Opening the “Black Box” Underlying Barriers to the Use of Canine Induced Pluripotent Stem Cells: A Narrative Review

Induced pluripotent stem cells (iPSCs) are produced by resetting the epigenetic and transcriptional landscapes of somatic cells to express the endogenous pluripotency network and revert them back to an undifferentiated state. The reduced ethical concerns associated with iPSCs and their capacity for extensive self-renewal and differentiation make them an unparalleled resource for drug discovery, disease modeling, and novel therapies. Canines (c) share many human diseases and environmental exposures, making them a superior translational model for drug screening and investigating human pathologies compared to other mammals. However, well-defined protocols for legitimate ciPSC production are lacking. Problems during canine somatic cell reprogramming (SCR) yield putative ciPSCs with incomplete pluripotency, at very low efficiencies. Despite the value of ciPSCs, the molecular mechanisms underlying their unsuccessful production and how these may be addressed have not been fully elucidated. Factors, including cost, safety, and feasibility, may also limit the widespread clinical adoption of ciPSCs for treating canine disease. The purpose of this narrative review is to identify barriers to canine SCR on molecular and cellular levels, using comparative research to inform potential solutions to their use in both research and clinical contexts. Current research is opening new doors for the application of ciPSCs in regenerative medicine for the mutual benefit of veterinary and human medicine.

[1]  F. Bressan,et al.  Neural Derivates of Canine Induced Pluripotent Stem Cells-Like Cells From a Mild Cognitive Impairment Dog , 2021, Frontiers in Veterinary Science.

[2]  S. Rodríguez-Fernández,et al.  Tips and tricks for successfully culturing and adapting human induced pluripotent stem cells , 2021, Molecular therapy. Methods & clinical development.

[3]  T. Dutt,et al.  Thin films of functionalized carbon nanotubes support long-term maintenance and cardio-neuronal differentiation of canine induced pluripotent stem cells. , 2021, Nanomedicine : nanotechnology, biology, and medicine.

[4]  Wei Huang,et al.  CRISPR activation of endogenous genes reprograms fibroblasts into cardiovascular progenitor cells for myocardial infarction therapy , 2021, Molecular therapy : the journal of the American Society of Gene Therapy.

[5]  Thomas K. Sears,et al.  Panobinostat Effectively Increases Histone Acetylation and Alters Chromatin Accessibility Landscape in Canine Embryonic Fibroblasts but Does Not Enhance Cellular Reprogramming , 2021, Frontiers in Veterinary Science.

[6]  Bokyung Kim,et al.  L-myc Gene Expression in Canine Fetal Fibroblasts Promotes Self-Renewal Capacity but Not Tumor Formation , 2021, Cells.

[7]  Yu Xin Wang,et al.  AP-1 is a temporally regulated dual gatekeeper of reprogramming to pluripotency , 2021, Proceedings of the National Academy of Sciences.

[8]  Kuldeep Kumar,et al.  In vitro propagation and cardiac differentiation of canine induced pluripotent stem cells on carbon nanotube substrates. , 2021, Tissue & cell.

[9]  D. Bottai,et al.  iPSC Preparation and Epigenetic Memory: Does the Tissue Origin Matter? , 2021, Cells.

[10]  A. Nagy,et al.  Human stem cells harboring a suicide gene improve the safety and standardisation of neural transplants in Parkinsonian rats , 2021, Nature Communications.

[11]  Hyun-Jai Cho,et al.  HLA DR Genome Editing with TALENs in Human iPSCs Produced Immune-Tolerant Dendritic Cells , 2021, Stem cells international.

[12]  K. Sugiura,et al.  Canine induced pluripotent stem cell maintenance under feeder‐free and chemically‐defined conditions , 2021, Molecular reproduction and development.

[13]  H. Okano,et al.  Non-viral derivation of a transgene-free induced pluripotent stem cell line from a male beagle dog. , 2021, Stem cell research.

[14]  H. Okano,et al.  Establishment of an induced pluripotent stem cell line from a female domestic ferret (Mustela putorius furo) with an X chromosome instability. , 2021, Stem cell research.

[15]  Hitoshi Takizawa,et al.  Fit-For-All iPSC-Derived Cell Therapies and Their Evaluation in Humanized Mice With NK Cell Immunity , 2021, Frontiers in Immunology.

[16]  H. Okano,et al.  Non-viral Induction of Transgene-free iPSCs from Somatic Fibroblasts of Multiple Mammalian Species , 2021, Stem cell reports.

[17]  J. Jelinek,et al.  DNA methylation landscape of 16 canine somatic tissues by methylation-sensitive restriction enzyme-based next generation sequencing , 2021, Scientific Reports.

[18]  S. Dow,et al.  Optimizing in vitro osteogenesis in canine autologous and induced pluripotent stem cell-derived mesenchymal stromal cells with dexamethasone and BMP-2. , 2020, Stem cells and development.

[19]  D. Song,et al.  Structurally-discovered KLF4 variants accelerate and stabilize reprogramming to pluripotency , 2020, iScience.

[20]  D. Fairlie,et al.  Taking the Myc out of cancer: toward therapeutic strategies to directly inhibit c-Myc , 2020, Molecular Cancer.

[21]  D. Betts,et al.  The use of induced pluripotent stem cells in domestic animals: a narrative review , 2020, BMC Veterinary Research.

[22]  K. Sugiura,et al.  Efficient Reprogramming of Canine Peripheral Blood Mononuclear Cells into Induced Pluripotent Stem Cells. , 2020, Stem cells and development.

[23]  A. Rosser,et al.  Induced pluripotent stem cells derived from the developing striatum as a potential donor source for cell replacement therapy for Huntington disease , 2020, Cytotherapy.

[24]  M. Settles,et al.  Chromatin accessibility in canine stromal cells and its implications for canine somatic cell reprogramming , 2020, Stem cells translational medicine.

[25]  S. Dow,et al.  Generation of Neural Progenitor Cells From Canine Induced Pluripotent Stem Cells and Preliminary Safety Test in Dogs With Spontaneous Spinal Cord Injuries , 2020, Frontiers in Veterinary Science.

[26]  C. Joshi,et al.  Positioning canine induced pluripotent stem cells (iPSCs) in the reprogramming landscape of naïve or primed state in comparison to mouse and human iPSCs. , 2020, Life sciences.

[27]  H. Taniguchi,et al.  Investigation of Clinical Safety of Human iPS Cell-Derived Liver Organoid Transplantation to Infantile Patients in Porcine Model , 2020, Cell transplantation.

[28]  S. Hyun,et al.  Optimized Approaches for the Induction of Putative Canine Induced Pluripotent Stem Cells from Old Fibroblasts Using Synthetic RNAs , 2020, Animals : an open access journal from MDPI.

[29]  J. LaMarre,et al.  Targeted expression profiling reveals distinct stages of early canine fibroblast reprogramming are regulated by 2-oxoglutarate hydroxylases , 2020, Stem cell research & therapy.

[30]  A. Cowin,et al.  Mesenchymal Stem Cell Secretome as an Emerging Cell-Free Alternative for Improving Wound Repair , 2020, International journal of molecular sciences.

[31]  Dong Sung Kim,et al.  Human kidney organoids model the tacrolimus nephrotoxicity and elucidate the role of autophagy , 2020, The Korean journal of internal medicine.

[32]  G. Pan,et al.  Glis1 facilitates induction of pluripotency via an epigenome–metabolome–epigenome signalling cascade , 2020, Nature Metabolism.

[33]  D. Ovchinnikov,et al.  Pluripotency and immunomodulatory signatures of canine induced pluripotent stem cell-derived mesenchymal stromal cells are similar to harvested mesenchymal stromal cells , 2020, Scientific Reports.

[34]  K. Sugiura,et al.  Generation of Footprint‐Free Canine Induced Pluripotent Stem Cells from Peripheral Blood Mononuclear Cells Using Sendai Virus Vector , 2020, Molecular reproduction and development.

[35]  René A. M. Dirks,et al.  In vitro capture and characterization of embryonic rosette-stage pluripotency between naive and primed states , 2020, Nature Cell Biology.

[36]  Carolyn E. Dundes,et al.  Bridging naïve and primed pluripotency , 2020, Nature Cell Biology.

[37]  T. Šarić,et al.  Research and therapy with induced pluripotent stem cells (iPSCs): social, legal, and ethical considerations , 2019, Stem Cell Research & Therapy.

[38]  Katja Schenke-Layland,et al.  Merging organoid and organ-on-a-chip technology to generate complex multi-layer tissue models in a human retina-on-a-chip platform , 2019, eLife.

[39]  Jean Tang,et al.  Induction of long-term allogeneic cell acceptance and formation of immune privileged tissue in immunocompetent hosts , 2019, bioRxiv.

[40]  D. Russell,et al.  Engineering universal cells that evade immune detection , 2019, Nature Reviews Immunology.

[41]  M. Avci-Adali,et al.  Generation of iPSCs by Nonintegrative RNA-Based Reprogramming Techniques: Benefits of Self-Replicating RNA versus Synthetic mRNA , 2019, Stem cells international.

[42]  K. Pinello,et al.  Incidence, characteristics and geographical distributions of canine and human non-Hodgkin's lymphoma in the Porto region (North West Portugal). , 2019, Veterinary journal.

[43]  Wojciech Zakrzewski,et al.  Stem cells: past, present, and future , 2019, Stem cell research & therapy.

[44]  Mark M. Davis,et al.  Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients , 2019, Nature Biotechnology.

[45]  E. Bach,et al.  JAK/STAT signaling in stem cells and regeneration: from Drosophila to vertebrates , 2019, Development.

[46]  K. Sugiura,et al.  Generation of Footprint-Free Canine Induced Pluripotent Stem Cells Using Auto-Erasable Sendai Virus Vector. , 2018, Stem cells and development.

[47]  Natalia B. Ivanova,et al.  Dppa2/4 Facilitate Epigenetic Remodeling during Reprogramming to Pluripotency. , 2018, Cell stem cell.

[48]  M. Huynen,et al.  Mammalian embryo comparison identifies novel pluripotency genes associated with the naïve or primed state , 2018, Biology Open.

[49]  Christoph Bock,et al.  Assessment of established techniques to determine developmental and malignant potential of human pluripotent stem cells , 2018, Nature Communications.

[50]  William A. Pastor,et al.  TFAP2C regulates transcription in human naive pluripotency by opening enhancers , 2018, Nature Cell Biology.

[51]  A. Koster,et al.  Renal Subcapsular Transplantation of PSC-Derived Kidney Organoids Induces Neo-vasculogenesis and Significant Glomerular and Tubular Maturation In Vivo , 2018, Stem cell reports.

[52]  Sheng Ding,et al.  CRISPR-Based Chromatin Remodeling of the Endogenous Oct4 or Sox2 Locus Enables Reprogramming to Pluripotency. , 2018, Cell stem cell.

[53]  Teresa J. Feo,et al.  Structural absorption by barbule microstructures of super black bird of paradise feathers , 2018, Nature Communications.

[54]  A. Hutchins,et al.  Chromatin Accessibility Dynamics during iPSC Reprogramming. , 2017, Cell stem cell.

[55]  S. Webb,et al.  Safety and immune regulatory properties of canine induced pluripotent stem cell-derived mesenchymal stem cells. , 2017, Stem cell research.

[56]  I. Hiratani,et al.  Epigenetic differences between naïve and primed pluripotent stem cells , 2017, Cellular and Molecular Life Sciences.

[57]  C. Gregory,et al.  In-vitro characterization of canine multipotent stromal cells isolated from synovium, bone marrow, and adipose tissue: a donor-matched comparative study , 2017, Stem Cell Research & Therapy.

[58]  L. Vallier,et al.  Variability of human pluripotent stem cell lines. , 2017, Current opinion in genetics & development.

[59]  E. Rulifson,et al.  Paracrine Effects of the Pluripotent Stem Cell-Derived Cardiac Myocytes Salvage the Injured Myocardium , 2017, Circulation research.

[60]  A. Kurisaki,et al.  Simple and effective generation of transgene-free induced pluripotent stem cells using an auto-erasable Sendai virus vector responding to microRNA-302. , 2017, Stem cell research.

[61]  J. Hanna,et al.  A multiplexed screening method for pluripotency. , 2017, Stem cell research.

[62]  J. Qin,et al.  Acetylation on histone H3 lysine 9 mediates a switch from transcription initiation to elongation , 2017, The Journal of Biological Chemistry.

[63]  H. Imai,et al.  Feeder‐independent canine induced pluripotent stem cells maintained under serum‐free conditions , 2017, Molecular reproduction and development.

[64]  C. Ambrósio,et al.  Generation of LIF-independent induced pluripotent stem cells from canine fetal fibroblasts. , 2017, Theriogenology.

[65]  A. Kurisaki,et al.  A Role for KLF4 in Promoting the Metabolic Shift via TCL1 during Induced Pluripotent Stem Cell Generation , 2017, Stem cell reports.

[66]  M. Moss,et al.  Translational models for vascular cognitive impairment: a review including larger species , 2017, BMC Medicine.

[67]  Jiesi Luo,et al.  Conserved Role of bFGF and a Divergent Role of LIF for Pluripotency Maintenance and Survival in Canine Pluripotent Stem Cells. , 2016, Stem cells and development.

[68]  L. Niklason,et al.  Human Pluripotent Stem Cells (iPSC) Generation, Culture, and Differentiation to Lung Progenitor Cells. , 2016, Methods in molecular biology.

[69]  Charles A. Gersbach,et al.  Targeted Epigenetic Remodeling of Endogenous Loci by CRISPR/Cas9-Based Transcriptional Activators Directly Converts Fibroblasts to Neuronal Cells. , 2016, Cell stem cell.

[70]  J. Elisseeff,et al.  Human iPSC-derived osteoblasts and osteoclasts together promote bone regeneration in 3D biomaterials , 2016, Scientific Reports.

[71]  Yoshimi Kawamura,et al.  Tumour resistance in induced pluripotent stem cells derived from naked mole-rats , 2016, Nature Communications.

[72]  H. Lohi,et al.  Canine models of human rare disorders , 2016, Rare diseases.

[73]  C. Verfaillie,et al.  Mesodermal iPSC-derived progenitor cells functionally regenerate cardiac and skeletal muscle. , 2015, The Journal of clinical investigation.

[74]  I. C. Tobias,et al.  Canine Pluripotent Stem Cells: Are They Ready for Clinical Applications? , 2015, Front. Vet. Sci..

[75]  Leigh G. Griffiths,et al.  Companion animals: Translational scientist’s new best friends , 2015, Science Translational Medicine.

[76]  Byeong-chun Lee,et al.  Effect of primary culture medium type for culture of canine fibroblasts on production of cloned dogs. , 2015, Theriogenology.

[77]  Xin Xie,et al.  Bromodeoxyuridine promotes full-chemical induction of mouse pluripotent stem cells , 2015, Cell Research.

[78]  T. Barsby,et al.  Derivation of Canine Induced Pluripotent Stem Cells. , 2015, Reproduction in domestic animals = Zuchthygiene.

[79]  Larry A Couture,et al.  Hurdles to clinical translation of human induced pluripotent stem cells. , 2015, The Journal of clinical investigation.

[80]  E. Sasaki,et al.  Non-Viral Generation of Marmoset Monkey iPS Cells by a Six-Factor-in-One-Vector Approach , 2015, PloS one.

[81]  William Ritchie,et al.  Genome-wide characterization of the routes to pluripotency , 2014, Nature.

[82]  Xin Li,et al.  A comparison of non-integrating reprogramming methods , 2014, Nature Biotechnology.

[83]  S. Balasubramanian,et al.  5-Hydroxymethylcytosine is a predominantly stable DNA modification. , 2014, Nature chemistry.

[84]  Stefan Liebau,et al.  A Comparative View on Human Somatic Cell Sources for iPSC Generation , 2014, Stem cells international.

[85]  D. Ovchinnikov,et al.  Derivation of mesenchymal stromal cells from canine induced pluripotent stem cells by inhibition of the TGFβ/activin signaling pathway. , 2014, Stem cells and development.

[86]  T. Hulsey,et al.  Association of Human Leukocyte Antigen Donor–Recipient Matching and Pediatric Heart Transplant Graft Survival , 2014, Circulation. Heart failure.

[87]  Michiko Nakamura,et al.  Induction of pluripotency in human somatic cells via a transient state resembling primitive streak-like mesendoderm , 2014, Nature Communications.

[88]  A. Goldrath,et al.  An effective approach to prevent immune rejection of human ESC-derived allografts. , 2014, Cell stem cell.

[89]  P W Nickerson,et al.  Class II HLA Epitope Matching—A Strategy to Minimize De Novo Donor‐Specific Antibody Development and Improve Outcomes , 2013, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons.

[90]  K. Hochedlinger,et al.  Chromatin dynamics during cellular reprogramming , 2013, Nature.

[91]  A. Fisher,et al.  DNA demethylation in pluripotency and reprogramming: the role of tet proteins and cell division. , 2013, Cell stem cell.

[92]  A. Muotri,et al.  Efficient generation of human iPSCs by a synthetic self-replicative RNA. , 2013, Cell stem cell.

[93]  S. Tsai,et al.  Growth requirements and chromosomal instability of induced pluripotent stem cells generated from adult canine fibroblasts. , 2013, Stem cells and development.

[94]  K. Plath,et al.  Epigenetics of Reprogramming to Induced Pluripotency , 2013, Cell.

[95]  H. Imai,et al.  Generation of functional platelets from canine induced pluripotent stem cells. , 2013, Stem cells and development.

[96]  P. Verma,et al.  Nanog Is an Essential Factor for Induction of Pluripotency in Somatic Cells from Endangered Felids , 2013, BioResearch open access.

[97]  J. C. Belmonte,et al.  Characterization of pluripotent stem cells , 2013, Nature Protocols.

[98]  S. Yamanaka,et al.  Epigenetic regulation in pluripotent stem cells: a key to breaking the epigenetic barrier , 2013, Philosophical Transactions of the Royal Society B: Biological Sciences.

[99]  Vincent C. Chen,et al.  Epigenetic stability, adaptability, and reversibility in human embryonic stem cells , 2012, Proceedings of the National Academy of Sciences.

[100]  Kisuk Yang,et al.  Engineering biomaterials for feeder-free maintenance of human pluripotent stem cells. , 2012, International journal of stem cells.

[101]  T. Böckers,et al.  Rat embryonic fibroblasts improve reprogramming of human keratinocytes into induced pluripotent stem cells. , 2012, Stem cells and development.

[102]  E. Wolvetang,et al.  Small Molecule Mesengenic Induction of Human Induced Pluripotent Stem Cells to Generate Mesenchymal Stem/Stromal Cells , 2012, Stem cells translational medicine.

[103]  D. Ovchinnikov,et al.  Generation and characterization of LIF-dependent canine induced pluripotent stem cells from adult dermal fibroblasts. , 2012, Stem cells and development.

[104]  G. Pan,et al.  The histone demethylases Jhdm1a/1b enhance somatic cell reprogramming in a vitamin-C-dependent manner. , 2011, Cell stem cell.

[105]  A. Yeung,et al.  Preclinical Derivation and Imaging of Autologously Transplanted Canine Induced Pluripotent Stem Cells* , 2011, The Journal of Biological Chemistry.

[106]  K. Cao,et al.  Intramyocardial Transplantation of Undifferentiated Rat Induced Pluripotent Stem Cells Causes Tumorigenesis in the Heart , 2011, PloS one.

[107]  S. Suhr,et al.  Generation of leukemia inhibitory factor and basic fibroblast growth factor-dependent induced pluripotent stem cells from canine adult somatic cells. , 2011, Stem cells and development.

[108]  A. Higuchi,et al.  Biomaterials for the feeder-free culture of human embryonic stem cells and induced pluripotent stem cells. , 2011, Chemical reviews.

[109]  E. Kirkness,et al.  Somatic coding mutations in human induced pluripotent stem cells , 2011, Nature.

[110]  S. Wood,et al.  Despite identifying some shared gene associations with human atopic dermatitis the use of multiple dog breeds from various locations limits detection of gene associations in canine atopic dermatitis. , 2010, Veterinary immunology and immunopathology.

[111]  Donald C. Chang,et al.  Regulation of somatic cell reprogramming through inducible mir-302 expression , 2010, Nucleic acids research.

[112]  Yi Zhang,et al.  Butyrate Promotes Induced Pluripotent Stem Cell Generation* , 2010, The Journal of Biological Chemistry.

[113]  A. G. Fadeev,et al.  Synthetic peptide-acrylate surfaces for long-term self-renewal and cardiomyocyte differentiation of human embryonic stem cells , 2010, Nature Biotechnology.

[114]  J. Lahann,et al.  Synthetic polymer coatings for long-term growth of human embryonic stem cells , 2010, Nature Biotechnology.

[115]  K. Chien,et al.  Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511 , 2010, Nature Biotechnology.

[116]  Tatsuo Nakamura,et al.  Generation of canine induced pluripotent stem cells by retroviral transduction and chemical inhibitors , 2009, Molecular reproduction and development.

[117]  Hideyuki Okano,et al.  Variation in the safety of induced pluripotent stem cell lines , 2009, Nature Biotechnology.

[118]  J. Nichols,et al.  Naive and primed pluripotent states. , 2009, Cell stem cell.

[119]  M. R. Hough,et al.  Derivation and Characterization of Canine Embryonic Stem Cell Lines with In Vitro and In Vivo Differentiation Potential , 2009, Stem cells.

[120]  A. Consiglio,et al.  Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes , 2008, Nature Biotechnology.

[121]  Katsushi Tokunaga,et al.  HLA-haplotype banking and iPS cells , 2008, Nature Biotechnology.

[122]  Wenjun Guo,et al.  Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds , 2008, Nature Biotechnology.

[123]  Michael Q. Zhang,et al.  Combinatorial patterns of histone acetylations and methylations in the human genome , 2008, Nature Genetics.

[124]  R. Clemmons,et al.  Nestin-positive spheres derived from canine bone marrow stromal cells generate cells with early neuronal and glial phenotypic characteristics , 2008, In Vitro Cellular & Developmental Biology - Animal.

[125]  M. Paoloni,et al.  Translation of new cancer treatments from pet dogs to humans , 2008, Nature Reviews Cancer.

[126]  A. Trounson,et al.  Embryonic stem cells in companion animals (horses, dogs and cats): present status and future prospects. , 2007, Reproduction, fertility, and development.

[127]  M. Trotter,et al.  Derivation of pluripotent epiblast stem cells from mammalian embryos , 2007, Nature.

[128]  E. Wolf,et al.  Canine Embryo‐Derived Stem Cells—Toward Clinically Relevant Animal Models for Evaluating Efficacy and Safety of Cell Therapies , 2007, Stem cells.

[129]  C. Allis,et al.  Epigenetics: A Landscape Takes Shape , 2007, Cell.

[130]  S. Yamanaka,et al.  Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors , 2006, Cell.

[131]  K. Sugiura,et al.  Isolation and characterization of embryonic stem‐like cells from canine blastocysts , 2006, Molecular reproduction and development.

[132]  M. Haskins,et al.  Effects of osteogenic inducers on cultures of canine mesenchymal stem cells. , 2005, American journal of veterinary research.

[133]  B. Gumbiner,et al.  Regulation of cadherin-mediated adhesion in morphogenesis , 2005, Nature Reviews Molecular Cell Biology.

[134]  C. Mellersh,et al.  Dogs really are man's best friend--canine genomics has applications in veterinary and human medicine! , 2005, Briefings in functional genomics & proteomics.

[135]  F. Gage,et al.  Human embryonic stem cells express an immunogenic nonhuman sialic acid , 2005, Nature Medicine.

[136]  R. Robinson,et al.  Dogs, zoonoses and immunosuppression , 2002, The journal of the Royal Society for the Promotion of Health.

[137]  Chunhui Xu,et al.  Feeder-free growth of undifferentiated human embryonic stem cells , 2001, Nature Biotechnology.

[138]  B. Lim,et al.  Ex uno plures: molecular designs for embryonic pluripotency. , 2015, Physiological reviews.

[139]  E. Matunis,et al.  JAK-STAT signaling in stem cells. , 2013, Advances in experimental medicine and biology.

[140]  D. H. Lee,et al.  285 GENERATION OF CANINE INDUCED PLURIPOTENT STEM CELLS FROM CANINE FETAL FIBROBLAST AND ADULT FIBROBLAST OF CLONED DOG , 2013 .