it was supposed that the bacteria use these
nucleases to cleave a foreign DNA wherein the
spacer sequence is used as an example of which ex-
actly DNA sequence is necessary to be cut [2, 3].
This hypothesis was experimentally proven in 2007
[4]. A detailed mechanism of the complex matura-
tion and directed cleavage of DNA was fi nally stud-
ied in 2011 [5] (Fig. 1, B). It was found that the
Cas9 enzyme formed a ternary complex with crRNA
and tracrRNA encoded in the cluster. This complex
unwinds DNA and introduce a double-stranded
break in DNA if crRNA binds complementary to
one of the DNA chains (Fig. 2, A). A year later it
was shown that crRNA can be joint to tracrRNA
and that this «single guide» RNA (sgRNA) together
with the Cas9 enzyme might be used to modify the
genome of eukaryotes [6] (Fig. 2, B)..This idea was
realized in 2013 and since then thousands of papers
where such technology was applied have been pub-
lished [7–9].
The approaches to targeted DNA breakage based
on the usage of zinc fi nger nucleases (ZFNs) and
TAL effector nucleases (TALENs) were developed
much earlier in 2003 and 2009 respectively. Both
methods are based on the DNA recognition by the
specifi c proteins designed individually for each par-
244
N. A. Lomov, V. V. Borunova, M. A. Rubtsov
ticular sequence [10, 11]. Compared to the these
technologies the CRISPR/Cas9 system has proved to
be much more simple, available, affordable and three
to four times more effi cient [12].
The basic principle
of CRISPR/Cas9 technology
At the heart of all the methods that use the CRISPR/
Cas9 system lies the possibility to introduce a break
in DNA in an exactly desired locus. It’s enough to
deliver the Cas9 gene (or its mRNA) together with
sgRNA into the cell, wherein sgRNA at its 5’-end
should have 20 nucleotide sequence complimentary
to a target DNA fragment. There is also easy way to
introduce the multiple breaks at the same time – just
to use several different sgRNAs [13].
Application of CRISPR/Cas9 technology
The fi rst example of application of the CRISPR/
Cas9 technology is the modeling of tumor-associat-
ed chromosomal translocations: CRISPR/Cas9 in-
duces the double-stranded DNA breaks (DSBs) in
the precise loci which are known to participate in the
oncogenic rearrangements. Typically, DSBs used to
be repaired by the mechanism of non-homologous
end joining (NHEJ). This error prone mechanism fre-
quently leads to joining the DNA ends belonging to
different chromosomes [14]. The cellular models of
certain types of cancer might be obtained using this
method. Inducing the human specifi c types of cancer
in laboratory animals is also possible. So, the mu rine
model of non-small-cell lung cancer (NSCLC) was
developed. Using lentiviral delivery the Cas9 gene
and sgRNA were brought into lung cells in order to
induce a cleavage of the murine endogenous Eml4
and Alk loci and to generate the Eml4-Alk fused gene
found recurrently in NSCLCs. The result was the de-
velopment of the disease in the treated mice [15]. To
date the murine in vivo models of human leukemia
(namely, AML) have also been developed [16, 17].
These and other similar murine models can be uti-
lized to study the carcinogenesis and to test new
treatment approaches.
The next purpose which might be successfully
achieved by application of the CRISPR/Cas9 technol-
ogy is knocking genes out. The DSB formation often
leads to the deletion or insertion of several nucleotides
in an initial sequence even in case of effi cient DSB
repair [18]. Thus, introducing a break into a particular
gene one may cause its knockout. So the Cas9-ex-
pressing cell line treated with various sgRNA can be
used for searching for the genes responsible for a spe-
cifi c disease. A few years earlier each gene knockout
was a complicated time-consuming procedure, but in
case of CRISPR/Cas9 it requires only to treat the
Cas9-expressing cells with a specifi c sgRNA [19].
In addition, one may insert the desired DNA frag-
ment into the specifi c broken DNA site. There are
Fig. 2. A – Targeted cleavage of DNA by a ternary complex of
Cas9 with crRNA and tracrRNA. B – Targeted cleavage of DNA
by a complex of Cas9 with «single guide» RNA (sgRNA)
Fig. 1. A – The overall structure of the CRISPR loci. B – Mech-
anism of CRISPR/Cas9 complex maturation
245
CRISPR/Cas9 technology for targeted genome editing
two major mechanisms of DSB repair: homology-
directed repair (HDR) and non-homologous end
joining (NHEJ). HDR requires a fragment of DNA
homologous to the damaged one as a template. Oth-
erwise, in the absence of such a fragment, NHEJ
takes place. It is possible to achieve insertion of a
desired DNA fragment in both cases. If a DNA frag-
ment with homology arms surrounding a desired
gene is available, it will be inserted via HDR. In case
of the NHEJ repair it is suffi cient to deliver a linear
DNA fragment with the ends protected from the ac-
tion of cellular exonucleases. There is a chance that
this fragment will be joined to the ends of the break.
So, the CRISPR/Cas9 technology makes possible
the insertion of the desired DNA fragment into the ge-
nome. It is widely used in scientifi c research and offers
great opportunities for the treatment of hereditary dis-
eases and viral infections. For example, this technology
allows generation of the stable cell lines with the de-
sired inserts in the genome. The CRISPR/Cas9 gene
knocking occurs with a much higher effi ciency than the
random integration of a plasmid into the genome and
solely at the particular pre-selected site in almost 100 %
of cases. Until recently the generation of a stable cell
line usually took at least few months, with the CRISPR/
Cas9 technology – up to one month [20].
Moreover, until recently, the gene therapy implied
the insertion of the desired gene into the random place
in the genome. In its turn, the CRISPR/Cas9 system
allows changes in a certain place, where one may not
only insert the gene but also remove or replace the
certain DNA fragments. Several in vivo studies in
mice have shown the possibility of gene therapy by
means of the CRISPR/Cas9 application. A deletion of
1 bp in the CRYGC gene is known to cause the cata-
ract in mice. After injection of the Cas9 mRNA,
sgRNA and the DNA fragment containing a wild-type
allele into the mouse embryos with this mutation the
mousekins were born healthy [21]. In a recent work
the researchers have successfully applied the CRISPR/
Cas9 system in order to reduce the level of cholesterol
in the blood of mice by turning off the PCSK9 gene in
the liver cells. They used adenovirus particles to de-
liver the Cas9 gene and sgRNA into hepatocytes. As a
result more than 50 % of hepatocytes were «edited»
and it resulted in a decrease in the cholesterol level to
a value that is typical for the PCSK9 knockout mice
[22]. Striking results were achieved with CRISPR/
Cas9 in the prevention of Duchene’s muscular dystro-
phy (DMD) in mice by correcting the dmd dystrophin
gene. A copy of the normal dmd gene was integrated
in the zygotes via the CRISPR/Cas9 system. The born
animals were mosaic in the dmd gene but nonetheless
even a partial gene editing has prevented the disease
development [23].
The ability to deliver the CRISPR/Cas9 system
elements into the brain cells has been also recently
demonstrated. Because of the packaging size limita-
tion of the adeno-associated viral (AAV) vectors, the
dual-vector system was designed composed of two se-
parate vectors that package the SpCas9 (AAV-SpCas9)
and sgRNA (AAV-SpGuide) expression cassettes.
However, the co-transfection effi ciency was shown
to be rather high [19].
Taken together, these in vivo studies on animals
prove the excellent prospects for the genome editing
via application of the CRISPR/Cas9 technology.
On the other hand, many studies ex vivo show the
ability to edit DNA in human cells. The particular
mutations in the CCR5 gene which occur in some
people are known to make it resistant to HIV. A re-
cent study has established that the CRISPR/Cas9-
based editing of CCR5 in the induced pluripotent
stem cells (iPSCs) may lead to the HIV-1 resistance
of descendant lymphocytes [12].
The possibility to treat the latent virus infection
has also been recently demonstrated: the patient-de-
rived cells from a Burkitt’s lymphoma with latent
Epstein–Barr virus infection showed a proliferation
arrest and a concomitant decrease in viral load after
targeting the viral genome by the CRISPR/Cas9 sys-
tem elements [24]. Finally, the healthy gene copy
was introduced in cells of the patients with cystic fi -
brosis via CRISPR/Cas9. As a result the cells dem-
onstrated a healthy phenotype [25].
Taken together with the fact demonstrating that
the «in vitro corrected» organs might be successfully
transplanted back into the body in mice [26], the
above experiments give us a hope for the successful
application of this experience in the treatment of pa-
246
N. A. Lomov, V. V. Borunova, M. A. Rubtsov
tients. This considerably strengthens the position of
gene therapy among the advanced treatments of the
genetic and viral diseases in humans.
An especial theme is the editing of the human em-
bryo genome. Such possibility opens up broad pros-
pects for the prenatal correction of the «broken»
genes. The researchers from Sun Yat-sen University,
China, have already tested this potential. They used
triplonuclear zygotes (for ethical reasons) and re-
placed the HBB gene using the CRISPR/Cas9 tech-
nique. As a result it was shown that: 1) the effective-
ness of the replacement of a normal gene copy by
means of HDR in these cells was relatively low; 2) the
edited embryos were mosaic; 3) the mutations in the
genome located in not-planned loci was identifi ed (so-
called «off-target» mutations); 4) another high ly ho-
mologous gene (HBD) could serve as a donor tem-
plate for HDR. However, despite the diffi culties this
study shows the fundamental possibility of the ge-
nome editing in human embryos [27].
Drawbacks of CRISPR/Cas9 technology
The common problem of all genome editing meth-
ods is so called «off-target» effects – when a nucle-
ase cleaves DNA not in the place where it was in-
tended. It may impair the function of any incidental
gene or may lead to integration of the desired gene in
the wrong place and genome surrounding. In case of
human embryos such a risk is absolutely unaccepta-
ble. In the abovementioned work with the human
embryos several off-target mutations have been fo-
und and documented, but later the authors noticed
that three out of four of them were polymorphisms
already presented in the germline and were not as-
sociated with the CRISPR/Cas9 action. The special
studies show that the level of off-target mutations in
case of the CRISPR/Cas9 application is in fact ex-
tremely low [28]. Today, several methods for meas-
uring the off-target editing have been developed. The
fi rst method is based on the fl uorescent in situ hy-
bridization (FISH): one fl uorescent probe anneals to
the insertion fragment and another fl uorescent probe
detects the insertion site. Normally these two probes
have to be colocalized otherwise it is the erroneous
insertion [29]. Other method (GUIDE-seq) is based
on the addition of short DNA fragments which would
be integrated at the breaks. These fragments of DNA
are then used as anchors when surrounding DNA is
amplifi ed by PCR and sequenced. So one we can see
all sites of insertion of these fragments and count
off-target mutations [30]. These eva luation methods
have shown that not all of the off-target sites might
be predicted by special computer software and that
the different sgRNA provides a different number of
these effects.
The various methods to improve the accuracy and
effi ciency of insertion were proposed. For example,
in order to obtain a higher specifi city a «double nick-
ing» method might be used [31]. It consists in spe-
cial modifi cation of Cas9 resulting in an ability of
the enzyme to introduce break in only one DNA
strand («nick»). Thus, for the formation of a double-
stranded break two such nickases should work vis-a-
vis. Furthermore, each of the enzymes has to bind its
own sgRNA which should recognize its own (mutu-
ally complementary) DNA sequence. Thus, the prob-
ability of double-stranded breakage in the off-target
sites reduces dramatically.
In order to increase the effi ciency of the insertion
in the genome the HDR mechanism should prevail.
To achieve this goal the NHEJ-inhibiting agents
might be used. In particular DNA ligase IV which
plays an important role in NHEJ might be turned off
by using the Scr7 agent. This may increase the effi -
cacy of DNA integration up to 19 times [32].
The selection of sgRNA sequences should be per-
formed in such a way to reduce the probability of the
off-target effects. The special algorithms have been
developed for this purpose (CRISPR Design Tool:
http://tools.genome-engineering.org). If there is need
for especial accuracy it is necessary to check a num-
ber of the off-target sites by applying the GUIDE-
seq technique.
Finally, the integration of new genes requires a
special attention when selecting the integration site.
Such target should be a locus where the inserted DNA
fragment would be «protected» against the epige-
netic effects, would work in a predictable manner
and would not cause negative effects on a cell, for
example, induce carcinogenesis. Such loci are called
247
CRISPR/Cas9 technology for targeted genome editing
«genomic safe harbors» (GSHs). They should be lo-
cated far from other genes especially protooncogenes
and at the same time should be outside the transcrip-
tion units and the ultra-conserved DNA regions. The
sites suitable as GSHs might be identifi ed by the bio-
informatics methods but also require experimental
confi rmation [33].
Conclusion
So far we can say with confi dence that the CRISPR/
Cas9-based technologies have revolutionized the bio-
medical science and are spreading at a remarkable
speed by virtue of their simplicity and effi ciency.
The CRISPR/Cas9 application dramatically facili-
tates the manipulation with DNA in terms of provid-
ing the targeted genome editing, foremost deletions
or gene replacement. Despite some drawbacks this
technology has a wide perspective in the fi elds of
basic research, biotechnology, translational and per-
sonalized medicine.
Acknowledgments
N. A. L., V. V. B., and M. A. R.: RFBR (grants 14-04-
93105-CNRS_a, 15-54-16007-CNRS_a).
Competing interests
The authors declare no competing interests.
REFERENCES
1. Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A.
Nucleotide sequence of the iap gene, responsible for alka-
line phosphatase isozyme conversion in Escherichia coli,
and identifi cation of the gene product. J Bacteriol. 1987;169
(12):5429–33.
2. Bolotin A, Quinquis B, Sorokin A, Ehrlich SD. Clustered re-
gularly interspaced short palindrome repeats (CRISPRs) ha-
ve spacers of extrachromosomal origin. Microbiology. 2005;
151(Pt 8):2551–61.
3. Makarova KS, Grishin NV, Shabalina SA, Wolf YI, Koonin EV.
A putative RNA-interference-based immune system in pro-
karyotes: computational analysis of the predicted enzymatic
machinery, functional analogies with eukaryotic RNAi, and
hypothetical mechanisms of action. Biol Direct. 2006;1:7.
4. Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P,
Moineau S, Romero DA, Horvath P. CRISPR provides acqu-
i red resistance against viruses in prokaryotes. Science. 2007;
315(5819):1709–12.
5. Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao
Y, Pirzada ZA, Eckert MR, Vogel J, Charpentier E. CRISPR
RNA maturation by trans-encoded small RNA and host fac-
tor RNase III. Nature. 2011;471(7340):602–7.
6. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Cha-
rpentier E. A programmable dual-RNA-guided DNA en do-
nuc lease in adaptive bacterial immunity. Science. 2012;337
(6096):816–21.
7. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD,
Wu X, Jiang W, Marraffi ni LA, Zhang F. Multiplex genome
engineering using CRISPR/Cas systems. Science. 2013; 339
(6121):819–23.
8. Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE,
Norville JE, Church GM. RNA-guided human genome en-
gineering via Cas9. Science. 2013;339(6121):823–6.
9. Jinek M, East A, Cheng A, Lin S, Ma E, Doudna J. RNA-
pro grammed genome editing in human cells. Elife. 2013; 2:
e00471.
10. Bibikova M, Beumer K, Trautman JK, Carroll D. Enhancing
gene targeting with designed zinc fi nger nucleases. Science.
2003;300(5620):764.
11. Christian M, Cermak T, Doyle EL, Schmidt C, Zhang F, Hu-
mmel A, Bogdanove AJ, Voytas DF. Targeting DNA double-
strand breaks with TAL effector nucleases. Genetics. 2010;
186(2):757–61.
12. Ye L, Wang J, Beyer AI, Teque F, Cradick TJ, Qi Z, Chang
JC, Bao G, Muench MO, Yu J, Levy JA, Kan YW. Seamless
modifi cation of wild-type induced pluripotent stem cells to
the natural CCR5Δ32 mutation confers resistance to HIV
infection. Proc Natl Acad Sci U S A. 2014;111(26):9591–6.
13. Kabadi AM, Ousterout DG, Hilton IB, Gersbach CA. Multi-
plex CRISPR/Cas9-based genome engineering from a sin-
gle lentiviral vector. Nucleic Acids Res. 2014;42(19):e147.
14. Lieber MR, Gu J, Lu H, Shimazaki N, Tsai AG. Nonhomolo-
gous DNA end joining (NHEJ) and chromosomal transloca-
tions in humans. Subcell Biochem. 2010;50:279–96.
15. Blasco RB, Karaca E, Ambrogio C, Cheong TC, Karayol E,
Minero VG, Voena C, Chiarle R. Simple and rapid in vivo
generation of chromosomal rearrangements using CRISPR/
Cas9 technology. Cell Rep. 2014;9(4):1219–27.
16. Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram
RV, McConkey ME, Thielke A, Aster JC, Regev A, Ebert BL.
Generation of mouse models of myeloid malignancy with
combinatorial genetic lesions using CRISPR-Cas9 genome
editing. Nat Biotechnol. 2014;32(9):941–6.
17. Chen C, Liu Y, Rappaport AR, Kitzing T, Schultz N, Zhao Z,
Shroff AS, Dickins RA, Vakoc CR, Bradner JE, Stock W,
LeBeau MM, Shannon KM, Kogan S, Zuber J, Lowe SW.
MLL3 is a haploinsuffi cient 7q tumor suppressor in acute
myeloid leukemia. Cancer Cell. 2014;25(5):652–65.
18. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F.
Genome engineering using the CRISPR-Cas9 system. Nat
Protoc. 2013;8(11):2281–308.
248
N. A. Lomov, V. V. Borunova, M. A. Rubtsov
19. Swiech L, Heidenreich M, Banerjee A, Habib N, Li Y, Trom-
betta J, Sur M, Zhang F. In vivo interrogation of gene func-
tion in the mammalian brain using CRISPR-Cas9. Nat Bio-
technol. 2015;33(1):102–6.
20. Merkle FT, Neuhausser WM, Santos D, Valen E, Gagnon JA,
Maas K, Sandoe J, Schier AF, Eggan K. Effi cient CRISPR-
Cas9-mediated generation of knockin human pluripotent stem
cells lacking undesired mutations at the targeted locus. Cell
Rep. 2015;11(6):875–83.
21. Wu Y, Liang D, Wang Y, Bai M, Tang W, Bao S, Yan Z, Li D,
Li J. Correction of a genetic disease in mouse via use of
CRISPR-Cas9. Cell Stem Cell. 2013;13(6):659–62.
22. Ding Q, Strong A, Patel KM, Ng SL, Gosis BS, Regan SN,
Cowan CA, Rader DJ, Musunuru K. Permanent alteration of
PCSK9 with in vivo CRISPR-Cas9 genome editing. Circ
Res. 2014;115(5):488–92.
23. Long C, McAnally JR, Shelton JM, Mireault AA, Bassel-
Duby R, Olson EN. Prevention of muscular dystrophy in
mice by CRISPR/Cas9-mediated editing of germline DNA.
Science. 2014;345(6201):1184–8.
24. Wang J, Quake SR. RNA-guided endonuclease provides a
therapeutic strategy to cure latent herpesviridae infection.
Proc Natl Acad Sci U S A. 2014;111(36):13157–62.
25. Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demir-
can T, Sasaki N, Boymans S, Cuppen E, van der Ent CK,
Nieuwenhuis EE, Beekman JM, Clevers H. Functional re-
pair of CFTR by CRISPR/Cas9 in intestinal stem cell orga-
noids of cystic fi brosis patients. Cell Stem Cell. 2013;13(6):
653–8.
26. Yui S, Nakamura T, Sato T, Nemoto Y, Mizutani T, Zheng X,
Ichinose S, Nagaishi T, Okamoto R, Tsuchiya K, Clevers H,
Watanabe M. Functional engraftment of colon epithelium
expanded in vitro from a single adult Lgr5+ stem cell. Nat
Med. 2012;18(4):618–23.
27. Liang P, Xu Y, Zhang X, Ding C, Huang R, Zhang Z, Lv J,
Xie X, Chen Y, Li Y, Sun Y, Bai Y, Songyang Z, Ma W, Zhou
C, Huang J. CRISPR/Cas9-mediated gene editing in human
tripronuclear zygotes. Protein Cell. 2015;6(5):363–72.
28. Iyer V, Shen B, Zhang W, Hodgkins A, Keane T, Huang X,
Skarnes WC. Off-target mutations are rare in Cas9-modifi ed
mice. Nat Methods. 2015;12(6):479.
29. Paulis M, Castelli A, Lizier M, Susani L, Lucchini F, Villa A,
Vezzoni P. A pre-screening FISH-based method to detect
CRISPR/Cas9 off-targets in mouse embryonic stem cells.
Sci Rep. 2015;5:12327.
30. Tsai SQ, Zheng Z, Nguyen NT, Liebers M, Topkar VV, Tha-
par V, Wyvekens N, Khayter C, Iafrate AJ, Le LP, Aryee MJ,
Joung JK. GUIDE-seq enables genome-wide profi ling of
off-target cleavage by CRISPR-Cas nucleases. Nat Biotech-
nol. 2015;33(2):187–97.
31. Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S,
Tre vino AE, Scott DA, Inoue A, Matoba S, Zhang Y, Zhang F.
Double nicking by RNA-guided CRISPR Cas9 for enhanced
genome editing specifi city. Cell. 2013;154(6): 1380–9.
32. Maruyama T, Dougan SK, Truttmann MC, Bilate AM, In-
gram JR, Ploegh HL. Increasing the effi ciency of precise
genome editing with CRISPR-Cas9 by inhibition of nonho-
mologous end joining. Nat Biotechnol. 2015;33(5):538–42.
CRISPR/Cas9 технологія
для цільового редагування геному
Н. А. Ломов, В. В. Борунова, М. А. Рубцов
CRISPRs (Clustered Regularly Interspaced Short Palindromic
Repeats) – послідовності в геномі прокариот, які складають-
ся з коротких повторів, що перемежовуються унікальними
послідовностями. Це система бактеріальної захисту від ві-
русної ДНК. Молекулярні компоненти даної системи з 2013
року використовуються як інструмент редагування еукаріо-
тічесого геному, хоча дана технологія і має деякі обмежен-
ня і недоліки. У даному огляді ми торкнемося історію за-
стосування системи CRISPR / Cas9 і обговоримо можливос-
ті, які дана технологія надає для дослідження і лікування
різних захворювань.
Ключевые слова: CRISPR/Cas9, тагретінг генома,
редагування геному, персоналізована терапія, хромосомні
транслокації, репарація ДНК.
CRISPR/Cas9 технология
для целевого редактирования генома
Н. А. Ломов, В. В. Борунова, М. А. Рубцов
CRISPRs (Clustered Regularly Interspaced Short Palindromic
Repeats) – последовательности в геноме прокариот, которые
состоят из коротких повторов, перемежающихся уникаль-
ными последовательностями. Это система бактериальной
защиты от вирусной ДНК. Молекулярные компоненты дан-
ной системы с 2013 года используются как инструмент ре-
дактирования эукариотического генома, хотя данная техно-
логия и имеет некоторые ограничения и недостатки. В дан-
ном обзоре мы затронем историю применения системы
CRISPR/Cas9 и обсудим возможности, которые данная тех-
нология предоставляет для исследования и лечения различ-
ных заболеваний.
Ключевые слова: CRISPR/Cas9, таргетинг генома,
редактирование генома, персонализированая терапия,
хромосоные транслокации, репарация ДНК.
Received 01.07.2015
|