Научная статья на тему 'CRISPR/CAS-EDITED PIGS FOR PERSONALIZED MEDICINE: MORE THAN PRECLINICAL TEST-SYSTEM'

CRISPR/CAS-EDITED PIGS FOR PERSONALIZED MEDICINE: MORE THAN PRECLINICAL TEST-SYSTEM Текст научной статьи по специальности «Биотехнологии в медицине»

CC BY
117
23
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
CRISPR-CAS / PERSONALIZED MODELS / XENOTRANSPLANTATION / PIG / BLASTOCYST COMPLEMENTATION / ANIMAL MODEL

Аннотация научной статьи по биотехнологиям в медицине, автор научной работы — Leonova E.I., Reshetnikov V.V., Sopova Ju.V.

Novel CRISPR-Cas-based genome editing tools made it feasible to introduce a variety of precise genomic modifications in the pig genome, including introducing multiple edits simultaneously, inserting long DNA sequences into specifically targeted loci, and performing nucleotide transitions and transversions. Pigs serve as a vital agricultural resource and animal model in biomedical studies, given their advantages over the other models. Pigs share high similarities to humans regarding body/organ size, anatomy, physiology, and a metabolic profile. The pig genome can be modified to carry the same genetic mutations found in humans to replicate inherited diseases to provide preclinical trials of drugs. Moreover, CRISPR-based modification of pigs antigen profile makes it possible to offer porcine organs for xenotransplantation with minimal transplant rejection responses. This review summarizes recent advances in endonuclease-mediated genome editing tools and research progress of genome-edited pigs as personalized test-systems for preclinical trials and as donors of organs with human-fit antigen profile.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «CRISPR/CAS-EDITED PIGS FOR PERSONALIZED MEDICINE: MORE THAN PRECLINICAL TEST-SYSTEM»



Research Results in Pharmacology

Research Results in Pharmacology 8(3): 87-98

UDC: 576.7

DOI 10.3897/rrpharmacology. 8.83872

CRISPR/Cas-edited pigs for personalized medicine: more than preclinical test-system

Elena I. Leonova1,2, Vasily V. Reshetnikov2, Julia V. Sopova1

1 St. Petersburg State University, 7-9 Universitetskaya Embankment, St. Petersburg 199034, Russia

2 Sirius University of Science and Technology, 1 Olympic Ave., Sochi 354340, Russia

Corresponding author: Julia V. Sopova (sopova@hotmail.com)

Academic editor: Alexey Deykin ♦ Received 18 March 2022 ♦ Accepted 3 August 2022 ♦ Published 13 September 2022

Citation: Leonova EI, Reshetnikov VV, Sopova JuV (2022) CRISPR/Cas-edited pigs for personalized medicine: more than preclinical test-system. Research Results in Pharmacology 8(3): 87-98. https://doi.org/10.3897/rrpharmacology.8.83872

Novel CRISPR-Cas-based genome editing tools made it feasible to introduce a variety of precise genomic modifications in the pig genome, including introducing multiple edits simultaneously, inserting long DNA sequences into specifically targeted loci, and performing nucleotide transitions and transversions. Pigs serve as a vital agricultural resource and animal model in biomedical studies, given their advantages over the other models. Pigs share high similarities to humans regarding body/organ size, anatomy, physiology, and a metabolic profile. The pig genome can be modified to carry the same genetic mutations found in humans to replicate inherited diseases to provide preclinical trials of drugs. Moreover, CRISPR-based modification of pigs antigen profile makes it possible to offer porcine organs for xenotransplantation with minimal transplant rejection responses. This review summarizes recent advances in endonuclease-mediated genome editing tools and research progress of genome-edited pigs as personalized test-systems for preclinical trials and as donors of organs with human-fit antigen profile.

Graphical abstract:

Copyright Leonova EIetal. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC-BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Keywords

CRISPR-Cas, personalized models, xenotransplantation, pig, blastocyst complementation, animal model

Introduction

Rapid advances in genome editing have made it possible to quickly and accurately create new animal models required to study the pathogenesis of human diseases and discover new pharmacological targets. Obviously, these revolutionary genome editing technologies have opened up new avenues for the design and development of promising novel human therapies. However, although some of the possible applications of genome editing technologies have been successfully introduced in the laboratories worldwide, some of them are still a long way off. For instance, genetically modified mice are the organisms routinely used to obtain new data on mammalian physiology, disease pathogenesis and to test new medicals (Bruter et al. 2021; Dolskiy et al. 2022). Some of species like rabbits and goats were proposed to generate bioreactors producing human proteins for pharmaceutics (Goldman et al. 2012; Maksimenko et al. 2013; Gurskiy et al. 2016). Nevertheless, despite numerous benefits for pharmacology and medicine, there are still a very limited number of studies focused on generation of genetically modified pigs.

In this review, we summarize recent advances in en-donuclease-mediated genome editing tools, the research progress of genome-edited pigs as models for nonclinical trials and as organ donors for xenotransplantation highlighting the prospects of their clinical applications.

Gene editing tools

The traditional gene targeting technique developed in mice required homologous recombination (HR) and manipulation of embryonic stem cells (ES) (Mansour et al. 1988). Due to the lack of "real" ES-cells, this technique has never been applicable in livestock species. The development of new synthetic, precise tools for the genetic modification of mammalian genomes, such as ZFN, TALENs, and CRISPR-Cas, rendered complex genetic modifications feasible and efficient in non-rodent mammals (Fig.1). The ability to introduce a single double-strand break (DSB) at a unique predetermined site enables genes to be inactivated by insertion or deletion mutations (Indels) via non-homologous end-joining (NHEJ) repair or by targeted sequence replacement via homology-direc-ted repair with an exogenous homologous DNA or single-stranded oligonucleotide fragment. The practicality and simplicity of gene editing have steadily improved in successive generations of endonuclease systems beginning with Zinc finger nucleases (ZFN) (Hauschild et al. 2011; Kwon et al. 2013), then transcription activator-like effector nucleases (TALENs) (Carlson et al. 2012,

2014) and, most recently, the CRISPR-Cas9 system. The CRISPR-Cas9 system has substantially revolutionized and accelerated the process of knocking out endogenous genes or knocking-in exogenous sequences at targeted sites within the genome (Gaj et al. 2013). With these improved skills and efficiencies of genetic modifications, the modifications of the pig genome can confer any desired, predetermined genetic changes. Protein-guided ZFN and TALEN, as well as the RNA-guided CRISPR-Cas9 (clustered regularly interspaced short palindromic repeat-associated protein 9) system have been successfully used for the establishment of genetically engineered pig lines (Tan et al. 2016). ZFNs and TALENs have now largely been superseded by CRISPR-Cas9, which is equally, if not more, efficient in inducing DSBs and in stimulating HR (Mali et al. 2013). The CRISPR-Cas9 system and, especially, its diverse derivates also offer improved target specificity, less off-target activity, and better prediction of off-target effects (Mali et al. 2013; Fu et al. 2014; Coelho et al. 2020). Highly efficient gene editing makes it possible to carry out genetic modification directly in zygotes and early-stage embryos and thus avoiding nuclear transfer, which needs a lot expertise and equipment. Since the discovery of DNA endonucleases, there have been numerous reports of potential porcine models of human diseases based on gene knockouts generated by the injection of CRISPR-Cas9 components into zygotes (Hai et al. 2014; Tanihara et al. 2019a; Koppes et al. 2020).

Production of gene-edited livestock

Gene transfer into the porcine genome was first carried out in 1985 via DNA microinjection into the pronuclei of fertilized oocytes (Hammer et al. 1985). Since then, sperm-mediated gene transfer, lentiviral transgenesis, transposon-based transgenesis, somatic cell nuclear transfer from genetically modified cells, as well as gene editing techniques based on designer nucleases, were used for the generation of mutant pig lines (Lai et al. 2002; Lavitrano et al. 2002; Ramsoondar et al. 2003; Dieckhoff et al. 2007; Ivics et al. 2014). Porcine embryonic stem (ES) cells or induced pluripotent stem cells showing comparable developmental capacity as mouse ES cells are not yet available (Soto and Ross 2016). This might change with the recent development of porcine expanded plu-ripotent stem cells (EPSCs), which have the potential to differentiate into all three germ layers (Gao et al. 2019). Their complete potential for genome editing in pigs still must be explored, but might be an essential part in the

future. Genetic modification of animals is mainly achieved by germline modification of the genome. Animals with germline modifications are used to produce stable mutant lines, making the model permanently and repro-ducibly available during all developmental stages. Genetically engineered animals are either transgenic animals harboring experimentally transferred DNA sequences or genetically modified animals without integrating foreign DNA, but a modification of endogenous gene sequences.

Genetic engineering of animals involves projects to analyze additional functions by over-and/or ectopic expression of a transgene and/or partial or complete loss of function of endogenous genes. This includes the inacti-vation of specific genomic sequences (knockout), defined genomic modifications (knock-in), specific suppression of the synthesis of gene products (knockdown, gene silencing), as well as random mutagenesis of the host genome (insertional mutagenesis).

Zygote modification (microinjection, electroporation)

Porcine in vivo produced embryos and embryos following in vitro fertilization (IVF) of in vitro matured (IVM) oocytes derived from slaughterhouse ovaries are used for zygote microinjection. Basal microinjection of DNA sequences as transgenes into the pronuclei of fertilized oocytes is still carried out in mice, but generally results in low efficiencies of transgene integration, random integration of multiple transgene copies, including the rearrangement of the host genome around the integration site, insertional mutagenesis, high numbers of transgenic mosaic founders, and possible genome position effects on the expression of the transgene (Ding et al. 2005; Kumar et al. 2009; Ivics et al. 2014; Li et al. 2014). The microinjection into the pronucleus of porcine zygotes is a much more difficult endeavor, as pig oocytes contain a high content of lipids, giving the cytoplasm a dark color, thus making it impossible to visualize the pronuclei without centrifugation of the embryos (Gil et al. 2017). Intracytoplasmic microinjection became a feasible option to genetically modify the genome of early pig embryos with the development of new vectors, such as transposon-based vectors and the CRISPR-Cas system (Fig 1.). The zygote first division is a critical time point to avoid mosaicism in the obtained offspring. Therefore, the use of Ribonucleic-protein complexes (gRNA + Cas-protein) is usually preferred, as they are immediately active and remain active only for a short time, which further reduces the risk of causing undesired mutations of the genome (off-targets) (Vakulskas and Be-hlke 2019; Naeem et al. 2020). Recently, electroporation of zygotes has been used for generating knock-out piglets. The efficiency of genome editing (especially, biallelic) after electroporation was significantly higher than after microinjection (Le et al. 2020). This method was used to create myostatin knock-out pigs (Wang et al. 2015), a pig model for diabetes with PDX-1 mutations (Tanihara et al.

2019b), CD1&3-edited pig (Tanihara et al. 2021b), pigs with point mutation in KRAS gene (Wittayarat et al. 2021), and pigs with targeted mutations in GGTA1, CMAH, and B4GALNT2 genes, generated by one-step electroporation (Tanihara et al. 2021a). Electroporation of porcine zygotes reaches biallelic editing efficiencies of up to 70%, depending on the Cas9 concentration and the target gene (Tanihara et al. 2019a, 2021a).

Somatic cell nuclear transfer

Somatic cell nuclear transfer (SCNT, cloning) is carried out to produce pigs carrying mutations (knock-in, knockout) in defined loci of the porcine genome after using genome editing tools (Betthauser et al. 2000; Onishi et al. 2000). The production of genetically modified animals by SCNT includes the transfection and selection of somatic donor cells in vitro, recovery and enucleation of recipient metaphase II oocytes, transfer of genetically modified somatic donor cells into the perivitelline space of the enucleated oocyte, electrical fusion of the donor cell and the oocyte, electrical activation of the reconstructed oocytes, and surgical embryo transfer to hormonally synchronized recipients (Holker et al. 2005). In the case when genetically modified donor cells are employed, 100% genetically modified founder animals without genetic mosaicism are obtained. Usually, 80- 400 embryos are transferred per recipient (Fig.1), but higher numbers of transferred embryos do not result in larger litters. The pig cloning efficiency is varying within relatively low values between 0.5-5% (offspring/transferred cloned embryos) (Betthauser et al. 2000; Onishi et al. 2000; Kolber-Simonds et al. 2004; Holker et al. 2005). The successful embryonic, fetal, and neonatal development of the transferred embryos derived from SCNT depends on the correct epigenetic reprogramming of the donor cell nucleus. Insufficient epi-genetic reprogramming may lead to an overall low cloning efficiency, as well as to peri- and neonatal health problems of cloned pigs, which might result in 50% stillborn piglets or piglets that die perinatally (own not published results). Abnormal phenotypes of cloned pigs occur less frequently than in other cloned mammals, and they are not transmitted to the offspring of affected clones (Estrada et al. 2007). Various somatic cell types were successfully used in the cloning procedure and numerous technical variations were established to increase the efficiency of porcine SCNT (Holker et al. 2005; Kurome et al. 2013).

Pigs as a test-system for preclinical trials

In general, because of their human-like anatomy, pigs are considered a very good test-system for pharmacological studies (Glauser 1966). High similarity of body size, mass indexes of internal organs and other physiological parameters between pigs and human beings, along with

their similar diet, determine the similarity of absorption, distribution, metabolism, and excretion (ADME) (Karar-li et al. 1995; Heinritz et al. 2013). Moreover, big body size permits to test the same routes for drug administration as in real patients. Anticipatedly, porcine models are highly more powerful than the rodent ones to translate the obtained data in clinical practice. However, on the other hand, porcine models are not mainstay because they are expensive and time-consuming.

Obviously, genetically modified pigs are not routine laboratory animals. Nowadays, the most widespread application of CRISPR/Cas-edited pigs is a field of oncological studies (Xu et al. 2019). Tumorigenesis in pigs resembles that in humans, which was first shown via au-tologous transplantation of primary porcine cells transformed with viral oncogenic cDNAs (Adam et al. 2007). Flisikowska et al. (2012, 2017). generated pigs that carry a translational stop signal at codon 1311 in porcine APC (APC1311), which is orthologous to the human APC1309 mutation responsible for a severe form of familial adeno-matous polyposis. These pigs develop polyps in the colon and rectum. As early as 4 months of age, these pigs were shown to display a clinical picture of the adenoma-carcinoma sequence. The same research group also generated pigs that carry a Cre-dependetly activated TP53R167H mutation in exon 5. Pigs heterozygous for the unindu-ced allele develop osteosarcomas after 16 months of age, while homozygotes show multiple osteosarcomas at 7-8 months of age (Leuchs et al. 2012; Saalfrank et al. 2016).

Additionally, CRISPR/Cas-mediated genome editing technologies were also utilized to generate porcine models of diabetes mellitus, Alzheimer's disease, Duchenne muscular atrophy, and cystic fibrosis (reviewed in Perleberg et al. 2018).

Pigs as organ donors

The growing shortage of available organs for allotransplantation has prompted scientists to search for alternative ways to treat end-stage organ failure. Pigs are now considered the optimal organ donor for xenotransplan-tation. The advantages of using pigs are that they share similar anatomical and physiological characteristics with humans, including cardiovascular, urinary, integumentary, and the digestive systems (Swindle et al. 2012; Ribitsch et al. 2020). Compared to other farm animals, pigs reach early puberty (at 5-8 months of age), breed well throughout the year, and have a significant number of offspring (an average of 10-14 piglets per litter) (Aigner et al. 2010; Ribitsch et al. 2020). Due to these characteristics, and the size of animals (young pigs have a size and body weight similar to humans), genetically modified pigs are widely used as models for xenotransplantation (Kahn et al. 1988; Chari et al. 1994; Martin et al. 1999; Spetzler et al. 2016; Vogel et al. 2017), human diseases and as a drug discovery platform (Dai et al. 2002; Swindle et al. 2012).

Generation of human organs in gene-edited pigs

One strategy to overcome the shortage of donor organs is the production of interspecies chimaeras by blastocyst complementation (Fig. 1). This allows obtaining both functional xenogeneic organs and tissues. The essence of the method lies in the fact that induced pluripotent stem cells (iPSCs) from a patient or a healthy donor are introduced into the pre-implantation embryo at the blastocyst stage, which theoretically should lead to the contribution of the donor stem cells to tissues and organs (Suchy and Nakauchi 2017). The use of zygotes homozygous for a knockout of a specific gene, which is evident for the formation of tissues or whole organs in recipient embryos, creates a tissue niche in blastocysts, which allows the formation of the corresponding organ from injected iPSCs. Through natural development in the embryo, all cell populations develop independently and lead to the development of specified organs and tissues during the embryo's normal development. This technology, created by Chen et al. (1993) has been shown to support organ formation after transferring normal heterologous pluripotent cells within and even between closely related species (Nagashima and Matsunari 2016). However, human iPSCs only made minor contributions to pig organs, mainly because pigs and humans are two distinct species that separated about 80 million years ago (Groenen et al. 2012). This results in distinct differences in the mechanisms of embryonic/fetal development, size and growth of the fetus, the timing of implantation, cell division rate, placental structure and so forth. Besides, pig cells will still contribute to the target human organ and form the endothelium. Therefore, such an organ would require genetic modification of the host endothelium by altering the pig embryo's genome or forming another niche that allows human stem cells to form the host's endothelium. Otherwise, the organ will trigger severe immune rejections (Freedman 2018). Pig embryos deficient in the etv2 gene, the main regulator of hema-toendothelial cells, might help to overcome this problem. After injection of human iPSCs into etv2-KO blastocys-ts, all vascular endothelial cells originated from human cells in day 17/18 embryos (Das et al. 2020). Another ethical concern remains the contribution of human cells to the parts of the pig other than the anticipated organ. For example, SALL1 knockout pig embryos were used to create a niche for human kidney formation. It is well known that the SALL1 protein plays a crucial role in the prenatal development of the kidneys and the hands, ears, anus, and other parts of the body (Choi et al. 2010). Thus, "wild-type" human iPSCs could contribute not only to the established organogenic niche, but might also contribute to other tissues and organs of the pig. Another work has shown more encouraging results using the blastocyst complementation method. The injection of porcine eGFP positive blastomeres into Hhex knockout porcine blasto-cysts resulted in a high degree of chimerism in the developing liver in day 25 fetuses (Ruiz-Estevez et al. 2021). The blastocyst complementation works to a certain extent

Figure 1. Approaches used to generate genetically modified pigs. The ZFN, TALEN, and CRISPR/Cas9 systems enable efficient gene targeting for modification of mammalian genomes. Microinjection or electroporation of ZFN/TALEN/CRISPR/Cas9 constructions results in one-step generation of genetically modified pigs. Interspecies blastocyst complementation is used for generation of human organs in pigs. The transfer of genetically modified somatic donor cells into oocyte is performed by electrical fusion of donor cell and oocyte. The surgical embryo transfer to hormonally synchronized recipients is necessary to receive genetically modified piglets. Created with BioRender.com.

within closely related species. Still, however, the xeno-geneic barrier is a major limitation, which is still poorly understood, and hinders progress in interspecies blasto-cyst complementation research. Further research in this area may one day lead to the cultivation of human organs inside animals for transplantation purposes. Nevertheless, blastocyst complementation remains unrealistic to supply enough transplantable organs to be an essential solution to the shortage of donor organs. This critical view is based on two columns: 1) the generated human organ has to be genetically edited to serve as a universal donor organ not causing rejection responses that cannot be addressed by immunosuppression, 2) every single organ would have to be produced by the very inefficient and currently unachievable way of blastocyst complementation as these pigs cannot be propagated by breeding or somatic cell nuclear transfer. Therefore, this research is somewhat of more interest to study developmental biology and the function of genes than a potential solution to the growing demand for transplantable organs.

Gene-edited pigs for xenotransplantation

Xenotransplantation could undoubtedly provide substantial benefits for human regenerative medicine. However, significant immunological and physiological incompatibilities must be addressed before xenotrans-plants becoming clinically effective xenogeneic grafts. Fortunately, our understanding of these barriers is multiplying, and rational strategies based on genome editing were developed to surmount them. Humoral rejection by preformed antibodies and incompatibilities between the blood coagulation systems present the most immediate obstacles. The most significant challenge comes from the adaptive immune response in the long term. An unmodified porcine organ transplanted into a human or primate recipient is confronted with a series of rejection responses. The first rejection response is the hyperacute rejection (HAR), followed by the acute humoral xenograft rejection (AHXR), also known as acute vascular rejection or delayed xenograft rejection. Both HAR and

AHXR are the ultimate result of antibodies binding to graft endothelial cell surface antigens.

In HAR, preformed antibodies in the human blood against the a1,3-galactosyl-galactose (aGal) epitope on the porcine endothelium cause the rapid formation of an antigen-antibody complex, which immediately activates the host complement system. The formation of the membrane attack complex (MAC) results in extensive hemorrhage, oedema, and thrombosis of small blood vessels, leading to the destruction of the graft within minutes to hours. The finding that aGal epitopes are not present in old-world primates (including humans) due to the non-functional a1,3-galactosyltransferase (GGTA1, also referred to as a1,3GT) gene, indicated the option to disable the orthologous gene in the porcine genome. Since the first reports of GGTA1 knockout in pigs (Dai et al. 2002; Lai et al. 2002), homozygous deficient animals have been generated (Phelps et al. 2003), and several independent herds have been established (Klymiuk et al. 2010). The benefit of organs from these pigs has been tested in numerous studies using pig-to-baboon organ transplantation models (Ekser et al. 2012) and revealed maximum survival times of three months for kidneys (Yamada et al. 2017), and 183 and 264 days after orthotopic transplantation of a pig heart (Langin et al. 2018; Mohiuddin et al. 2021).

AHXR presents the next immunological obstacle once HAR has been overcome. Hearts from aGal-KO pigs transplanted into baboons exhibited widespread thrombotic mi-croangiopathy, ischemia, focal hemorrhage, and necrosis because of progressive humoral rejection and disordered thromboregulation (Ezzelarab et al. 2009). The underlying mechanisms are incompletely understood, but the binding of non-Gal antibodies and activation of the endothelium played a crucial role, leading to a procoagulant state (Diswall et al. 2010; Byrne et al. 2011). The major non-Gal antigens are Neu5GC encoded by the CMAH gene (Cytidine monophospho-N-acetylneuraminic acid hydroxylase), and Sda, a product of B4GalNT2 (Beta-1,4 N-acetylgalactosa-minyltransferase 2) (Padler-Karavani et al. 2008; Byrne et al. 2018). Therefore, the common sense in the xenotransplantation field is that knockouts of GGTA1, CMAH and B4GalNT2 are essential to achieve long-term survival after xenotransplantation. This is further supported by the increasing survival times after pig-to-baboon xenotransplanta-tions of pig hearts carrying a triple or quadruple knockout (including knockout of the porcine growth hormone receptor (GHR) to keep pigs smaller). Various transgenic pigs expressing human transgenes on the vascular endothelium have been generated and tested. Transgenes that modulate endothelial activation in the xenograft, e.g. heme oxygenase 1 (HO-1) or tumor necrosis factor-a-induced protein (A20), have been produced and have proven their efficacy to protect the endothelium from being activated after xeno-perfusion with human blood (Oropeza et al. 2009; Petersen et al. 2011). In addition to the antibody-mediated activation of the xenograft endothelium, incompatibilities in the coagulation components in the human bloodstream and the porcine vessel wall have been found to contribute to the formation of microthrombi (Cowan 2007). Expression of

human regulators of blood coagulation, such as tissue factor pathway inhibitor (TFPI), endothelial protein C receptor (EPCR) or thrombomodulin (THBD), can help overcome these incompatibilities (Petersen et al. 2009).

The requirements continue to be refined as knowledge increases, but the current scientific agreement on genetic modifications required in donor pigs to combat HAR and AHXR are:

- Removal of the major xenoreactive surface antigens: aGal and non-Gal epitopes Neu5GC and Sda

- One or more abundantly expressed human complement regulatory genes: e.g. CD46, CD55, CD59

- A human anti-thrombotic and/or anticoagulant gene: e.g. THBD, EPCR, and others

- A gene with vascular protective properties: e.g. HO-1, hA20, and others.

Porcine endogenous retroviruses (PERVs) are gamma retroviruses expressed in various organs and found in the genome of all pig lines (Bittmann et al. 2012). Compared to other known retroviruses, such as the human immunodeficiency virus (HIV) and human T-cell lymphotropic virus, integration of PERVs into the patient's genome has the potential to lead to immunodeficiency and tumorigenesis (Liu et al. 2020). No evidence of PERV transmission has been found in preclinical trials of porcine cell and organ transplantation in non-human primates and in clinical trials of transplantation of encapsulated porcine islets (Specke et al. 2001; Denner 2021). The reason for such discrepancies is not completely clear; however, the existing preclinical and clinical trials have a number of limitations (lack of functional PERV receptors in most model animals, the use of encapsulated islet cells in human clinical trials) regarding the interpretation of PERV transmission and, thus, cannot give an accurate answer about the safety of xenotransplantation (Denner 2018, 2021). Various strategies have been used to inactivate PERVs, including antiretroviral drug administration, vaccination and RNA interference (Denner 2021), but complete inactivation was only achieved with CRISPR-Cas9 in 2017 (Niu et al. 2017). Twenty-five copies of functional PERVs in a porcine primary cell line and generated PERV-inactivated pigs via somatic cell nuclear transfer have been studied. Pigs with all PERVs inactivated can also be genetically engineered to eliminate transplant rejection and these pigs exhibit normal physiology, fertility and germline transmission and cannot be infected with PERVs from other pigs including their mother (Yue et al. 2021). Taken together, new knowledge and advances in the field of porcine viruses and pathogens indicate that there are no fundamentally insoluble problems for infection safety in xenotransplantation.

Conclusions and discussion

New technologies for targeted modification of the animal genome considerably facilitate the creation of human disease models for preclinical research and gene therapy.

Remarkable results were obtained in mouse models, making it possible to obtain more advanced models of large animals, including genetically modified pigs. However, genome editing of pigs opens up even more significant avenues for medicine. The feasibility to use genetically modified pigs as donors can be a silver bullet to solve one of the dramatic problems with the deficiency of organs for transplantation.

Restoration and replacement of damaged tissues and organs are some of the priorities in the development of health care. Human tissues and organs, despite their advantages, could not overcome the global acute shortage of donor materials. Heart and kidney transplantation remains the only approach for end-stage organ failure. Allotransplantation of cadaveric organs, preparation of the donor and the recipient, and the surgical procedure are necessarily carried out under severe time pressure. Consequently, infections can be transmitted to the recipient (Morris et al. 2010). The average waiting time for a cadaveric kidney is five years, which significantly reduces the prospects for patients eventually receiving a donated kidney because graft survival drops substantially after extended dialysis. Although some guidelines have been developed to optimize the outcome of transplantations relying on deceased donors (Domínguez-Gil et al. 2011), tissues or organs are frequently taken from marginal donors, resulting in impaired graft function (Zimmerhackl et al. 2010).

The limitations of allotransplantation have prompted worldwide search for alternative treatments. Two different approaches offer potential solutions for solving the problem - the creation of human organs by blastocysts complementation and the transplantation of grafts from non-human donors. Both approaches provide the opportunity to closely examine donor tissue and thoroughly prepare the recipient before surgery. The strategy of growing organs in interspecies chimeras of pigs by complementing the blastocyst resulted in limited success, as grown organs can still cause immunological rejection in further transplantation to humans. It has become evident that the pig (Sus scrofa) is the donor of choice for xenotransplantation for several reasons: (i) similarities in size, anatomy, nutrition, and physiology as well as low genetic distance to man; (ii) short generation intervals (12 months) and high fertility (10-14 offspring per litter); (iii) well established and economic housing and breeding conditions at high hygienic standards; (iv) availability of advanced reproductive biotechnologies and genetic engineering techniques; (v) minor concerns regarding the slaughter of pigs, at least in western countries, because they are raised for meat production on an industrial scale; (vi) the risk of zoonosis is considered to be a minimal risk (strict monitoring, gnotobiotic environment), e.g. PERVs can

be eliminated from the porcine genome and PERV-C free pigs are available. Therefore, the creation of GMO pigs for xenotransplantation is one of the promising ways to cope with the problem of organ shortage.

Despite these clear advantages, the selection of optimal immunosuppression therapy in each transplantation case needs further study. Moreover, the results of studies performed by different researchers is difficult to compare due to a large number of factors (organ type, pig used, etc.). In addition, there are other serious ethical concerns in society regarding the use of pigs as donors, and these need to be dispelled (Cozzi et al. 2009).

Nevertheless, recently, xenotransplantation has made great progress with the first heart transplanted from the genetically modified swine source that had 10 individual gene edits to a human in the US (Griffith et al. 2022). The recipient of the pig heart was in good condition after 5-week post-transplantation, which is longer than the survival of the first patient who received a cardiac allotrans-plant (Brink et al. 2017; Graham 2022). Unfortunately, sudden diastolic thickening and failure of the xenograft occurred on day 49 after transplantation, and life support was withdrawn on day 60. However, subsequent full in-vestigitaion revealed that the patient's heart was affected by porcine cytomegalovirus, a preventable infection that is linked to devastating effects on transplants (Regalado 2022). Interestingly, this case emphasizes the need of considering swine genes responsible for the viral sensitivity to be also modified besides the other ones.

Additionally, genetically modified porcine kidneys have been connected to the blood system of two brain-dead patients. These experiments were terminated after 53 and 77h without any signs of rejection and fully functional kidneys with normal urine production (Porrett et al. 2022). These three experiments clearly show that xenot-ransplantation has started to enter the clinical stage and has the potential to overcome the shortfall of available human donor organs.

Conflict of interests

The authors declare no conflict of interests.

Funding

This work was supported by St. Petersburg State University grant for the development of scientific research (ID 92561695) and by Sirius University project GEN-RND-2017.

References

■ Adam SJ, Rund LA, Kuzmuk KN, Zachary JF, Schook LB, Counter CM (2007) Genetic induction of tumorigenesis in swine. Oncogene 26(7): 1038-1045. https://doi.org/10.1038/sj.onc.1209892 [PubMed]

■ Aigner B, Renner S, Kessler B, Klymiuk N, Kurome M, Wünsch A, Wolf E (2010) Transgenic pigs as models for translational biomedical research. Journal of Molecular Medicine 88(7): 653-664. https:// doi.org/10.1007/s00109-010-0610-9 [PubMed]

■ Betthauser J, Forsberg E, Augenstein M, Childs L, Eilertsen K, Enos J, Forsythe T, Golueke P, Jurgella G, Koppang R, Lesmeister T, Mal-lon K, Mell G, Misica P, Pace M, Pfister-Genskow M, Strelchenko N, Voelker G, Watt S, Thompson S, Bishop M (2000) Production of cloned pigs from in vitro systems. Nature Biotechnology 18(10): 1055-1059. https://doi.org/10.1038/80242 [PubMed]

■ Bittmann I, Mihica D, Plesker R, Denner J (2012) Expression of porcine endogenous retroviruses (PERV) in different organs of a pig. Virology 433(2): 329-336. https://doi.org/10.1016/j.vi-rol.2012.08.030 [PubMed]

■ Brink J, Pennel T, Seele K, Zilla P (2017) The world's first human-to-human heart transplant at Groote Schuur Hospital: 50 years later. South African Medical Journal 107(12): 1035-1036. https:// doi.org/10.7196/SAMJ.2017.v107i12.12960 [PubMed]

■ Bruter A, Korshunova DS, Kubekina MV (2021) Novel transgenic mice with Cre-dependent co-expression of GFP and human ACE2: a safe tool for study of COVID-19 pathogenesis. Transgenic Research 30(3): 289-301. https://doi.org/10.1007/s11248-021-00249-8 [PubMed] [PMC]

■ Byrne G, Ahmad-Villiers S, Du Z, McGregor C (2018) B4GAL-NT2 and xenotransplantation: A newly appreciated xenogeneic antigen. Xenotransplantation 25(5): e12394. https://doi.org/10.1111/ xen.12394 [PubMed] [PMC]

■ Byrne GW, Stalboerger PG, Du Z, Davis TR, McGregor CGA (2011) Identification of new carbohydrate and membrane protein antigens in cardiac xenotransplantation. Transplantation 91(3): 287-292. https://doi.org/10.1097/TP.0b013e318203c27d [PubMed]

■ Carlson DF, Tan W, Hackett PB, Fahrenkrug SC (2014) Editing livestock genomes with site-specific nucleases. Reproduction, Fertility and Development 26(1): 74-82. https://doi.org/10.1071/RD13260 [PubMed]

■ Carlson DF, Tan W, Lillico SG, Stverakova D, Proudfoot C, Christian M, Voytas DF, Long CR, Whitelaw CBA, Fahrenkrug SC (2012) Efficient TALEN-mediated gene knockout in livestock. Proceedings of the National Academy of Sciences 109(43): 17382-17387. https:// doi.org/10.1073/pnas.1211446109 [PubMed]

■ Chari RS, Collins BH, Magee JC, DiMaio JM, Kirk AD, Harland RC, McCann RL, Platt JL, Meyers WC (1994) Treatment of hepatic failure with ex vivo pig-liver perfusion followed by liver transplantation. New England Journal of Medicine 331: 234-237. https://doi. org/10.1056/NEJM199407283310404

■ Chen J, Lansford R, Stewart V, Young F, Alt FW (1993) RAG-2-defi-cient blastocyst complementation: an assay of gene function in lymphocyte development. Proceedings of the National Academy of Sciences 90(10): 4528-4532. https://doi.org/10.1073/pnas.90.10.4528 [PubMed] [PMC]

■ Choi WI, Kim JH, Yoo HW, Oh SH (2010) A family with Townes-Brocks syndrome with congenital hypothyroidism and a novel mutation of the SALL1 gene. Korean Journal of Pediatrics 53(12): 1018-1021. https://doi.org/10.3345/kjp.2010.53.12.1018 [PubMed] [PMC]

■ Coelho MA, De Braekeleer E, Firth M, Bista M, Lukasiak S, Cuomo ME, Taylor BJM (2020) CRISPR GUARD protects off-target sites from Cas9 nuclease activity using short guide RNAs. Nature Communications 11(1): 4132. https://doi.org/10.1038/s41467-020-17952-5 [PubMed] [PMC]

■ Cowan PJ (2007) Coagulation and the xenograft endothelium. Xenotransplantation 14(1): 7-12. https://doi.org/10.1111/j.1399-3089.2006.00368.x [PubMed]

■ Cozzi NV, Gopalakrishnan A, Anderson LL, Feih JT, Shulgin AT, Daley PF, Ruoho AE (2009) Dimethyltryptamine and other hallucinogenic tryptamines exhibit substrate behavior at the serotonin uptake transporter and the vesicle monoamine transporter. Journal of Neural Transmission 116(12): 1591-1599. https://doi.org/10.1007/ s00702-009-0308-8 [PubMed]

■ Dai Y, Vaught TD, Boone J, Chen S-H, Phelps CJ, Ball S, Monahan JA, Jobst PM, McCreath KJ, Lamborn AE, Cowell-Lucero JL, Wells KD, Colman A, Polejaeva IA, Ayares DL (2002) Targeted disruption of the a1,3-galactosyltransferase gene in cloned pigs. Nature Biotechnology 20(3): 251-255. https://doi.org/10.1038/nbt0302-251 [PubMed]

■ Das S, Koyano-Nakagawa N, Gafni O, Maeng G, Singh BN, Ras-mussen T, Pan X, Choi K-D, Mickelson D, Gong W, Pota P, Weaver CV, Kren S, Hanna JH, Yannopoulos D, Garry MG, Garry DJ (2020) Generation of human endothelium in pig embryos deficient in ETV2. Nature Biotechnology 38(3): 297-302. https://doi.org/10.1038/ s41587-019-0373-y [PubMed]

■ Denner J (2018) Why was PERV not transmitted during preclinical and clinical xenotransplantation trials and after inoculation of animals? Retrovirology 15(1): 28. https://doi.org/10.1186/s12977-018-0411-8 [PubMed] [PMC]

■ Denner J (2021) Porcine endogenous retroviruses and xenotrans-plantation. Viruses 13(11): 2156. https://doi.org/10.3390/v13112156 [PubMed] [PMC]

■ Dieckhoff B, Karlas A, Hofmann A, Kues WA, Petersen B, Pfeifer A, Niemann H, Kurth R, Denner J (2007) Inhibition of porcine endogenous retroviruses (PERVs) in primary porcine cells by RNA interference using lentiviral vectors. Archives of Virology 152(3): 629-634. https://doi.org/10.1007/s00705-006-0868-y [PubMed]

■ Ding S, Wu X, Li G, Han M, Zhuang Y, Xu T (2005) Efficient transposition of the piggyBac (PB) transposon in mammalian cells and mice. Cell 122(3): 473-483. https://doi.org/10.1016/j. cell.2005.07.013 [PubMed]

■ Diswall M, Ängström J, Karlsson H, Phelps CJ, Ayares D, Teneberg S, Breimer ME (2010) Structural characterization of a1,3-galactosyl-transferase knockout pig heart and kidney glycolipids and their reactivity with human and baboon antibodies. Xenotransplantation 17(1): 48-60. https://doi.org/10.1111/j.1399-3089.2009.00564.x [PubMed]

■ Dolskiy AA, Gudymo AS, Taranov OS, Grishchenko IV, Shitik EM, Prokopov DY, Soldatov VO, Sobolevskaya EV, Bodnev SA, Danilchenko NV, Moiseeva AA, Torzhkova PY, Bulanovich YA, Onhonova GS, Ivleva EK, Kubekina MV, Belykh AE, Tregubchak TV, Ryzhikov AB, Gavrilova EV, Maksyutov RA, Deykin AV, Yud-kin DV (2022) The tissue distribution of SARS-CoV-2 in trans-genic mice with inducible ubiquitous expression of hACE2. Frontiers in Molecular Biosciences 8: 821506. https://doi.org/10.3389/ fmolb.2021.821506 [PubMed] [PMC]

■ Domínguez-Gil B, Haase-Kromwijk B, Van Leiden H, Neuberger J, Coene L, Morel P, Corinne A, Muehlbacher F, Brezovsky P, Costa AN, Rozental R, Matesanz R, on behalf of the European Committee (Partial Agreement) on Organ Transplantation. Council of Europe (CD-P-TO) (2011) Current situation of donation after circulatory death in European countries: Donation after circulatory death in the council of Europe. Transplant International 24(7): 676-686. https:// doi.org/10.1111/j.1432-2277.2011.01257.x [PubMed]

■ Ekser B, Ezzelarab M, Hara H, van der Windt DJ, Wijkstrom M, Bottino R, Trucco M, Cooper DK (2012) Clinical xenotransplan-tation: the next medical revolution? Lancet 379(9816): 672-683. https://doi.org/10.1016/S0140-6736(11)61091-X [PubMed]

■ Estrada J, Sommer J, Collins B, Mir B, Martin A, York A, Petters RM, Piedrahita JA (2007) Swine generated by somatic cell nuclear transfer have increased incidence of intrauterine growth restriction (IUGR). Cloning and Stem Cells 9(2): 229-236. https://doi. org/10.1089/clo.2006.0079 [PubMed]

■ Ezzelarab M, Garcia B, Azimzadeh A, Sun H, Lin CC, Hara H, Ke-lishadi S, Zhang T, Lin YJ, Tai H-C, Wagner R, Thacker J, Murase N, McCurry K, Barth RN, Ayares D, Pierson RN, Cooper DKC (2009) The innate immune response and activation of coagulation in a1,3-galactosyltransferase gene-knockout xenograft recipients. Transplantation 87(6): 805-812. https://doi.org/10.1097/TP. 0b013e318199c34f [PubMed] [PMC]

■ Flisikowska T, Merkl C, Landmann M, Eser S, Rezaei N, Cui X, Ku-rome M, Zakhartchenko V, Kessler B, Wieland H (2012) A porcine model of familial adenomatous polyposis. Gastroenterology 143(5): 1173-1175. https://doi.org/10.1053/j.gastro.2012.07.110 [PubMed]

■ Flisikowska T, Stachowiak M, Xu H, Wagner A, Hernandez-Cace-res A, Wurmser C, Perleberg C, Pausch H, Perkowska A, Fischer K (2017) Porcine familial adenomatous polyposis model enables systematic analysis of early events in adenoma progression. Scientific Reports 7(1): 6613. https://doi.org/10.1038/s41598-017-06741-8 [PubMed] [PMC]

■ Freedman BS (2018) Hopes and difficulties for blastocyst complementation. Nephron 138(1): 42-47. https://doi.org/10.1159/000480370 [PubMed] [PMC]

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

■ Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK (2014) Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nature Biotechnology 32(3): 279-284. https://doi.org/10.1038/ nbt.2808 [PubMed] [PMC]

■ Gaj T, Gersbach CA, Barbas CF (2013) ZFN, TALEN, and CRISPR/ Cas-based methods for genome engineering. Trends in Biotechnology 31(7): 397-405. https://doi.org/10.1016Zj.tibtech.2013.04.004 [PubMed] [PMC]

■ Gao X, Nowak-Imialek M, Chen X, Chen D, Herrmann D, Ruan D, Chen ACH, Eckersley-Maslin MA, Ahmad S, Lee YL, Kobayashi T, Ryan D, Zhong J, Zhu J, Wu J, Lan G, Petkov S, Yang J, Antunes L, Campos LS, Fu B, Wang S, Yong Y, Wang X, Xue S-G, Ge L, Liu Z, Huang Y, Nie T, Li P, Wu D, Pei D, Zhang Y, Lu L, Yang F, Kimber SJ, Reik W, Zou X, Shang Z, Lai L, Surani A, Tam PPL, Ahmed A, Yeung WSB, Teichmann SA, Niemann H, Liu P (2019) Establishment of porcine and human expanded potential stem cells. Nature Cell Biology 21(6): 687-699. https://doi.org/10.1038/s41556-019-0333-2 [PubMed] [PMC]

■ Gil MA, Martinez CA, Nohalez A, Parrilla I, Roca J, Wu J, Ross PJ, Cuello C, Izpisua JC, Martinez EA (2017) Developmental competence of porcine genome-edited zygotes. Molecular Reproduction and Development 84(9): 814-821. https://doi.org/10.1002/ mrd.22829 [PubMed]

■ Glauser EM (1966) Advantages of piglets as experimental animals in pediatric research. Experimental Medicine and Surgery 24(2): 181-190. [PubMed]

■ Goldman IL, Georgieva SG, Gurskiy YG, Krasnov AN, Deykin AV, Popov AN, Ermolkevich TG, Budzevich AI, Chernousov AD, Sadchikova ER (2012) Production of human lactoferrin in animal milk. Biochemistry and Cell Biology 90(3): 513-519. https://doi. org/10.1139/o11-088 [PubMed]

■ Graham F (2022) Daily briefing: First transplant of a gene-edited pig heart. Nature: d41586-022-00085-8. https://doi.org/10.1038/ d41586-022-00085-8 [PubMed]

■ Griffith BP, Goerlich CE, Singh AK, Rothblatt M, Lau CL, Shah A, Lorber M, Grazioli A, Saharia KK, Hong SN, Joseph SM, Ayares D, Mohiuddin MM (2022) Genetically modified porcine-to-human cardiac xenotransplantation. The New England Journal of Medicine 387(1): 35-44. https://doi.org/10.1056/NEJMoa2201422 [PubMed]

■ Groenen MAM, Archibald AL, Uenishi H, Tuggle CK, Takeuchi Y, Rothschild MF, Rogel-Gaillard C, Park C, Milan D, Megens H-J, Li S, Larkin DM, Kim H, Frantz LAF, Caccamo M, Ahn H, Aken BL, Anselmo A, Anthon C, Auvil L, Badaoui B, Beattie CW, Bendixen C, Berman D, Blecha F, Blomberg J, Bolund L, Bosse M, Botti S, Bujie Z, Bystrom M, Capitanu B, Carvalho-Silva D, Chardon P, Chen C, Cheng R, Choi S-H, Chow W, Clark RC, Clee C, Crooijmans RPMA, Dawson HD, Dehais P, De Sapio F, Dibbits B, Drou N, Du Z-Q, Eversole K, Fadista J, Fairley S, Faraut T, Faulkner GJ, Fowler KE, Fredholm M, Fritz E, Gilbert JGR, Giuffra E, Gorodkin J, Griffin DK, Harrow JL, Hayward A, Howe K, Hu Z-L, Humphray SJ, Hunt T, Hornsh0j H, Jeon J-T, Jern P, Jones M, Jurka J, Kanamori H, Kapeta-novic R, Kim J, Kim J-H, Kim K-W, Kim T-H, Larson G, Lee K, Lee K-T, Leggett R, Lewin HA, Li Y, Liu W, Loveland JE, Lu Y, Lunney JK, Ma J, Madsen O, Mann K, Matthews L, McLaren S, Morozumi T, Murtaugh MP, Narayan J, Truong Nguyen D, Ni P, Oh S-J, Onteru S, Panitz F, Park E-W, Park H-S, Pascal G, Paudel Y, Perez-Enciso M, Ramirez-Gonzalez R, Reecy JM, Rodriguez-Zas S, Rohrer GA, Rund L, Sang Y, Schachtschneider K, Schraiber JG, Schwartz J, Sco-bie L, Scott C, Searle S, Servin B, Southey BR, Sperber G, Stadler P, Sweedler JV, Tafer H, Thomsen B, Wali R, Wang J, Wang J, White S, Xu X, Yerle M, Zhang G, Zhang J, Zhang J, Zhao S, Rogers J, Churcher C, Schook LB (2012) Analyses of pig genomes provide insight into porcine demography and evolution. Nature 491(7424): 393-398. https://doi.org/10.1038/nature11622 [PubMed] [PMC]

■ Gurskiy YG, Garbuz DG, Soshnikova NV, Krasnov AN, Deikin A, Lazarev VF, Sverchinskyi D, Margulis BA, Zatsepina OG, Karpov VL, Belzhelarskaya SN, Feoktistova E, Georgieva SG, Evgen'ev MB (2016) The development of modified human Hsp70 (HSPA1A) and its production in the milk oftransgenic mice. Cell Stress Chaperones 21(6): 10551064. https://doi.org/10.1007/s12192-016-0729-x [PubMed] [PMC]

■ Hai T, Teng F, Guo R, Li W, Zhou Q (2014) One-step generation of knockout pigs by zygote injection of CRISPR/Cas system. Cell Research 24(3): 372-375. https://doi.org/10.1038/cr.2014.11 [PubMed] [PMC]

■ Hammer RE, Pursel VG, Rexroad CE, Wall RJ, Bolt DJ, Ebert KM, Palmiter RD, Brinster RL (1985) Production of transgenic rabbits, sheep and pigs by microinjection. Nature 315(6021): 680-683. https://doi.org/10.1038/315680a0 [PubMed]

■ Hauschild J, Petersen B, Santiago Y, Queisser A-L, Carnwath JW, Lucas-Hahn A, Zhang L, Meng X, Gregory PD, Schwinzer R, Cost GJ, Niemann H (2011) Efficient generation of a biallelic knockout in pigs using zinc-finger nucleases. Proceedings of the National Academy of Sciences 108(29): 12013-12017. https://doi.org/10.1073/ pnas.1106422108 [PubMed] [PMC]

■ Heinritz SN, Mosenthin R, Weiss E (2013) Use of pigs as a potential model for research into dietary modulation of the human gut mi-crobiota. Nutrition Research Reviews 26(2): 191-209. https://doi. org/10.1017/S0954422413000152 [PubMed]

■ Hölker M, Petersen B, Hassel P, Kues WA, Lemme E, Lucas-Hahn A, Niemann H (2005) Duration of in vitro maturation of recipient oocytes affects blastocyst development of cloned porcine embryos. Cloning and Stem Cells 7(1): 35-44. https://doi.org/10.1089/ clo.2005.7.35 [PubMed]

■ Ivics Z, Garrels W, Mates L, Yau TY, Bashir S, Zidek V, Landa V, Geurts A, Pravenec M, Rülicke T, Kues WA, Izsvak Z (2014) Germline transgenesis in pigs by cytoplasmic microinjection of Sleeping Beauty transposons. Nature Protocols 9(4): 810-827. https://doi. org/10.1038/nprot.2014.010 [PubMed]

■ Kahn D, Hickman R, Terblanche J, von Sommoggy St (1988) Partial hepatectomy and liver regeneration in pigs - The response to different resection sizes. Journal of Surgical Research 45(2): 176-180. https://doi.org/10.1016/0022-4804(88)90062-5 [PubMed]

■ Kararli TT (1995) Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals. Biopharmaceuticals and Drug Disposition 16(5): 351-380. https://doi.org/10.1002/bdd.2510160502 [PubMed]

■ Klymiuk N, Aigner B, Brem G, Wolf E (2010) Genetic modification of pigs as organ donors for xenotransplantation: Modification of pigs as organ donors. Molecular Reproduction and Development 77(3): 209-221. https://doi.org/10.1002/mrd.21127 [PubMed]

■ Kolber-Simonds D, Lai L, Watt SR, Denaro M, Arn S, Augenstein ML, Betthauser J, Carter DB, Greenstein JL, Hao Y, Im G-S, Liu Z, Mell GD, Murphy CN, Park K-W, Rieke A, Ryan DJJ, Sachs DH, Forsberg EJ, Prather RS, Hawley RJ (2004) Production of -1,3-ga-lactosyltransferase null pigs by means of nuclear transfer with fi-broblasts bearing loss of heterozygosity mutations. Proceedings of the National Academy of Sciences 101(19): 7335-7340. https://doi. org/10.1073/pnas.0307819101 [PubMed] [PMC]

■ Koppes EA, Redel BK, Johnson MA, Skvorak KJ, Ghaloul-Gon-zalez L, Yates ME, Lewis DW, Gollin SM, Wu YL, Christ SE, Yer-le M, Leshinski A, Spate LD, Benne JA, Murphy SL, Samuel MS, Walters EM, Hansen SA, Wells KD, Lichter-Konecki U, Wagner RA, Newsome JT, Dobrowolski SF, Vockley J, Prather RS, Nich-olls RD (2020) A porcine model of phenylketonuria generated by CRISPR/Cas9 genome editing. JCI Insight 5(20): e141523. https:// doi.org/10.1172/jci.insight.141523 [PubMed] [PMC]

■ Kumar TR, Larson M, Wang H, McDermott J, Bronshteyn I (2009) Transgenic mouse technology: Principles and methods. Methods in Molecular Biology 590: 335-362. https://doi.org/10.1007/978-1-60327-378-7_22 [PubMed] [PMC]

■ Kurome M, Geistlinger L, Kessler B, Zakhartchenko V, Klymiuk N, Wuensch A, Richter A, Baehr A, Kraehe K, Burkhardt K, Flisi-kowski K, Flisikowska T, Merkl C, Landmann M, Durkovic M, Tschukes A, Kraner S, Schindelhauer D, Petri T, Kind A, Nagashima H, Schnieke A, Zimmer R, Wolf E (2013) Factors influencing the efficiency of generating genetically engineered pigs by nuclear transfer: multi-factorial analysis of a large data set. BMC Biotechnology 13: 43. https://doi.org/10.1186/1472-6750-13-43 [PubMed] [PMC]

■ Kwon D-N, Lee K, Kang M-J, Choi Y-J, Park C, Whyte JJ, Brown AN, Kim J-H, Samuel M, Mao J, Park K-W, Murphy CN, Prather RS, Kim J-H (2013) Production of biallelic CMP-Neu5Ac hydroxylase knock-out pigs. Scientific Reports 3: 1981. https://doi. org/10.1038/srep01981 [PubMed] [PMC]

■ Lai L, Kolber-Simonds D, Park K-W, Cheong H-T, Greenstein JL, Im G-S, Samuel M, Bonk A, Rieke A, Day BN, Murphy CN, Carter DB, Hawley RJ, Prather RS (2002) Production of a-1,3-galactosyltrans-ferase knockout pigs by nuclear transfer cloning. Science 295(5557): 1089-1092. https://doi.org/10.1126/science.1068228 [PubMed]

■ Längin M, Mayr T, Reichart B, Michel S, Buchholz S, Guethoff S, Dashkevich A, Baehr A, Egerer S, Bauer A, Mihalj M, Panelli A, Issl L, Ying J, Fresch AK, Buttgereit I, Mokelke M, Radan J, Wer-

ner F, Lutzmann I, Steen S, Sjöberg T, Paskevicius A, Qiuming L, Sfriso R, Rieben R, Dahlhoff M, Kessler B, Kemter E, Kurome M, Zakhartchenko V, Klett K, Hinkel R, Kupatt C, Falkenau A, Reu S, Ellgass R, Herzog R, Binder U, Wich G, Skerra A, Ayares D, Kind

A, Schönmann U, Kaup F-J, Hagl C, Wolf E, Klymiuk N, Brenner P, Abicht J-M (2018) Consistent success in life-supporting porcine cardiac xenotransplantation. Nature 564(7736): 430-433. https:// doi.org/10.1038/s41586-018-0765-z [PubMed]

■ Lavitrano M, Bacci ML, Forni M, Lazzereschi D, Di Stefano C, Fio-retti D, Giancotti P, Marfe G, Pucci L, Renzi L, Wang H, Stoppac-ciaro A, Stassi G, Sargiacomo M, Sinibaldi P, Turchi V, Giovannoni R, Della Casa G, Seren E, Rossi G (2002) Efficient production by sperm-mediated gene transfer of human decay accelerating factor (hDAF) transgenic pigs for xenotransplantation. Proceedings of the National Academy of Sciences 99(22): 14230-14235. https://doi. org/10.1073/pnas.222550299 [PubMed] [PMC]

■ Le QA, Hirata M, Nguyen NT, Takebayashi K, Wittayarat M, Sato Y, Namula Z, Nii M, Tanihara F, Otoi T (2020) Effects of electro-poration treatment using different concentrations of Cas9 protein with gRNA targeting Myostatin (MSTN) genes on the development and gene editing of porcine zygotes. Animal Science Journal 91(1): e13386. https://doi.org/10.1111/asj.13386 [PubMed]

■ Leuchs S, Saalfrank A, Merkl C, Flisikowska T, Edlinger M, Durkovic M, Rezaei N, Kurome M, Zakhartchenko V, Kessler B (2012) Inactivation and inducible oncogenic mutation of p53 in gene targeted pigs. PLoS ONE 7(10): e43323. https://doi.org/10.1371/ journal.pone.0043323 [PubMed] [PMC]

■ Li M, Yang H, Zhao J, Fang L, Shi H, Li M, Sun Y, Zhang X, Jiang D, Zhou L, Wang D (2014) Efficient and heritable gene targeting in tilapia by CRISPR/Cas9. Genetics 197(2): 591-599. https://doi. org/10.1534/genetics.114.163667 [PubMed] [PMC]

■ Liu R, Simonetti FR, Ho Y-C (2020) The forces driving clonal expansion of the HIV-1 latent reservoir. Virology Journal 17(1): 4. https://doi.org/10.1186/s12985-019-1276-8 [PubMed] [PMC]

■ Maksimenko OG, Deykin AV, Khodarovich YM, Georgiev PG (2013) Use of transgenic animals in biotechnology: prospects and problems. Acta Naturae 5(1): 33-46. https://doi.org/10.32607/20758251-2013-5-1-33-46 [PubMed] [PMC]

■ Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M, Kosuri S, Yang L, Church GM (2013) CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nature Biotechnology 31(9): 833-838. https:// doi.org/10.1038/nbt.2675 [PubMed] [PMC]

■ Mansour SL, Thomas KR, Capecchi MR (1988) Disruption of the proto-oncogene int-2 in mouse embryo-derived stem cells: a general strategy for targeting mutations to non-selectable genes. Nature 336(6197): 348-352. https://doi.org/10.1038/336348a0 [PubMed]

■ Martin J, Sarai K, Yoshitake M, Haberstroh J, Takahashi N, Lutter G, Beyersdorf F (1999) Successful orthotopic pig heart transplantation from non-heart-beating donors. The Journal of Heart and Lung Transplantation 18(6): 597-606. https://doi.org/10.1016/S1053-2498(98)00017-5 [PubMed]

■ Mohiuddin M, Goerlich C, Singh A, Zhang T, Tatarov I, Lewis B, Sentz F, Hershfeld A, Braileanu G, Odonkor P, Strauss E, Williams

B, Burke A, Hittman J, Bhutta A, Tabatabai A, Gupta A, Vaught T, Sorrells L, Kuravi K, Dandro A, Eyestone W, Kaczorowski D, Ayares D, Griffith B (2021) Progressive genetic modifications with growth hormone receptor knockout extends cardiac xenograft sur-

vival to 9 months. Xenotransplantation 29(3): e12744.https://doi. org/10.21203/rs.3.rs-382870/v1 [PubMed]

■ Morris C, Drake M, Apperley J, Iacobelli S, van Biezen A, Bjork-strand B, Goldschmidt H, Harousseau J-L, Morgan G, de Witte T, Niederwieser D, Gahrton G, for the Myeloma Subcommittee of the Chronic Leukaemia Working Party of the EBMT (2010) Efficacy and outcome of autologous transplantation in rare myelomas. Haematologica 95(12): 2126-2133. https://doi.org/10.3324/haema-tol.2010.022848 [PubMed] [PMC]

■ Naeem M, Majeed S, Hoque MZ, Ahmad I (2020) Latest developed strategies to minimize the off-target effects in CRISPR-cas-me-diated genome editing. Cells 9(7): 1608. https://doi.org/10.3390/ cells9071608 [PubMed] [PMC]

■ Nagashima H, Matsunari H (2016) Growing human organs in pigs - A dream or reality? Theriogenology 86(1): 422-426. https://doi. org/10.1016/j.theriogenology.2016.04.056 [PubMed]

■ Niu D, Wei H-J, Lin L, George H, Wang T, Lee I-H, Zhao H-Y, Wang Y, Kan Y, Shrock E, Lesha E, Wang G, Luo Y, Qing Y, Jiao D, Zhao H, Zhou X, Wang S, Wei H, Güell M, Church GM, Yang L (2017) Inactivation of porcine endogenous retrovirus in pigs using CRIS-PR-Cas9. Science 357(6357): 1303-1307. https://doi.org/10.1126/ science.aan4187 [PubMed] [PMC]

■ Onishi A, Iwamoto M, Akita T, Mikawa S, Takeda K, Awata T, Hanada H, Perry ACF (2000) Pig cloning by microinjection of fetal fibroblast nuclei. Science 289(5482): 1188-1190. https://doi.org/10.1126/ science.289.5482.1188 [PubMed]

■ Oropeza M, Petersen B, Carnwath JW, Lucas-Hahn A, Lemme E, Hassel P, Herrmann D, Barg-Kues B, Holler S, Queisser A-L, Schwinzer R, Hinkel R, Kupatt C, Niemann H (2009) Transgenic expression of the human A20 gene in cloned pigs provides protection against apoptotic and inflammatory stimuli. Xenotransplantation 16(6): 522-534. https://doi.org/10.1111/j.1399-3089.2009.00556.x [PubMed]

■ Padler-Karavani V, Yu H, Cao H, Chokhawala H, Karp F, Varki N, Chen X, Varki A (2008) Diversity in specificity, abundance, and composition of anti-Neu5Gc antibodies in normal humans: Potential implications for disease. Glycobiology 18(10): 818-830. https://doi. org/10.1093/glycob/cwn072 [PubMed] [PMC]

■ Perleberg C, Kind A, Schnieke A (2018) Genetically engineered pigs as models for human disease. Disease Models and Mechanisms 11(1): dmm030783. https://doi.org/10.1242/dmm.030783 [PubMed] [PMC]

■ Petersen B, Ramackers W, Tiede A, Lucas-Hahn A, Herrmann D, Barg-Kues B, Schuettler W, Friedrich L, Schwinzer R, Winkler M, Niemann H (2009) Pigs transgenic for human thrombomodulin have elevated production of activated protein C. Xenotransplantation 16(6): 486-495. https://doi.org/10.1111/j.1399-3089.2009.00537.x [PubMed]

■ Petersen B, Ramackers W, Lucas-Hahn A, Lemme E, Hassel P, Queißer A-L, Herrmann D, Barg-Kues B, Carnwath JW, Klose J, Tiede A, Friedrich L, Baars W, Schwinzer R, Winkler M, Niemann H (2011) Transgenic expression of human heme oxygenase-1 in pigs confers resistance against xenograft rejection during ex vivo perfusion of porcine kidneys: Human heme oxygenase-1 in pigs. Xenotransplantation 18(6): 355-368. https://doi.org/10.1111/j.1399-3089.2011.00674.x [PubMed]

■ Phelps CJ, Koike C, Vaught TD, Boone J, Wells KD, Chen S-H, Ball S, Specht SM, Polejaeva IA, Monahan JA, Jobst PM, Sharma SB, Lamborn AE, Garst AS, Moore M, Demetris AJ, Rudert WA, Bottino

R, Bertera S, Trucco M, Starzl TE, Dai Y, Ayares DL (2003) Production of a1,3-galactosyltransferase-deficient pigs. Science 299(5605): 411-414. https://doi.org/10.1126/science.1078942 [PubMed] [PMC]

■ Porrett PM, Orandi BJ, Kumar V, Houp J, Anderson D, Cozette Killian A, Hauptfeld-Dolejsek V, Martin DE, Macedon S, Budd N, Stegner KL, Dandro A, Kokkinaki M, Kuravi KV, Reed RD, Fatima H, Killian JT, Baker G, Perry J, Wright ED, Cheung MD, Erman EN, Kraebber K, Gamblin T, Guy L, George JF, Ayares D, Locke JE (2022) First clinical-grade porcine kidney xenotransplant using a human decedent model. American Journal of Transplantation 22(4): 1037-1053. https://doi.org/10.1111/ajt.16930 [PubMed]

■ Ramsoondar JJ, Machâty Z, Costa C, Williams BL, Fodor WL, Bon-dioli KR (2003) Production of a1,3-galactosyltransferase-knockout cloned pigs expressing human a1,2-fucosylosyltransferase1. Biology of Reproduction 69(2): 437-445. https://doi.org/10.1095/biolre-prod.102.014647 [PubMed]

■ Ribitsch I, Baptista PM, Lange-Consiglio A, Melotti L, Patruno M, Jenner F, Schnabl-Feichter E, Dutton LC, Connolly DJ, van Steen-beek FG, Dudhia J, Penning LC (2020) Large animal models in regenerative medicine and tissue engineering: to do or not to do. Frontiers in Bioengineering and Biotechnology 8: 972. https://doi. org/10.3389/fbioe.2020.00972 [PubMed] [PMC]

■ Regalado A (2022) The gene-edited pig heart given to a dying patient was infected with a pig virus. MIT Technology Review. https://www. technologyreview.com/2022/05/04/1051725/xenotransplant-pa-tient-died-received-heart-infected-with-pig-virus/

■ Ruiz-Estevez M, Crane AT, Rodriguez-Villamil P, Ongaratto FL, You Y, Steevens AR, Hill C, Goldsmith T, Webster DA, Sherry L, Lim S, Denman N, Low WC, Carlson DF, Dutton JR, Steer CJ, Gafni O (2021) Liver development is restored by blastocyst complementation of HHEX knockout in mice and pigs. Stem Cell Research & Therapy 12(1): 292. https://doi.org/10.1186/s13287-021-02348-z [PubMed] [PMC]

■ Saalfrank A, Janssen KP, Ravon M, Flisikowski K, Eser S, Steiger K, Flisikowska T, Muller-Fliedner P, Schulze E, Bronner C (2016) A porcine model of osteosarcoma. Oncogenesis 5(3): e210 https://doi. org/10.1038/oncsis.2016.19 [PubMed] [PMC]

■ Soto DA, Ross PJ (2016) Pluripotent stem cells and livestock genetic engineering. Transgenic Research 25(3): 289-306. https://doi. org/10.1007/s11248-016-9929-5 [PubMed] [PMC]

■ Specke V, Rubant S, Denner J (2001) Productive infection of human primary cells and cell lines with porcine endogenous retroviruses. Virology 285(2): 177-180. https://doi.org/10.1006/viro.2001.0934 [PubMed]

■ Spetzler VN, Goldaracena N, Echiverri J, Kaths JM, Louis KS, Ad-eyi OA, Yip PM, Grant DR, Selzner N, Selzner M (2016) Subnor-mothermic ex vivo liver perfusion is a safe alternative to cold static storage for preserving standard criteria grafts: ex vivo liver perfusion for HBD grafts. Liver Transplantation 22(1): 111-119. https://doi. org/10.1002/lt.24340 [PubMed]

■ Suchy F, Nakauchi H (2017) Lessons from interspecies mammalian chimeras. Annual Review of Cell and Developmental Biology 33: 203-217. https://doi.org/10.1146/annurev-cellbio-100616-060654 [PubMed]

■ Swindle MM, Makin A, Herron AJ, Clubb FJ, Frazier KS (2012) Swine as models in biomedical research and toxicology testing. Veterinary Pathology 49(2): 344-356. https://doi. org/10.1177/0300985811402846 [PubMed]

■ Tan W, Proudfoot C, Lillico SG, Whitelaw CBA (2016) Gene targeting, genome editing: from Dolly to editors. Transgenic Research 25(3): 273-287. https://doi.org/10.1007/s11248-016-9932-x [PubMed] [PMC]

■ Tanihara F, Hirata M, Nguyen NT, Le QA, Hirano T, Otoi T (2019a) Effects of concentration of CRISPR/Cas9 components on genetic mosaicism in cytoplasmic microinjected porcine embryos. Journal of Reproduction and Development 65(3): 209-214. https://doi. org/10.1262/jrd.2018-116 [PubMed] [PMC]

■ Tanihara F, Hirata M, Nguyen NT, Sawamoto O, Kikuchi T, Otoi T (2021a) One-step generation of multiple gene-edited pigs by electro-poration of the CRISPR/Cas9 system into zygotes to reduce xenoanti-gen biosynthesis. International Journal of Molecular Sciences 22(5): 2249. https://doi.org/10.3390/ijms22052249 [PubMed] [PMC]

■ Tanihara F, Hirata M, Nguyen NT, Le QA, Wittayarat M, Fahrudin M, Hirano T, Otoi T (2021b) Generation of CD163-edited pig via electroporation of the CRISPR/Cas9 system into porcine in vitro-fer-tilized zygotes. Animal Biotechnology 32(2): 147-154. https://doi.or g/10.1080/10495398.2019.1668801 [PubMed]

■ Tanihara F, Hirata M, Nguyen NT, Le QA, Hirano T, Takemoto T, Nakai M, Fuchimoto D, Otoi T (2019b) Generation of PDX-1 mutant porcine blastocysts by introducing CRISPR/Cas9-system into porcine zygotes via electroporation. Animal Science Journal 90(1): 55-61. https://doi.org/10.1111/asj.13129 [PubMed]

■ Vakulskas CA, Behlke MA (2019) Evaluation and reduction of CRISPR off-target cleavage events. Nucleic Acid Therapeutics 29(4): 167-174. https://doi.org/10.1089/nat.2019.0790 [PubMed] [PMC]

■ Vogel T, Brockmann JG, Pigott D, Neil DAH, Muthusamy ASR, Coussios CC, Friend PJ (2017) Successful transplantation of porcine liver grafts following 48-hour normothermic preservation. PLoS ONE 12(11): e0188494. https://doi.org/10.1371/journal. pone.0188494 [PubMed] [PMC]

■ Wang K, Ouyang H, Xie Z, Yao C, Guo N, Li M, Jiao H, Pang D (2015) Efficient generation of myostatin mutations in pigs using the CRISPR/Cas9 system. Scientific Reports 5: 16623. https://doi. org/10.1038/srep16623 [PubMed] [PMC]

■ Wittayarat M, Hirata M, Namula Z, Sato Y, Nguyen NT, Le QA, Lin Q, Takebayashi K, Tanihara F, Otoi T (2021) Introduction of a point mutation in the KRAS gene of in vitro fertilized porcine zygotes via electroporation of the CRISPR/Cas9 system with single-stranded oligodeoxynucleotides. Animal Science Journal 92(1):e13534. https://doi.org/10.1111/asj.13534 [PubMed]

■ Xu C, Wu S, Schook LB, Schachtschneider KM (2019) Translating human cancer sequences into personalized porcine cancer models. Frontiers in Oncology 9: 105. https://doi.org/10.3389/ fonc.2019.00105 [PubMed] [PMC]

■ Yamada K, Shah JA, Tanabe T, Lanaspa MA, Johnson RJ (2017) Xenotransplantation: Where are we with potential kidney recipients? Recent progress and potential future clinical trials. Current Transplantation Reports 4(2): 101-109. https://doi.org/10.1007/s40472-017-0149-6 [PubMed] [PMC]

■ Yue Y, Xu W, Kan Y, Zhao H-Y, Zhou Y, Song X, Wu J, Xiong J, Goswami D, Yang M, Lamriben L, Xu M, Zhang Q, Luo Y, Guo J, Mao S, Jiao D, Nguyen TD, Li Z, Layer JV, Li M, Paragas V, Youd ME, Sun Z, Ding Y, Wang W, Dou H, Song L, Wang X, Le L, Fang X, George H, Anand R, Wang SY, Westlin WF, Güell M, Markmann J, Qin W, Gao Y, Wei H-J, Church GM, Yang L (2021) Extensive germline genome engineering in pigs. Nature Biomedical Engineering 5(2): 134-143. https://doi.org/10.1038/s41551-020-00613-9 [PubMed]

■ Zimmerhackl LB, Hofer J, Cortina G, Mark W, Würzner R, Jun-graithmayr TC, Khursigara G, Kliche KO, Radauer W (2010) Prophylactic eculizumab after renal transplantation in atypical hemo-lytic-uremic syndrome. New England Journal of Medicine 362(18): 1746-1748. https://doi.org/10.1056/NEJMc1001060 [PubMed]

Author contributions

■ Elena I. Leonova, PhD in Biology, Head of the Center for Transgenesis and Genome Editing of St. Petersburg State University, Scientific Center for Genetics and Life Sciences of Sirius University of Science and Technology, e-mail: e.leonova@spbu.ru, ORCID ID https://orcid.org/0000-0002-0236-3302. The author provided the idea of research, analyzed the material, results and made the conclusions, wrote and edited the text of the article.

■ Vasily V. Reshetnikov, researcher, Department of Biotechnology, Sirius University of Science and Technology, e-mail: reshetnikov.vv@talantiuspeh.ru The author defined the idea of research, analyzed the material, results and conclusions, wrote the text of the article and presented picture

■ Julia V. Sopova, PhD in Biology, Center of Tansgenesic and Genome Editing, Institute of Translational Biomedici-ne, St. Petersburg State University, e-mail: y.sopova@ spbu.ru, ORCID ID https://orcid.org/0000-0002-7825-273X. The author defined the idea of research, analyzed the material, results and conclusions, wrote the text of the article.

i Надоели баннеры? Вы всегда можете отключить рекламу.