1
|
Na Q, Zhang S, Shao P, Jia Y, Wang Y, Wei M, Chen Y, Chen C, Zhao L, Wang Z, Song Y, Wu B, Bao S, Li X. In vitro generation of trophoblast like stem cells from goat pluripotent stem cells. Theriogenology 2024; 226:120-129. [PMID: 38878464 DOI: 10.1016/j.theriogenology.2024.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024]
Abstract
Since the first mouse induced pluripotent stem cells (iPSCs) was derived, the in vitro culture of domestic iPSCs functionally and molecularly comparable with mouse iPSCs has been a challenge. Here, we established dairy goat iPSCs (giPSCs) from goat ear fibroblast cells with mouse iPSCs morphology, the expression of pluripotent markers and differentiation ability in vitro delivered by piggyBac transposon with nine Dox-inducible exogenous reprogramming factors. These reprogramming factors were bOMSK (bovine OCT4, CMYC, SOX2, and KLF4), pNhL (porcine NANOG and human LIN28), hRL (human RARG and LRH1), and SV40 Large T. Notably, AF-giPSCs (induced in activin A and bFGF condition) were capable of differentiation in embryoid bodies in vitro and could contribute to interspecies chimerism in mouse E6.5 embryos in vitro, demonstrating that AF-giPSCs have the developmental capability to generate some embryonic cell lineages. Moreover, Wnt/β-catenin signaling has an important role in driving goat induced trophoblast-like stem cells (giTLSCs) from Dox-independent giPSCs. This study will support further establishment of the stable giPSC lines without any integration of exogenous genes.
Collapse
Affiliation(s)
- Qin Na
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Siyu Zhang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Peng Shao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Yu Jia
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Yanqiu Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Mengyi Wei
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Yanglin Chen
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Chen Chen
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Lixia Zhao
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Zixin Wang
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animal, 011517, Hohhot, China
| | - Yongli Song
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Baojiang Wu
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China
| | - Siqin Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China.
| | - Xihe Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, 010020, Hohhot, China; Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, 010020, Hohhot, China; Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animal, 011517, Hohhot, China.
| |
Collapse
|
2
|
Zeng R, Huang X, Fu W, Ji W, Cai W, Xu M, Lan D. Construction of Lentiviral Vectors Carrying Six Pluripotency Genes in Yak to Obtain Yak iPSC Cells. Int J Mol Sci 2024; 25:9431. [PMID: 39273379 PMCID: PMC11394755 DOI: 10.3390/ijms25179431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/02/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Yak is an excellent germplasm resource on the Tibetan Plateau and is able to live in high-altitude areas with hypoxic, cold, and harsh environments. Studies on induced pluripotent stem cells (iPSCs) in large ruminants commonly involve a combination strategy involving six transcription factors, Oct4, Sox2, Klf4, c-Myc, Nanog, and Lin28 (OSKMNL). This strategy tends to utilize genes from the same species to optimize pluripotency maintenance. In this study, we cloned the six pluripotency genes (OSKMNL) from yak and constructed a multi-cistronic lentiviral vector carrying these genes. This vector efficiently delivered the genes into yak fibroblasts, aiming to promote the reprogramming process. We verified that the treated cells had several pluripotency characteristics, marking the first successful construction of a lentiviral system carrying yak pluripotency genes. This achievement lays the foundation for subsequent establishment of yak iPSCs and holds significant implications for yak-breed improvement and germplasm-resource conservation.
Collapse
Affiliation(s)
- Ruilin Zeng
- College of Animal & Verterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Xianpeng Huang
- College of Animal & Verterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Wei Fu
- College of Animal & Verterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Wenhui Ji
- College of Animal & Verterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Wenyi Cai
- College of Animal & Verterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Meng Xu
- College of Animal & Verterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Daoliang Lan
- College of Animal & Verterinary Sciences, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Qinghai-Tibet Plateau Animal Genetic Resource and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
3
|
Schrade L, Mah N, Bandrowski A, Chen Y, Dewender J, Diecke S, Hiepen C, Lancaster MA, Marques-Bonet T, Martinez S, Mueller SC, Navara C, Prigione A, Seltmann S, Sochacki J, Sutcliffe MA, Zywitza V, Hildebrandt TB, Kurtz A. A Standardized Nomenclature Design for Systematic Referencing and Identification of Animal Cellular Material. Animals (Basel) 2024; 14:1541. [PMID: 38891588 PMCID: PMC11171381 DOI: 10.3390/ani14111541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
The documentation, preservation and rescue of biological diversity increasingly uses living biological samples. Persistent associations between species, biosamples, such as tissues and cell lines, and the accompanying data are indispensable for using, exchanging and benefiting from these valuable materials. Explicit authentication of such biosamples by assigning unique and robust identifiers is therefore required to allow for unambiguous referencing, avoid identification conflicts and maintain reproducibility in research. A predefined nomenclature based on uniform rules would facilitate this process. However, such a nomenclature is currently lacking for animal biological material. We here present a first, standardized, human-readable nomenclature design, which is sufficient to generate unique and stable identifying names for animal cellular material with a focus on wildlife species. A species-specific human- and machine-readable syntax is included in the proposed standard naming scheme, allowing for the traceability of donated material and cultured cells, as well as data FAIRification. Only when it is consistently applied in the public domain, as publications and inter-institutional samples and data are exchanged, distributed and stored centrally, can the risks of misidentification and loss of traceability be mitigated. This innovative globally applicable identification system provides a standard for a sustainable structure for the long-term storage of animal bio-samples in cryobanks and hence facilitates current as well as future species conservation and biomedical research.
Collapse
Affiliation(s)
- Lisa Schrade
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research (IZW), 10315 Berlin, Germany
| | - Nancy Mah
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Anita Bandrowski
- Department of Neuroscience, FAIR Data Informatics Lab, University of California San Diego, San Diego, CA 92093, USA
- SciCrunch Inc., San Diego, CA 92192, USA
| | - Ying Chen
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Johannes Dewender
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Sebastian Diecke
- Technology Platform Pluripotent Stem Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Christian Hiepen
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Madeline A. Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Tomas Marques-Bonet
- Institute of Evolutionary Biology, Pompeu Fabra University—Spanish National Research Council, ICREA, 08003 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Centro Nacional de Analisis Genomico (CNAG), 08028 Barcelona, Spain
- Catalan Institute of Palaeontology Miquel Crusafont, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Sira Martinez
- Institute of Evolutionary Biology, Pompeu Fabra University—Spanish National Research Council, ICREA, 08003 Barcelona, Spain
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003 Barcelona, Spain
| | - Sabine C. Mueller
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Christopher Navara
- San Antonio Cellular Therapeutics Institute, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Duesseldorf University Hospital, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Stefanie Seltmann
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Jaroslaw Sochacki
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003 Barcelona, Spain
| | | | - Vera Zywitza
- Technology Platform Pluripotent Stem Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Thomas B. Hildebrandt
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research (IZW), 10315 Berlin, Germany
- Faculty of Veterinary Medicine, Free University of Berlin, 14163 Berlin, Germany
| | - Andreas Kurtz
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
- Berlin Institute of Health (BIH), Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany
| |
Collapse
|
4
|
Li Z, Li Y, Zhang Q, Ge W, Zhang Y, Zhao X, Hu J, Yuan L, Zhang W. Establishment of Bactrian Camel Induced Pluripotent Stem Cells and Prediction of Their Unique Pluripotency Genes. Int J Mol Sci 2023; 24:ijms24031917. [PMID: 36768240 PMCID: PMC9916525 DOI: 10.3390/ijms24031917] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/05/2023] [Accepted: 01/15/2023] [Indexed: 01/21/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) can differentiate into all types of cells and can be used in livestock for research on biological development, genetic breeding, and in vitro genetic resource conservation. The Bactrian camel is a large domestic animal that inhabits extreme environments and holds value in the treatment of various diseases and the development of the local economy. Therefore, we transferred four mouse genes (Oct4, Sox2, Klf4, and c-Myc) into Bactrian camel fetal fibroblasts (BCFFs) using retroviruses with a large host range to obtain Bactrian camel induced pluripotent stem cells (bciPSCs). They were comprehensively identified based on cell morphology, pluripotency gene and marker expression, chromosome number, transcriptome sequencing, and differentiation potential. The results showed the pluripotency of bciPSCs. However, unlike stem cells of other species, late formation of stem cell clones was observed; moreover, the immunofluorescence of SSEA1, SSEA3, and SSEA4 were positive, and teratoma formation took four months. These findings may be related to the extremely long gestation period and species specificity of Bactrian camels. By mining RNA sequence data, 85 potential unique pluripotent genes of Bactrian camels were predicted, which could be used as candidate genes for the production of bciPSC in the future. Among them, ASF1B, DTL, CDCA5, PROM1, CYTL1, NUP210, Epha3, and SYT13 are more attractive. In conclusion, we generated bciPSCs for the first time and obtained their transcriptome information, expanding the iPSC genetic information database and exploring the applicability of iPSCs in livestock. Our results can provide an experimental basis for Bactrian camel ESC establishment, developmental research, and genetic resource conservation.
Collapse
Affiliation(s)
- Zongshuai Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China
| | - Yina Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China
| | - Qiran Zhang
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Wenbo Ge
- Chinese Academy of Agricultural Sciences Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Lanzhou 730070, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China
- Correspondence:
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China
| | - Junjie Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Gansu Agricultural University, Lanzhou 730070, China
| | - Ligang Yuan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Wangdong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
5
|
Widerspick L, Steffen JF, Tappe D, Muñoz-Fontela C. Animal Model Alternatives in Filovirus and Bornavirus Research. Viruses 2023; 15:158. [PMID: 36680198 PMCID: PMC9863967 DOI: 10.3390/v15010158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
The order Mononegavirales contains a variety of highly pathogenic viruses that may infect humans, including the families Filoviridae, Bornaviridae, Paramyxoviridae, and Rhabodoviridae. Animal models have historically been important to study virus pathogenicity and to develop medical countermeasures. As these have inherent shortcomings, the rise of microphysiological systems and organoids able to recapitulate hallmarks of the diseases caused by these viruses may have enormous potential to add to or partially replace animal modeling in the future. Indeed, microphysiological systems and organoids are already used in the pharmaceutical R&D pipeline because they are prefigured to overcome the translational gap between model systems and clinical studies. Moreover, they may serve to alleviate ethical concerns related to animal research. In this review, we discuss the value of animal model alternatives in human pathogenic filovirus and bornavirus research. The current animal models and their limitations are presented followed by an overview of existing alternatives, such as organoids and microphysiological systems, which might help answering open research questions.
Collapse
Affiliation(s)
- Lina Widerspick
- Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Riems, 38124 Braunschweig, Germany
| | | | - Dennis Tappe
- Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
- National Reference Center for Tropical Pathogens, Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - César Muñoz-Fontela
- Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Riems, 38124 Braunschweig, Germany
| |
Collapse
|
6
|
Weeratunga P, Harman RM, Van de Walle GR. Induced pluripotent stem cells from domesticated ruminants and their potential for enhancing livestock production. Front Vet Sci 2023; 10:1129287. [PMID: 36891466 PMCID: PMC9986305 DOI: 10.3389/fvets.2023.1129287] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023] Open
Abstract
Ruminant livestock, including cattle, sheep, goat, and buffalo, are essential for global food security and serve valuable roles in sustainable agricultural systems. With the limited availability of embryonic stem cells (ESCs) from these species, ruminant induced pluripotent stem cells (iPSCs) and iPSC-like cells provide a valuable research tool for agricultural, veterinary, biomedical, and pharmaceutical applications, as well as for the prospect of translation to human medicine. iPSCs are generated by reprogramming of adult or fetal cells to an ESC-like state by ectopic expression of defined transcription factors. Despite the slow pace the field has evolved in livestock species compared to mice and humans, significant progress has been made over the past 15 years in using different cell sources and reprogramming protocols to generate iPSCs/iPSC-like cells from ruminants. This mini review summarizes the current literature related to the derivation of iPSCs/iPSC-like cells from domesticated ruminants with a focus on reprogramming protocols, characterization, associated limitations, and potential applications in ruminant basic science research and production.
Collapse
Affiliation(s)
- Prasanna Weeratunga
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
7
|
Botigelli RC, Pieri NCG, Bessi BW, Machado LS, Bridi A, de Souza AF, Recchia K, Neto PF, Ross PJ, Bressan FF, Nogueira MFG. Acquisition and maintenance of pluripotency are influenced by fibroblast growth factor, leukemia inhibitory factor, and 2i in bovine-induced pluripotent stem cells. Front Cell Dev Biol 2022; 10:938709. [PMID: 36187479 PMCID: PMC9515551 DOI: 10.3389/fcell.2022.938709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/11/2022] [Indexed: 12/03/2022] Open
Abstract
Several opportunities for embryo development, stem cell maintenance, cell fate, and differentiation have emerged using induced pluripotent stem cells (iPSCs). However, the difficulty in comparing bovine iPSCs (biPSCs) with embryonic stem cells (ESCs) was a challenge for many years. Here, we reprogrammed fetal fibroblasts by transient expression of the four transcription factors (Oct4, Sox2, Klf4, and c-Myc, collectively termed “OSKM” factors) and cultured in iPSC medium, supplemented with bFGF, bFGF2i, leukemia inhibitory factor (LIF), or LIF2i, and then compared these biPSC lines with bESC to evaluate the pluripotent state. biPSC lines were generated in all experimental groups. Particularly, reprogrammed cells treated with bFGF were more efficient in promoting the acquisition of pluripotency. However, LIF2i treatment did not promote continuous self-renewal. biPSCs (line 2) labeled with GFP were injected into early embryos (day 4.5) to assess the potential to contribute to chimeric blastocysts. The biPSC lines show a pluripotency state and are differentiated into three embryonic layers. Moreover, biPSCs and bESCs labeled with GFP were able to contribute to chimeric blastocysts. Additionally, biPSCs have shown promising potential for contributing to chimeric blastocysts and for future studies.
Collapse
Affiliation(s)
- Ramon Cesar Botigelli
- Multiuser Facility (FitoFarmaTec), Department of Pharmacology, Biosciences Institute (IBB), São Paulo State University (UNESP), Botucatu, Brazil
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
- Correspondence: Ramon Cesar Botigelli, ; Marcelo Fábio Gouveia Nogueira,
| | - Naira Carolina Godoy Pieri
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
| | - Brendon William Bessi
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
| | - Lucas Simões Machado
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
- Paulista School of Medicine (EPM), Laboratory of Neurobiology, Department of Biochemistry, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Alessandra Bridi
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
| | - Aline Fernanda de Souza
- Laboratory Biomedical Science, Department of Biomedical Science, Ontario Veterinary College (OVC), University of Guelph, Guelph, ON, Canada
| | - Kaiana Recchia
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
| | - Paulo Fantinato Neto
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
| | - Pablo Juan Ross
- Laboratory Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Fabiana Fernandes Bressan
- Laboratory of Molecular Morphophysiology and Development (LMMD), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering (FZEA), University of São Paulo (USP), Pirassununga, Brazil
| | - Marcelo Fábio Gouveia Nogueira
- Multiuser Facility (FitoFarmaTec), Department of Pharmacology, Biosciences Institute (IBB), São Paulo State University (UNESP), Botucatu, Brazil
- School of Sciences and Languages, Laboratory of Embryonic Micromanipulation, Department of Biological Sciences, São Paulo State University (UNESP), Assis, Brazil
- Correspondence: Ramon Cesar Botigelli, ; Marcelo Fábio Gouveia Nogueira,
| |
Collapse
|
8
|
Knežić T, Janjušević L, Djisalov M, Yodmuang S, Gadjanski I. Using Vertebrate Stem and Progenitor Cells for Cellular Agriculture, State-of-the-Art, Challenges, and Future Perspectives. Biomolecules 2022; 12:699. [PMID: 35625626 PMCID: PMC9138761 DOI: 10.3390/biom12050699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/07/2022] [Indexed: 12/19/2022] Open
Abstract
Global food systems are under significant pressure to provide enough food, particularly protein-rich foods whose demand is on the rise in times of crisis and inflation, as presently existing due to post-COVID-19 pandemic effects and ongoing conflict in Ukraine and resulting in looming food insecurity, according to FAO. Cultivated meat (CM) and cultivated seafood (CS) are protein-rich alternatives for traditional meat and fish that are obtained via cellular agriculture (CA) i.e., tissue engineering for food applications. Stem and progenitor cells are the building blocks and starting point for any CA bioprocess. This review presents CA-relevant vertebrate cell types and procedures needed for their myogenic and adipogenic differentiation since muscle and fat tissue are the primary target tissues for CM/CS production. The review also describes existing challenges, such as a need for immortalized cell lines, or physical and biochemical parameters needed for enhanced meat/fat culture efficiency and ways to address them.
Collapse
Affiliation(s)
- Teodora Knežić
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Ljiljana Janjušević
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Mila Djisalov
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Supansa Yodmuang
- Research Affairs, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd, Pathumwan, Bangkok 10330, Thailand;
| | - Ivana Gadjanski
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| |
Collapse
|
9
|
Feeding role of mouse embryonic fibroblast cells is influenced by genetic background, cell passage and day of isolation. ZYGOTE 2022; 30:550-560. [PMID: 35485762 DOI: 10.1017/s0967199421000083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Mouse embryonic fibroblast (MEF) cells are commonly used as feeder cells to maintain the pluripotent state of stem cells. MEFs produce growth factors and provide adhesion molecules and extracellular matrix (ECM) compounds for cellular binding. In the present study, we compared the expression levels of Fgf2, Bmp4, ActivinA, Lif and Tgfb1 genes at the mRNA level and the level of Fgf2 protein secretion and Lif cytokine secretion at passages one, three and five of MEFs isolated from 13.5-day-old and 15.5-day-old embryos of NMRI and C57BL/6 mice using real-time PCR and enzyme-linked immunosorbent assay. We observed differences in the expression levels of the studied genes and secretion of the two growth factors in the three passages of MEFs isolated from 13.5-day-old and 15.5-day-old embryos, respectively. These differences were also observed between the NMRI and C57BL/6 strains. The results of this study suggested that researchers should use mice embryos that have different genetic backgrounds and ages, in addition to different MEF passages, when producing MEFs based on the application and type of their study.
Collapse
|
10
|
Liu M, Zhao L, Wang Z, Su H, Wang T, Yang G, Chen L, Wu B, Zhao G, Guo J, Yang Z, Zhang J, Hao C, Ma T, Song Y, Bao S, Zuo Y, Li X, Cao G. Generation of Sheep Induced Pluripotent Stem Cells With Defined DOX-Inducible Transcription Factors via piggyBac Transposition. Front Cell Dev Biol 2021; 9:785055. [PMID: 34977028 PMCID: PMC8716767 DOI: 10.3389/fcell.2021.785055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022] Open
Abstract
Pluripotent stem cells (PSCs) have the potential to differentiate to all cell types of an adult individual and are useful for studying mammalian development. Establishing induced pluripotent stem cells (iPSCs) capable of expressing pluripotent genes and differentiating to three germ layers will not only help to explain the mechanisms underlying somatic reprogramming but also lay the foundation for the establishment of sheep embryonic stem cells (ESCs) in vitro. In this study, sheep somatic cells were reprogrammed in vitro into sheep iPSCs with stable morphology, pluripotent marker expression, and differentiation ability, delivered by piggyBac transposon system with eight doxycycline (DOX)-inducible exogenous reprogramming factors: bovine OCT4, SOX2, KLF4, cMYC, porcine NANOG, human LIN28, SV40 large T antigen, and human TERT. Sheep iPSCs exhibited a chimeric contribution to the early blastocysts of sheep and mice and E6.5 mouse embryos in vitro. A transcriptome analysis revealed the pluripotent characteristics of somatic reprogramming and insights into sheep iPSCs. This study provides an ideal experimental material for further study of the construction of totipotent ESCs in sheep.
Collapse
Affiliation(s)
- Moning Liu
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Hohhot, China
| | - Lixia Zhao
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
- Inner Mongolia Saikexing Institutes of Breeding and Reproductive Biotechnologies in Domestic Animal, Hohhot, China
| | - Zixin Wang
- Inner Mongolia Saikexing Institutes of Breeding and Reproductive Biotechnologies in Domestic Animal, Hohhot, China
| | - Hong Su
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Hohhot, China
| | - Tong Wang
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Hohhot, China
| | - Guang Yang
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Lu Chen
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Hohhot, China
| | - Baojiang Wu
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Gaoping Zhao
- Inner Mongolia Saikexing Institutes of Breeding and Reproductive Biotechnologies in Domestic Animal, Hohhot, China
| | - Jitong Guo
- Inner Mongolia Saikexing Institutes of Breeding and Reproductive Biotechnologies in Domestic Animal, Hohhot, China
| | - Zhiqing Yang
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jia Zhang
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Chunxia Hao
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Teng Ma
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Hohhot, China
| | - Yongli Song
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Siqin Bao
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yongchun Zuo
- China State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xihe Li
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, China
- Inner Mongolia Saikexing Institutes of Breeding and Reproductive Biotechnologies in Domestic Animal, Hohhot, China
- *Correspondence: Guifang Cao, ; Xihe Li,
| | - Guifang Cao
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, College of Veterinary, Inner Mongolia Agricultural University, Hohhot, China
- *Correspondence: Guifang Cao, ; Xihe Li,
| |
Collapse
|
11
|
Martínez-Falguera D, Iborra-Egea O, Gálvez-Montón C. iPSC Therapy for Myocardial Infarction in Large Animal Models: Land of Hope and Dreams. Biomedicines 2021; 9:1836. [PMID: 34944652 PMCID: PMC8698445 DOI: 10.3390/biomedicines9121836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction is the main driver of heart failure due to ischemia and subsequent cell death, and cell-based strategies have emerged as promising therapeutic methods to replace dead tissue in cardiovascular diseases. Research in this field has been dramatically advanced by the development of laboratory-induced pluripotent stem cells (iPSCs) that harbor the capability to become any cell type. Like other experimental strategies, stem cell therapy must meet multiple requirements before reaching the clinical trial phase, and in vivo models are indispensable for ensuring the safety of such novel therapies. Specifically, translational studies in large animal models are necessary to fully evaluate the therapeutic potential of this approach; to empirically determine the optimal combination of cell types, supplementary factors, and delivery methods to maximize efficacy; and to stringently assess safety. In the present review, we summarize the main strategies employed to generate iPSCs and differentiate them into cardiomyocytes in large animal species; the most critical differences between using small versus large animal models for cardiovascular studies; and the strategies that have been pursued regarding implanted cells' stage of differentiation, origin, and technical application.
Collapse
Affiliation(s)
- Daina Martínez-Falguera
- Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain;
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Oriol Iborra-Egea
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| |
Collapse
|
12
|
Zhang Y, He Y, Wu P, Hu S, Zhang Y, Chen C. miR-200c-141 Enhances Sheep Kidney Cell Reprogramming into Pluripotent Cells by Targeting ZEB1. Int J Stem Cells 2021; 14:423-433. [PMID: 34456193 PMCID: PMC8611307 DOI: 10.15283/ijsc21080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/31/2021] [Accepted: 06/11/2021] [Indexed: 11/19/2022] Open
Abstract
Background and Objectives Sheep-induced pluripotent stem cells (siPSCs) have low reprogramming efficiency, thereby hampering their use in biotechnology and agriculture. Several studies have shown that some microRNAs play an important role in promoting somatic reprogramming in mouse and human. In this study, we investigated the effect of miR-200c-141 on somatic reprogramming in sheep and explored the mechanism of promoting the reprogramming. Methods and Results The lentivirus system driven by tetracycline (TET)-on carrying Oct4, Sox2, c-Myc, Klf4, Nanog, Lin28, hTERT, and SV40LT (OSKMNLST) could reprogram sheep kidney cells into pluripotent cells. Overexpression of miR-200c-141 in combination with OSKMNLST could significantly improve the efficiency of sheep iPSC generation (p<0.01). Sheep iPSCs derived from miR-200c-141 showed embryonic stem cell (ESC)-like pluripotent properties, were positive for alkaline phosphatase and some pluripotent markers by quantitative real-time PCR (qRT-PCR) and immunofluorescence, and were able to differentiate into three germ layers in vitro. Oar-miR-200c was transfected into HEK293FT cells and was able to target the zinc finger E-box-binding homeobox 1 (ZEB1) 3’UTR using dual luciferase reporting analysis. Overexpression of oar-miR-200c in SKCs significantly reduced the expression of ZEB1, but increased the expression of E-cadherin by qRT-PCR and western blotting analysis. Conclusions These results suggest that miR-200c-141 can promote the reprogramming of sheep somatic cells to iPSCs, and oar-miR-200c targeted ZEB1 3’UTR, significantly decreased expression of ZEB1, and increased expression of E-cadherin. Oar-miR-200c may improve the MET process by affecting the TGF-β signaling pathway, thus improving the efficiency of somatic cell reprogramming in sheep.
Collapse
Affiliation(s)
- Yunfeng Zhang
- College of Animal Science and Technology, Shihezi University, Xinjiang, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious, Shihezi, China.,State Key Laboratory of Sheep Genetic Improvement and Healthy Production/Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | - Yanhua He
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production/Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | - Peng Wu
- College of Life Technology, Shihezi University, Xinjiang, China
| | - Shengwei Hu
- College of Life Technology, Shihezi University, Xinjiang, China
| | - Yanyan Zhang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production/Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University, Xinjiang, China.,Collaborative Innovation Center for Prevention and Control of High Incidence Zoonotic Infectious, Shihezi, China
| |
Collapse
|
13
|
Reiss J, Robertson S, Suzuki M. Cell Sources for Cultivated Meat: Applications and Considerations throughout the Production Workflow. Int J Mol Sci 2021; 22:7513. [PMID: 34299132 PMCID: PMC8307620 DOI: 10.3390/ijms22147513] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Cellular agriculture is an emerging scientific discipline that leverages the existing principles behind stem cell biology, tissue engineering, and animal sciences to create agricultural products from cells in vitro. Cultivated meat, also known as clean meat or cultured meat, is a prominent subfield of cellular agriculture that possesses promising potential to alleviate the negative externalities associated with conventional meat production by producing meat in vitro instead of from slaughter. A core consideration when producing cultivated meat is cell sourcing. Specifically, developing livestock cell sources that possess the necessary proliferative capacity and differentiation potential for cultivated meat production is a key technical component that must be optimized to enable scale-up for commercial production of cultivated meat. There are several possible approaches to develop cell sources for cultivated meat production, each possessing certain advantages and disadvantages. This review will discuss the current cell sources used for cultivated meat production and remaining challenges that need to be overcome to achieve scale-up of cultivated meat for commercial production. We will also discuss cell-focused considerations in other components of the cultivated meat production workflow, namely, culture medium composition, bioreactor expansion, and biomaterial tissue scaffolding.
Collapse
Affiliation(s)
- Jacob Reiss
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
14
|
Bessi BW, Botigelli RC, Pieri NCG, Machado LS, Cruz JB, de Moraes P, de Souza AF, Recchia K, Barbosa G, de Castro RVG, Nogueira MFG, Bressan FF. Cattle In Vitro Induced Pluripotent Stem Cells Generated and Maintained in 5 or 20% Oxygen and Different Supplementation. Cells 2021; 10:cells10061531. [PMID: 34204517 PMCID: PMC8234940 DOI: 10.3390/cells10061531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022] Open
Abstract
The event of cellular reprogramming into pluripotency is influenced by several factors, such as in vitro culture conditions (e.g., culture medium and oxygen concentration). Herein, bovine iPSCs (biPSCs) were generated in different levels of oxygen tension (5% or 20% of oxygen) and supplementation (bFGF or bFGF + LIF + 2i-bFL2i) to evaluate the efficiency of pluripotency induction and maintenance in vitro. Initial reprogramming was observed in all groups and bFL2i supplementation initially resulted in a superior number of colonies. However, bFL2i supplementation in low oxygen led to a loss of self-renewal and pluripotency maintenance. All clonal lines were positive for alkaline phosphatase; they expressed endogenous pluripotency-related genes SOX2, OCT4 and STELLA. However, expression was decreased throughout the passages without the influence of oxygen tension. GLUT1 and GLUT3 were upregulated by low oxygen. The biPSCs were immunofluorescence-positive stained for OCT4 and SOX2 and they formed embryoid bodies which differentiated in ectoderm and mesoderm (all groups), as well as endoderm (one line from bFL2i in high oxygen). Our study is the first to compare high and low oxygen environments during and after induced reprogramming in cattle. In our conditions, a low oxygen environment did not favor the pluripotency maintenance of biPSCs.
Collapse
Affiliation(s)
- Brendon Willian Bessi
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
| | - Ramon Cesar Botigelli
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
- Department of Pharmacology, Institute of Biosciences (IBB), São Paulo State University (UNESP), Botucatu 18618-689, Brazil
- Correspondence: (R.C.B.); (F.F.B.)
| | - Naira Caroline Godoy Pieri
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
- Department of Animal Reproduction, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo (USP), São Paulo 05508-270, Brazil
| | - Lucas Simões Machado
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
| | - Jessica Brunhara Cruz
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
| | - Pamela de Moraes
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
| | - Aline Fernanda de Souza
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
| | - Kaiana Recchia
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
| | - Gabriela Barbosa
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
| | - Raquel Vasconcelos Guimarães de Castro
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
- Department of Pathology, Reproduction and One Health, Faculty of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Botucatu 14884-900, Brazil
| | - Marcelo Fábio Gouveia Nogueira
- Department of Biological Science, School of Sciences, Humanities and Languages, São Paulo State University (UNESP), Assis 19806-900, Brazil;
| | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo (USP), Pirassununga 13635-000, Brazil; (B.W.B.); (N.C.G.P.); (L.S.M.); (J.B.C.); (P.d.M.); (A.F.d.S.); (K.R.); (G.B.); (R.V.G.d.C.)
- Correspondence: (R.C.B.); (F.F.B.)
| |
Collapse
|
15
|
Pain B, Baquerre C, Coulpier M. Cerebral organoids and their potential for studies of brain diseases in domestic animals. Vet Res 2021; 52:65. [PMID: 33941270 PMCID: PMC8090903 DOI: 10.1186/s13567-021-00931-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
The brain is a complex organ and any model for studying it in its normal and pathological aspects becomes a tool of choice for neuroscientists. The mastering and dissemination of protocols allowing brain organoids development have paved the way for a whole range of new studies in the field of brain development, modeling of neurodegenerative or neurodevelopmental diseases, understanding tumors as well as infectious diseases that affect the brain. While studies are so far limited to the use of human cerebral organoids, there is a growing interest in having similar models in other species. This review presents what is currently developed in this field, with a particular focus on the potential of cerebral organoids for studying neuro-infectious diseases in human and domestic animals.
Collapse
Affiliation(s)
- Bertrand Pain
- Univ Lyon, Université Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, USC1361, Bron, France.
| | - Camille Baquerre
- Univ Lyon, Université Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, USC1361, Bron, France
| | - Muriel Coulpier
- UMR1161 Virologie, Anses, INRAE, École Nationale Vétérinaire D'Alfort, Université Paris-Est, Maisons-Alfort, France
| |
Collapse
|
16
|
Abstract
Organoids are three-dimensional structures that are derived from the self-organization of stem cells as they differentiate in vitro. The plasticity of stem cells is one of the major criteria for generating organoids most similar to the tissue structures they intend to mimic. Stem cells are cells with unique properties of self-renewal and differentiation. Depending on their origin, a distinction is made between pluripotent (embryonic) stem cells (PSCs), adult (or tissue) stem cells (ASCs), and those obtained by somatic reprogramming, so-called induced pluripotent stem cells (iPSCs). While most data since the 1980s have been acquired in the mouse model, and then from the late 1990s in humans, the process of somatic reprogammation has revolutionized the field of stem cell research. For domestic animals, numerous attempts have been made to obtain PSCs and iPSCs, an approach that makes it possible to omit the use of embryos to derive the cells. Even if the plasticity of the cells obtained is not always optimal, the recent progress in obtaining reprogrammed cells is encouraging. Along with PSCs and iPSCs, many organoid derivations in animal species are currently obtained from ASCs. In this study, we present state-of-the-art stem cell research according to their origins in the various animal models developed.
Collapse
Affiliation(s)
- Bertrand Pain
- Univ Lyon, Université Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, CSC USC1361, Bron, France.
| |
Collapse
|
17
|
Li Z, Ge W, Li Y, Zhang Y, Zhao X, Hu J. Valproic Acid Enhance Reprogramming of Bactrian Camel Cells through Promoting the Expression of Endogenous Gene c-Myc and the Process of Angiogenesis. Int J Stem Cells 2021; 14:191-202. [PMID: 33632993 PMCID: PMC8138656 DOI: 10.15283/ijsc20213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 11/30/2022] Open
Abstract
Background and Objectives Induced pluripotent stem cells (iPSCs) are usually generated by reprogramming differentiated cells through the introduction of specific transcription factors, but this is a difficult and inefficient process. Valproic acid (VPA) is a histone deacetylase inhibitor that significantly improves the efficiency of iPSC generation. But its role and mechanism are still unclear. Methods and Results We transduced Bactrian camel fetal fibroblasts (BCFFs) with retroviruses carrying defined factors (OCT4, SOX2, KLF4, c-MYC and EGFP; OSKMG) in the presence of VPA. Cells were collected (Day 7) and analyzed using RNA-seq technology. Afterwards, different groups of cells and transcriptomics results were detected by PCR and qRT-PCR technology. The results showed that VPA promoted the expression of the endogenous gene c-Myc and inhibited cell proliferation; at the same time, it promoted the expression of VEGF and other genes related to angiogenesis. Conclusions When VPA is added to the culture medium, only the cells that have begun to reprogram can break the G2/M repression through the expression of the endogenous gene c-Myc, and use the nutrients and space in the culture dish to proliferate normally, which can achieve the purpose of directly improving the efficiency of reprogramming. Another new discovery for Bactrian camels, VPA significantly increased the expression of VEGFC and other genes, promoting the transformation of fibroblasts to endothelial cells (different from the mesenchymal-to-epithelial transition process of other species) to accelerate the early induction of Bactrian camels iPSc process. Overall, this study proved the new mechanism of VPA in enhancing the induction of pluripotency from the transcriptome level.
Collapse
Affiliation(s)
- Zongshuai Li
- Department of Veterinary Obstetrics, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Wenbo Ge
- Department of Veterinary Obstetrics, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yina Li
- Department of Veterinary Obstetrics, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yong Zhang
- Department of Veterinary Obstetrics, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xingxu Zhao
- Department of Veterinary Obstetrics, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Junjie Hu
- Department of Veterinary Obstetrics, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
18
|
Su Y, Zhu J, Salman S, Tang Y. Induced pluripotent stem cells from farm animals. J Anim Sci 2021; 98:5937369. [PMID: 33098420 DOI: 10.1093/jas/skaa343] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
The development of the induced pluripotent stem cells (iPSCs) technology has revolutionized the world on the establishment of pluripotent stem cells (PSCs) across a great variety of animal species. Generation of iPSCs from domesticated animals would provide unrestricted cell resources for the study of embryonic development and cell differentiation of these species, for screening and establishing desired traits for sustainable agricultural production, and as veterinary and preclinical therapeutic tools for animal and human diseases. Induced PSCs from domesticated animals thus harbor enormous scientific, economical, and societal values. Although much progress has been made toward the generation of PSCs from these species, major obstacles remain precluding the exclamation of the establishment of bona fide iPSCs. The most prominent of them remain the inability of these cells to silence exogenous reprogramming factors, the obvious reliance on exogenous factors for their self-renewal, and the restricted development potential in vivo. In this review, we summarize the history and current progress in domestic farm animal iPSC generation, with a focus on swine, ruminants (cattle, ovine, and caprine), horses, and avian species (quails and chickens). We also discuss the problems associated with the farm animal iPSCs and potential future directions toward the complete reprogramming of somatic cells from farm animals.
Collapse
Affiliation(s)
- Yue Su
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Jiaqi Zhu
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Saleh Salman
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Young Tang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT
| |
Collapse
|
19
|
Kumar D, Talluri TR, Selokar NL, Hyder I, Kues WA. Perspectives of pluripotent stem cells in livestock. World J Stem Cells 2021; 13:1-29. [PMID: 33584977 PMCID: PMC7859985 DOI: 10.4252/wjsc.v13.i1.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The recent progress in derivation of pluripotent stem cells (PSCs) from farm animals opens new approaches not only for reproduction, genetic engineering, treatment and conservation of these species, but also for screening novel drugs for their efficacy and toxicity, and modelling of human diseases. Initial attempts to derive PSCs from the inner cell mass of blastocyst stages in farm animals were largely unsuccessful as either the cells survived for only a few passages, or lost their cellular potency; indicating that the protocols which allowed the derivation of murine or human embryonic stem (ES) cells were not sufficient to support the maintenance of ES cells from farm animals. This scenario changed by the innovation of induced pluripotency and by the development of the 3 inhibitor culture conditions to support naïve pluripotency in ES cells from livestock species. However, the long-term culture of livestock PSCs while maintaining the full pluripotency is still challenging, and requires further refinements. Here, we review the current achievements in the derivation of PSCs from farm animals, and discuss the potential application areas.
Collapse
Affiliation(s)
- Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India.
| | - Thirumala R Talluri
- Equine Production Campus, ICAR-National Research Centre on Equines, Bikaner 334001, India
| | - Naresh L Selokar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India
| | - Iqbal Hyder
- Department of Physiology, NTR College of Veterinary Science, Gannavaram 521102, India
| | - Wilfried A Kues
- Department of Biotechnology, Friedrich-Loeffler-Institute, Federal Institute of Animal Health, Neustadt 31535, Germany
| |
Collapse
|
20
|
Scarfone RA, Pena SM, Russell KA, Betts DH, Koch TG. The use of induced pluripotent stem cells in domestic animals: a narrative review. BMC Vet Res 2020; 16:477. [PMID: 33292200 PMCID: PMC7722595 DOI: 10.1186/s12917-020-02696-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are undifferentiated stem cells characterized by the ability to differentiate into any cell type in the body. iPSCs are a relatively new and rapidly developing technology in many fields of biology, including developmental anatomy and physiology, pathology, and toxicology. These cells have great potential in research as they are self-renewing and pluripotent with minimal ethical concerns. Protocols for their production have been developed for many domestic animal species, which have since been used to further our knowledge in the progression and treatment of diseases. This research is valuable both for veterinary medicine as well as for the prospect of translation to human medicine. Safety, cost, and feasibility are potential barriers for this technology that must be considered before widespread clinical adoption. This review will analyze the literature pertaining to iPSCs derived from various domestic species with a focus on iPSC production and characterization, applications for tissue and disease research, and applications for disease treatment.
Collapse
Affiliation(s)
- Rachel A Scarfone
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Samantha M Pena
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Keith A Russell
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Dean H Betts
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Thomas G Koch
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada.
| |
Collapse
|
21
|
Endo Y, Kamei KI, Inoue-Murayama M. Genetic Signatures of Evolution of the Pluripotency Gene Regulating Network across Mammals. Genome Biol Evol 2020; 12:1806-1818. [PMID: 32780791 PMCID: PMC7643368 DOI: 10.1093/gbe/evaa169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2020] [Indexed: 01/01/2023] Open
Abstract
Mammalian pluripotent stem cells (PSCs) have distinct molecular and biological characteristics among species, but to date we lack a comprehensive understanding of regulatory network evolution in mammals. Here, we carried out a comparative genetic analysis of 134 genes constituting the pluripotency gene regulatory network across 48 mammalian species covering all the major taxonomic groups. We report that mammalian genes in the pluripotency regulatory network show a remarkably high degree of evolutionary stasis, suggesting the conservation of fundamental biological process of mammalian PSCs across species. Nevertheless, despite the overall conservation of the regulatory network, we discovered rapid evolution of the downstream targets of the core regulatory elements and specific amino acid residues that have undergone positive selection. Our data indicate development of lineage-specific pluripotency regulating networks that may explain observed variations in some characteristics of mammalian PSCs. We further revealed that positively selected genes could be associated with species' unique adaptive characteristics that were not dedicated to regulation of PSCs. These results provide important insight into the evolution of the pluripotency gene regulatory network underlying variations in characteristics of mammalian PSCs.
Collapse
Affiliation(s)
| | - Ken-ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Japan
| | - Miho Inoue-Murayama
- Wildlife Research Center, Kyoto University, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Japan
- Wildlife Genome Collaborative Research Group, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan
| |
Collapse
|
22
|
Navarro M, Soto DA, Pinzon CA, Wu J, Ross PJ. Livestock pluripotency is finally captured in vitro. Reprod Fertil Dev 2020; 32:11-39. [PMID: 32188555 DOI: 10.1071/rd19272] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pluripotent stem cells (PSCs) have demonstrated great utility in improving our understanding of mammalian development and continue to revolutionise regenerative medicine. Thanks to the improved understanding of pluripotency in mice and humans, it has recently become feasible to generate stable livestock PSCs. Although it is unlikely that livestock PSCs will be used for similar applications as their murine and human counterparts, new exciting applications that could greatly advance animal agriculture are being developed, including the use of PSCs for complex genome editing, cellular agriculture, gamete generation and invitro breeding schemes.
Collapse
Affiliation(s)
- Micaela Navarro
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA 95616, USA
| | - Delia A Soto
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA 95616, USA
| | - Carlos A Pinzon
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Pablo J Ross
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA 95616, USA; and Corresponding author.
| |
Collapse
|
23
|
Kumar D, Anand T, Talluri TR, Kues WA. Potential of transposon-mediated cellular reprogramming towards cell-based therapies. World J Stem Cells 2020; 12:527-544. [PMID: 32843912 PMCID: PMC7415244 DOI: 10.4252/wjsc.v12.i7.527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/09/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem (iPS) cells present a seminal discovery in cell biology and promise to support innovative treatments of so far incurable diseases. To translate iPS technology into clinical trials, the safety and stability of these reprogrammed cells needs to be shown. In recent years, different non-viral transposon systems have been developed for the induction of cellular pluripotency, and for the directed differentiation into desired cell types. In this review, we summarize the current state of the art of different transposon systems in iPS-based cell therapies.
Collapse
Affiliation(s)
- Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar 125001, India.
| | - Taruna Anand
- NCVTC, ICAR-National Research Centre on Equines, Hisar 125001, India
| | - Thirumala R Talluri
- Equine Production Campus, ICAR-National Research Centre on Equines, Bikaner 334001, India
| | - Wilfried A Kues
- Friedrich-Loeffler-Institut, Institute of Farm Animal Genetics, Department of Biotechnology, Mariensee 31535, Germany
| |
Collapse
|
24
|
Bressan FF, Bassanezze V, de Figueiredo Pessôa LV, Sacramento CB, Malta TM, Kashima S, Fantinato Neto P, Strefezzi RDF, Pieri NCG, Krieger JE, Covas DT, Meirelles FV. Generation of induced pluripotent stem cells from large domestic animals. Stem Cell Res Ther 2020; 11:247. [PMID: 32586372 PMCID: PMC7318412 DOI: 10.1186/s13287-020-01716-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/23/2020] [Accepted: 05/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) have enormous potential in developmental biology studies and in cellular therapies. Although extensively studied and characterized in human and murine models, iPSCs from animals other than mice lack reproducible results. METHODS Herein, we describe the generation of robust iPSCs from equine and bovine cells through lentiviral transduction of murine or human transcription factors Oct4, Sox2, Klf4, and c-Myc and from human and murine cells using similar protocols, even when different supplementations were used. The iPSCs were analyzed regarding morphology, gene and protein expression of pluripotency factors, alkaline phosphatase detection, and spontaneous and induced differentiation. RESULTS Although embryonic-derived stem cells are yet not well characterized in domestic animals, generation of iPS cells from these species is possible through similar protocols used for mouse or human cells, enabling the use of pluripotent cells from large animals for basic or applied purposes. Herein, we also infer that bovine iPS (biPSCs) exhibit similarity to mouse iPSCs (miPSCs), whereas equine iPSs (eiPSCs) to human (hiPSCs). CONCLUSIONS The generation of reproducible protocols in different animal species will provide an informative tool for producing in vitro autologous pluripotent cells from domestic animals. These cells will create new opportunities in animal breeding through transgenic technology and will support a new era of translational medicine with large animal models.
Collapse
Affiliation(s)
- Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
- Postgraduate Program in Anatomy of Domestic and Wild Animals, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
- Center for Cell-Based Therapy, Regional Blood Center, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Vinícius Bassanezze
- Heart Institute (INCOR), Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Present Address: Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Laís Vicari de Figueiredo Pessôa
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
- Postgraduate Program in Anatomy of Domestic and Wild Animals, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Chester Bittencourt Sacramento
- Heart Institute (INCOR), Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Present Address: Weill Cornell Medicine, Cornell University, Ithaca, USA
| | - Tathiane Maistro Malta
- Center for Cell-Based Therapy, Regional Blood Center, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP Brazil
| | - Simone Kashima
- Center for Cell-Based Therapy, Regional Blood Center, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Paulo Fantinato Neto
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
- Postgraduate Program in Anatomy of Domestic and Wild Animals, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ricardo De Francisco Strefezzi
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Naira Caroline Godoy Pieri
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - José Eduardo Krieger
- Heart Institute (INCOR), Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-Based Therapy, Regional Blood Center, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Flávio Vieira Meirelles
- Department of Veterinary Medicine, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
- Postgraduate Program in Anatomy of Domestic and Wild Animals, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
- Center for Cell-Based Therapy, Regional Blood Center, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
25
|
Moradi S, Mahdizadeh H, Šarić T, Kim J, Harati J, Shahsavarani H, Greber B, Moore JB. Research and therapy with induced pluripotent stem cells (iPSCs): social, legal, and ethical considerations. Stem Cell Res Ther 2019; 10:341. [PMID: 31753034 PMCID: PMC6873767 DOI: 10.1186/s13287-019-1455-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 10/04/2019] [Accepted: 10/15/2019] [Indexed: 02/08/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) can self-renew indefinitely in culture and differentiate into all specialized cell types including gametes. iPSCs do not exist naturally and are instead generated (“induced” or “reprogrammed”) in culture from somatic cells through ectopic co-expression of defined pluripotency factors. Since they can be generated from any healthy person or patient, iPSCs are considered as a valuable resource for regenerative medicine to replace diseased or damaged tissues. In addition, reprogramming technology has provided a powerful tool to study mechanisms of cell fate decisions and to model human diseases, thereby substantially potentiating the possibility to (i) discover new drugs in screening formats and (ii) treat life-threatening diseases through cell therapy-based strategies. However, various legal and ethical barriers arise when aiming to exploit the full potential of iPSCs to minimize abuse or unauthorized utilization. In this review, we discuss bioethical, legal, and societal concerns associated with research and therapy using iPSCs. Furthermore, we present key questions and suggestions for stem cell scientists, legal authorities, and social activists investigating and working in this field.
Collapse
Affiliation(s)
- Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, 47138-18983, Babol, Iran.
| | - Hamid Mahdizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for NeurophysiologyMedical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Johnny Kim
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Javad Harati
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Hosein Shahsavarani
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran.,Department of Cellular and Molecular Sciences, Faculty of Bioscience and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Boris Greber
- RheinCell Therapeutics GmbH, 40764, Langenfeld, Germany
| | - Joseph B Moore
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA.,The Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY, USA
| |
Collapse
|
26
|
Cong X, Zhang SM, Ellis MW, Luo J. Large Animal Models for the Clinical Application of Human Induced Pluripotent Stem Cells. Stem Cells Dev 2019; 28:1288-1298. [PMID: 31359827 DOI: 10.1089/scd.2019.0136] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology offers a practically infinite and ethically acceptable source to obtain a variety of somatic cells. Coupled with the biotechnologies of cell therapy or tissue engineering, iPSC technology will enormously contribute to human regenerative medicine. Before clinical application, such human iPSC (hiPSC)-based therapies should be assessed using large animal models that more closely match biological or biomechanical properties of human patients. Therefore, it is critical to generate large animal iPSCs, obtain their iPSC-derived somatic cells, and preclinically evaluate their therapeutic efficacy and safety in large animals. During the past decade, the establishment of iPSC lines of a series of large animal species has been documented, and the acquisition and preclinical evaluation of iPSC-derived somatic cells has also been reported. Despite this progress, significant obstacles, such as obtaining or preserving the bona fide pluripotency of large animal iPSCs, have been encountered. Simultaneously, studies of large animal iPSCs have been overlooked in comparison with those of mouse and hiPSCs, and this field deserves more attention and support due to its important preclinical relevance. Herein, this review will focus on the large animal models of pigs, dogs, horses, and sheep/goats, and summarize current progress, challenges, and potential future directions of research on large animal iPSCs.
Collapse
Affiliation(s)
- Xiaoqiang Cong
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut.,Department of Cardiology, Bethune First Hospital of Jilin University, Changchun, China
| | - Shang-Min Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Matthew W Ellis
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut.,Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
| | - Jiesi Luo
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut.,Yale Stem Cell Center, New Haven, Connecticut
| |
Collapse
|
27
|
Pessôa LVDF, Bressan FF, Freude KK. Induced pluripotent stem cells throughout the animal kingdom: Availability and applications. World J Stem Cells 2019; 11:491-505. [PMID: 31523369 PMCID: PMC6716087 DOI: 10.4252/wjsc.v11.i8.491] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023] Open
Abstract
Up until the mid 2000s, the capacity to generate every cell of an organism was exclusive to embryonic stem cells. In 2006, researchers Takahashi and Yamanaka developed an alternative method of generating embryonic-like stem cells from adult cells, which they coined induced pluripotent stem cells (iPSCs). Such iPSCs possess most of the advantages of embryonic stem cells without the ethical stigma associated with derivation of the latter. The possibility of generating “custom-made” pluripotent cells, ideal for patient-specific disease models, alongside their possible applications in regenerative medicine and reproduction, has drawn a lot of attention to the field with numbers of iPSC studies published growing exponentially. IPSCs have now been generated for a wide variety of species, including but not limited to, mouse, human, primate, wild felines, bovines, equines, birds and rodents, some of which still lack well-established embryonic stem cell lines. The paucity of robust characterization of some of these iPSC lines as well as the residual expression of transgenes involved in the reprogramming process still hampers the use of such cells in species preservation or medical research, underscoring the requirement for further investigations. Here, we provide an extensive overview of iPSC generated from a broad range of animal species including their potential applications and limitations.
Collapse
Affiliation(s)
- Laís Vicari de Figueiredo Pessôa
- Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-000, São Paulo, Brazil
| | - Kristine Karla Freude
- Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| |
Collapse
|
28
|
Augustyniak J, Bertero A, Coccini T, Baderna D, Buzanska L, Caloni F. Organoids are promising tools for species-specific in vitro toxicological studies. J Appl Toxicol 2019; 39:1610-1622. [PMID: 31168795 DOI: 10.1002/jat.3815] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/05/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022]
Abstract
Organoids are three-dimensional self-aggregating structures generated from stem cells (SCs) or progenitor cells in a process that recapitulates molecular and cellular stages of early organ development. The differentiation process leads to the appearance of specialized mature cells and is connected with changes in the organoid internal structure rearrangement and self-organization. The formation of organ-specific structures in vitro with highly ordered architecture is also strongly influenced by the extracellular matrix. These features make organoids as a powerful model for in vitro toxicology. Nowadays this technology is developing very quickly. In this review we present, from a toxicological and species-specific point of view, the state of the art of organoid generation from adult SCs and pluripotent SCs: embryonic SCs or induced pluripotent SCs. The current culture organoid techniques are discussed for their main advantages, disadvantages and limitations. In the second part of the review, we concentrated on the characterization of species-specific organoids generated from tissue-specific SCs of different sources: mammary (bovine), epidermis (canine), intestinal (porcine, bovine, canine, chicken) and liver (feline, canine).
Collapse
Affiliation(s)
- Justyna Augustyniak
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Alessia Bertero
- Department of Veterinary Medicine (DIMEVET), Università degli Studi di Milano, Milan, Italy
| | - Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, Toxicology Unit, ICS Maugeri SpA-SB, IRCCS Pavia, Pavia, Italy
| | - Diego Baderna
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Italy
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Francesca Caloni
- Department of Veterinary Medicine (DIMEVET), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
29
|
Barboni B, Russo V, Berardinelli P, Mauro A, Valbonetti L, Sanyal H, Canciello A, Greco L, Muttini A, Gatta V, Stuppia L, Mattioli M. Placental Stem Cells from Domestic Animals: Translational Potential and Clinical Relevance. Cell Transplant 2019; 27:93-116. [PMID: 29562773 PMCID: PMC6434480 DOI: 10.1177/0963689717724797] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The field of regenerative medicine is moving toward clinical practice in veterinary science. In this context, placenta-derived stem cells isolated from domestic animals have covered a dual role, acting both as therapies for patients and as a valuable cell source for translational models. The biological properties of placenta-derived cells, comparable among mammals, make them attractive candidates for therapeutic approaches. In particular, stemness features, low immunogenicity, immunomodulatory activity, multilineage plasticity, and their successful capacity for long-term engraftment in different host tissues after autotransplantation, allo-transplantation, or xenotransplantation have been demonstrated. Their beneficial regenerative effects in domestic animals have been proven using preclinical studies as well as clinical trials starting to define the mechanisms involved. This is, in particular, for amniotic-derived cells that have been thoroughly studied to date. The regenerative role arises from a mutual tissue-specific cell differentiation and from the paracrine secretion of bioactive molecules that ultimately drive crucial repair processes in host tissues (e.g., anti-inflammatory, antifibrotic, angiogenic, and neurogenic factors). The knowledge acquired so far on the mechanisms of placenta-derived stem cells in animal models represent the proof of concept of their successful use in some therapeutic treatments such as for musculoskeletal disorders. In the next future, legislation in veterinary regenerative medicine will be a key element in order to certify those placenta-derived cell-based protocols that have already demonstrated their safety and efficacy using rigorous approaches and to improve the degree of standardization of cell-based treatments among veterinary clinicians.
Collapse
Affiliation(s)
- B Barboni
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Russo
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - P Berardinelli
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Mauro
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Valbonetti
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - H Sanyal
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Canciello
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Greco
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Muttini
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Gatta
- 1 Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - L Stuppia
- 2 Medical Genetics, University "G. d'Annunzio" of Chieti Pescara, Chieti, Italy
| | - M Mattioli
- 3 Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale," Teramo, Italy
| |
Collapse
|
30
|
Kim SJ, Kwon HS, Kwon DK, Koo OJ, Moon JH, Park EJ, Yum SY, Lee BC, Jang G. Production of Transgenic Porcine Embryos Reconstructed with Induced Pluripotent Stem-Like Cells Derived from Porcine Endogenous Factors Using piggyBac System. Cell Reprogram 2019; 21:26-36. [DOI: 10.1089/cell.2018.0036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Su-Jin Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Hee-Sun Kwon
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Dae-kee Kwon
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | | | - Joon-Ho Moon
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Eun-Jung Park
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Soo-Young Yum
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Byeong-Chun Lee
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Goo Jang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Research Institute of Veterinary Science, Seoul National University, Seoul, Republic of Korea
- BK21 Plus program, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Emergence Center for Food-Medicine Personalized Therapy System, Advanced Institutes of Convergence Technology, Seoul National University, Gyeonggi-do, Republic of Korea
| |
Collapse
|
31
|
Li D, Secher J, Hyttel P, Ivask M, Kolko M, Hall VJ, Freude KK. Generation of transgene-free porcine intermediate type induced pluripotent stem cells. Cell Cycle 2018; 17:2547-2563. [PMID: 30457474 DOI: 10.1080/15384101.2018.1548790] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Physiologically and anatomically, humans and pigs share many similarities, which make porcine induced pluripotent stem cells (piPSCs) very attractive for modeling human cell therapy as well as for testing safety of iPSC based cell replacement therapies. To date, several integrative and non-integrative strategies have been reported to successfully generate piPSCs, but all resulting piPSCs had integration of transgenes. The use of integrative methods has the disadvantage of potential lack of silencing or inappropriate re-activation of these genes during differentiation, as well as uncertainty regarding disruption of important genomic regions caused by integration. In our study, we performed a non-integrative vector based reprogramming approach using porcine fetal fibroblasts. The resulting four piPSC lines were positive for pluripotency marker and when subjected to in vitro and in vivo differentiation assays, all four lines formed embryoid bodies, capable to differentiate into all three germ layers, and three out of the four cell lines formed teratomas. PCR analysis on genomic and plasmid DNA revealed that the episomal vectors were undetectable in six out of eight subclones derived from one of the piPSC lines (piPSC1) above passage 20. These piPSCs could potentially be ideal cell lines for the generation of porcine in vitro and in vivo models. Furthermore, subsequent analyses of our new transgene independent piPSCs could provide novel insights on the genetic and epigenetic necessities to achieve and maintain piPSCs.
Collapse
Affiliation(s)
- Dong Li
- a Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Frederiksberg C , Denmark
| | - Jan Secher
- b Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Taastrup , Denmark
| | - Poul Hyttel
- a Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Frederiksberg C , Denmark
| | - Marilin Ivask
- c Institute of Biomedicine and Translational Medicine , University of Tartu , Tartu , Estonia.,d Institute of Veterinary Medicine and Animal Sciences , Estonian University of Life Sciences , Tartu , Estonia
| | - Miriam Kolko
- e Department of Drug Design and Pharmacology , University of Copenhagen , Copenhagen O , Denmark.,f Department of Ophthalmology , Rigshospital-Glostrup , Glostrup , Denmark
| | - Vanessa Jane Hall
- a Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Frederiksberg C , Denmark
| | - Kristine K Freude
- a Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Frederiksberg C , Denmark
| |
Collapse
|
32
|
Gugjoo MB, Amarpal. Mesenchymal stem cell research in sheep: Current status and future prospects. Small Rumin Res 2018. [DOI: 10.1016/j.smallrumres.2018.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
NANOG Is Required for the Long-Term Establishment of Avian Somatic Reprogrammed Cells. Stem Cell Reports 2018; 11:1272-1286. [PMID: 30318291 PMCID: PMC6235669 DOI: 10.1016/j.stemcr.2018.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 01/16/2023] Open
Abstract
Somatic reprogramming, which was first identified in rodents, remains poorly described in non-mammalian species. Here, we generated avian reprogrammed cells by reprogramming of chicken and duck primary embryonic fibroblasts. The efficient generation of long-term proliferating cells depends on the method of delivery of reprogramming factors and the addition of NANOG and LIN28 to the canonical OCT4, SOX2, KLF4, and c-MYC gene combination. The reprogrammed cells were positive for several key pluripotency-associated markers including alkaline phosphatase activity, telomerase activity, SSEA1 expression, and specific cell cycle and epigenetic markers. Upregulated endogenous pluripotency-associated genes included POU5F3 (POUV) and KLF4, whereas cells failed to upregulate NANOG and LIN28A. However, cells showed a tumorigenic propensity when injected into recipient embryos. In conclusion, although the somatic reprogramming process is active in avian primary cells, it needs to be optimized to obtain fully reprogrammed cells with similar properties to those of chicken embryonic stem cells. NANOG is required for avian somatic reprogramming NANOG is necessary for long-term establishment of avian reprogrammed cells Avian reprogrammed cells express pluripotency markers Avian cells are only partially reprogrammed
Collapse
|
34
|
Saadeldin IM, Abdel-Aziz Swelum A, Alzahrani FA, Alowaimer AN. The current perspectives of dromedary camel stem cells research. Int J Vet Sci Med 2018; 6:S27-S30. [PMID: 30761317 PMCID: PMC6161867 DOI: 10.1016/j.ijvsm.2018.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/06/2018] [Indexed: 12/17/2022] Open
Abstract
Camels have cultural value in the Arab society and are considered one of the most important animals in the Arabian Peninsula and arid environments, due to the distinct characteristics of their meat and milk. Moreover, there is a great interest in camel racing and beauty shows. Therefore, treatment of elite animals, increasing the number of camels as well as genetic improvement is an essential demand. Because there are unique camels for milk production, meat, or in racing, the need to propagate genetically superior camels is urgent. Recent biotechnological approaches such as stem cells hold great promise for biomedical research, genetic engineering, and as a model for studying early mammalian developmental biology. Establishment of stem cells lines from camels would tremendously facilitate regenerative medicine for genetically superior camels, permit the gene targeting of the camel genome and the generation of genetically modified animal and be a mean for genome conservation for the elite breeds. In this mini-review, we show the current research, future horizons and potential applications for camel stem cells.
Collapse
Affiliation(s)
- Islam M Saadeldin
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451 Riyadh, Saudi Arabia.,Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Ayman Abdel-Aziz Swelum
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451 Riyadh, Saudi Arabia.,Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Faisal A Alzahrani
- Department of Biological Sciences, Rabigh College of Science and Arts, King Abdulaziz University, Rabigh Branch, Rabigh 21911, Saudi Arabia
| | - Abdullah N Alowaimer
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451 Riyadh, Saudi Arabia
| |
Collapse
|
35
|
Kehl D, Generali M, Görtz S, Geering D, Slamecka J, Hoerstrup SP, Bleul U, Weber B. Amniotic Fluid Cells Show Higher Pluripotency-Related Gene Expression Than Allantoic Fluid Cells. Stem Cells Dev 2017; 26:1424-1437. [DOI: 10.1089/scd.2016.0352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Debora Kehl
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Melanie Generali
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Sabrina Görtz
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Diego Geering
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
| | - Jaroslav Slamecka
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Ulrich Bleul
- Clinic of Reproductive Medicine, Department of Farm Animals, Vetsuisse-Faculty University of Zurich, Zurich, Switzerland
| | - Benedikt Weber
- Institute for Regenerative Medicine (IREM), Center for Therapy Development and Good Manufacturing Practice, University of Zurich, Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Wu Y, Li O, He C, Li Y, Li M, Liu XL, Wang Y, He Y. Generation and characterization of induced pluripotent stem cells from guinea pig fetal fibroblasts. Mol Med Rep 2017; 15:3690-3698. [PMID: 28393187 PMCID: PMC5436227 DOI: 10.3892/mmr.2017.6431] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/09/2017] [Indexed: 12/18/2022] Open
Abstract
Induced pluripotent stem cells (iPS) represent an important tool to develop disease-modeling assays, drug testing assays and cell-based replacement therapies. The application of iPS in these fields requires the development of suitable animal models. Of the suitable species, guinea pigs are particularly important and offer significant advantages. Successful iPS generation has been accomplished in a number of species; however, it has not been reported in the guinea pig. The present study successfully generated iPS from guinea pigs (giPS) using single polycistronic virus transduction with mouse octamer-binding transcription factor 4 (Oct4), sex determining region Y-box 2 (Sox2), Kruppel-like factor 4 and c-Myc. The giPS cell lines were cultured in media containing leukemia inhibitory factor and guinea pig fibroblast cells were used as feeder cells. These cultures were expanded under feeder-free culture conditions using ESGRO Complete Plus Clonal Grade medium containing 15% fetal bovine serum on gelatin-coated dishes. The resultant cells had a normal karyotype, exhibited alkaline phosphatase activity and expressed the pluripotency markers Oct4, Sox2 and Nanog. The cells differentiated in vivo to form teratomas that contained all three germ layers of the tissue cells. The generation of giPS may facilitate future studies investigating the mechanisms underlying innate immunity, particularly for tuberculosis. These experiments provide proof of principle that iPS technology may be adapted to use the guinea pig as a model of human diseases.
Collapse
Affiliation(s)
- Yuehong Wu
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Ouyang Li
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Chengwen He
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Department of Biochemistry and Molecular Biology, College of Life Science, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Yong Li
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Department of Biochemistry and Molecular Biology, College of Life Science, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Min Li
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Department of Biochemistry and Molecular Biology, College of Life Science, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Xiaoming Liu Liu
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Department of Biochemistry and Molecular Biology, College of Life Science, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Department of Biochemistry and Molecular Biology, College of Life Science, Ningxia University, Yinchuan, Ningxia 750021, P.R. China
| | - Yulong He
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| |
Collapse
|
37
|
Wei Q, Xi Q, Liu X, Meng K, Zhao X, Ma B. Characterization of goat inner cell mass derived cells in double kinase inhibition condition. Biochem Biophys Res Commun 2017; 483:325-331. [DOI: 10.1016/j.bbrc.2016.12.144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 01/01/2023]
|
38
|
Song H, Li H, Huang M, Xu D, Wang Z, Wang F. Big Animal Cloning Using Transgenic Induced Pluripotent Stem Cells: A Case Study of Goat Transgenic Induced Pluripotent Stem Cells. Cell Reprogram 2016; 18:37-47. [PMID: 26836033 DOI: 10.1089/cell.2015.0035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Using of embryonic stem cells (ESCs) could improve production traits and disease resistance by improving the efficiency of somatic cell nuclear transfer (SCNT) technology. However, robust ESCs have not been established from domestic ungulates. In the present study, we generated goat induced pluripotent stem cells (giPSCs) and transgenic cloned dairy goat induced pluripotent stem cells (tgiPSCs) from dairy goat fibroblasts (gFs) and transgenic cloned dairy goat fibroblasts (tgFs), respectively, using lentiviruses that contained hOCT4, hSOX2, hMYC, and hKLF4 without chemical compounds. The giPSCs and tgiPSCs expressed endogenous pluripotent markers, including OCT4, SOX2, MYC, KLF4, and NANOG. Moreover, they were able to maintain a normal karyotype and differentiate into derivatives from all three germ layers in vitro and in vivo. Using SCNT, tgFs and tgiPSCs were used as donor cells to produce embryos, which were named tgF-Embryos and tgiPSC-Embryos. The fusion rates and cleavage rates had no significant differences between tgF-Embryos and tgiPSC-Embryos. However, the expression of IGF-2, which is an important gene associated with embryonic development, was significantly lower in tgiPSC-Embryos than in tgF-Embryos and was not significantly different from vivo-Embryos.
Collapse
Affiliation(s)
- Hui Song
- 1 Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University , Nanjing, 210095, P.R. China .,2 Department of Medical Genetic and Cell Biology, Ningxia Medical University , Yinchuan, 750004, China
| | - Hui Li
- 1 Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University , Nanjing, 210095, P.R. China .,2 Department of Medical Genetic and Cell Biology, Ningxia Medical University , Yinchuan, 750004, China
| | - Mingrui Huang
- 1 Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University , Nanjing, 210095, P.R. China
| | - Dan Xu
- 3 Stanford University School of Medicine , Stanford, CA, 94305
| | - Ziyu Wang
- 1 Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University , Nanjing, 210095, P.R. China
| | - Feng Wang
- 1 Jiangsu Engineering Technology Research Center of Meat Sheep & Goat Industry, Nanjing Agricultural University , Nanjing, 210095, P.R. China
| |
Collapse
|
39
|
Singh VK, Saini A, Kalsan M, Kumar N, Chandra R. Describing the Stem Cell Potency: The Various Methods of Functional Assessment and In silico Diagnostics. Front Cell Dev Biol 2016; 4:134. [PMID: 27921030 PMCID: PMC5118841 DOI: 10.3389/fcell.2016.00134] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Stem cells are defined by their capabilities to self-renew and give rise to various types of differentiated cells depending on their potency. They are classified as pluripotent, multipotent, and unipotent as demonstrated through their potential to generate the variety of cell lineages. While pluripotent stem cells may give rise to all types of cells in an organism, Multipotent and Unipotent stem cells remain restricted to the particular tissue or lineages. The potency of these stem cells can be defined by using a number of functional assays along with the evaluation of various molecular markers. These molecular markers include diagnosis of transcriptional, epigenetic, and metabolic states of stem cells. Many reports are defining the particular set of different functional assays, and molecular marker used to demonstrate the developmental states and functional capacities of stem cells. The careful evaluation of all these methods could help in generating standard identifying procedures/markers for them.
Collapse
Affiliation(s)
- Vimal K Singh
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Manisha Kalsan
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Neeraj Kumar
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi Delhi, India
| |
Collapse
|
40
|
Ogorevc J, Orehek S, Dovč P. Cellular reprogramming in farm animals: an overview of iPSC generation in the mammalian farm animal species. J Anim Sci Biotechnol 2016; 7:10. [PMID: 26900466 PMCID: PMC4761155 DOI: 10.1186/s40104-016-0070-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/11/2016] [Indexed: 12/19/2022] Open
Abstract
Establishment of embryonic stem cell (ESC) lines has been successful in mouse and human, but not in farm animals. Development of direct reprogramming technology offers an alternative approach for generation of pluripotent stem cells, applicable also in farm animals. Induced pluripotent stem cells (iPSCs) represent practically limitless, ethically acceptable, individuum-specific source of pluripotent cells that can be generated from different types of somatic cells. iPSCs can differentiate to all cell types of an organism’s body and have a tremendous potential for numerous applications in medicine, agriculture, and biotechnology. However, molecular mechanisms behind the reprogramming process remain largely unknown and hamper generation of bona fide iPSCs and their use in human clinical practice. Large animal models are essential to expand the knowledge obtained on rodents and facilitate development and validation of transplantation therapies in preclinical studies. Additionally, transgenic animals with special traits could be generated from genetically modified pluripotent cells, using advanced reproduction techniques. Despite their applicative potential, it seems that iPSCs in farm animals haven’t received the deserved attention. The aim of this review was to provide a systematic overview on iPSC generation in the most important mammalian farm animal species (cattle, pig, horse, sheep, goat, and rabbit), compare protein sequence similarity of pluripotency-related transcription factors in different species, and discuss potential uses of farm animal iPSCs. Literature mining revealed 32 studies, describing iPSC generation in pig (13 studies), cattle (5), horse (5), sheep (4), goat (3), and rabbit (2) that are summarized in a concise, tabular format.
Collapse
Affiliation(s)
- J Ogorevc
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domžale, Slovenia
| | - S Orehek
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domžale, Slovenia
| | - P Dovč
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domžale, Slovenia
| |
Collapse
|
41
|
Pluripotent stem cells and livestock genetic engineering. Transgenic Res 2016; 25:289-306. [PMID: 26894405 DOI: 10.1007/s11248-016-9929-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/06/2016] [Indexed: 01/12/2023]
Abstract
The unlimited proliferative ability and capacity to contribute to germline chimeras make pluripotent embryonic stem cells (ESCs) perfect candidates for complex genetic engineering. The utility of ESCs is best exemplified by the numerous genetic models that have been developed in mice, for which such cells are readily available. However, the traditional systems for mouse genetic engineering may not be practical for livestock species, as it requires several generations of mating and selection in order to establish homozygous founders. Nevertheless, the self-renewal and pluripotent characteristics of ESCs could provide advantages for livestock genetic engineering such as ease of genetic manipulation and improved efficiency of cloning by nuclear transplantation. These advantages have resulted in many attempts to isolate livestock ESCs, yet it has been generally concluded that the culture conditions tested so far are not supportive of livestock ESCs self-renewal and proliferation. In contrast, there are numerous reports of derivation of livestock induced pluripotent stem cells (iPSCs), with demonstrated capacity for long term proliferation and in vivo pluripotency, as indicated by teratoma formation assay. However, to what extent these iPSCs represent fully reprogrammed PSCs remains controversial, as most livestock iPSCs depend on continuous expression of reprogramming factors. Moreover, germline chimerism has not been robustly demonstrated, with only one successful report with very low efficiency. Therefore, even 34 years after derivation of mouse ESCs and their extensive use in the generation of genetic models, the livestock genetic engineering field can stand to gain enormously from continued investigations into the derivation and application of ESCs and iPSCs.
Collapse
|
42
|
Shi H, Fu Q, Li G, Ren Y, Hu S, Ni W, Guo F, Shi M, Meng L, Zhang H, Qiao J, Guo Z, Chen C. Roles of p53 and ASF1A in the Reprogramming of Sheep Kidney Cells to Pluripotent Cells. Cell Reprogram 2015; 17:441-52. [PMID: 26580119 DOI: 10.1089/cell.2015.0039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Since the first report of induced pluripotent stem cells (iPSCs) by Takahashi and Yamanaka, numerous attempts have been made to derive iPSCs from other species via the ectopic expression of defined factors. Sheep iPSCs (siPSCs) have significant potential for biotechnology and agriculture. Although several groups have described siPSCs, the reprogramming efficiency was extremely low. The exogenous transgenes could be not silenced in the iPSCs, which hampered their development and application. Here, we report that p53 knockdown and antisilencing function 1A (ASF1A) overexpression promoted iPSC generation from sheep kidney cells (SKCs). Compared with transduction with eight human defined transcription factors (Oct4, Sox2, Klf4, c-Myc, Nanog, Lin28, hTERT, and SV40LT), the additional introduction of p53 RNA interference (RNAi) and/or ASF1A in the presence of small-molecule compounds [vitamin C (Vc) and valproic acid (VPA)] greatly improved the efficiency of sheep iPSC generation. The siPSCs exhibited morphological features similar to mouse embryonic stem cells (ESCs) and were positive for alkaline phosphatase and, pluripotent marker genes (Oct4, Nanog, Sox2, Rex1, TRA-1-60, TRA-1-81, and E-cadherin). Furthermore, these cells exhibited a normal karyotype of 54 chromosomes and were able to differentiate into all three germ layers both in vitro and in vivo. Moreover, the exogenous genes were silenced in siPSCs when p53 small hairpin RNA (shRNA) and ASF1A were added. Our results may help to reveal the role of p53 and ASF1A in sheep somatic cell reprogramming and provide an efficient approach to reprogramming sheep somatic cells.
Collapse
Affiliation(s)
- Huijun Shi
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China .,2 College of Veterinary Medicine, Xinjiang Agricultural University , Urumqi 830052, Xinjiang, China .,5 These authors contributed equally to this work
| | - Qiang Fu
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China .,2 College of Veterinary Medicine, Xinjiang Agricultural University , Urumqi 830052, Xinjiang, China .,5 These authors contributed equally to this work
| | - Guozhong Li
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China .,5 These authors contributed equally to this work
| | - Yan Ren
- 3 College of Medicine, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Shengwei Hu
- 4 College of Life Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Wei Ni
- 4 College of Life Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Fei Guo
- 3 College of Medicine, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Mengting Shi
- 4 College of Life Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Luping Meng
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Hui Zhang
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Jun Qiao
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Zhiru Guo
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| | - Chuangfu Chen
- 1 College of Animal Science and Technology, Shihezi University , Shihezi 832003, Xinjiang, China
| |
Collapse
|
43
|
Abstract
This review deals with the latest advances in the study of embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) from domesticated species, with a focus on pigs, cattle, sheep, goats, horses, cats, and dogs. Whereas the derivation of fully pluripotent ESC from these species has proved slow, reprogramming of somatic cells to iPSC has been more straightforward. However, most of these iPSC depend on the continued expression of the introduced transgenes, a major drawback to their utility. The persistent failure in generating ESC and the dependency of iPSC on ectopic genes probably stem from an inability to maintain the stability of the endogenous gene networks necessary to maintain pluripotency. Based on work in humans and rodents, achievement of full pluripotency will likely require fine adjustments in the growth factors and signaling inhibitors provided to the cells. Finally, we discuss the future utility of these cells for biomedical and agricultural purposes.
Collapse
Affiliation(s)
- Toshihiko Ezashi
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211; , ,
| | - Ye Yuan
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211; , ,
| | - R Michael Roberts
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, Missouri 65211; , ,
| |
Collapse
|
44
|
Betts DH, Tobias IC. Canine Pluripotent Stem Cells: Are They Ready for Clinical Applications? Front Vet Sci 2015; 2:41. [PMID: 26664969 PMCID: PMC4672225 DOI: 10.3389/fvets.2015.00041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/21/2015] [Indexed: 12/20/2022] Open
Abstract
The derivation of canine embryonic stem cells and generation of canine-induced pluripotent stem cells are significant achievements that have unlocked the potential for developing novel cell-based disease models, drug discovery platforms, and transplantation therapies in the dog. A progression from concept to cure in this clinically relevant companion animal will not only help our canine patients but also help advance human regenerative medicine. Nevertheless, many issues remain to be resolved before pluripotent cells can be used clinically in a safe and reproducible manner.
Collapse
Affiliation(s)
- Dean H Betts
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario , London, ON , Canada ; Children's Health Research Institute, Lawson Health Research Institute , London, ON , Canada
| | - Ian C Tobias
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario , London, ON , Canada
| |
Collapse
|
45
|
Kumar D, Talluri TR, Anand T, Kues WA. Induced pluripotent stem cells: Mechanisms, achievements and perspectives in farm animals. World J Stem Cells 2015; 7:315-328. [PMID: 25815117 PMCID: PMC4369489 DOI: 10.4252/wjsc.v7.i2.315] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/19/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Pluripotent stem cells are unspecialized cells with unlimited self-renewal, and they can be triggered to differentiate into desired specialized cell types. These features provide the basis for an unlimited cell source for innovative cell therapies. Pluripotent cells also allow to study developmental pathways, and to employ them or their differentiated cell derivatives in pharmaceutical testing and biotechnological applications. Via blastocyst complementation, pluripotent cells are a favoured tool for the generation of genetically modified mice. The recently established technology to generate an induced pluripotency status by ectopic co-expression of the transcription factors Oct4, Sox2, Klf4 and c-Myc allows to extending these applications to farm animal species, for which the derivation of genuine embryonic stem cells was not successful so far. Most induced pluripotent stem (iPS) cells are generated by retroviral or lentiviral transduction of reprogramming factors. Multiple viral integrations into the genome may cause insertional mutagenesis and may increase the risk of tumour formation. Non-integration methods have been reported to overcome the safety concerns associated with retro and lentiviral-derived iPS cells, such as transient expression of the reprogramming factors using episomal plasmids, and direct delivery of reprogramming mRNAs or proteins. In this review, we focus on the mechanisms of cellular reprogramming and current methods used to induce pluripotency. We also highlight problems associated with the generation of iPS cells. An increased understanding of the fundamental mechanisms underlying pluripotency and refining the methodology of iPS cell generation will have a profound impact on future development and application in regenerative medicine and reproductive biotechnology of farm animals.
Collapse
|
46
|
Li M, Li L, Zhang J, Verma V, Liu Q, Shi D, Huang B. An Insight on Small Molecule Induced Foot-Print Free Naive Pluripotent Stem Cells in Livestock. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/scd.2015.51001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
47
|
Nagy K, Nagy A. Derivation of Equine-Induced Pluripotent Stem Cell Lines Using a piggyBac Transposon Delivery System and Temporal Control of Transgene Expression. Methods Mol Biol 2015; 1330:79-88. [PMID: 26621591 DOI: 10.1007/978-1-4939-2848-4_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) has had a transforming effect on our understanding of biology and has brought an enormous promise to regenerative medicine. It has opened up a magnitude of unprecedented possibilities to study disease processes in vitro, model them in animal systems, and develop patient-specific cell-based regenerative therapies. iPSCs derived from other than the human species will be instrumental for bringing these prospects to fruition by providing preclinical models and novel treatments for veterinary medicine. In this chapter, we describe the derivation of iPSCs from equine embryonic fibroblasts using a non-viral method developed in our laboratory and originally applied to the murine and human systems (Woltjen et al., Nature 458:766-770, 2009). We will detail the procedures involved and discuss potential pitfalls as well as elaborate on possible variations and future improvements of this technique.
Collapse
Affiliation(s)
- Kristina Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 60 Murray St., Toronto, ON, Canada, M5T 3X7
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 60 Murray St., Toronto, ON, Canada, M5T 3X7.
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada.
- Lunenfeld-Tanenbaum, Research Institute, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Ave., Toronto, ON, Canada, M5G 1X5.
| |
Collapse
|
48
|
German SD, Campbell KHS, Thornton E, McLachlan G, Sweetman D, Alberio R. Ovine induced pluripotent stem cells are resistant to reprogramming after nuclear transfer. Cell Reprogram 2014; 17:19-27. [PMID: 25513856 DOI: 10.1089/cell.2014.0071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) share similar characteristics of indefinite in vitro growth with embryonic stem cells (ESCs) and may therefore serve as a useful tool for the targeted genetic modification of farm animals via nuclear transfer (NT). Derivation of stable ESC lines from farm animals has not been possible, therefore, it is important to determine whether iPSCs can be used as substitutes for ESCs in generating genetically modified cloned farm animals. We generated ovine iPSCs by conventional retroviral transduction using the four Yamanaka factors. These cells were basic fibroblast growth factor (bFGF)- and activin A-dependent, showed persistent expression of the transgenes, acquired chromosomal abnormalities, and failed to activate endogenous NANOG. Nonetheless, iPSCs could differentiate into the three somatic germ layers in vitro. Because cloning of farm animals is best achieved with diploid cells (G1/G0), we synchronized the iPSCs in G1 prior to NT. Despite the cell cycle synchronization, preimplantation development of iPSC-NT embryos was lower than with somatic cells (2% vs. 10% blastocysts, p<0.01). Furthermore, analysis of the blastocysts produced demonstrated persistent expression of the transgenes, aberrant expression of endogenous SOX2, and a failure to activate NANOG consistently. In contrast, gene expression in blastocysts produced with the parental fetal fibroblasts was similar to those generated by in vitro fertilization. Taken together, our data suggest that the persistent expression of the exogenous factors and the acquisition of chromosomal abnormalities are incompatible with normal development of NT embryos produced with iPSCs.
Collapse
Affiliation(s)
- Sergio D German
- 1 Division of Animal Sciences, School of Biosciences, University of Nottingham , Loughborough, LE12 5RD, United Kingdom
| | | | | | | | | | | |
Collapse
|
49
|
Whitworth DJ, Banks TA. Stem cell therapies for treating osteoarthritis: prescient or premature? Vet J 2014; 202:416-24. [PMID: 25457267 DOI: 10.1016/j.tvjl.2014.09.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 09/22/2014] [Accepted: 09/25/2014] [Indexed: 01/23/2023]
Abstract
There has been unprecedented interest in recent years in the use of stem cells as therapy for an array of diseases in companion animals. Stem cells have already been deployed therapeutically in a number of clinical settings, in particular the use of mesenchymal stem cells to treat osteoarthritis in horses and dogs. However, an assessment of the scientific literature highlights a marked disparity between the purported benefits of stem cell therapies and their proven abilities as defined by rigorously controlled scientific studies. Although preliminary data generated from clinical trials in human patients are encouraging, therapies currently available to treat animals are supported by very limited clinical evidence, and the commercialisation of these treatments may be premature. This review introduces the three main types of stem cells relevant to veterinary applications, namely, embryonic stem cells, induced pluripotent stem cells, and mesenchymal stem cells, and draws together research findings from in vitro and in vivo studies to give an overview of current stem cell therapies for the treatment of osteoarthritis in animals. Recent advances in tissue engineering, which is proposed as the future direction of stem cell-based therapy for osteoarthritis, are also discussed.
Collapse
Affiliation(s)
- Deanne J Whitworth
- School of Veterinary Science, University of Queensland, Gatton, Queensland 4343, Australia.
| | - Tania A Banks
- School of Veterinary Science, University of Queensland, Gatton, Queensland 4343, Australia
| |
Collapse
|
50
|
Romli F, Alitheen NB, Hamid M, Ismail R, Abd Rahman NMAN. Current techniques in reprogramming cell potency. J Cell Biochem 2014; 114:1230-7. [PMID: 23239017 DOI: 10.1002/jcb.24477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/05/2012] [Indexed: 11/11/2022]
Abstract
The first successful attempt to reprogram somatic cell into embryonic-like stem cell was achieved on 2006. Since then, it had sparked a race against time to bring this wonderful invention from bench to bedside but it is not easily achieved due to severe problems in term of epigenetic and genomic. With each problem arise, new technique and protocol will be constructed to try to overcome it. This review addresses the various techniques made available to create iPSC with problems hogging down the technique.
Collapse
Affiliation(s)
- Firdaus Romli
- Faculty of Biotechnology and Biomolecular Sciences, Department of Cell and Molecular Biology, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | | | | | | |
Collapse
|