You are on page 1of 6

Genetics: Current Commentary

Genetically Modified Babies and a First


Application of Clustered Regularly
Interspaced Short Palindromic
Repeats (CRISPR-Cas9)
Downloaded from http://journals.lww.com/greenjournal by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8KKGKV0Ymy+78= on 04/21/2021

Bruce I. Rose, MD, PhD, and Samuel Brown, MD

require to enter cells.1 This genetic modification


The world’s first babies with CRISPR-Cas9 (Clustered
was done on the embryos to protect the resulting
Regularly Interspaced Short Palindromic Repeats)–
children from contracting HIV. The children’s father
edited genes were born on November 25, 2018. Dr.
had HIV.
Jiankui He of Southern University of Science and Tech-
Although this experiment has not yet been
nology in Shenzhen performed this gene editing. Dr. He’s
presented in a peer-reviewed scientific journal, it
objectives and an assessment of how well they were
achieved are discussed in the context of existing research
has been extensively discussed in both lay and
in this area. science-oriented media. It seems that genetic engi-
(Obstet Gynecol 2019;134:157–62)
neering of human embryos is “here” despite its lim-
DOI: 10.1097/AOG.0000000000003327
ited public support. Ed Yong reported in The
Atlantic2 that modifying germ cells before human
implantation is illegal in 15 of 22 countries in West-

O n November 26, 2018, at a gene editing con-


ference in Hong Kong, Dr. Jiankui He
announced the delivery of twins Lulu and Nina, con-
ern Europe. In the United States, the U.S. Food and
Drug Administration is prohibited from even review-
ing any drug or biological product involving the
ceived with embryos that had been genetically mod- modification of a human embryo.3 The scientific
ified using Clustered Regularly Interspaced Short community has recently expressed hesitation about
Palindromic Repeats (CRISPR-Cas9). Dr. He re- the long-term safety of modifying germ cells that will
ported that he altered the gene CCR5 in those become babies and has recommended proceeding
embryos. This gene produces a protein that most cautiously.4–6 The discovery of CRISPR-Cas9 has
strains of human immunodeficiency virus (HIV) made genetic modifications easier to engineer and
less expensive to perform than previously used meth-
From the Brown Fertility and the Department of Obstetrics and Gynecology, ods such as zinc finger nucleases and transcription
University of Florida College of Medicine, Jacksonville, Florida activator-like effector nucleases. These older meth-
The authors thank Kate Harfe, PhD, for her review of an earlier version of this ods had more complex requirements for program-
manuscript and for her suggestions on how to improve this paper. ming them to bind to DNA at the targeted site.4,5
Each author has confirmed compliance with the journal’s requirements for Further development of this molecular tool is likely
authorship.
to make genetic modifications of cells common for
Corresponding author: Bruce I. Rose, MD, PhD, Brown Fertility, LLC, preventing and treating human diseases,3,4 for
Jacksonville, FL; email: brose@brownfertility.com.
manufacturing commercial products that use live or-
Financial Disclosure
The authors did not report any potential conflicts of interest.
ganisms,4,5,7 for exploring the intricacies of the
human genome,8 and even for novel applications
© 2019 The Author(s). Published by Wolters Kluwer Health, Inc. This is an
open-access article distributed under the terms of the Creative Commons such as the rescue of extinct species.9 We will explain
Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), what CRISPR-Cas9 is and how it was used by Dr. He
where it is permissible to download and share the work provided it is properly to prenatally change the genome of a baby. We will
cited. The work cannot be changed in any way or used commercially without
permission from the journal. then look more deeply into the details of this
ISSN: 0029-7844/19 experiment.

VOL. 134, NO. 1, JULY 2019 OBSTETRICS & GYNECOLOGY 157


BACKGROUND finger nucleases and transcription activator-like effec-
The human genome is contained in 23 chromosome tor nucleases have more complex attachments, which
pairs, primarily consisting of DNA. Each DNA make them harder to engineer. For example, it takes
molecule is a dimer containing about six billion many months to create zinc finger nuclease and tran-
nucleotides connected by a phosphate backbone. scription activator-like effector nuclease editing tools,
Only four different nucleosides occur in this very whereas it takes days to create a CRISPR-Cas9 tool
long sequence: adenine (A), guanine (G), thymine (T) (with a cost reduction of similar magnitude).4
and, cytosine (C). Each of these four nucleotides can Once CRISPR-Cas9 has attached to the DNA
form hydrogen bonds with only one other nucleotide molecule, the CRISPR-Cas9–associated nuclease
(A with T and G with C). This is the basis for the Cas9 makes a double-stranded cut in a defined loca-
genetic continuity of cells during mitosis or meiosis. tion on the DNA molecule. To make this precise cut,
Each strand of the DNA molecule can exactly dictate CRISPR-Cas9 must also simultaneously bind to
a short DNA sequence (called a PAM sequence).
the opposite subunit by acting as a template in which
The cut is made two or three nucleotides away from
each nucleotide in the sequence establishes hydrogen
the PAM sequence.5,13 PAM sequences differ and
bonds with its unique mate and also forms a covalent
depend on the bacteria from which the CRISPR-
bond to the proceeding nucleotide to create an
Cas9 tool was isolated.
elongated phosphate spine reconstituting the double-
CRISPR-Cas9 is guided to its binding site by an
stranded molecule.
engineered piece of RNA created to attach to the
Most life contains DNA, including bacteria and
target location on the DNA on the sense strand.14 The
many viruses. Viruses that attack bacteria, or bacter-
double-stranded break created by CRISPR-Cas9
iophages, are very numerous.10 Bacteria have devel-
needs to be repaired rapidly by the cell or the cell is
oped defensive weapons that attach to and destroy
likely to die. Prevalent exonucleases will delete nu-
invading bacteriophages by cutting apart their DNA.
cleotides from bare cut ends of DNA. Random nu-
In their study of bacteria, scientists have identified
cleotides may attach to the bare ends. These
some of these weapons (referred to as restriction en- deletion and insertion mutations may make the gene’s
zymes) and have used them routinely as tools in the protein product nonfunctional. The investigator may
study of DNA. From different species of bacteria, also supply small fragments of DNA that enable the
more than 3,000 of these bacterial weapons have been cell to rebuild a targeted cutting site by using these
identified. Different restriction enzymes bind to differ- pieces as a template in its repair.
ent short sequences of DNA (usually 4–8 nucleotides CRISPR-Cas9 may be used to cut out a long
long) and then make double-stranded cuts in the DNA sequence of DNA or to remove an entire gene from
near those oligonucleotides. a cell’s DNA by using two CRISPR-Cas9 molecules
A more recent discovery in bacteria is a defensive
tool called CRISPR-Cas9 (CRISPR stands for Clus-
tered Regularly Interspaced Short Palindromic Re-
peats.) As with restriction enzymes, CRISPR-Cas9
variants can be found in many bacteria.11 CRISPR-
Cas9 acts like an immune system for its bacterium.12
If a bacterial cell survives an attack by a bacteriophage,
CRISPR-Cas9 retains a small piece of that virus’s
DNA. This retained memory of prior exposure ena-
bles that bacterium to rapidly defend itself, if it is
attacked again by the same species of bacteriophage.
Fig. 1. Diagram of the CRISPR-Cas9 molecule. Both trans-
The CRISPR-Cas9 system (Fig. 1), adapted for activating CRISPR RNA (tracrRNA) and CRISPR (crRNA)
genetic engineering differs from restriction enzymes occur in nature. The CRISPR component tracrRNA is
in that its binding–recognition site is defined by a binding scaffold to the Cas nuclease. The crRNA molecule
a longer sequence of about 20 nucleotides. Because is a small molecule that attaches to the RNA derived from
a phage that attacked the bacteria (one of the “spacers” in
restriction enzyme binding sequences are short, they
CRISPR), which together constitute the guide RNA. Licensed
bind at many sites and make many cuts in a long from Mari-Leaf/Shutterstock.com(SSTK-066F1-A564). Used
DNA molecule. The binding site of CRISPR-Cas9 with permission. PAM, protospacer adjacent motif.
molecules is long enough to be used for precise bind- Rose and Brown. Genetically Modified Babies Using CRISPR-Cas9.
ing if the CRISPR-Cas9 tool is well designed. Zinc Obstet Gynecol 2019.

158 Rose and Brown Genetically Modified Babies Using CRISPR-Cas9 OBSTETRICS & GYNECOLOGY
designed to cut the DNA on both sides of the unde- Dr. He’s approach was to delete a specific 32 base
sired sequence. However, cutting or disabling an pair segment of the CCR5 gene. This specifically tar-
entire gene from a cell’s functional DNA may have geted deletion is called the CCR5 gene’s delta 32
a negative effect on that organism, because many mutation. It occurs naturally in about 10% of Euro-
genes code for more than one protein; genes and their peans.1 Creating a delta 32 mutation as opposed to
products may have unexpected interactions to other some other mutation, would be reassuring in that the
genes and proteins, and genes may play different roles planned genetic alteration occurs naturally and there-
when expressed in different parts of the body. fore would be unlikely to cause unexpected adverse
health effects in the resulting children. However, it is
DR. JIANKUI HE known that the delta 32 mutation is not entirely
benign. Having a nonfunctional CCR5 gene increases
Dr. He received his PhD from Rice University in
a person’s susceptibility to West Nile virus and Japa-
2010, which was followed by postgraduate work at
nese encephalitis.21 It also may increase the likelihood
Stanford University. After training in the United
of an individual dying from influenza.22,23
States, Dr. He was recruited back to China as part
One may question why Dr. He chose to modify
of China’s “Thousand Talents Plan” (www.1000plan.
the CCR5 gene. Human immunodeficiency virus is
org/en/young.html). Genetics is one of many areas of
treatable, and the knowledge of how to decrease the
interest to the Chinese government.15 It is rumored
risk of contracting it is widespread. Perhaps the choice
that China will spend $9.2 billion on precision
to modify CCR5 gene was made because this modifi-
(genetic) medicine over the next 15 years.16 In the
cation had been used successfully in somatic cells to
next year, Sichuan University’s West China Hospital
make CD4+ T cells resistant to HIV infection (using
alone plans to sequence the genome of more than one
zinc finger nuclease technology).24 Finding a worth-
million people.16 Meghna Kataria, writing in BioN-
while candidate gene to modify in germline cells may
ews,17 reported that China has already treated 86 adult
be difficult. Established technology, namely preim-
patients with advanced cancer using CRISPR-Cas9
plantation genetic testing, can be used to avoid most
technologies. A description of at least 11 ongoing clin-
identified mutations that cause recessive, X-linked, or
ical trials in China, involving treatment of cancer and
dominantly inherited diseases and still enable carriers
HIV, using CRISPR-Cas9, can be found in the U.S.
of these abnormal genes to have genetically related
clinical trial registry (www.clinicaltrials.gov). These
children.25 Preimplantation genetic testing screens
trials go back as far as 2015 and have a projected
embryos created with in vitro fertilization for the gene
enrollment of more than 200 patients. Several Chi-
of interest so that only nonaffected embryos are trans-
nese teams have previously edited human embryos
ferred back to the patient. At this early stage of devel-
in experiments to eventually treat important diseases,
opment, genetic editing of human embryos needs to
but without the intent of using them for human
prevent diseases that cannot be prevented with exist-
implantation.18,19
ing technology.
Through conferences and travel, Dr. He main-
In 2017, at a conference held at Cold Springs
tained communication with many U.S. scientists
Harbor Laboratory titled “Cancer and Stem Cells”
involved in genetics and genetic engineering. He
(available on YouTube), Dr. He presented research
discussed his interest in modifying the genome of
on mouse, monkey, and human embryos. In this
embryos with several U.S. scientists, although, accord-
research he looked at the problems of avoiding mosa-
ing to news reports,20 these scientists did not believe
icism and off-target DNA changes using CRISPR-
that he would proceed with this project at that time.
Cas9 on the CCR5 gene.
Reporters from The Wall Street Journal were able
EMBRYO EDITING to question Dr. He’s spokesperson after his talk
Dr. He intended to create embryos that would be announcing this birth at the Hong Kong conference.26
resistant to most strains of HIV. The gene CCR5 co- Five women underwent in vitro fertilization, and
des for a protein that most HIV strains use to enter a total of 22 oocytes were retrieved. Each father’s
cells.1 His objective was to eliminate production of sperm was prepared in a manner to limit or eliminate
this protein in individuals born from the modified HIV in the sperm specimen. Reportedly, a sperm and
embryos. He initially recruited eight couples in which the CRISPR-Cas9/Cas9 enzymes were injected into
the father had HIV and the couple wished to inter- 18 of the unfertilized oocytes. Thirteen edited
vene so that their child would be prevented from ever embryos were transferred back into the five women.
contracting HIV. Two of the four embryos from one couple contained

VOL. 134, NO. 1, JULY 2019 Rose and Brown Genetically Modified Babies Using CRISPR-Cas9 159
modifications of the CCR5 gene. According to Yong ported the University had turned down Dr. He’s
reporting for The Atlantic,2 one of those transferred research proposal.2 The University subsequently is-
embryos was known to still contain one copy of a nor- sued a statement condemning Dr. He’s experiment
mal CCR5 gene. This embryo transfer led to the birth and denying any knowledge of it (various news sour-
of Lulu and Nina. Lucas Laursen reported in Fortune ces report that he was subsequently fired.) Dr. He had
that there is at least one other pregnancy with a mod- taken a 3-year unpaid leave from the University start-
ified genome that is still ongoing.27 ing in February of 2018. He also worked at HarMon-
How to best perform gene editing of embryos iCare Women and Children’s Hospital in Shenzhen,
efficiently and avoid creating mosaic embryos while which is independent of Southern University of Sci-
modifying the genes inherited from both the mother ence and Technology.26 Yong also reported that Dr.
and father is a subject of active research. The first He obtained institutional review board approval from
Chinese team using CRISPR-Cas9 in 2015, and that hospital.2 The approval was published in the
editing genes of nonviable embryos, was able to China Clinical Trial Registry, but the hospital claims
change the genome as desired in only 4 out of 86 that the signature on the approval form was forged.
embryos.19 More recently, a team at Oregon Health As reported in the Atlantic, the Associated Press,
and Science University edited genes of donated and The Wall Street Journal,2,20,26 much of the scientific
embryos (without the intention of implanting them) community was shocked at the risks intrinsic to this
with markedly better results.28 The CRISPR-Cas9 en- undertaking. Although Dr. He achieved a “first”;
zymes were likely injected into zygotes as close to the there are tangible risks involved in modifying
time of fertilization as possible. embryos at this time. Several studies have suggested
Dr. He was successful in modifying the genes of that off-target mutations after the use of CRISPR-Cas9
embryos that subsequently became babies. However, remain a significant problem and active area of
it is troubling that he did not achieve what he set out research.14,30–33 Gene editing in animals using both
to do. At the Hong Kong conference where the births CRISPR-Cas9 and prior techniques has demonstrated
were announced (and in the YouTube video that Dr. occasional unintended consequences. For example,
He produced, dated November 25, 2018), he reported there are a number of animal experiments of commer-
that the genome of each child had been sequenced cial value in which an MSTN-like gene was deleted
and showed no changes other than in the CCR5 genes. (using CRISPR-Cas9 and prior tools). This gene codes
This suggests that this use of CRISPR-Cas9 made no for myostatin, which limits how large muscles can
unintended changes in the embryo’s genome. How- grow in humans.34 Animal gene editing experiments
ever, because Lulu has one normal CCR5 gene that to delete this gene had the intended effect of doubling
will still produce the normal protein, she will not be or tripling muscle mass, but genetically modified cat-
resistant to HIV infection. Of greater concern, accord- tle were created that were too big to exit though the
ing to an (unnamed) expert who attended the confer- birth canal,35 some pigs were born with extra thoracic
ence where the births were announced, the slides vertebrae,7 and rabbits and pigs were born with
presented did not show a delta 32 mutation in any enlarged tongues.36 Gene function is difficult to fully
of the CCR5 genes.2 The mutations that were created deduce and some applications of CRISPR-Cas9 result
in the CCR5 genes of Nina may or may not produce in unintended genetic code changes with uncertain
functional proteins. If this report is accurate, the gene effect.14,30–33
modifications actually performed may not prevent Ni-
na from contracting HIV and may or may not have CONCLUSION
other health consequences. In addition, even monitor- Dr. Jiankui He was the first scientist involved in the
ing the health of Lulu and Nina over their lifetimes birth of a baby with edited genes. He chose to edit
will not necessarily demonstrate that the objective of a gene related to a disease, HIV, which could both be
preventing HIV infection was met, because they may avoided and treated with established therapies. If the
never be exposed to HIV. recipient of the gene modification does not contract
Most countries regulate or ban research on HIV, it would not demonstrate the efficacy of this
human embryos.29 China has guidelines for such gene modification because the individual may never
research. Investigators must present their research be exposed to HIV. He also chose to modify a gene to
proposal to the institutional review board for approval eliminate a gene product that did not completely
at the institution where the research is to take place. protect the resulting child from the disease of concern.
Dr. He was on the faculty of Shenzhen-based South- The child could still be infected by strains of HIV that
ern University of Science and Technology. Yong re- used a different binding protein. Successfully

160 Rose and Brown Genetically Modified Babies Using CRISPR-Cas9 OBSTETRICS & GYNECOLOGY
eliminating this gene product by creating a delta 32 13. Anders C, Niewoehner O, Duerst A, Jinek M. Structural basis
mutation, as planned, was known to create alternative of PAM-dependent target DNA recognition by the Cas9 endo-
nuclease. Nature 2014;513:569–73.
health issues for the recipient of the mutation. He
14. Doench JG, Fusi N, Sullender M, Hegde M, Vaimberg EW,
aided in the birth of a child who was heterozygotic for Donovan KF, et al. Optimized sgRNA design to maximize
a mutated CCR5 gene and who therefore would not be activity and minimize off-target effects of CRISPR-Cas 9. Nat
immune to HIV infection. The other child born had Biotechnol 2016;34:184–91.
mutations of her CCR5 genes that might still produce 15. Zweig D, Wang H. Can China bring back the best? The Com-
functional proteins, or as seen in animal experiments, munist Party Organizes China’s Search for Talent. China Q
2013;215:590–615.
might present with unintended health outcomes.
16. Cyranoski D. China embraces precision medicine on a massive
More concisely, although the thought of gene
scale. Nature 2016;529:9–10.
editing of embryos is an exciting prospect, our present
17. Kataria M. China has treated 86 people with CRISPR genome
experience using gene editing for the treatment of editing. BioNews January 29, 2018.
adults with severe disease or for beneficial genome 18. Kang X, He W, Huang Y, Yu Q, Chen Y, Gao X, et al. Intro-
modification of animal populations is limited. Many ducing precise genetic modifications into human 3PN embryos
aspects of the experiment undertaken by Dr. He were by CRISPR/Cas-mediated genome editing. J Assisted Reprod
troubling. Even with the discovery of CRISPR-Cas9, Genet 2016;33:581–8.
suboptimal control of molecular tools for gene editing 19. Liang P, Xu Y, Zhang X, Ding C, Huang R. CRISPR/Cas9-
mediated gene editing in human tripronuclear zygotes. Protein
and a review of the history of gene editing suggest the
Cell 2015;6:363–72.
need for more caution and more collaboration before
20. Marchione M. Chinese researcher claims first gene-edited ba-
undertaking additional attempts to modify germline bies. AP November 26, 2018.
cells to create babies. 21. Glass WG, McDermott DH, Lim JK, Lekhong S, You SF, Frank
WA, et al. CCR5 deficiency increases risk of symptomatic West
Nile virus infection. J Exp Med 2006;203:35–40.
REFERENCES
1. Martinson JJ, Chapman NH, Rees DC, Luis Y-T, Clegg JB. 22. Tyner JW, Uchida O, Kajiwara N, Kim EY, Patel AC,
Global distribution of the CCR5 gene 32-basepair deletion. O’Sullivan MP, et al. CCL5-CCR5 interaction provides antia-
Nat Genet 1997;16:100–3. poptotic signals for macrophage survival during viral infection.
Nat Med 2008;11:1180–7.
2. Yong E. The CRISPR baby scandal gets worse by the day. The
Atlantic December 3, 2018. 23. Dawson TC, Beck MA, Kaziel WA, Henderson F, Maeda N.
Contrasting effects of CCR5 and CCR2 deficiency in the pul-
3. Cohen IG, Adashi EY. The FDA is prohibited from going monary inflammation response to influenza A virus. Am J Path-
germline. Science 2016;353:545–6. ol 2000;156:1951–9.
4. Doudna J, Sternberg SH. A crack in creation: gene editing and 24. Didigu CA, Wilen CB, Wang J, Duong J, Secreto AJ, Danet-
the unthinkable power to control evolution. New York (NY):
Desnoyers GA, et al. Simultaneous zinc-finger nuclease editing
Houghton Mifflin; 2017.
of the HIV coreceptors ccr5 and cxcr4 protects CD4+ T cells
5. National Academies of Sciences, Engineering, and Medicine. from HIV-1 infection. Blood 2014;123:61–9.
Human genome editing: science, ethics, and governance. Wash-
25. Brezina PR, Kutteh WH. Clinical applications of preimplanta-
ington, DC: The National Academies Press; 2017.
tion genetic testing. BMJ 2015;350:7611–20.
6. Baltimore D, Berg P, Botchan M, Carroll D, Charo RA, Church G,
26. Rana P, Fan W. Chinese scientist claims world’s first genetically
et al. Biotechnology. A prudent path forward for genomic
engineering and germline gene modification. Science 2015; modified babies. Wall Street Journal November 26, 2018.
348:36–8. 27. Laursen L. Chinese scientist defends his gene-edited babies,
7. Qian L, Tang M, Yang J, Wang Q, Cai C, et al. Targeted claims a third is on the way. Fortune November 28, 2018.
mutations in myostatin by zinc-finger nucleases result in 28. Ma H, Marti-Gutierrez N, Park S-W, Wu J, Lee Y, Suzuki K,
double-muscled phenotype in Meishar pigs. Scientific Rep et al. Correction of a pathological gene mutation in human
2015;5:14435. embryos. Nature 2017;548:413–19.
8. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, 29. Araki M, Ishii T. International regulatory landscape and inte-
Arkin AP, et al. Repurposing CRISPR as an RNA-guided plat- gration of corrective genome editing into in vitro fertilization.
form for sequence specific control of gene expression. Cell Reprod Biol Endocrinol 2014;12:108–19.
2013;152:1173–83.
30. Mon H, Smith JL, Peng L, Yin H, Moore J, Zhang XO, et al.
9. Shapiro B. Mammoth 2.0: will genome engineering resurrect CRISPR/Cas 9 mediated genome editing induces exon skip-
extinct species? Genome Biol 2015;16:228. ping by alternate splicing on exon deletion. Genome Biol
10. Ackermann H-W. Bacteriophage taxonomy. Microbiol Aust 2017;18:108–15.
2011;32:90–4. 31. Kosicki M, Tomberg K, Bradley A. Repair of double-strand
11. Markarva KS, Haft DH, Barrangou R, Broins SJ, Charpentier breaks induced by CRISPR-Cas 9 leads to large deletions and
E, Horvath P, et al. Evolution and classification of CRISPR-Cas complex rearrangements. Nat Biotechnol 2018;36:765–71.
systems. Nat Rev Microbiol 2011;9:467–77. 32. Fu Y, Foder JA, Khayter C, Maeder ML, Reyon D. High-frequency
12. Horvath P, Barrangou R. CRISPR/Cas, the immune system of off-target mutagenesis induced by CRISPR-Cas nucleases in human
bacteria and archaea. Science 2010;327:167–70. cells. Nat Biotechnol 2013;32:822–6.

VOL. 134, NO. 1, JULY 2019 Rose and Brown Genetically Modified Babies Using CRISPR-Cas9 161
33. Cradick JJ, Fine EJ, Antico CJ, Bao G. CRISPR/Cas9 systems 36. Qingyan L, Yuan L, Deng J, Chen M, Wang Y, Zeng J, et al.
targeting beta-globin and CCR5 genes have substantial off tar- Efficient generation of myostatin gene mutated rabbit by
get activity. Nucleic Acid Res 2013;41:9584–92. CRISPR/Cas9. Scientific Rep 2016; 6:25029.
34. Lee SJ, Reed LA, Davies MV, Girgenrath S, Goad MEP, Tom-
kinson KN, et al. Regulation of muscle growth by multiple
ligand signaling through activin type II receptors. Proc Natl
Acad Sci U S A 2005;102:18117–22. PEER REVIEW HISTORY
35. Bellinge RHS, Liberies DA, Laschi SPA, O’Brian PA, Jay GK. Received January 22, 2019. Received in revised form April 1, 2019.
Myostatin and its implications on animal breeding: a review. Accepted April 4, 2019. Peer reviews and author correspondence
Anim Genet 2005;36:1–6. are available at http://links.lww.com/AOG/B409.

162 Rose and Brown Genetically Modified Babies Using CRISPR-Cas9 OBSTETRICS & GYNECOLOGY

You might also like