Open access peer-reviewed chapter

Induced Pluripotent Stem Cells from Animal Models: Applications on Translational Research

Written By

Laís Vicari de Figueiredo Pessôa, Naira Caroline Godoy Pieri, Kaiana Recchia and Fabiana Fernandes Bressan

Submitted: 22 June 2020 Reviewed: 25 September 2020 Published: 27 October 2020

DOI: 10.5772/intechopen.94199

From the Edited Volume

Novel Perspectives of Stem Cell Manufacturing and Therapies

Edited by Diana Kitala and Ana Colette Maurício

Chapter metrics overview

816 Chapter Downloads

View Full Metrics

Abstract

Over the history of humankind, knowledge acquisition regarding the human body, health, and the development of new biomedical techniques have run through some animal model at some level. The mouse model has been primarily used as the role model for a long time; however, it is severely hampered regarding its feasibility for translational outcomes, in particular, to preclinical and clinical studies. Herein we aim to discuss how induced pluripotent stem cells generated from non-human primates, pigs and dogs, all well-known as adequate large biomedical models, associated or not with gene editing tools, can be used as models on in vivo or in vitro translational research, specifically on regenerative medicine, drug screening, and stem cell therapy.

Keywords

  • pluripotency
  • regenerative medicine
  • stem cell
  • therapy
  • domestic animals
  • non-human primates

1. Introduction

For centuries, animal models have been used to aid on the quest for knowledge regarding human anatomy, physiology, and health, at first by simple observation, progressing to a proper investigation, selection of adequate models for given conditions and resuming on the development of specific transgenic animal models [1]. A recent concern regarding welfare and animal rights [2] has highlighted the relevance of in vitro models, such as pluripotent and adult stem cells. Here we describe the recent advances of biomedical research using induced pluripotent stem cell (iPSCs) models isolated from non-human primates, pigs, and dogs. Due to anatomical, physiologic, genetic, environmental, and other similarities to humans and conditions, those animals are considered highly relevant models for translational studies, each presenting specific advantages and drawbacks. Herein we discuss the advantages of using iPSCs, associated or not with gene editing tools, to enlarge the value and possible applications for pharmaceutical development and therapeutic approaches in these models.

Advertisement

2. Stem cells from animal models: applications on translational research

2.1 Non-human primates: most promising although challenging model?

Although non-human primates (NH-primates) represent only a small share of the animals used in medical research, the significance of those studies for human health, especially pharmaceuticals and new therapeutic approaches, is prominent [3, 4]. NH-primates are often the most suitable model for assessing the safety and efficiency of said drugs prior to human trials [5] and supply information to connect data from other relevant clinical models, such as rodents, to humans [6].

As study models, NH-primates are highly attractive due to longevity, behavioral, anatomical, genetic, physiological, and immunological similarities with humans [57, 8, 9, 10]. Over the past decades, NH-primates have been used on studies and research to prevent or cure human conditions, through the development of vaccines and drugs or treatment for cancer, diabetes, obesity, Parkinson’s and other neurodegenerative, respiratory and cardiovascular diseases [4, 5, 11, 12], as well as methods to prevent mother-fetus transmission of diseases such as HIV [13], amongst other conditions and illnesses. Moreover, it has recently been shown that some NH-primates present a working memory capacity similar to that of human children [14], which highlights their importance for cognitive and neurological studies.

Stem cells are also considered an excellent tool for disease modeling and drug screening [15]. Although pluripotent cells derived from embryos, also called embryonic stem cells (ESCs), and multipotent adult stem cells (ASCs) are relevant and have been widely used on stem cell research and therapy purposes [16, 17, 18, 19, 20, 21, 22, 23], ESCs limited sources and ASCs limited proliferation, and differentiation potentials have hampered their use. The advent of inducing pluripotency in vitro on virtually any somatic cell from any species reported since 2006, led to an entire flock of biotechnological and therapeutic applications. Thus, since the debut of induced pluripotent stem cells (iPSCs) [24], it is possible to produce patient-specific pluripotent stem cells that are highly valuable as models [15]. Furthermore, supported by age-related changes on the immune system of both humans and NH-primates [25], the use of said animals modeling human diseases associated with stem cell research might provide remarkable insight on translational stem cell-based therapy and transplantation [6].

Since iPSCs were first reported, these cells are now available for a variety of wild and domestic animal species (reviewed by [26]). Amongst NH-primates, they include but are not limited to the rhesus [27]; drill [28]; cynomolgus monkey [2930]; marmoset [31]; baboon [32]; orangutans [33]; Japanese macaque [34]. These cells were mainly generated from fibroblasts and integrative methods, but more recently, they were produced through non-integrative methods, such as Sendai-virus and episomal vectors [10, 35, 36, 37]. NH-primate-derived iPSCs have been used in research related to or as models for neurological [38, 39, 40, 41], cardiac [3642, 43], reproductive [44], hematopoietic conditions [37, 45], transplantation and grafting [30, 46] and others.

As previously stated, similarities between humans and NH-primates make them essential models to assess the safety of drugs and therapeutic methodologies before human trials [5]. Immunologic similarities were considered when multiple NH-primate species were chosen as models to establish an iPSCs-derived multipotential hematopoietic progenitor cell differentiation protocol [37] and baboon enucleated red blood cells derived from iPSCs [45], aiming at blood disease and others preclinical testing. Cell transplantation is a relevant therapeutic methodology for some cardiac conditions leading to heart failure [42]. NH-primates iPSCs-derived cardiomyocytes were generated from rhesus monkeys [36, 43] and cynomolgus monkeys [42, 47] to assess drug screening, regenerative therapy, grafting viability, and immune rejection potential.

Aside from immunologic, physiologic, and genetic similarities, NH-primates cognitive capacity and longevity drag special attention for these animals as models for mental illness, age-related or not. Huntington’s disease transgenic animals iPSCs have been used for generating neural progenitor cells that may be addressed for drug screening [48] pathogenesis modeling [40] and epigenetic and transcriptional profile analyses [49]. iPSCs and iPSCs-derived neural stem cells have also been generated from other NH-primate species aiming to develop regenerative therapy methods and modeling other neurological conditions, such as Alzheimer’s and Parkinson’s Disease [10, 36, 50, 51, 52].

Nevertheless, another possibility is the generation of custom-made specific transgenic disease models, by injecting retroviruses expressing target genes or gene editing techniques. The most known gene editing tools are zinc finger nuclease (ZFN), transcription activator-like effectors nuclease (TALEN), and clustered regularly interspaced short palindromic repeats (CRISPR). More recently, ZFNs and TALENs have been superseded by CRISPR/Cas9, which is equally, if not more efficient in inducing double-strand breaks (DSBs) and in stimulating homology-directed repair (HDR) [5354], also offering improved target specificity, prediction of off-target effects and activity [53, 55, 56]. Those approaches have been successfully applied to generate various NH-primate models (Reviewed by [5758]), including the above mentioned Huntington’s disease transgenic monkey [59], Parkinson’s [41, 60], neurodevelopmental disorders [61], Duchenne muscular dystrophy [62], severe combined immunodeficiency [63], and others.

Those models represent a significant scientific advance, allowing more faithful models than rodents previously used [58]. Although the use of NH-primate as research models is notable, some issues still need to be addressed. The greatly developed social skills of those animals implicate in environmental and social requirements to be met to keep NH-primates in an ethical and healthy environment ([64] art. 17), which implicates in high costs. Furthermore, results obtained from NH-primates studies are often not translatable to human research [65], highlighting the need for other research models, such as porcine and canine.

2.2 Swine: a large model in an already optimized production system

The domestication of swine (Sus scrofa domesticus) as a farm animal in established and controlled housing conditions, including specific conditions free of pathogens, has led to an important wide public acceptance that requires only minor adaptations for research [66]. The swine reproductive maturity is relatively fast compared to other large species (6–8 months), and they present a short gestation period (115 days) associated with the capability to produce large litters, with around 8–16 piglets per litter. Also, the swine body size, anatomy, physiology, and genetic homology are compatible with humans [66, 67, 68, 69]. Hence, they are one of the most exciting species as a translational model for regenerative medicine research, and probably the most similar physiological model for humans apart from NH-primates.

The swine has already been explored as a biomedical model to develop diagnostic methods, studies, and treatment for several different conditions and diseases. For example, immunology studies and allergy models [70], and respiratory and cardiovascular conditions, such as pulmonary surfactant function, reperfusion injury, pulmonary hypertension, and asthma [71, 72, 73]. Similar to humans, swine are omnivores, reassuring its adequacy in studies examining the gastrointestinal system: transit time of pharmaceuticals [74], inflammatory bowel disease [75], gastric dilation [76] and metabolic disorders that influence of endocrine system [77, 78, 79]. The swine model has also been used to study neurological and neurodegenerative human disorders, such as amyotrophic lateral sclerosis, Alzheimer’s Disease [80, 81], and Huntington’s Disease [82].

For the advancement of regenerative medicine, specifically regarding cellular therapies, it is of great importance to study swine stem cells aiming to prove its efficacy and safety. Researchers have already demonstrated the effectiveness of treatment in the swine model using ASCs such as bone marrow mesenchymal stem cells (BM-MSCs) for the repair of myocardial infarction [83] and also for autologous therapy for disc degeneration [84].

However, cellular therapies using multipotent stem cells are restricted to specific diseases due to the limited capacity for differentiation to specific types of cells. Pluripotent stem cells, nevertheless, circumvents such drawback by presenting the ability to differentiate into several cells from the endoderm, mesoderm, or ectoderm origin, thus expanding the possibility translational studies for regenerative medicine [67].

ESCs are often studied and divided into two pluripotency states: naïve or primed. Naïve ESCs are found in the pre-implantation embryo, in the inner cell mass (ICM), and primed ESCs are found in the post-implantation stage in the epiblast [85, 86]. It is known that the mice ESCs cultured and maintained in vitro are considered “naïve”, are collected from ICM and supplemented in culture with LIF, although human ECSs are collected from the epiblast and maintained in vitro with bFGF supplementation (for more details, refer to [87, 88]). For animal models including swine, the establishment of robust pluripotent ESCs using a straightforward and conventional approach has not yet been reported, and protocols regarding naive or primed pluripotency state characterization have not been consistent in the last decades [89].

Hence, the generation of iPSCs has shown to provide critical advantages over ESCs, particularly, when animal models are used. The iPSCs were already derived in the swine model (pig iPSCs or piPSCs) and reported in over 25 studies. The majority of those studies have used integrative methodologies to reprogram cells derived from embryonic, fetal, or adult fibroblasts. Although more efficient then non-integrative methods, integration of reprogramming factors onto the cell’s genome might lead to the persistent expression of said factors, which can generate tumors and become unfavorable for cell therapy [90, 91]. Pluripotency induction using non-integrative vectors would greatly assist their use in cellular therapy [92]; however, piPSCs produced by episomal non-integrative methodology were until now only considered iPSCs-like [93].

piPSCs have already been induced to differentiate into several lineages: cardiomyocytes [94], hepatocytes [95], and even neuronal precursors cells [80, 96]. Kim et al. [96] for example, reported the derivation of piPSCs using porcine embryonic fibroblasts (PEFs) with four doxycycline-inducible human factors inserted into the cell by lentivirus, and the iPSCs generated were induced into neuronal progenitor cells (NPCs), positive for neuronal cells markers (PLAG1, NESTIN, and VIMENTIN). The differentiation protocol of iPSCs into NPCs can assist in future studies on animal models for neurodegenerative diseases, and the transplantation of these cells may provide details regarding the regenerative potency in vivo.

In particular, the swine is an attractive model to study human genetic diseases due to the genetic homology found between the species [97, 98, 99]. The extension of genetic editing tools to the piPSCs could significantly increase their value as a biomedical model, motivating efforts to develop safe and efficient genome editing technologies in this model, aiming to replicate human disease and develop therapeutic approaches [100].

In swine, gene editing tools are more prone to be effective and accepted once reproductive biotechnologies (such as embryo manipulation and microinjection and somatic cell nuclear cloning – SCNT) are far more studied than other models such as NH-primates and dogs [101]. The use of CRISPR/Cas9 injection into swine zygotes, for example, has been reported as an exciting model for human disease based on gene knock-out [102, 103, 104], in special, presenting high efficiency and without detection of off-targets [105].

Gene editing is highly explored in human iPSCs for cardiovascular, neurodegenerative diseases like Alzheimer’s and Parkinson’s, and degenerative muscular dystrophy (DMD), however, its applicability in autologous therapies is still restricted. Thus, gene editing in piPSCs to study diseases and their treatments [106] and transplant these cells or even to generate new entire edited organisms is a game-changer in the regenerative medicine field. Yu [104] edited swine zygotes using CRISPR/Cas9 for DMD the piglets born had the disease in skeletal muscle, heart and decreased smooth muscle thickness in the stomach and intestine. These models would enable, trough gene editing on piPSCs, to test autologous therapies for DMD.

Apart from the use of edited cells for cellular therapy, the technology would also be useful to the production of human organs by interspecies blastocyst-iPSCs complementation [68, 107]. Wu [108, 109] described the chimeras’ production through the complementation of hiPSCs in swine zygotes genetically edited via CRISPR/Cas9. Researchers also reported to efficiently disable pancreatogenesis in pig embryos via zygotic co-delivery of Cas9 mRNA and dual sgRNAs targeting the PDX1 gene. When combined with chimeric-competent human pluripotent stem cells, the authors inferred that these results would provide a suitable platform for the xeno-generation of human tissues and organs in pigs [108, 109].

Bypassing the ethical problems of possible humanization of the swine during the embryo complementation process, another option for producing patient-specific organs is to recellularize swine organ scaffolds with hiPSCs. The selected organ goes through the decellularization process that completely removes cells and organic components of tissue, such as lipids, DNA, and antigenic proteins, but maintains the extracellular matrix (ECM). Recently, Goldfracht [110] combined hiPSCS-cardiomyocytes (hiPSC-CMs) with extracellular-matrix (ECM) derived from decellularized swine hearts, developing an ECM-derived engineered heart tissues (ECM-EHTs) model. Ohata and Ott [111] decellularized the lungs of human, swine, and NH-primates, the structure kept the original bronchial tree, vascular network, and most of the ECM composition and bioreactors were used to recellularize the lungs, and successful cell growth was achieved with perfusion culture.

Organ engineering based on recellularization with patient-derived iPSCs offers the unique potential to promote autologous treatment, and are also promising as tools for animal production, once piPSCs differentiated into germinative cells could be used to re-colonize depleted ovaries or testicles in order to spread the desired genetics in other animals [112]. Also, the use of muscle differentiation from iPSCs in scaffolds would benefit not only the cellular therapy for injured muscles in general, but opens new possibilities regarding in vitro meat production. The production of “animal-free” meat offers a reduction in environmental pollution and allows disease-free meat production due to its controllable and manipulative production system. However, technical challenges and intense research are still needed to establish such “animal-free” meat culture system [68, 113].

Although the complete reprogramming of iPSCs in the swine model is not yet fully elucidated as it is for human and murine reprogramming, the technology has the clear benefit of improving animal production and reproduction, opening new perspectives to study genetic diseases or develop cellular transplantation therapies. Studies are still needed to optimize the production of non-transgenic piPSCs and their association with other biotechnologies in the swine model, and the inter-species difference regarding pluripotency acquisition is important in order to define proper culture conditions to maintain the pluripotency and the reprogramming protocols in this model.

2.3 Canines: closer to humans than ever

The dog (Canis lupus familiaris) is considered a well-suited animal model for many diseases, drug development, and regenerative therapies. Like humans, dogs present a great phenotypic diversity and a well-mixed gene pool because of centuries of random breeding [114], and they also exhibit metabolic, physiological, and anatomical similarities to humans [115]. More than 200 known hereditary canine diseases have an equivalent human disease, including cardiomyopathies, muscular dystrophy, and cancer. Moreover, the dog was the most prevalently used species in early transplantation research, including bone marrow transplantation and gene therapy [116], due to their similarities to humans concerning stem cell kinetics, hematopoietic demand, and responsiveness to cytokines [117, 118].

Because of the many similar cancer characteristics in dogs and humans, including histological features, genetics, behavior, and response to conventional therapies [119], dogs are amongst the leading models for human cancer studies. Notably, the number of dogs that are diagnosed and managed with cancer is estimated to be over 6 million per year in the United States [119]. Such conditions triggered researchers’ interest and efforts to identify cancer-associated genes, study the environmental risk factors, understand tumor biology and progression, and develop of novel cancer therapeutics [120]. Different researchers described similar types of cancer in dogs and humans that include prostate, skin, mammary, lymphoid neoplasia, and others [121, 122, 123, 124]. Nonetheless, it should be recognized that just as in other models, both similarities and dissimilarities exist [119], including disparities concerning genomic factors, clinical behavior, and prevalence. For example, the BRAF gene’s somatic mutation occurs in nearly 60% of melanoma from humans, but only in approximately 6% of dogs [125]. Also, while osteosarcoma most typically affects the appendicular skeleton and metastasizes to lungs in humans and dogs, peak onset occurs at a young age in humans, but more often at an advanced age in dogs [126].

Stem cell research is a recent and increasing field for canines, unraveling the development of novel cell-based disease models, drug discovery, and therapies. Some research with ASCs has been performed dogs due to their regeneration properties. Canine MSCs (cMSCs), for example, successfully recovered damaged spinal cord neurons [127], increased tubular epithelial cell proliferation in cisplatin-induced kidney damage [128], successfully treated osteonecrosis [129], repaired infarcted myocardial tissue [130], are capable of chondrogenic differentiation [131] and suppression of inflammation of ruptured crucial ligament [132].

As previously discussed, ASCs cells have limitations when considered for therapy or regenerative medicine, such as limited proliferation, expansion, and differentiation potentials. In contrast, pluripotent stem cells can fill a critical void in regenerative medicine by allowing autologous studies or gene editing for in vivo or in vitro disease modeling. Similar to pigs, but in particular in dogs, isolation of genuinely pluripotent cells has been challenging. According to [133] six studies from 2007 until 2009 derived ESCs from blastocysts that expressed the core pluripotency markers and were capable of differentiating into representative lineages of all three germ layers in vitro; however, a limited proliferative potential and differentiation in germ cells layers were observed in all [134, 135, 136, 137, 138, 139]. Moreover, a consensus regarding typical morphology and cell culture conditions are still unreported [136, 138].

iPSCs generation in canines has evolved quickly. Some studies have shown that the generation of canine induced pluripotency stem cells (ciPSCs) from fetal or adult cells through retroviral transduction of dog, human, or mouse factors [140, 141, 142, 143, 144, 145, 146, 147, 148, 149]. The pluripotency state of the ciPSCs (naïve, primed, or other) has been discussed, and the proper characterization of these cells lacks consensus. These studies tested different medium and supplement combinations in culture and reported different pluripotency acquisition requirements and maintenance (different culture supplementation and different cell surface markers detection). Interestingly, [141] obtained ciPSCs derived from adipose multipotent stromal cells that showed similarity to human ESCs regarding morphology, pluripotency markers expression, and the ability to differentiate into all three derivatives germ layers in vitro (endoderm, ectoderm, and mesoderm).

Remarkably, dogs develop breed-associated genetic predispositions to particular disorders and suffer from many of the same maladies as humans. Many genetic diseases, such as Alzheimer’s disease, retinal atrophy, muscular dystrophy, cancer, obesity, cardiovascular diseases, and diabetes mellitus, affect dogs and humans [121, 135, 150]. For instance, the neurobehavioral syndrome called canine cognitive dysfunction (CCD), which affects 14.2–22.5% of dogs over eight years old, shares many clinical and neuropathological similarities with human aging and early stages of Alzheimer’s Disease [151, 152, 153, 154]. Recently, Hyttel and collaborators [155] aimed to characterize the CCD condition in iPSC-derived neurons from aged demented and healthy dogs, allowing the comparison of CCD with human Alzheimer’s at the cellular level. Canine iPSCs have also been tested in other studies, as researchers transplanted autologous iPSCs into the myocardial wall of dogs to examine the potential for myocardial infarct treatment, and the stem cell population were tracked regarding distribution, migration, engraftment, survival, proliferation, and differentiation [142].

Although biotechnological techniques and tools for the dog are less developed than for other species such as swine, the progress on gene editing technologies that can correct genetic defects, thereby offering potential treatment of some inherited diseases, is of great interest in canines due to the genetic proximity to humans described before [156, 157]. In 2015, [158] explored the feasibility of producing gene knockout (KO) dogs using gene editing by CRISPR/CAS9. The study focused to knock out the myostatin gene (MSTN), that is a negative regulator of skeletal muscle mass and demonstrated for the first time that a single injection of Cas9 mRNA and sgRNA corresponding to a particular gene into zygotes, combined with an embryo transfer strategy, efficiently generated site-specific genome-modified dogs [158, 159].

Recent studies also focused on using CRISPR/Cas9 edition for canine cancer models [160, 161]. Eun et al. [161], reported the attempt to optimize the CRISPR/Cas9 system to target canine tumor protein 53 (TP53), one of the most important tumor suppressor genes. The establishment of TP53 knockout canine cells could generate a useful platform to reveal novel oncogenic functions and effects of developing anti-cancer therapeutics [161].

Whereas one of the key benefits of using ciPSCs in disease modeling is the already discussed advantage over murine models, mostly due to the higher similarity between dogs to humans, another important perspective about ciPSCs is its potential use in clinical applications to improve the health and welfare of dogs themselves, an important aspect to be considered, in particular, due to the increasing inclusion of pets inside families and their overall importance to the One Health concept.

Advertisement

3. Future perspectives and final considerations

Herein advantages and hurdles of using of induced pluripotent stem cells were discussed concerning ongoing and future applications in large animal models, summarized in Figure 1. While true ESCs have only been described in mouse and rat models, it is widely accepted that these models are not the most adequate for studies on cellular therapies in regenerative medicine. Therefore, progress on translational medicine relies on the development of pluripotent-based technologies in suitable environments such as NH-primates, swine, or canine organisms. The alliance between in vitro induced pluripotency and gene editing tools opens a new road to suitable and experimental preclinical protocols. Besides the in vivo or in vitro disease modeling, the validation of pluripotency in domestic and wild animals holds great promise to contribute to animal production, preservation, and health by enabling, for example, the generation of gene editing and improved gametes, embryos, and animals.

Figure 1.

Biomedical and regenerative possibilities for translational use of induced pluripotent stem cells derived from large animal models.

Advertisement

Acknowledgments

Funding was provided by FAPESP (São Paulo Research Foundation) grants no. 2013/08135-2 and 2015/26818-5.

References

  1. 1. Ericsson AC, Crim MJ, Franklin CL. A brief history of animal modeling. Mo Med [Internet]. 110(3):201-5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23829102
  2. 2. ANDERSEN ML, WINTER LMF. Animal models in biological and biomedical research - experimental and ethical concerns. An Acad Bras Cienc [Internet]. 2019;91(suppl 1). Available from: http://www.scielo.br/scielo.php?script=sci_arttext&pid=S0001-37652019000200701&tlng=en
  3. 3. Chan AWS, Cheng P-H, Neumann A, Yang J-J. Reprogramming Huntington Monkey Skin Cells into Pluripotent Stem Cells. Cell Reprogram [Internet]. 2010 Oct;12(5):509-517 Available from: http://www.liebertpub.com/doi/10.1089/cell.2010.0019
  4. 4. Friedman H, Ator N, Haigwood N, Newsome W, Allen JS, Golos TG, et al. The Critical Role of Nonhuman Primates in Medical Research - White Paper. Pathog Immun [Internet]. 2017 Aug 21;2(3):352. Available from: http://paijournal.com/index.php/paijournal/article/view/186
  5. 5. Harding JD. Nonhuman Primates and Translational Research: Progress, Opportunities, and Challenges. ILAR J [Internet]. 2017 Dec 1;58(2):141-150 Available from: https://academic.oup.com/ilarjournal/article/58/2/141/4745719
  6. 6. Daadi MM, Barberi T, Shi Q , Lanford RE. Nonhuman Primate Models in Translational Regenerative Medicine. Stem Cells Dev [Internet]. 2014 Dec;23(S1):83-87 Available from: https://www.liebertpub.com/doi/10.1089/scd.2014.0374
  7. 7. Cox LA, Comuzzie AG, Havill LM, Karere GM, Spradling KD, Mahaney MC, et al. Baboons as a model to study genetics and epigenetics of human disease. ILAR J [Internet]. 2013;54(2):106-21. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24174436
  8. 8. Mora-Bermúdez F, Badsha F, Kanton S, Camp JG, Vernot B, Köhler K, et al. Differences and similarities between human and chimpanzee neural progenitors during cerebral cortex development. Elife [Internet]. 2016;5. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27669147
  9. 9. Camacho P, Fan H, Liu Z, He J-Q . Large Mammalian Animal Models of Heart Disease. J Cardiovasc Dev Dis [Internet]. 2016 Oct 5;3(4). Available from: http://www.ncbi.nlm.nih.gov/pubmed/29367573
  10. 10. Yang G, Hong H, Torres A, Malloy KE, Choudhury GR, Kim J, et al. Standards for Deriving Nonhuman Primate-Induced Pluripotent Stem Cells, Neural Stem Cells and Dopaminergic Lineage. Int J Mol Sci [Internet]. 2018 Sep 17;19(9). Available from: http://www.ncbi.nlm.nih.gov/pubmed/30227600
  11. 11. Dudley DM, Aliota MT, Mohr EL, Weiler AM, Lehrer-Brey G, Weisgrau KL, et al. A rhesus macaque model of Asian-lineage Zika virus infection. Nat Commun [Internet]. 2016 Nov 28;7(1):12204. Available from: http://www.nature.com/articles/ncomms12204
  12. 12. Seo J, Lee Y, Kim BS, Park J, Yang S, Yoon H-J, et al. A non-human primate model for stable chronic Parkinson’s disease induced by MPTP administration based on individual behavioral quantification. J Neurosci Methods [Internet]. 2019 Jan;311:277-87. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0165027018303510
  13. 13. Hessell AJ, Jaworski JP, Epson E, Matsuda K, Pandey S, Kahl C, et al. Early short-term treatment with neutralizing human monoclonal antibodies halts SHIV infection in infant macaques. Nat Med [Internet]. 2016 Apr 21;22(4):362-368 Available from: http://www.nature.com/articles/nm.4063
  14. 14. Völter CJ, Mundry R, Call J, Seed AM. Chimpanzees flexibly update working memory contents and show susceptibility to distraction in the self-ordered search task. Proc R Soc B Biol Sci [Internet]. 2019 Jul 24;286(1907):20190715. Available from: https://royalsocietypublishing.org/doi/10.1098/rspb.2019.0715
  15. 15. Li T, Ai Z, Ji W. Primate stem cells: bridge the translation from basic research to clinic application. Sci China Life Sci [Internet]. 2019 Jan 9;62(1):12-21 Available from: http://link.springer.com/10.1007/s11427-018-9334-2
  16. 16. Thomson JA, Kalishman J, Golos TG, Durning M, Harris CP, Becker RA, et al. Isolation of a primate embryonic stem cell line. Proc Natl Acad Sci [Internet]. 1995 Aug 15;92(17):7844-7848 Available from: http://www.pnas.org/cgi/doi/10.1073/pnas.92.17.7844
  17. 17. Branco E, Miranda CMFC, Lima AR, Silva KSM, Cabral RM, Miranda MS, et al. Bone marrow mononuclear cells versus mesenchymal stem cells from adipose tissue on bone healing in an Old World primate: can this be extrapolated to humans? Arq Bras Med Veterinária e Zootec [Internet]. 2019 Jun;71(3):917-928 Available from: http://www.scielo.br/scielo.php?script=sci_arttext&pid=S0102-09352019000300917&tlng=en
  18. 18. Pogozhykh O, Pogozhykh D, Neehus A-L, Hoffmann A, Blasczyk R, Müller T. Molecular and cellular characteristics of human and non-human primate multipotent stromal cells from the amnion and bone marrow during long term culture. Stem Cell Res Ther [Internet]. 2015 Dec 22;6(1):150. Available from: http://stemcellres.com/content/6/1/150
  19. 19. Devine SM, Bartholomew AM, Mahmud N, Nelson M, Patil S, Hardy W, et al. Mesenchymal stem cells are capable of homing to the bone marrow of non-human primates following systemic infusion. Exp Hematol [Internet]. 2001 Feb;29(2):244-255 Available from: https://linkinghub.elsevier.com/retrieve/pii/S0301472X00006354
  20. 20. Sasaki E, Hanazawa K, Kurita R, Akatsuka A, Yoshizaki T, Ishii H, et al. Establishment of Novel Embryonic Stem Cell Lines Derived from the Common Marmoset ( Callithrix jacchus ). Stem Cells [Internet]. 2005 Oct;23(9):1304-1313 Available from: http://doi.wiley.com/10.1634/stemcells.2004-0366
  21. 21. Suemori H, Tada T, Torii R, Hosoi Y, Kobayashi K, Imahie H, et al. Establishment of embryonic stem cell lines from cynomolgus monkey blastocysts produced by IVF or ICSI. Dev Dyn [Internet]. 2001 Oct;222(2):273-279. Available from: http://doi.wiley.com/10.1002/dvdy.1191
  22. 22. Shimozawa N, Nakamura S, Takahashi I, Hatori M, Sankai T. Characterization of a novel embryonic stem cell line from an ICSI-derived blastocyst in the African green monkey. REPRODUCTION [Internet]. 2010 Mar;139(3):565-573 Available from: https://rep.bioscientifica.com/view/journals/rep/139/3/565.xml
  23. 23. Simerly CR, Navara CS, Castro CA, Turpin JC, Redinger CJ, Mich-Basso JD, et al. Establishment and characterization of baboon embryonic stem cell lines: An Old World Primate model for regeneration and transplantation research. Stem Cell Res [Internet]. 2009 May;2(3):178-187 Available from: https://linkinghub.elsevier.com/retrieve/pii/S1873506109000221
  24. 24. Takahashi K, Yamanaka S. Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. 2006;2:663-76.
  25. 25. Grow DA, McCarrey JR, Navara CS. Advantages of nonhuman primates as preclinical models for evaluating stem cell-based therapies for Parkinson’s disease. Stem Cell Res [Internet]. 2016 Sep;17(2):352-366 Available from: https://linkinghub.elsevier.com/retrieve/pii/S187350611630112X
  26. 26. Pessôa LV de F, Bressan FF, Freude KK. Induced pluripotent stem cells throughout the animal kingdom: Availability and applications. World J Stem Cells [Internet]. 2019 Aug 26;11(8):491-505 Available from: https://www.wjgnet.com/1948-0210/full/v11/i8/491.htm
  27. 27. Liu H, Zhu F, Yong J, Zhang P, Hou P, Li H, et al. Correspondence Generation of Induced Pluripotent Stem Cells from Adult Rhesus Monkey Fibroblasts. Stem Cell [Internet]. 2008;3(6):587-590 Available from: http://dx.doi.org/10.1016/j.stem.2008.10.014
  28. 28. Ben-Nun IF, Montague SC, Houck ML, Tran HT, Garitaonandia I, Leonardo TR, et al. Induced pluripotent stem cells from highly endangered species. Nat Methods [Internet]. 2011 Sep 4;8(10):829-31. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21892153
  29. 29. Shimozawa N, Ono R, Shimada M, Shibata H, Takahashi I, Inada H, et al. Cynomolgus monkey induced pluripotent stem cells established by using exogenous genes derived from the same monkey species. Differentiation [Internet]. 85(4-5):131-9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23792767
  30. 30. Domingues S, Masson Y, Marteyn A, Allouche J, Perrier AL, Peschanski M, et al. Differentiation of nonhuman primate pluripotent stem cells into functional keratinocytes. Stem Cell Res Ther [Internet]. 2017 Dec 19;8(1):285. Available from: https://stemcellres.biomedcentral.com/articles/10.1186/s13287-017-0741-9
  31. 31. Tomioka I, Maeda T, Shimada H, Kawai K, Okada Y, Igarashi H, et al. Generating induced pluripotent stem cells from common marmoset (Callithrix jacchus) fetal liver cells using defined factors, including Lin28. Genes Cells [Internet]. 2010 Sep 1;15(9):959-69. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20670273
  32. 32. Navara CS, Hornecker J, Grow D, Chaudhari S, Hornsby PJ, Ichida JK, et al. Derivation of induced pluripotent stem cells from the baboon: a nonhuman primate model for preclinical testing of stem cell therapies. Cell Reprogram [Internet]. 2013 Dec;15(6):495-502. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24182315
  33. 33. Ramaswamy K, Yik WY, Wang X-M, Oliphant EN, Lu W, Shibata D, et al. Derivation of induced pluripotent stem cells from orangutan skin fibroblasts. BMC Res Notes [Internet]. 2015 Oct 16;8:577. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26475477
  34. 34. Nakai R, Ohnuki M, Kuroki K, Ito H, Hirai H, Kitajima R, et al. Derivation of induced pluripotent stem cells in Japanese macaque (Macaca fuscata). Sci Rep [Internet]. 2018 Dec 15;8(1):12187. Available from: http://www.nature.com/articles/s41598-018-30734-w
  35. 35. Thoma EC, Heckel T, Keller D, Giroud N, Leonard B, Christensen K, et al. Establishment of a translational endothelial cell model using directed differentiation of induced pluripotent stem cells from Cynomolgus monkey. Sci Rep [Internet]. 2016;6:35830. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27779219
  36. 36. Zhang X, Cao H, Bai S, Huo W, Ma Y. Differentiation and characterization of rhesus monkey atrial and ventricular cardiomyocytes from induced pluripotent stem cells. Stem Cell Res [Internet]. 2017 Apr;20:21-9. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1873506117300156
  37. 37. D’Souza SS, Maufort J, Kumar A, Zhang J, Smuga-Otto K, Thomson JA, et al. GSK3β Inhibition Promotes Efficient Myeloid and Lymphoid Hematopoiesis from Non-human Primate-Induced Pluripotent Stem Cells. Stem Cell Reports [Internet]. 2016 Feb;6(2):243-256 Available from: https://linkinghub.elsevier.com/retrieve/pii/S2213671115003756
  38. 38. Hallett PJ, Deleidi M, Astradsson A, Smith GA, Cooper O, Osborn TM, et al. Successful Function of Autologous iPSC-Derived Dopamine Neurons following Transplantation in a Non-Human Primate Model of Parkinson’s Disease. Cell Stem Cell [Internet]. 2015 Mar;16(3):269-274 Available from: https://linkinghub.elsevier.com/retrieve/pii/S1934590915000569
  39. 39. Aron Badin R, Bugi A, Williams S, Vadori M, Michael M, Jan C, et al. MHC matching fails to prevent long-term rejection of iPSC-derived neurons in non-human primates. Nat Commun [Internet]. 2019 Dec 25;10(1):4357. Available from: http://www.nature.com/articles/s41467-019-12324-0
  40. 40. Cho IK, Yang B, Forest C, Qian L, Chan AWS. Amelioration of Huntington’s disease phenotype in astrocytes derived from iPSC-derived neural progenitor cells of Huntington’s disease monkeys. PLoS One [Internet]. 2019;14(3):e0214156. Available from: http://www.ncbi.nlm.nih.gov/pubmed/30897183
  41. 41. Vermilyea SC, Babinski A, Tran N, To S, Guthrie S, Kluss JH, et al. In Vitro CRISPR/Cas9-Directed Gene Editing to Model LRRK2 G2019S Parkinson’s Disease in Common Marmosets. Sci Rep [Internet]. 2020 Feb 26;10(1):3447. Available from: http://www.ncbi.nlm.nih.gov/pubmed/32103062
  42. 42. Kawamura T, Miyagawa S, Fukushima S, Maeda A, Kashiyama N, Kawamura A, et al. Cardiomyocytes Derived from MHC-Homozygous Induced Pluripotent Stem Cells Exhibit Reduced Allogeneic Immunogenicity in MHC-Matched Non-human Primates. Stem Cell Reports [Internet]. 2016 Mar;6(3):312-320 Available from: https://linkinghub.elsevier.com/retrieve/pii/S2213671116000291
  43. 43. Zhao X, Chen H, Xiao D, Yang H, Itzhaki I, Qin X, et al. Comparison of Non-human Primate versus Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Treatment of Myocardial Infarction. Stem Cell Reports [Internet]. 2018 Feb;10(2):422-435 Available from: https://linkinghub.elsevier.com/retrieve/pii/S2213671118300286
  44. 44. Sosa E, Chen D, Rojas EJ, Hennebold JD, Peters KA, Wu Z, et al. Differentiation of primate primordial germ cell-like cells following transplantation into the adult gonadal niche. Nat Commun [Internet]. 2018 Dec 17;9(1):5339. Available from: http://www.nature.com/articles/s41467-018-07740-7
  45. 45. Olivier EN, Wang K, Grossman J, Mahmud N, Bouhassira EE. Differentiation of Baboon (Papio anubis) Induced-Pluripotent Stem Cells into Enucleated Red Blood Cells. Cells [Internet]. 2019 Oct 19;8(10):1282. Available from: https://www.mdpi.com/2073-4409/8/10/1282
  46. 46. McGill TJ, Stoddard J, Renner LM, Messaoudi I, Bharti K, Mitalipov S, et al. Allogeneic iPSC-Derived RPE Cell Graft Failure Following Transplantation Into the Subretinal Space in Nonhuman Primates. Investig Opthalmology Vis Sci [Internet]. 2018 Mar 8;59(3):1374. Available from: http://iovs.arvojournals.org/article.aspx?doi=10.1167/iovs.17-22467
  47. 47. Shiba Y, Gomibuchi T, Seto T, Wada Y, Ichimura H, Tanaka Y, et al. Allogeneic transplantation of iPS cell-derived cardiomyocytes regenerates primate hearts. Nature [Internet]. 2016 Oct 10;538(7625):388-391 Available from: http://www.nature.com/articles/nature19815
  48. 48. Kunkanjanawan T, Carter R, Ahn K-S, Yang J, Parnpai R, Chan AWS. Induced Pluripotent HD Monkey Stem Cells Derived Neural Cells for Drug Discovery. SLAS Discov Adv Sci Drug Discov [Internet]. 2017 Jul 27;22(6):696-705 Available from: http://journals.sagepub.com/doi/10.1177/2472555216685044
  49. 49. Goodnight A V., Kremsky I, Khampang S, Jung YH, Billingsley JM, Bosinger SE, et al. Chromatin accessibility and transcription dynamics during in vitro astrocyte differentiation of Huntington’s Disease Monkey pluripotent stem cells. Epigenetics Chromatin [Internet]. 2019 Dec 13;12(1):67. Available from: https://epigeneticsandchromatin.biomedcentral.com/articles/10.1186/s13072-019-0313-6
  50. 50. Vermilyea SC, Guthrie S, Meyer M, Smuga-Otto K, Braun K, Howden S, et al. Induced Pluripotent Stem Cell-Derived Dopaminergic Neurons from Adult Common Marmoset Fibroblasts. Stem Cells Dev [Internet]. 2017 Sep;26(17):1225-1235 Available from: https://www.liebertpub.com/doi/10.1089/scd.2017.0069
  51. 51. Emborg ME, Liu Y, Xi J, Zhang X, Yin Y, Lu J, et al. Induced Pluripotent Stem Cell-Derived Neural Cells Survive and Mature in the Nonhuman Primate Brain. Cell Rep [Internet]. 2013 Mar;3(3):646-650 Available from: https://linkinghub.elsevier.com/retrieve/pii/S2211124713000752
  52. 52. Hong H, Roy-Choudhury G, Kim J, Daadi MM. Isolation and Differentiation of Self-Renewable Neural Stem Cells from Marmoset-Induced Pluripotent Stem Cells. In 2019. p. 199-204. Available from: http://link.springer.com/10.1007/978-1-4939-9007-8_15
  53. 53. Yang L, Esvelt KM, Aach J, Guell M, Dicarlo JE, Norville JE, et al. RNA-Guided Human Genome. 2013;(February):823-7.
  54. 54. Wu M, Wei C, Lian Z, Liu R, Zhu C, Wang H, et al. Rosa26-targeted sheep gene knock-in via CRISPR-Cas9 system. Sci Rep. 2016;6(September 2015):1-7
  55. 55. Cho SW, Kim S, Kim Y, Kweon J, Kim HS, Bae S, et al. Sup2. Cold Spring Harb Lab Press Method. 2014;132-41.
  56. 56. Fu Y, Sander JD, Reyon D, Cascio VM, Joung JK. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat Biotechnol. 2014;32(3):279-284
  57. 57. Niu Y, Li T, Ji W. Paving the road for biomedicine: genome editing and stem cells in primates. Natl Sci Rev [Internet]. 2017 Jul 1;4(4):543-549 Available from: https://academic.oup.com/nsr/article/4/4/543/4093906
  58. 58. Chen Y, Niu Y, Ji W. Genome editing in nonhuman primates: approach to generating human disease models. J Intern Med [Internet]. 2016 Sep;280(3):246-251 Available from: http://doi.wiley.com/10.1111/joim.12469
  59. 59. Yang S-H, Cheng P-H, Banta H, Piotrowska-Nitsche K, Yang J-J, Cheng ECH, et al. Towards a transgenic model of Huntington’s disease in a non-human primate. Nature [Internet]. 2008 Jun 18;453(7197):921-924 Available from: http://www.nature.com/articles/nature06975
  60. 60. Niu Y, Guo X, Chen Y, Wang C-E, Gao J, Yang W, et al. Early Parkinson’s disease symptoms in -synuclein transgenic monkeys. Hum Mol Genet [Internet]. 2015 Apr 15;24(8):2308-2317 Available from: https://academic.oup.com/hmg/article-lookup/doi/10.1093/hmg/ddu748
  61. 61. Liu H, Chen Y, Niu Y, Zhang K, Kang Y, Ge W, et al. TALEN-Mediated Gene Mutagenesis in Rhesus and Cynomolgus Monkeys. Cell Stem Cell [Internet]. 2014 Mar;14(3):323-328 Available from: https://linkinghub.elsevier.com/retrieve/pii/S1934590914000484
  62. 62. Chen Y, Zheng Y, Kang Y, Yang W, Niu Y, Guo X, et al. Functional disruption of the dystrophin gene in rhesus monkey using CRISPR/Cas9. Hum Mol Genet [Internet]. 2015 Jul 1;24(13):3764-3774 Available from: https://academic.oup.com/hmg/article-lookup/doi/10.1093/hmg/ddv120
  63. 63. Sato K, Oiwa R, Kumita W, Henry R, Sakuma T, Ito R, et al. Generation of a Nonhuman Primate Model of Severe Combined Immunodeficiency Using Highly Efficient Genome Editing. Cell Stem Cell [Internet]. 2016 Jul;19(1):127-138 Available from: https://linkinghub.elsevier.com/retrieve/pii/S1934590916301539
  64. 64. EU (2010) Directive 2010/63/EU of the European Parliament and of the Council of 22 September 2010 on the protection of animals used for scientific purposes (text with EEA relevance). OJ L 276/33.
  65. 65. Carvalho C, Gaspar A, Knight A, Vicente L. Ethical and Scientific Pitfalls Concerning Laboratory Research with Non-Human Primates, and Possible Solutions. Animals [Internet]. 2018 Dec 29;9(1):12. Available from: http://www.mdpi.com/2076-2615/9/1/12
  66. 66. Perleberg C, Kind A, Schnieke A. Genetically engineered pigs as models for human disease. DMM Dis Model Mech. 2018;11(1).
  67. 67. Roberts RM, Telugu BPVL, Ezashi T. Induced pluripotent stem cells from swine (Sus scrofa): Why they may prove to be important. Cell Cycle. 2009;8(19):3078-3081
  68. 68. Michael Roberts R, Yuan Y, Genovese N, Ezashi T. Livestock models for exploiting the promise of pluripotent stem cells. ILAR J. 2015;56(1):74-82
  69. 69. Groenen MAM, Archibald AL, Uenishi H, Tuggle CK, Takeuchi Y, Rothschild MF, et al. Europe PMC Funders Group Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts Analyses of pig genomes provide insight into porcine demography and evolution. 2012;491(7424):393-8.
  70. 70. Schmied J, Rupa P, Garvie S, Wilkie B. Immune response phenotype of allergic versus clinically tolerant pigs in a neonatal swine model of allergy. Vet Immunol Immunopathol. 2013;154(1-2):17-24
  71. 71. Thomas DJ, Husmann RJ, Villamar M, Winship TR, Buck RH, Zuckermann FA. Lactobacillus rhamnosus HN001 attenuates allergy development in a pig model. PLoS One. 2011;6(2).
  72. 72. Turner DJ, Noble PB, Lucas MP, Mitchell HW. Decreased airway narrowing and smooth muscle contraction in hyperresponsive pigs. J Appl Physiol. 2002;93(4):1296-300.
  73. 73. Brandler MD, Powell SC, Craig DM, Quick G, McMahon TJ, Goldberg RN, et al. A novel inhaled organic nitrate that affects pulmonary vascular tone in a piglet model of hypoxia-induced pulmonary hypertension. Pediatr Res. 2005;58(3):531-536
  74. 74. Davis SS, Illum L, Hinchcliffe M. Gastrointestinal transit of dosage forms in the pig. J Pharm Pharmacol. 2001;53(1):33-39
  75. 75. Bassaganya-Riera J, Hontecillas R. CLA and n-3 PUFA differentially modulate clinical activity and colonic PPAR-responsive gene expression in a pig model of experimental IBD. Clin Nutr. 2006;25(3):454-465
  76. 76. Nocca D, Gagner M, Abente FC, Del Genio GM, Ueda K, Assalia A, et al. Laparoscopic gastric bypass with silicone band in a pig model: Prevention of anastomotic dilatation - Feasibility study. Obes Surg. 2005;15(4):523-527
  77. 77. Badin JK, Progar V, Pareddy A, Cagle J, Alloosh M, Sturek M. Effect of Age on Diabetogenicity of Alloxan in Ossabaw Miniature Swine. Comp Med. 2019;69(2):114-122
  78. 78. van den Heuvel M, Sorop O, van Ditzhuijzen NS, de Vries R, van Duin RWB, Peters I, et al. The effect of bioresorbable vascular scaffold implantation on distal coronary endothelial function in dyslipidemic swine with and without diabetes. Int J Cardiol. 2018;252:44-51
  79. 79. Torres-Rovira L, Astiz S, Caro A, Lopez-Bote C, Ovilo C, Pallares P, et al. Diet-induced swine model with obesity/leptin resistance for the study of metabolic syndrome and type 2 diabetes. Sci World J. 2012;2012
  80. 80. Dolezalova D, Hruska-Plochan M, Bjarkam CR, Sørensen JCH, Cunningham M, Weingarten D, et al. Pig models of neurodegenerative disorders: Utilization in cell replacement-based preclinical safety and efficacy studies. J Comp Neurol. 2014;522(12):2784-2801
  81. 81. Lorson MA, Spate LD, Samuel MS, Murphy CN, Lorson CL, Prather RS, et al. Disruption of the Survival Motor Neuron (SMN) gene in pigs using ssDNA. Transgenic Res. 2011;20(6):1293-1304
  82. 82. Yan S, Tu Z, Liu Z, Fan N, Yang H, Yang S, et al. A huntingtin knock-in pig model recapitulates features of selective neurodegeneration in Huntington’s disease. 2019;173(4):989-1002.
  83. 83. Cai M, Shen R, Song L, Lu M, Wang J, Zhao S, et al. Bone Marrow Mesenchymal Stem Cells (BM-MSCs) Improve Heart Function in Swine Myocardial Infarction Model through Paracrine Effects. Sci Rep. 2016;6(January):1-11.
  84. 84. Omlor GW, Lorenz S, Nerlich AG, Guehring T, Richter W. Disc cell therapy with bone-marrow-derived autologous mesenchymal stromal cells in a large porcine disc degeneration model. Eur Spine J. 2018;27(10):2639-2649
  85. 85. Nichols J, Smith A. Naive and Primed Pluripotent States. Cell Stem Cell. 2009;4(6):487-492
  86. 86. Hanna J, Saha K, Jaenisch R. Somatic cell reprogramming and transitions between pluripotent states: facts, hypotheses, unresolved issues. Cell. 2010;143(4):508-525
  87. 87. Davidson KC, Mason EA, Pera MF. The pluripotent state in mouse and human. Development [Internet]. 2015 Sep 15;142(18):3090-3099 Available from: http://dev.biologists.org/cgi/doi/10.1242/dev.116061
  88. 88. Kumari D. States of Pluripotency: Naïve and Primed Pluripotent Stem Cells. In: Pluripotent Stem Cells - From the Bench to the Clinic [Internet]. InTech; 2016. Available from: http://www.intechopen.com/books/pluripotent-stem-cells-from-the-bench-to-the-clinic/states-of-pluripotency-na-ve-and-primed-pluripotent-stem-cells
  89. 89. Nowak-Imialek M, Niemann H. Pluripotent cells in farm animals: state of the art and future perspectives. Reprod Fertil Dev [Internet]. 2013;25(1):103. Available from: http://www.publish.csiro.au/?paper=RD12265
  90. 90. Du X, Feng T, Yu D, Wu Y, Zou H, Ma S, et al. Barriers for Deriving Transgene-Free Pig iPS Cells with Episomal Vectors. Stem Cells. 2015 Nov;33(11):3228-3238
  91. 91. Yu J, Hu K, Smuga-otto K, Tian S, Stewart R, Igor I, et al. Human Induced Pluripotent Stem Cell Free of Vector Transgene Sequences. Science (80- ). 2009;324(5928):797-801.
  92. 92. Okita K, Matsumura Y, Sato Y, Okada A, Morizane A, Okamoto S, et al. A more efficient method to generate integration-free human iPS cells. Nat Methods. 2011;8(5):409-412
  93. 93. Li D, Secher J, Hyttel P, Ivask M, Kolko M, Hall VJ, et al. Generation of transgene-free porcine intermediate type induced pluripotent stem cells. Cell Cycle. 2018;17(23):2547-2563
  94. 94. Montserrat N, Bahima EG, Batlle L, Häfner S, Rodrigues AMC, González F, et al. Generation of pig iPS cells: A model for cell therapy. J Cardiovasc Transl Res. 2011;4(2):121-130
  95. 95. Ao Y, Mich-Basso JD, Lin B, Yang L. High efficient differentiation of functional hepatocytes from porcine induced pluripotent stem cells. PLoS One. 2014;9(6):1-11
  96. 96. Kim E, Kim M, Hwang SU, Kim J, Lee G, Park YS, et al. Neural induction of porcine-induced pluripotent stem cells and further differentiation using glioblastoma-cultured medium. J Cell Mol Med. 2019;23(3):2052-2063
  97. 97. Larsen NJ, Marklund S, Kelly KA, Malek M, Tuggle CK, Yerle M, et al. New insights into porcine-human synteny conservation. Mamm Genome. 1999;10(5):488-491
  98. 98. Jiang Z, He H, Hamasima N, Suzuki H, Verrinder Gibbins AM. Comparative mapping of Homo sapiens chromosome 4 (HSA4) and Sus scrofa chromosome 8 (SSC8) using orthologous genes representing different cytogenetic bands as landmarks. Genome. 2002;45(1):147-156
  99. 99. Lahbib-Mansais Y, Karlskov-Mortensen P, Mompart F, Milan D, Jørgensen CB, Cirera S, et al. A high-resolution comparative map between pig chromosome 17 and human chromosomes 4, 8, and 20: Identification of synteny breakpoints. Genomics. 2005;86(4):405-413
  100. 100. Cheng LT, Sun LT, Tada T. Genome editing in induced pluripotent stem cells. Genes to Cells. 2012;17(6):431-438
  101. 101. Kikuchi K, Kashiwazaki N, Nagai T, Nakai M, Somfai T, Noguchi J, et al. Selected Aspects of Advanced Porcine Reproductive Technology. Reprod Domest Anim [Internet]. 2008 Jul;43:401-6. Available from: http://doi.wiley.com/10.1111/j.1439-0531.2008.01191.x
  102. 102. Hai T, Teng F, Guo R, Li W, Zhou Q . One-step generation of knockout pigs by zygote injection of CRISPR/Cas system. Cell Res. 2014;24(3):372-375
  103. 103. Whitworth KM, Lee K, Benne JA, Beaton BP, Spate LD, Murphy SL, et al. Use of the CRISPR/Cas9 System to Produce Genetically Engineered Pigs from In Vitro-Derived Oocytes and Embryos1. Biol Reprod. 2014;91(3):1-13
  104. 104. Yu HH, Zhao H, Qing YB, Pan WR, Jia BY, Zhao HY, et al. Porcine zygote injection with Cas9/sgRNAResults in DMD-modified pig with muscle dystrophy. Int J Mol Sci. 2016;17(10).
  105. 105. Zhou X, Wang L, Du Y, Xie F, Li L, Liu Y, et al. Efficient Generation of Gene-Modified Pigs Harboring Precise Orthologous Human Mutation via CRISPR/Cas9-Induced Homology-Directed Repair in Zygotes. Hum Mutat. 2016;37(1):110-118
  106. 106. Li H, Yang Y, Hong W, Huang M, Wu M, Zhao X. Applications of genome editing technology in the targeted therapy of human diseases: mechanisms, advances and prospects. Signal Transduct Target Ther. 2020;5(1).
  107. 107. Hryhorowicz M, Zeyland J, Słomski R, Lipiński D. Genetically Modified Pigs as Organ Donors for Xenotransplantation. Mol Biotechnol. 2017;59(9-10):435-444
  108. 108. Wu J, Platero-Luengo A, Sakurai M, Sugawara A, Gil MA, Yamauchi T, et al. Interspecies Chimerism with Mammalian Pluripotent Stem Cells. Cell. 2017;168(3):473-486.e15.
  109. 109. Wu J, Vilarino M, Suzuki K, Okamura D, Bogliotti YS, Park I, et al. CRISPR-Cas9 mediated one-step disabling of pancreatogenesis in pigs. Sci Rep [Internet]. 2017 Dec 5;7(1):10487. Available from: http://www.nature.com/articles/s41598-017-08596-5
  110. 110. Goldfracht I, Efraim Y, Shinnawi R, Kovalev E, Huber I, Gepstein A, et al. Engineered heart tissue models from hiPSC-derived cardiomyocytes and cardiac ECM for disease modeling and drug testing applications. Acta Biomater. 2019;92:145-159
  111. 111. Ohata K, Ott HC. Human-scale lung regeneration based on decellularized matrix scaffolds as a biologic platform. Surg Today. 2020;50(7):633-643
  112. 112. Alshaikh AB, Padma AM, Dehlin M, Akouri R, Song MJ, Brännström M, et al. Decellularization and recellularization of the ovary for bioengineering applications; studies in the mouse. Reprod Biol Endocrinol [Internet]. 2020 Dec 23;18(1):75. Available from: https://rbej.biomedcentral.com/articles/10.1186/s12958-020-00630-y
  113. 113. Bhat ZF, Kumar S, Bhat HF. In vitro meat: A future animal-free harvest. Crit Rev Food Sci Nutr. 2017;57(4):782-789
  114. 114. Ostrander EA. Wayne RK. Genome Research: The canine genome; 2005
  115. 115. Tsai KL, Clark LA, Murphy KE. Understanding hereditary diseases using the dog and human as companion model systems. Mammalian Genome. 2007.
  116. 116. Kirk AD. Crossing the bridge: Large animal models in translational transplantation research. Immunological Reviews. 2003
  117. 117. Horn PA, Morris JC, Neff T, Kiem HP. Stem cell gene transfer - Efficacy and safety in large animal studies. Molecular Therapy. 2004.
  118. 118. Schneider MR, Wolf E, Braun J, Kolb HJ, Adler H. Canine embryonic stem cells: State of the art. Theriogenology. 2010;74(4):492-497
  119. 119. Lucroy MD, Suckow MA. Predictive modeling for cancer drug discovery using canine models. Expert Opin Drug Discov. 2020;15(6):731-738
  120. 120. Paoloni M. Khanna C. Nature Reviews Cancer: Translation of new cancer treatments from pet dogs to humans; 2008
  121. 121. Rivera P, von Euler H. Molecular biological aspects on canine and human mammary tumors. Vet Pathol. 2011
  122. 122. Bongiovanni L, Brachelente C, Moreno E, Welle MM. Canine epithelial skin tumours: Expression of the stem cell markers Lgr5, Lgr6 and Sox9 in light of new cancer stem cell theories. Vet Sci. 2020
  123. 123. Liu W, Sender S, Kong W, Beck J, Sekora A, Bornemann-Kolatzki K, et al. Establishment and characterization of stable red, far-red (fR) and near infra-red (NIR) transfected canine prostate cancer cell lines. Cancer Cell Int. 2020
  124. 124. Villarnovo D, McCleary-Wheeler AL, Richards KL. Barking up the right tree: Advancing our understanding and treatment of lymphoma with a spontaneous canine model. Current Opinion in Hematology. 2017
  125. 125. Mochizuki H, Kennedy K, Shapiro SG, Breen MB. BRAF mutations in canine cancers. PLoS One. 2015
  126. 126. Fenger JM, London CA, Kisseberth WC. Canine osteosarcoma: A naturally occurring disease to inform pediatric oncology. ILAR J. 2014
  127. 127. Ryu HH, Kang BJ, Park SS, Kim Y, Sung GJ, Woo HM, et al. Comparison of mesenchymal stem cells derived from fat, bone marrow, Wharton’s jelly, and umbilical cord blood for treating spinal cord injuries in dogs. Journal of Veterinary Medical Science. 2012.
  128. 128. Lim CY, Han JI, Kim SG, Lee CM, Park HM. Evaluation of autologous bone marrow-derived mesenchymal stem cells on renal regeneration after experimentally induced acute kidney injury in dogs. Am J Vet Res. 2016;
  129. 129. Hang D, Li F, Che W, Wu X, Wan Y, Wang J, et al. One-Stage Positron Emission Tomography and Magnetic Resonance Imaging to Assess Mesenchymal Stem Cell Survival in a Canine Model of Intervertebral Disc Degeneration. Stem Cells Dev. 2017;
  130. 130. Vela DC, Silva GV, Assad JAR, Sousa ALS, Coulter S, Fernandes MR, et al. Histopathological study of healing after allogenic mesenchymal stem cell delivery in myocardial infarction in dogs. J Histochem Cytochem. 2009
  131. 131. Reich CM, Raabe O, Wenisch S, Bridger PS, Kramer M, Arnhold S. Isolation, culture and chondrogenic differentiation of canine adipose tissue- and bone marrow-derived mesenchymal stem cells-A comparative study. Vet Res Commun. 2012;
  132. 132. Muir P, Hans EC, Racette M, Volstad N, Sample SJ, Heaton C, et al. Autologous bone marrow-derived mesenchymal stem cells modulate molecular markers of inflammation in dogs with cruciate ligament rupture. PLoS One. 2016;
  133. 133. Betts DH, Tobias IC. Canine pluripotent stem cells: Are they ready for clinical applications? Frontiers in Veterinary Science. 2015.
  134. 134. Hatoya S, Torii R, Kondo Y, Okuno T, Kobayashi K, Wijewardana V, et al. Isolation and characterization of embryonic stem-like cells from canine blastocysts. Mol Reprod Dev. 2006;
  135. 135. Schneider MR, Wolf E, Braun J, Kolb HJ, Adler H. Canine embryo-derived stem cells and models for human diseases. Hum Mol Genet. 2008;
  136. 136. Hayes B, Fagerlie SR, Ramakrishnan A, Baran S, Harkey M, Graf L, et al. Derivation, characterization, and in vitro differentiation of canine embryonic stem cells. Stem Cells. 2008;26(2):465-473
  137. 137. Vaags AK, Rosic-Kablar S, Gartley CJ, Zheng YZ, Chesney A, Villagómez DAF, et al. Derivation and Characterization of Canine Embryonic Stem Cell Lines with In Vitro and In Vivo Differentiation Potential. Stem Cells. 2009;
  138. 138. Wilcox JT, Semple E, Gartley C, Brisson B a, Perrault SD, Villagómez D a F, et al. Characterization of canine embryonic stem cell lines derived from different niche microenvironments. Stem Cells Dev. 2009 Oct;18(8):1167-78.
  139. 139. Wilcox JT, Lai JKY, Semple E, Brisson BA, Gartley C, Armstrong JN, et al. Synaptically-competent neurons derived from canine embryonic stem cells by lineage selection with EGF and noggin. PLoS One. 2011;
  140. 140. Luo J, Cibelli JB. Conserved Role of bFGF and a Divergent Role of LIF for Pluripotency Maintenance and Survival in Canine Pluripotent Stem Cells. Stem Cells Dev. 2016;25(21):1670-1680
  141. 141. Baird A, Barsby T, Guest D. Derivation of Canine Induced Pluripotent Stem Cells. Reprod Domest Anim [Internet]. 2015 Aug;50(4):669-676 Available from: http://doi.wiley.com/10.1111/rda.12562
  142. 142. Lee AS, Xu D, Plews JR, Nguyen PK, Nag D, Lyons JK, et al. Preclinical Derivation and Imaging of Autologously Transplanted Canine Induced Pluripotent Stem Cells. J Biol Chem [Internet]. 2011 Sep 16;286(37):32697-32704 Available from: http://www.jbc.org/lookup/doi/10.1074/jbc.M111.235739
  143. 143. Gonçalves NJN, Bressan FF, Roballo KCS, Meirelles FV, Xavier PLP, Fukumasu H, et al. Generation of LIF-independent induced pluripotent stem cells from canine fetal fibroblasts. Theriogenology [Internet]. 2017 Apr;92:75-82. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0093691X17300249
  144. 144. Koh S, Thomas R, Tsai S, Bischoff S, Lim J-H, Breen M, et al. Growth Requirements and Chromosomal Instability of Induced Pluripotent Stem Cells Generated from Adult Canine Fibroblasts. Stem Cells Dev [Internet]. 2013 Mar 15;22(6):951-963 Available from: https://www.liebertpub.com/doi/10.1089/scd.2012.0393
  145. 145. Shimada H, Nakada A, Hashimoto Y, Shigeno K, Shionoya Y, Nakamura T. Generation of canine induced pluripotent stem cells by retroviral transduction and chemical inhibitors. Mol Reprod Dev [Internet]. 2009 Nov 4;77(1):2-2 Available from: http://doi.wiley.com/10.1002/mrd.21117
  146. 146. Luo J, Suhr ST, Chang EA, Wang K, Ross PJ, Nelson LL, et al. Generation of leukemia inhibitory factor and basic fibroblast growth factor-dependent induced pluripotent stem cells from canine adult somatic cells. Stem Cells Dev. 2011;
  147. 147. Whitworth DJ, Ovchinnikov D. a, Wolvetang EJ. Generation and characterization of LIF-dependent canine induced pluripotent stem cells from adult dermal fibroblasts. Stem Cells Dev. 2012 Aug;21(12):2288-2297
  148. 148. Nishimura T, Hatoya S, Kanegi R, Sugiura K, Wijewardana V, Kuwamura M, et al. Generation of functional platelets from canine induced pluripotent stem cells. Stem Cells Dev. 2013;
  149. 149. Shimada H, Nakada A, Hashimoto Y, Shigeno K, Shionoya Y, Nakamura T. Generation of canine-induced pluripotent stem cells by retroviral transduction and chemical inhibitors. Molecular Reproduction and Development. 2010.
  150. 150. Lindblad-Toh K, Wade CM, Mikkelsen TS, Karlsson EK, Jaffe DB, Kamal M, et al. Genome sequence, comparative analysis and haplotype structure of the domestic dog. Nature. 2005;
  151. 151. Studzinski CM, Araujo JA, Milgram NW. The canine model of human cognitive aging and dementia: Pharmacological validity of the model for assessment of human cognitive-enhancing drugs. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2005
  152. 152. Cotman CW, Head E. The canine (dog) model of human aging and disease: Dietary, environmental and immunotherapy approaches. J Alzheimer’s Dis. 2008;15(4):685-707
  153. 153. Azkona G, García-Belenguer S, Chacón G, Rosado B, León M, Palacio J. Prevalence and risk factors of behavioural changes associated with age-related cognitive impairment in geriatric dogs: PAPER. J Small Anim Pract. 2009
  154. 154. Salvin HE, McGreevy PD, Sachdev PS, Valenzuela MJ. Under diagnosis of canine cognitive dysfunction: A cross-sectional survey of older companion dogs. Vet J. 2010
  155. 155. Hyttel P, Pessôa LV de F, Secher JBM, Dittlau KS, Freude K, Hall VJ, et al. Oocytes, embryos and pluripotent stem cells from a biomedical perspective. Anim Reprod. 2019;16(3):508-23.
  156. 156. Cebrian-Serrano A, Stout T, Dinnyes A. Veterinary applications of induced pluripotent stem cells: Regenerative medicine and models for disease? Veterinary Journal. 2013.
  157. 157. Ebert AD, Liang P, Wu JC. Induced pluripotent stem cells as a disease modeling and drug screening platform. Journal of Cardiovascular Pharmacology. 2012.
  158. 158. Zou Q , Wang X, Liu Y, Ouyang Z, Long H, Wei S, et al. Generation of gene-target dogs using CRISPR / Cas 9 system. 2015;7:580-3.
  159. 159. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-β superfamily member. Nature. 1997;
  160. 160. Grüntzig K, Graf R, Boo G, Guscetti F, Hässig M, Axhausen KW, et al. Swiss Canine Cancer Registry 1955-2008: Occurrence of the Most Common Tumour Diagnoses and Influence of Age, Breed, Body Size, Sex and Neutering Status on Tumour Development. J Comp Pathol. 2016;
  161. 161. Eun K, Park MG, Jeong YW, Jeong YI, Hyun S-H, Hwang WS, et al. Establishment of TP53-knockout canine cells using optimized CRIPSR/Cas9 vector system for canine cancer research. BMC Biotechnol. 2019;19(1):1-11

Written By

Laís Vicari de Figueiredo Pessôa, Naira Caroline Godoy Pieri, Kaiana Recchia and Fabiana Fernandes Bressan

Submitted: 22 June 2020 Reviewed: 25 September 2020 Published: 27 October 2020