1
|
Anwised P, Moorawong R, Samruan W, Somredngan S, Srisutush J, Laowtammathron C, Aksoy I, Parnpai R, Savatier P. An expedition in the jungle of pluripotent stem cells of non-human primates. Stem Cell Reports 2023; 18:2016-2037. [PMID: 37863046 PMCID: PMC10679654 DOI: 10.1016/j.stemcr.2023.09.013] [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: 06/16/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/22/2023] Open
Abstract
For nearly three decades, more than 80 embryonic stem cell lines and more than 100 induced pluripotent stem cell lines have been derived from New World monkeys, Old World monkeys, and great apes. In this comprehensive review, we examine these cell lines originating from marmoset, cynomolgus macaque, rhesus macaque, pig-tailed macaque, Japanese macaque, African green monkey, baboon, chimpanzee, bonobo, gorilla, and orangutan. We outline the methodologies implemented for their establishment, the culture protocols for their long-term maintenance, and their basic molecular characterization. Further, we spotlight any cell lines that express fluorescent reporters. Additionally, we compare these cell lines with human pluripotent stem cell lines, and we discuss cell lines reprogrammed into a pluripotent naive state, detailing the processes used to attain this. Last, we present the findings from the application of these cell lines in two emerging fields: intra- and interspecies embryonic chimeras and blastoids.
Collapse
Affiliation(s)
- Preeyanan Anwised
- University Lyon, University Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, 69500 Bron, France; Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Ratree Moorawong
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Worawalan Samruan
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Sirilak Somredngan
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Jittanun Srisutush
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Irene Aksoy
- University Lyon, University Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, 69500 Bron, France.
| | - Rangsun Parnpai
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| | - Pierre Savatier
- University Lyon, University Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, 69500 Bron, France.
| |
Collapse
|
2
|
Bayurova E, Zhitkevich A, Avdoshina D, Kupriyanova N, Kolyako Y, Kostyushev D, Gordeychuk I. Common Marmoset Cell Lines and Their Applications in Biomedical Research. Cells 2023; 12:2020. [PMID: 37626830 PMCID: PMC10453182 DOI: 10.3390/cells12162020] [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: 05/29/2023] [Revised: 07/19/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Common marmosets (Callithrix jacchus; CMs) are small New World primates widely used in biomedical research. Early stages of such research often include in vitro experiments which require standardized and well-characterized CM cell cultures derived from different tissues. Despite the long history of laboratory work with CMs and high translational potential of such studies, the number of available standardized, well-defined, stable, and validated CM cell lines is still small. While primary cells and immortalized cell lines are mostly used for the studies of infectious diseases, biochemical research, and targeted gene therapy, the main current applications of CM embryonic stem cells and induced pluripotent stem cells are regenerative medicine, stem cell research, generation of transgenic CMs, transplantology, cell therapy, reproductive physiology, oncology, and neurodegenerative diseases. In this review we summarize the data on the main advantages, drawbacks and research applications of CM cell lines published to date including primary cells, immortalized cell lines, lymphoblastoid cell lines, embryonic stem cells, and induced pluripotent stem cells.
Collapse
Affiliation(s)
- Ekaterina Bayurova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
| | - Alla Zhitkevich
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
| | - Daria Avdoshina
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
| | - Natalya Kupriyanova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
- Institute for Translational Medicine and Biotechnology, Sechenov University, 117418 Moscow, Russia
| | - Yuliya Kolyako
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
- Institute for Translational Medicine and Biotechnology, Sechenov University, 117418 Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119435 Moscow, Russia;
- Scientific Center for Genetics and Life Sciences, Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ilya Gordeychuk
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia; (E.B.); (A.Z.); (D.A.); (N.K.); (Y.K.)
- Institute for Translational Medicine and Biotechnology, Sechenov University, 117418 Moscow, Russia
| |
Collapse
|
3
|
Rodriguez-Polo I, Behr R. Non-human primate pluripotent stem cells for the preclinical testing of regenerative therapies. Neural Regen Res 2022; 17:1867-1874. [PMID: 35142660 PMCID: PMC8848615 DOI: 10.4103/1673-5374.335689] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Non-human primates play a key role in the preclinical validation of pluripotent stem cell-based cell replacement therapies. Pluripotent stem cells used as advanced therapy medical products boost the possibility to regenerate tissues and organs affected by degenerative diseases. Therefore, the methods to derive human induced pluripotent stem cell and embryonic stem cell lines following clinical standards have quickly developed in the last 15 years. For the preclinical validation of cell replacement therapies in non-human primates, it is necessary to generate non-human primate pluripotent stem cell with a homologous quality to their human counterparts. However, pluripotent stem cell technologies have developed at a slower pace in non-human primates in comparison with human cell systems. In recent years, however, relevant progress has also been made with non-human primate pluripotent stem cells. This review provides a systematic overview of the progress and remaining challenges for the generation of non-human primate induced pluripotent stem cells/embryonic stem cells for the preclinical testing and validation of cell replacement therapies. We focus on the critical domains of (1) reprogramming and embryonic stem cell line derivation, (2) cell line maintenance and characterization and, (3) application of non-human primate pluripotent stem cells in the context of selected preclinical studies to treat cardiovascular and neurodegenerative disorders performed in non-human primates.
Collapse
|
4
|
Park JE, Sasaki E. Assisted Reproductive Techniques and Genetic Manipulation in the Common Marmoset. ILAR J 2021; 61:286-303. [PMID: 33693670 PMCID: PMC8918153 DOI: 10.1093/ilar/ilab002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/27/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Genetic modification of nonhuman primate (NHP) zygotes is a useful method for the development of NHP models of human diseases. This review summarizes the recent advances in the development of assisted reproductive and genetic manipulation techniques in NHP, providing the basis for the generation of genetically modified NHP disease models. In this study, we review assisted reproductive techniques, including ovarian stimulation, in vitro maturation of oocytes, in vitro fertilization, embryo culture, embryo transfer, and intracytoplasmic sperm injection protocols in marmosets. Furthermore, we review genetic manipulation techniques, including transgenic strategies, target gene knock-out and knock-in using gene editing protocols, and newly developed gene-editing approaches that may potentially impact the production of genetically manipulated NHP models. We further discuss the progress of assisted reproductive and genetic manipulation techniques in NHP; future prospects on genetically modified NHP models for biomedical research are also highlighted.
Collapse
Affiliation(s)
- Jung Eun Park
- Department of Neurobiology, University of Pittsburgh, School of Medicine in Pittsburgh, Pennsylvania, USA
| | - Erika Sasaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals in Kawasaki, Kanagawa, Japan
| |
Collapse
|
5
|
Hanna M, Sahito RGA, Rateb M, Kachiwal AB, Seddiek HA, Bhutto B, Hescheler J. Generation of transgene-free induced pluripotent stem cells from cardiac fibroblasts of goat embryos. J Stem Cells Regen Med 2020; 16:34-43. [PMID: 33414579 DOI: 10.46582/jsrm.1602007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/11/2020] [Indexed: 11/19/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold a great potential for therapeutic regenerative medicine. The aim of this study was to generate induced pluripotent stem cells from goat embryonic cardiac tissue derived fibroblasts. The isolated cardiac fibroblasts from the cardiac tissue of goat embryos were positive for alfa smooth muscle actin, vimentin and discoidin domain receptor2. From these cells, we generated transgene free iPSCs using piggyBac transposons / transposase using five transcription factors (Oct4, Sox2, Klf, Myc and Lin 28). The generated iPSCs were SSEA1, SSEA4 and Oct4 positive. They were cultured on neofeeders using 20% Serum replacement - IMDM with bFGF. They could form cystic and compact embryoid bodies that showed differentiated ectodermal and mesodermal like cells when cultured using 20% FBS-IMDM without bFGF. The iPSCs, generated in the frame of this approach were produced without the use of integrating virus and the reprogramming transgenes were removed at the end of the process. Though there were limitations in the approach used, a substantial sign of reprogramming was obtained.
Collapse
Affiliation(s)
- Mira Hanna
- Institute of Neurophysiology, University of Cologne, Robert-Koch-Strasse 39, 50931 Cologne, Germany.,Department of physiology, Faculty of medicine (Kasr El-Aini) Cairo University, El-Maniel, Cairo 11451, Egypt
| | | | - Moshira Rateb
- Department of physiology, Faculty of medicine (Kasr El-Aini) Cairo University, El-Maniel, Cairo 11451, Egypt
| | - Allah Bux Kachiwal
- Department of Veterinary Physiology and Biochemistry, Sindh Agriculture University Tandojam, Pakistan
| | - Hanan A Seddiek
- Department of physiology, Faculty of medicine (Kasr El-Aini) Cairo University, El-Maniel, Cairo 11451, Egypt
| | - Bachal Bhutto
- Department of Veterinary Parasitology, Sindh Agriculture University Tandojam, Pakistan
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, Robert-Koch-Strasse 39, 50931 Cologne, Germany
| |
Collapse
|
6
|
Utility of Common Marmoset ( Callithrix jacchus) Embryonic Stem Cells in Liver Disease Modeling, Tissue Engineering and Drug Metabolism. Genes (Basel) 2020; 11:genes11070729. [PMID: 32630053 PMCID: PMC7397002 DOI: 10.3390/genes11070729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
The incidence of liver disease is increasing significantly worldwide and, as a result, there is a pressing need to develop new technologies and applications for end-stage liver diseases. For many of them, orthotopic liver transplantation is the only viable therapeutic option. Stem cells that are capable of differentiating into all liver cell types and could closely mimic human liver disease are extremely valuable for disease modeling, tissue regeneration and repair, and for drug metabolism studies to develop novel therapeutic treatments. Despite the extensive research efforts, positive results from rodent models have not translated meaningfully into realistic preclinical models and therapies. The common marmoset Callithrix jacchus has emerged as a viable non-human primate model to study various human diseases because of its distinct features and close physiologic, genetic and metabolic similarities to humans. C. jacchus embryonic stem cells (cjESC) and recently generated cjESC-derived hepatocyte-like cells (cjESC-HLCs) could fill the gaps in disease modeling, liver regeneration and metabolic studies. They are extremely useful for cell therapy to regenerate and repair damaged liver tissues in vivo as they could efficiently engraft into the liver parenchyma. For in vitro studies, they would be advantageous for drug design and metabolism in developing novel drugs and cell-based therapies. Specifically, they express both phase I and II metabolic enzymes that share similar substrate specificities, inhibition and induction characteristics, and drug metabolism as their human counterparts. In addition, cjESCs and cjESC-HLCs are advantageous for investigations on emerging research areas, including blastocyst complementation to generate entire livers, and bioengineering of discarded livers to regenerate whole livers for transplantation.
Collapse
|
7
|
Shiozawa S, Nakajima M, Okahara J, Kuortaki Y, Kisa F, Yoshimatsu S, Nakamura M, Koya I, Yoshimura M, Sasagawa Y, Nikaido I, Sasaki E, Okano H. Primed to Naive-Like Conversion of the Common Marmoset Embryonic Stem Cells. Stem Cells Dev 2020; 29:761-773. [DOI: 10.1089/scd.2019.0259] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Seiji Shiozawa
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Mayutaka Nakajima
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Junko Okahara
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Yoko Kuortaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Fumihiko Kisa
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
- Discovery Research Laboratories I, Minase Research Institute, Ono Pharmaceutical Co., Ltd., Mishima, Japan
| | - Sho Yoshimatsu
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Mari Nakamura
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Ikuko Koya
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Mika Yoshimura
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Wako, Japan
| | - Yohei Sasagawa
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Wako, Japan
| | - Itoshi Nikaido
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Wako, Japan
- Bioinformatics Course, Master's/Doctoral Program in Life Science Innovation (T-LSI), School of Integrative and Global Majors (SIGMA), University of Tsukuba, Wako, Japan
| | - Erika Sasaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Japan
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
8
|
Kropp J, Di Marzo A, Golos T. Assisted reproductive technologies in the common marmoset: an integral species for developing nonhuman primate models of human diseases. Biol Reprod 2018; 96:277-287. [PMID: 28203717 DOI: 10.1095/biolreprod.116.146514] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 12/31/2022] Open
Abstract
Generation of nonhuman primate models of human disease conditions will foster the development of novel therapeutic strategies. Callithrix jacchus, or the common marmoset, is a New World, nonhuman primate species that exhibits great reproductive fitness in captivity with an ovarian cycle that can be easily managed with pharmacological agents. This characteristic, among others, provides an opportunity to employ assisted reproductive technologies to generate embryos that can be genetically manipulated to create a variety of nonhuman primate models for human disease. Here, we review methods to synchronize the marmoset ovarian cycle and stimulate oocyte donors, and compare various protocols for in vitro production of embryos. In light of advances in genomic editing, recent approaches used to generate transgenic or genetically edited embryos in the marmoset and also future perspective are reviewed.
Collapse
Affiliation(s)
- Jenna Kropp
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrea Di Marzo
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Thaddeus Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
9
|
Ke M, He Q, Hong D, Li O, Zhu M, Ou WB, He Y, Wu Y. Leukemia inhibitory factor regulates marmoset induced pluripotent stem cell proliferation via a PI3K/Akt‑dependent Tbx‑3 activation pathway. Int J Mol Med 2018; 42:131-140. [PMID: 29620145 PMCID: PMC5979829 DOI: 10.3892/ijmm.2018.3610] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/28/2018] [Indexed: 01/22/2023] Open
Abstract
Leukemia inhibitory factor (LIF) is the most pleiotropic cytokine of the interleukin-6 family, and is widely used to establish and maintain pluripotent stem cells, particularly mouse pluripotent stem cells. However, no reports have fully elucidated the application of LIF in marmoset induced pluripotent stem cell (iPSC) culture, particularly the underlying mechanisms. To demonstrate the feasibility of the application of LIF to marmoset iPSCs, the present study assessed these cells in the presence of LIF. Cell proliferation was measured using MTT assay, cell apoptosis was determined by flow cytometric analysis of fluorescein isothiocyanate Annexin V staining and the differentially expressed genes were analysed using Digital Gene Expression (DGE) analysis. The altered expression of pluripotency-associated genes was confirmed by reverse transcription-quantitative polymerase chain reaction and western blot analysis. Furthermore, following treatment with LY294002, cell proliferation was measured by MTT assay and protein levels were confirmed by western blot analysis. The results showed that LIF significantly promoted the number of proliferating cells, but had no effect on apoptosis. Digital Gene Expression analysis was used to examine the differentially expressed genes of marmoset iPSCs in the presence of LIF. The results showed that the pluripotency-associated transcription factor-encoding gene T-box 3 (Tbx-3) was activated by LIF. Notably, LIF increased the levels of phosphorylated (p-)AKT and Tbx-3 in the marmoset iPSCs. Furthermore, pretreatment with LY294002, an inhibitor of phosphoinositide 3-kinase (PI3K), significantly impaired the LIF-induced upregulation of p-AKT and Tbx-3 in the marmoset iPSCs, suggesting that the PI3K/Akt signaling pathway is involved in this regulation. Taken together, the results suggested that LIF is effective in maintaining marmoset iPSCs in cultures, which is associated with the activation of Tbx-3 through regulation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Minxia Ke
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Quan He
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Danping Hong
- 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
| | - Mengyi Zhu
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Wen-Bin Ou
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, 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
| | - Yuehong Wu
- Department of Biochemistry and Molecular Biology, College of Life Science, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| |
Collapse
|
10
|
Jin JX, Lee S, Setyawan EMN, Taweechaipaisankul A, Kim GA, Han HJ, Ahn C, Lee BC. A potential role of knockout serum replacement as a porcine follicular fluid substitute for in vitro maturation: Lipid metabolism approach. J Cell Physiol 2018; 233:6984-6995. [PMID: 29345310 DOI: 10.1002/jcp.26489] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/17/2018] [Indexed: 02/06/2023]
Abstract
The use of supplements, such as porcine follicular fluid (pFF), fetal bovine serum and human serum albumin are widely used during in vitro maturation (IVM) in different species but these supplements contain undefined components that cause technical difficulties in standardization and influence the efficiency of IVM. Knockout serum replacement (KSR) is a synthetic protein source, without any undefined growth factors or differentiation-promoting factors. Therefore, it is feasible to use KSR as a defined component for avoiding effects of unknown molecules in an IVM system. In this study, the rates of oocyte maturation and blastocyst formation after parthenogenetic activation (PA), somatic cell nuclear transfer (SCNT) and in vitro fertilization (IVF) were significantly higher in the 5% KSR supplemented group than in the unsupplemented control group and more similar to those of the 10% pFF supplemented group. Moreover, the intensity of GDF9, BMP15, ROS, GSH, BODIPY-LD, BODIPY-FA, and BODIPY-ATP staining showed similar values between 5% KSR and 10% pFF, which have significant difference with control group. Most of the gene expression related to lipid metabolism with both supplements exhibited similar patterns. In conclusion, 5% KSR upregulated lipid metabolism and thereby provides an essential energy source to sustain and improve oocyte quality and subsequent embryo development after PA, SCNT, and IVF. These indications support the idea that KSR used as a defined serum supplement for oocyte IVM might be universally used in other species.
Collapse
Affiliation(s)
- Jun-Xue Jin
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea.,Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Sanghoon Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Erif Maha Nugraha Setyawan
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Anukul Taweechaipaisankul
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Geon A Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Curie Ahn
- Designed Animal Resource Center and Biotransplant Research Institute, Seoul National University Green-Bio Research Complex, Gangwon-do, Korea.,Division of Nephrology, Seoul National University College of Medicine, Seoul, Korea.,Transplantation Center, Seoul National University Hospital, Seoul, Korea
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
11
|
Rodriguez-Polo I, Nielsen M, Debowski K, Behr R. The ubiquitin ligase c-CBL is expressed in undifferentiated marmoset monkey pluripotent stem cells but is not a general stem cell marker. Primate Biol 2017; 4:231-240. [PMID: 32110709 PMCID: PMC7041541 DOI: 10.5194/pb-4-231-2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/26/2017] [Indexed: 11/23/2022] Open
Abstract
The protein c-CBL is a ubiquitin ligase. It catalyzes the last step of the
transfer of ubiquitin to target proteins. Upon completion of
polyubiquitination, the target proteins are degraded. Clinically, it is
important that c-CBL is mutated in a subset of patients who develop myeloid
malignancies, which are diseases of the hematopoietic stem or progenitor
cells. c-CBL has also been shown to be expressed by human spermatogonia. The
whole spermatogonial cell population possesses a subset that comprises also
the spermatogonial stem cells. Based on these findings we hypothesized that
c-CBL might be a general stem cell marker. To test this, we first validated
the antibody using marmoset bone marrow and adult testis. In both tissues,
the expected staining pattern was observed. Western blot analysis revealed
only one band of the expected size. Then, we examined the expression of c-CBL
in marmoset monkey embryonic stem (ES) cells, induced pluripotent stem (iPS)
cells and adult stem cells. We found that c-CBL is strongly expressed in
undifferentiated marmoset iPS cells and ES cells. However, adult stem cells
in the gut and the stomach did not express c-CBL, indicating that c-CBL is not
a general stem cell marker. In summary, c-CBL is strongly expressed in
pluripotent stem cells of the marmoset monkey as well as in selected adult
stem cell types. Future studies will define the function of c-CBL in
pluripotent stem cells.
Collapse
Affiliation(s)
- Ignacio Rodriguez-Polo
- Platform Degenerative Diseases, German Primate Center (DPZ), Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.,DZHK, German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany.,These authors contributed equally to this work
| | - Maike Nielsen
- Platform Degenerative Diseases, German Primate Center (DPZ), Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.,These authors contributed equally to this work
| | - Katharina Debowski
- Platform Degenerative Diseases, German Primate Center (DPZ), Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.,present address: STEMCELL Technologies Germany GmbH, Stolberger Str. 200, 50933 Cologne, Germany.,These authors contributed equally to this work
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center (DPZ), Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.,DZHK, German Center for Cardiovascular Research, Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Neehus AL, Wistuba J, Ladas N, Eiz-Vesper B, Schlatt S, Müller T. Gene conversion of the major histocompatibility complex class I Caja-G in common marmosets (Callithrix jacchus). Immunology 2016; 149:343-352. [PMID: 27450742 PMCID: PMC5046058 DOI: 10.1111/imm.12652] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 06/30/2016] [Accepted: 07/13/2016] [Indexed: 01/13/2023] Open
Abstract
Currently, the amount of sequenced and classified MHC class I genes of the common marmoset is limited, in spite of the wide use of this species as an animal model for biomedical research. In this study, 480 clones of MHC class I G locus (Caja‐G) cDNA sequences were obtained from 21 common marmosets. Up to 10 different alleles were detected in each common marmoset, leading to the assumption that the Caja‐G loci duplicated in the marmoset genome. In the investigated population, four alleles occurred more often, giving evidence for higher immunological advantage of these alleles. In contrast to the human non‐classical MHC class I genes, Caja‐G shows high rates of polymorphism at the relevant peptide‐binding sites, despite its phylogenetic relationship to the non‐classical HLA‐G. Our results provide information for better understanding of the immunological properties of the common marmoset and confirm the theory of a gene conversion of the Caja‐G due to its detected plasticity and the absence of any known HLA‐A equivalent.
Collapse
Affiliation(s)
- Anna-Lena Neehus
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Joachim Wistuba
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Münster, Münster, Germany
| | - Nektarios Ladas
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Stefan Schlatt
- Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Münster, Münster, Germany
| | - Thomas Müller
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany. .,Synlab Medical Care Centre Weiden Ltd, Weiden, Germany.
| |
Collapse
|
13
|
Debowski K, Drummer C, Lentes J, Cors M, Dressel R, Lingner T, Salinas-Riester G, Fuchs S, Sasaki E, Behr R. The transcriptomes of novel marmoset monkey embryonic stem cell lines reflect distinct genomic features. Sci Rep 2016; 6:29122. [PMID: 27385131 PMCID: PMC4935898 DOI: 10.1038/srep29122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 06/13/2016] [Indexed: 12/21/2022] Open
Abstract
Embryonic stem cells (ESCs) are useful for the study of embryonic development. However, since research on naturally conceived human embryos is limited, non-human primate (NHP) embryos and NHP ESCs represent an excellent alternative to the corresponding human entities. Though, ESC lines derived from naturally conceived NHP embryos are still very rare. Here, we report the generation and characterization of four novel ESC lines derived from natural preimplantation embryos of the common marmoset monkey (Callithrix jacchus). For the first time we document derivation of NHP ESCs derived from morula stages. We show that quantitative chromosome-wise transcriptome analyses precisely reflect trisomies present in both morula-derived ESC lines. We also demonstrate that the female ESC lines exhibit different states of X-inactivation which is impressively reflected by the abundance of the lncRNA X inactive-specific transcript (XIST). The novel marmoset ESC lines will promote basic primate embryo and ESC studies as well as preclinical testing of ESC-based regenerative approaches in NHP.
Collapse
Affiliation(s)
- Katharina Debowski
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Charis Drummer
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Jana Lentes
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Maren Cors
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen (UMG), Humboldtallee 34, 37073 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Germany
| | - Thomas Lingner
- Microarray and Deep-Sequencing Core Facility, University Medical Center Göttingen (UMG), Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Gabriela Salinas-Riester
- Microarray and Deep-Sequencing Core Facility, University Medical Center Göttingen (UMG), Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Sigrid Fuchs
- Department of Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Erika Sasaki
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, 3-25-12 Tonomachi Kawasaki-ku, Kawasaki, 210-0821 Japan.,Keio Advanced Research Center, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Germany
| |
Collapse
|
14
|
Fereydouni B, Salinas-Riester G, Heistermann M, Dressel R, Lewerich L, Drummer C, Behr R. Long-Term Oocyte-Like Cell Development in Cultures Derived from Neonatal Marmoset Monkey Ovary. Stem Cells Int 2015; 2016:2480298. [PMID: 26664406 PMCID: PMC4655298 DOI: 10.1155/2016/2480298] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/28/2015] [Accepted: 07/28/2015] [Indexed: 11/17/2022] Open
Abstract
We use the common marmoset monkey (Callithrix jacchus) as a preclinical nonhuman primate model to study reproductive and stem cell biology. The neonatal marmoset monkey ovary contains numerous primitive premeiotic germ cells (oogonia) expressing pluripotent stem cell markers including OCT4A (POU5F1). This is a peculiarity compared to neonatal human and rodent ovaries. Here, we aimed at culturing marmoset oogonia from neonatal ovaries. We established a culture system being stable for more than 20 passages and 5 months. Importantly, comparative transcriptome analysis of the cultured cells with neonatal ovary, embryonic stem cells, and fibroblasts revealed a lack of germ cell and pluripotency genes indicating the complete loss of oogonia upon initiation of the culture. From passage 4 onwards, however, the cultured cells produced large spherical, free-floating cells resembling oocyte-like cells (OLCs). OLCs strongly expressed several germ cell genes and may derive from the ovarian surface epithelium. In summary, our novel primate ovarian cell culture initially lacked detectable germ cells but then produced OLCs over a long period of time. This culture system may allow a deeper analysis of early phases of female primate germ cell development and-after significant refinement-possibly also the production of monkey oocytes.
Collapse
Affiliation(s)
- Bentolhoda Fereydouni
- Stem Cell Biology Unit, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Gabriela Salinas-Riester
- Microarray and Deep-Sequencing Core Facility, University Medical Center Göttingen (UMG), Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Ralf Dressel
- Department of Cellular and Molecular Immunology, University of Göttingen, Humboldtallee 34, 37073 Göttingen, Germany
| | - Lucia Lewerich
- Stem Cell Biology Unit, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Charis Drummer
- Stem Cell Biology Unit, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Rüdiger Behr
- Stem Cell Biology Unit, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| |
Collapse
|
15
|
Lauterboeck L, Hofmann N, Mueller T, Glasmacher B. Active control of the nucleation temperature enhances freezing survival of multipotent mesenchymal stromal cells. Cryobiology 2015; 71:384-90. [PMID: 26499840 DOI: 10.1016/j.cryobiol.2015.10.145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 10/15/2015] [Accepted: 10/19/2015] [Indexed: 02/03/2023]
Abstract
Cryopreservation is a technique that has been extensively used for storage of multipotent mesenchymal stromal cells (MSCs) in regenerative medicine. Therefore, improving current cryopreservation procedures in terms of increasing cell viability and functionality is important. In this study, we optimized the cryopreservation protocol of MSCs derived from the common marmoset Callithrix jacchus (cj), which can be used as a non-human primate model in various pathological and transplantation studies and have a great potential for regenerative medicine. We have investigated the effect of the active control of the nucleation temperature using induced nucleation at a broad range of temperatures and two different dimethylsulfoxide concentrations (Me2SO, 5% (v/v) and 10%, (v/v)) to evaluate the overall effect on the viability, metabolic activity and recovery of cells after thawing. Survival rate and metabolic activity displayed an optimum when ice formation was induced at -10 °C. Cryomicroscopy studies indicated differences in ice crystal morphologies as well as differences in intracellular ice formation with different nucleation temperatures. High subzero nucleation temperatures resulted in larger extracellular ice crystals and cellular dehydration, whereas low subzero nucleation temperatures resulted in smaller ice crystals and intracellular ice formation.
Collapse
Affiliation(s)
- L Lauterboeck
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany.
| | - N Hofmann
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany.
| | - T Mueller
- Service Unit Embryonic Stem Cells, Institute for Transfusion Medicine, Medical School Hannover, Germany.
| | - B Glasmacher
- Institute for Multiphase Processes, Leibniz Universität Hannover, Hannover, Germany.
| |
Collapse
|
16
|
Multipotent stromal cells derived from common marmoset Callithrix jacchus within alginate 3D environment: Effect of cryopreservation procedures. Cryobiology 2015; 71:103-11. [PMID: 25980899 DOI: 10.1016/j.cryobiol.2015.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 11/23/2022]
Abstract
Multipotent stromal cells derived from the common marmoset monkey Callithrix jacchus (cjMSCs) possess high phylogenetic similarity to humans, with a great potential for preclinical studies in the field of regenerative medicine. Safe and effective long-term storage of cells is of great significance to clinical and research applications. Encapsulation of such cell types within alginate beads that can mimic an extra-cellular matrix and provide a supportive environment for cells during cryopreservation, has several advantages over freezing of cells in suspension. In this study we have analysed the effect of dimethyl sulfoxide (Me2SO, 2.5-10%, v/v) and pre-freeze loading time of alginate encapsulated cjMSCs in Me2SO (0-45 min) on the viability and metabolic activity of the cells after freezing using a slow cooling rate (-1°C/min). It was found that these parameters affect the stability and homogeneity of alginate beads after thawing. Moreover, the cjMSCs can be frozen in alginate beads with lower Me2SO concentration of 7.5% after 30 min of loading, while retaining high cryopreservation outcome. We demonstrated the maximum viability, membrane integrity and metabolic activity of the cells under optimized, less cytotoxic conditions. The results of this study are another step forward towards the application of cryopreservation for the long-term storage and subsequent applications of transplants in cell-based therapies.
Collapse
|
17
|
Debowski K, Warthemann R, Lentes J, Salinas-Riester G, Dressel R, Langenstroth D, Gromoll J, Sasaki E, Behr R. Non-viral generation of marmoset monkey iPS cells by a six-factor-in-one-vector approach. PLoS One 2015; 10:e0118424. [PMID: 25785453 PMCID: PMC4365012 DOI: 10.1371/journal.pone.0118424] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/01/2014] [Indexed: 02/07/2023] Open
Abstract
Groundbreaking studies showed that differentiated somatic cells of mouse and human origin could be reverted to a stable pluripotent state by the ectopic expression of only four proteins. The resulting pluripotent cells, called induced pluripotent stem (iPS) cells, could be an alternative to embryonic stem cells, which are under continuous ethical debate. Hence, iPS cell-derived functional cells such as neurons may become the key for an effective treatment of currently incurable degenerative diseases. However, besides the requirement of efficacy testing of the therapy also its long-term safety needs to be carefully evaluated in settings mirroring the clinical situation in an optimal way. In this context, we chose the long-lived common marmoset monkey (Callithrix jacchus) as a non-human primate species to generate iPS cells. The marmoset monkey is frequently used in biomedical research and is gaining more and more preclinical relevance due to the increasing number of disease models. Here, we describe, to our knowledge, the first-time generation of marmoset monkey iPS cells from postnatal skin fibroblasts by non-viral means. We used the transposon-based, fully reversible piggyback system. We cloned the marmoset monkey reprogramming factors and established robust and reproducible reprogramming protocols with a six-factor-in-one-construct approach. We generated six individual iPS cell lines and characterized them in comparison with marmoset monkey embryonic stem cells. The generated iPS cells are morphologically indistinguishable from marmoset ES cells. The iPS cells are fully reprogrammed as demonstrated by differentiation assays, pluripotency marker expression and transcriptome analysis. They are stable for numerous passages (more than 80) and exhibit euploidy. In summary, we have established efficient non-viral reprogramming protocols for the derivation of stable marmoset monkey iPS cells, which can be used to develop and test cell replacement therapies in preclinical settings.
Collapse
Affiliation(s)
- Katharina Debowski
- Stem Cell Biology Unit, German Primate Center—Leibniz Institute for Primate Research, Göttingen, Germany
- * E-mail: (KD); (RB)
| | - Rita Warthemann
- Stem Cell Biology Unit, German Primate Center—Leibniz Institute for Primate Research, Göttingen, Germany
| | - Jana Lentes
- Stem Cell Biology Unit, German Primate Center—Leibniz Institute for Primate Research, Göttingen, Germany
| | - Gabriela Salinas-Riester
- Microarray and Deep-Sequencing Core Facility, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Ralf Dressel
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Germany
| | - Daniel Langenstroth
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Jörg Gromoll
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Erika Sasaki
- Department of Applied Developmental Biology, Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan, Keio Advanced Research Center, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Rüdiger Behr
- Stem Cell Biology Unit, German Primate Center—Leibniz Institute for Primate Research, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
- * E-mail: (KD); (RB)
| |
Collapse
|
18
|
Sasaki E. Prospects for genetically modified non-human primate models, including the common marmoset. Neurosci Res 2015; 93:110-5. [PMID: 25683291 DOI: 10.1016/j.neures.2015.01.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 01/01/2023]
Abstract
Genetically modified mice have contributed much to studies in the life sciences. In some research fields, however, mouse models are insufficient for analyzing the molecular mechanisms of pathology or as disease models. Often, genetically modified non-human primate (NHP) models are desired, as they are more similar to human physiology, morphology, and anatomy. Recent progress in studies of the reproductive biology in NHPs has enabled the introduction of exogenous genes into NHP genomes or the alteration of endogenous NHP genes. This review summarizes recent progress in the production of genetically modified NHPs, including the common marmoset, and future perspectives for realizing genetically modified NHP models for use in life sciences research.
Collapse
Affiliation(s)
- Erika Sasaki
- Advanced Research Center, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Center of Applied Developmental Biology, Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki, Kanagawa 210-0821, Japan.
| |
Collapse
|
19
|
Walker L, Baumgartner L, Keller KC, Ast J, Trettner S, Zur Nieden NI. Non-human primate and rodent embryonic stem cells are differentially sensitive to embryotoxic compounds. Toxicol Rep 2014; 2:165-174. [PMID: 28962348 PMCID: PMC5598278 DOI: 10.1016/j.toxrep.2014.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/27/2014] [Accepted: 11/21/2014] [Indexed: 01/08/2023] Open
Abstract
Many industrial chemicals and their respective by-products need to be comprehensively evaluated for toxicity using reliable and efficient assays. In terms of teratogenicity evaluations, the murine-based embryonic stem cell test (EST) offers a promising solution to screen for multiple tissue endpoints. However, use of a mouse model in the EST can yield only a limited understanding of human development, anatomy, and physiology. Non-human primate or human in vitro models have been suggested to be a pharmacologically and pathophysiologically desirable alternative to murine in vitro models. Here, we comparatively evaluated the sensitivity of embryonic stem cells (ESCs) of a non-human primate to skeletal teratogens with mouse ESCs hypothesizing that inclusion of non-human primate cells in in vitro tests would increase the reliability of safety predictions for humans. First, osteogenic capacity was compared between ESCs from the mouse and a New World monkey, the common marmoset. Then, cells were treated with compounds that have been previously reported to induce bone teratogenicity. Calcification and MTT assays evaluated effects on osteogenesis and cell viability, respectively. Our data indicated that marmoset ESCs responded differently than mouse ESCs in such embryotoxicity screens with no obvious dependency on chemical or compound classes and thus suggest that embryotoxicity screening results could be affected by species-driven response variation. In addition, ESCs derived from rhesus monkey, an Old World monkey, and phylogenetically closer to humans than the marmoset, were observed to respond differently to test compounds than marmoset ESCs. Together these results indicate that there are significant differences in the responses of non-human primate and mouse ESC to embryotoxic agents.
Collapse
Affiliation(s)
- Lauren Walker
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA 92521, USA.,Environmental Toxicology Graduate Program, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA 92521, USA
| | - Laura Baumgartner
- Fraunhofer Institute for Cell Therapy & Immunology, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Kevin C Keller
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA 92521, USA
| | - Julia Ast
- Fraunhofer Institute for Cell Therapy & Immunology, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Susanne Trettner
- Fraunhofer Institute for Cell Therapy & Immunology, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Nicole I Zur Nieden
- Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA 92521, USA.,Environmental Toxicology Graduate Program, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA 92521, USA.,Fraunhofer Institute for Cell Therapy & Immunology, Perlickstrasse 1, 04103 Leipzig, Germany
| |
Collapse
|
20
|
Encapsulating non-human primate multipotent stromal cells in alginate via high voltage for cell-based therapies and cryopreservation. PLoS One 2014; 9:e107911. [PMID: 25259731 PMCID: PMC4178041 DOI: 10.1371/journal.pone.0107911] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/21/2014] [Indexed: 01/24/2023] Open
Abstract
Alginate cell-based therapy requires further development focused on clinical application. To assess engraftment, risk of mutations and therapeutic benefit studies should be performed in an appropriate non-human primate model, such as the common marmoset (Callithrix jacchus). In this work we encapsulated amnion derived multipotent stromal cells (MSCs) from Callithrix jacchus in defined size alginate beads using a high voltage technique. Our results indicate that i) alginate-cell mixing procedure and cell concentration do not affect the diameter of alginate beads, ii) encapsulation of high cell numbers (up to 10×106 cells/ml) can be performed in alginate beads utilizing high voltage and iii) high voltage (15–30 kV) does not alter the viability, proliferation and differentiation capacity of MSCs post-encapsulation compared with alginate encapsulated cells produced by the traditional air-flow method. The consistent results were obtained over the period of 7 days of encapsulated MSCs culture and after cryopreservation utilizing a slow cooling procedure (1 K/min). The results of this work show that high voltage encapsulation can further be maximized to develop cell-based therapies with alginate beads in a non-human primate model towards human application.
Collapse
|
21
|
Aeckerle N, Drummer C, Debowski K, Viebahn C, Behr R. Primordial germ cell development in the marmoset monkey as revealed by pluripotency factor expression: suggestion of a novel model of embryonic germ cell translocation. Mol Hum Reprod 2014; 21:66-80. [PMID: 25237007 PMCID: PMC4275041 DOI: 10.1093/molehr/gau088] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Primordial germ cells (PGCs) are the embryonic progenitors of sperm and egg cells. Mammalian PGCs are thought to actively migrate from the yolk sac endoderm over long distances across the embryo to reach the somatic genital ridges. The general principles of mammalian PGC development were discovered in mice. In contrast, little is known about PGC development in primates due to extremely limited access to primate embryos. Here, we analyzed 12 well preserved marmoset monkey (Callithrix jacchus) embryos covering the phase from PGC emergence in the endoderm to the formation of the sexually differentiated gonad (embryonic day (E) 50 to E95). We show using immunohistochemistry that the pluripotency factors OCT4A and NANOG specifically mark PGCs throughout the period studied. In contrast, SALL4 and LIN28 were first expressed ubiquitously and only later down-regulated in somatic tissues. We further show, for the first time, that PGCs are located in the endoderm in E50 embryos in close spatial proximity to the prospective genital ridge, making a long-range migration of PGCs dispensable. At E65, PGCs are already present in the primitive gonad, while significantly later embryonic stages still exhibit PGCs at their original endodermal site, revealing a wide spatio-temporal window of PGC distribution. Our findings challenge the ‘dogma’ of active long-range PGC migration from the endoderm to the gonads. We therefore favor an alternative model based primarily on passive translocation of PGCs from the mesenchyme that surrounds the gut to the prospective gonad through the intercalar expansion of mesenchymal tissue which contains the PGCs. In summary, we (i) show differential pluripotency factor expression during primate embryo development and (ii) provide a schematic model for embryonic PGC translocation.
Collapse
Affiliation(s)
- N Aeckerle
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - C Drummer
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - K Debowski
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - C Viebahn
- Department of Anatomy and Embryology, Center of Anatomy, University of Göttingen, Kreuzbergring 36, 37075 Göttingen, Germany
| | - R Behr
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| |
Collapse
|
22
|
Irie N, Tang WWC, Azim Surani M. Germ cell specification and pluripotency in mammals: a perspective from early embryogenesis. Reprod Med Biol 2014; 13:203-215. [PMID: 25298745 PMCID: PMC4182624 DOI: 10.1007/s12522-014-0184-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/19/2014] [Indexed: 12/01/2022] Open
Abstract
Germ cells are unique cell types that generate a totipotent zygote upon fertilization, giving rise to the next generation in mammals and many other multicellular organisms. How germ cells acquire this ability has been of considerable interest. In mammals, primordial germ cells (PGCs), the precursors of sperm and oocytes, are specified around the time of gastrulation. PGCs are induced by signals from the surrounding extra-embryonic tissues to the equipotent epiblast cells that give rise to all cell types. Currently, the mechanism of PGC specification in mammals is best understood from studies in mice. Following implantation, the epiblast cells develop as an egg cylinder while the extra-embryonic ectoderm cells which are the source of important signals for PGC specification are located over the egg cylinder. However, in most cases, including humans, the epiblast cells develop as a planar disc, which alters the organization and the source of the signaling for cell fates. This, in turn, might have an effect on the precise mechanism of PGC specification in vivo as well as in vitro using pluripotent embryonic stem cells. Here, we discuss how the key early embryonic differences between rodents and other mammals may affect the establishment of the pluripotency network in vivo and in vitro, and consequently the basis for PGC specification, particularly from pluripotent embryonic stem cells in vitro.
Collapse
Affiliation(s)
- Naoko Irie
- Wellcome Trust/Cancer Research UK, Gurdon InstituteUniversity of CambridgeTennis Court RoadCB2 1QNCambridgeUK
| | - Walfred W. C. Tang
- Wellcome Trust/Cancer Research UK, Gurdon InstituteUniversity of CambridgeTennis Court RoadCB2 1QNCambridgeUK
| | - M. Azim Surani
- Wellcome Trust/Cancer Research UK, Gurdon InstituteUniversity of CambridgeTennis Court RoadCB2 1QNCambridgeUK
| |
Collapse
|
23
|
Fereydouni B, Drummer C, Aeckerle N, Schlatt S, Behr R. The neonatal marmoset monkey ovary is very primitive exhibiting many oogonia. Reproduction 2014; 148:237-47. [PMID: 24840529 PMCID: PMC4086814 DOI: 10.1530/rep-14-0068] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oogonia are characterized by diploidy and mitotic proliferation. Human and mouse oogonia express several factors such as OCT4, which are characteristic of pluripotent cells. In human, almost all oogonia enter meiosis between weeks 9 and 22 of prenatal development or undergo mitotic arrest and subsequent elimination from the ovary. As a consequence, neonatal human ovaries generally lack oogonia. The same was found in neonatal ovaries of the rhesus monkey, a representative of the old world monkeys (Catarrhini). By contrast, proliferating oogonia were found in adult prosimians (now called Strepsirrhini), which is a group of ‘lower’ primates. The common marmoset monkey (Callithrix jacchus) belongs to the new world monkeys (Platyrrhini) and is increasingly used in reproductive biology and stem cell research. However, ovarian development in the marmoset monkey has not been widely investigated. Herein, we show that the neonatal marmoset ovary has an extremely immature histological appearance compared with the human ovary. It contains numerous oogonia expressing the pluripotency factors OCT4A, SALL4, and LIN28A (LIN28). The pluripotency factor-positive germ cells also express the proliferation marker MKI67 (Ki-67), which has previously been shown in the human ovary to be restricted to premeiotic germ cells. Together, the data demonstrate the primitiveness of the neonatal marmoset ovary compared with human. This study may introduce the marmoset monkey as a non-human primate model to experimentally study the aspects of primate primitive gonad development, follicle assembly, and germ cell biology in vivo.
Collapse
Affiliation(s)
- B Fereydouni
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - C Drummer
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - N Aeckerle
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - S Schlatt
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - R Behr
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| |
Collapse
|
24
|
Trettner S, Findeisen A, Taube S, Horn PA, Sasaki E, zur Nieden NI. Osteogenic induction from marmoset embryonic stem cells cultured in feeder-dependent and feeder-independent conditions. Osteoporos Int 2014; 25:1255-66. [PMID: 24531422 DOI: 10.1007/s00198-013-2566-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/22/2013] [Indexed: 12/20/2022]
Abstract
UNLABELLED Embryonic stem cells (ESCs) have become increasingly attractive for cell replacement therapies of osteodegenerative diseases; however, pre-clinical studies in large animal models to repair diseased or injured bone are lacking. As a first step into this direction, we describe here the feeder-free cultivation and directed osteogenic differentiation of marmoset ESCs. INTRODUCTION Owing to their potential to self-renew and their enormous differentiation capability, ESCs are an adequate cell source for cell replacement therapies. To implement stem cell technology clinically, standardized cultivation and differentiation protocols and appropriate animal models are needed. Here, we describe the feeder-free cultivation of Callithrix jacchus ESCs (cESCs) in a chemically defined medium and their subsequent osteogenic differentiation. METHODS cESCs were maintained on mouse embryonic fibroblast feeder layers or in feeder-free conditions with activin A and basic fibroblast growth factor. Differentiation into mature osteoblasts was steered with ascorbic acid, β-glycerophosphate and 1α,25-(OH)2 vitamin D3 employing various induction strategies. RESULTS In feeder-free conditions, cESCs maintained pluripotency as indicated by Oct-4 and Nanog expression, positive immunostaining for typical primate ESC markers and high telomerase activity. Cells also remained karyotypically normal after 40 passages without feeder cells. The hanging drop protocol as well as omitting the embryoid body step proved unsuccessful to initiate osteogenic differentiation. The highest degree of osteogenesis was achieved by formation of embryoid bodies employing the cell cluster technique as indicated by the amount of deposited calcium and bone marker gene expression. Early addition of retinoic acid further improved the yield of osteoblasts and led to an increase in calcium deposition. CONCLUSIONS The osteogenic differentiation potential of feeder-free cESCs was equal if not higher compared to cells grown on feeders. These findings open the field for near clinical transplantation studies in primate models to evaluate the effectiveness of ESC-derived osteoblasts.
Collapse
Affiliation(s)
- S Trettner
- Group Applied Stem Cell Technology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Analysis of essential pathways for self-renewal in common marmoset embryonic stem cells. FEBS Open Bio 2014; 4:213-9. [PMID: 24649403 PMCID: PMC3958738 DOI: 10.1016/j.fob.2014.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 02/12/2014] [Accepted: 02/12/2014] [Indexed: 11/20/2022] Open
Abstract
Common marmoset (CM) is widely recognized as a useful non-human primate for disease modeling and preclinical studies. Thus, embryonic stem cells (ESCs) derived from CM have potential as an appropriate cell source to test human regenerative medicine using human ESCs. CM ESCs have been established by us and other groups, and can be cultured in vitro. However, the growth factors and downstream pathways for self-renewal of CM ESCs are largely unknown. In this study, we found that basic fibroblast growth factor (bFGF) rather than leukemia inhibitory factor (LIF) promoted CM ESC self-renewal via the activation of phosphatidylinositol-3-kinase (PI3K)-protein kinase B (AKT) pathway on mouse embryonic fibroblast (MEF) feeders. Moreover, bFGF and transforming growth factor β (TGFβ) signaling pathways cooperatively maintained the undifferentiated state of CM ESCs under feeder-free condition. Our findings may improve the culture techniques of CM ESCs and facilitate their use as a preclinical experimental resource for human regenerative medicine.
Collapse
Key Words
- AKT, protein kinase B
- CM, common marmoset
- Common marmoset
- EB, embryoid body
- ERK, extracellular signal-regulated kinase
- ESCs, embryonic stem cells
- Embryonic stem cells
- EpiSCs, epiblast stem cells
- FCM, flow cytometry
- JAK, janus kinase
- KSR, knockout serum replacement
- LIF, leukemia inhibitory factor
- MEFs, mouse embryonic fibroblasts
- MEK, mitogen-activated protein/extracellular signal-regulated kinase kinase
- PI3K, phosphatidylinositol-3-kinase
- RT-PCR, reverse transcription-polymerase chain reaction
- SMAD2/3, mothers against decapentaplegic homolog 2/3
- STAT3, signal transducer and activator of transcription 3
- Self-renewal
- TGFβ
- TGFβ, transforming growth factor β
- bFGF
- bFGF, basic fibroblast growth factor
- iPSCs, induced pluripotent stem cells
Collapse
|
26
|
Ben-Yehudah A, Campanaro BM, Wakefield LM, Kinney TN, Brekosky J, Eisinger VM, Castro CA, Carlisle DL. Nicotine exposure during differentiation causes inhibition of N-myc expression. Respir Res 2013; 14:119. [PMID: 24499207 PMCID: PMC3828478 DOI: 10.1186/1465-9921-14-119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 10/11/2013] [Indexed: 11/10/2022] Open
Abstract
Background The ability of chemicals to disrupt neonatal development can be studied using embryonic stem cells (ESC). One such chemical is nicotine. Prenatal nicotine exposure is known to affect postnatal lung function, although the mechanisms by which it has this effect are not clear. Since fibroblasts are a critical component of the developing lung, providing structure and secreting paracrine factors that are essential to epithelialization, this study focuses on the differentiation of ESC into fibroblasts using a directed differentiation protocol. Methods Fibroblasts obtained from non-human primate ESC (nhpESC) differentiation were analyzed by immunohistochemistry, immunostaining, Affymetrix gene expression array, qPCR, and immunoblotting. Results Results of these analyses demonstrated that although nhpESCs differentiate into fibroblasts in the presence of nicotine and appear normal by some measures, including H&E and SMA staining, they have an altered gene expression profile. Network analysis of expression changes demonstrated an over-representation of cell-cycle related genes with downregulation of N-myc as a central regulator in the pathway. Further investigation demonstrated that cells differentiated in the presence of nicotine had decreased N-myc mRNA and protein expression and longer doubling times, a biological effect consistent with downregulation of N-myc. Conclusions This study is the first to use primate ESC to demonstrate that nicotine can affect cellular differentiation from pluripotency into fibroblasts, and in particular, mediate N-myc expression in differentiating ESCs. Given the crucial role of fibroblasts throughout the body, this has important implications for the effect of cigarette smoke exposure on human development not only in the lung, but in organogenesis in general.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Diane L Carlisle
- Division of Developmental and Regenerative Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
27
|
Navara CS, Hornecker J, Grow D, Chaudhari S, Hornsby PJ, Ichida JK, Eggan K, McCarrey JR. Derivation of induced pluripotent stem cells from the baboon: a nonhuman primate model for preclinical testing of stem cell therapies. Cell Reprogram 2013; 15:495-502. [PMID: 24182315 DOI: 10.1089/cell.2012.0093] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Development of effective pluripotent stem cell-based therapies will require safety and efficacy testing in a clinically relevant preclinical model such as nonhuman primates (NHPs). Baboons and macaques are equally similar to humans genetically and both have been extensively used for biomedical research. Macaques are preferred for human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) research whereas baboons are preferred for transplantation studies because of the greater similarity of their anatomy and immunogenetic system to those of humans. We generated four induced pluripotent stem cell (iPSC) lines from skin cells of the olive baboon (Papio anubis). Each line shows the distinct morphology of primate pluripotent stem cells, including flat colonies with well-defined borders and a high nuclear/cytoplasm ratio. Each is positive for the pluripotency markers OCT4, SOX2, NANOG, and SSEA4. Pluripotency was confirmed in two lines by teratoma formation with representative tissues from each germ layer, whereas a third produced cells from all three germ layers following embryoid body differentiation. Three lines have a normal male karyotype and the fourth is missing the short arm of one copy of chromosome 18. This may serve as an in vitro model for the human developmental disorder 18p-, which impacts 1 in 50,000 births/year. These iPSC lines represent the first step toward establishing the baboon as a NHP model for developing stem cell-based therapies.
Collapse
Affiliation(s)
- Christopher S Navara
- 1 Department of Biology, University of Texas at San Antonio , San Antonio, TX 78249
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Imamura M, Hikabe O, Lin ZYC, Okano H. Generation of germ cells in vitro in the era of induced pluripotent stem cells. Mol Reprod Dev 2013; 81:2-19. [PMID: 23996404 DOI: 10.1002/mrd.22259] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 08/21/2013] [Indexed: 01/15/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are stem cells that can be artificially generated via "cellular reprogramming" using gene transduction in somatic cells. iPSCs have enormous potential in stem-cell biology as they can give rise to numerous cell lineages, including the three germ layers. An evaluation of germ-line competency by blastocyst injection or tetraploid complementation, however, is critical for determining the developmental potential of mouse iPSCs towards germ cells. Recent studies have demonstrated that primordial germ cells obtained by the in vitro differentiation of iPSCs produce functional gametes as well as healthy offspring. These findings illustrate not only that iPSCs are developmentally similar to embryonic stem cells (ESCs), but also that somatic cells from adult tissues can produce gametes in vitro, that is, if they are reprogrammed into iPSCs. In this review, we discuss past and recent advances in the in vitro differentiation of germ cells using pluripotent stem cells, with an emphasis on ESCs and iPSCs. While this field of research is still at a stage of infancy, it holds great promises for investigating the mechanisms of germ-cell development, especially in humans, and for advancing reproductive and developmental engineering technologies in the future.
Collapse
Affiliation(s)
- Masanori Imamura
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
29
|
Wiedemann A, Hemmer K, Bernemann I, Göhring G, Pogozhykh O, Figueiredo C, Glage S, Schambach A, Schwamborn JC, Blasczyk R, Müller T. Induced pluripotent stem cells generated from adult bone marrow-derived cells of the nonhuman primate (Callithrix jacchus) using a novel quad-cistronic and excisable lentiviral vector. Cell Reprogram 2013. [PMID: 23194452 DOI: 10.1089/cell.2012.0036] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Regenerative medicine is in need of solid, large animal models as a link between rodents and humans to evaluate the functionality, immunogenicity, and clinical safety of stem cell-derived cell types. The common marmoset (Callithrix jacchus) is an excellent large animal model, genetically close to humans and readily used worldwide in clinical research. Until now, only two groups showed the generation of induced pluripotent stem cells (iPSCs) from the common marmoset using integrating retroviral vectors. Therefore, we reprogrammed bone marrow-derived mesenchymal cells (MSCs) of adult marmosets in the presence of TAV, SB431542, PD0325901, and ascorbic acid via a novel, excisable lentiviral spleen focus-forming virus (SFFV)-driven quad-cistronic vector system (OCT3/4, KLF4, SOX2, C-MYC). Endogenous pluripotency markers like OCT3/4, KLF4, SOX2, C-MYC, LIN28, NANOG, and strong alkaline phosphatase signals were detected. Exogenous genes were silenced and additionally the cassette was removed with a retroviral Gag precursor system. The cell line could be cultured in absence of leukemia inhibitory factor (LIF) and basic fibroblast growth factor (bFGF) and could be successfully differentiated into embryoid bodies and teratomas with presence of all three germ layers. Directed differentiation generated neural progenitors, megakaryocytes, adipocytes, chondrocytes, and osteogenic cells. Thus, all criteria for fully reprogrammed bone marrow-MSCs of a nonhuman primate with a genetically sophisticated construct could be demonstrated. These cells will be a promising tool for future autologous transplantations.
Collapse
Affiliation(s)
- Anastasia Wiedemann
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Virant-Klun I, Stimpfel M, Cvjeticanin B, Vrtacnik-Bokal E, Skutella T. Small SSEA-4-positive cells from human ovarian cell cultures: related to embryonic stem cells and germinal lineage? J Ovarian Res 2013; 6:24. [PMID: 23570331 PMCID: PMC3660272 DOI: 10.1186/1757-2215-6-24] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 03/24/2013] [Indexed: 01/06/2023] Open
Abstract
Background It has already been found that very small embyronic-like stem cells (VSELs) are present in adult human tissues and organs. The aim of this study was to find if there exists any similar population of cells in cell cultures of reproductive tissues and embryonic stem cells, and if these cells have any relation to pluripotency and germinal lineage. Methods and results Here we report that a population of small SSEA-4-positive cells with diameters of up to 4 μm was isolated by fluorescence-activated cell sorting (FACS) from the human ovarian cell cultures after enzymatic degradation of adult cortex tissues. These small cells – putative ovarian stem cells – were also observed during cell culturing of up to 6 months and more. In general, small putative ovarian stem cells, isolated by FACS, showed a relatively low gene expression profile when compared to human embryonic stem cells (hESCs) and human adult fibroblasts; this may reflect the quiescent state of these cells. In spite of that, small putative ovarian stem cells expressed several genes related to primordial germ cells (PGCs), pluripotency and germinal lineage, including VASA. The PGC-related gene PRDM1 was strongly expressed in small putative ovarian stem cells; in both hESCs and fibroblasts it was significantly down-regulated. In addition, putative ovarian stem cells expressed other PGC-related genes, such as PRDM14 and DPPA3. Most of the pluripotency and germinal lineage-related genes were up-regulated in hESCs (except VASA). When compared to fibroblasts, there were several pluripotency-related genes, which were up-regulated in small putative ovarian stem cells. Similar populations of small cells were also isolated by FACS from human testicular and hESC cultures. Conclusions Our results confirm the potential embryonic-like character of small putative stem cells isolated from human adult ovaries and their possible relation to germinal lineage.
Collapse
Affiliation(s)
- Irma Virant-Klun
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, Slajmerjeva 3, Ljubljana, 1000, Republic of Slovenia
| | - Martin Stimpfel
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, Slajmerjeva 3, Ljubljana, 1000, Republic of Slovenia
| | - Branko Cvjeticanin
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, Slajmerjeva 3, Ljubljana, 1000, Republic of Slovenia
| | - Eda Vrtacnik-Bokal
- Department of Obstetrics and Gynecology, University Medical Centre Ljubljana, Slajmerjeva 3, Ljubljana, 1000, Republic of Slovenia
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Im Neuenheimer Feld 307, Heidelberg, 69120, Germany
| |
Collapse
|
31
|
Diekmann U, Naujok O, Blasczyk R, Müller T. Embryonic stem cells of the non-human primate Callithrix jacchus can be differentiated into definitive endoderm by Activin-A but not IDE-1/2. J Tissue Eng Regen Med 2013; 9:473-9. [PMID: 23418163 DOI: 10.1002/term.1709] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 11/01/2012] [Accepted: 12/20/2012] [Indexed: 12/14/2022]
Abstract
Pluripotent stem cells hold great promise for regenerative medicine, due to their unlimited self-renewal potential and the ability to differentiate into all somatic cell types. Differences between the rodent disease models and the situation in humans can be narrowed down with non-human primate models. The common marmoset monkey (Callithrix jacchus) is an interesting model for biomedical research because these animals are easy to breed, get relatively old (≤ 13 years), are small in size, are relatively cost-effective and have a high genetic proximity to the human. In particular, diseases of the liver and pancreas are interesting for cell replacement therapies but the in vitro differentiation of ESCs into the definitive endoderm germ layer is still a demanding task. Membrane-permeable, chemically defined small molecules can possibly replace recombinant growth factors used in most directed differentiation protocols. However, the potent small molecules IDE-1 and IDE-2 were not able to induce definitive endoderm-like cells when ESCs from the common marmoset were treated with these compounds, whereas the recombinant growth factor Activin A could force the differentiation into this lineage. Our results indicate that ESCs from the common marmoset are less sensitive or even insensitive to these small molecules. Thus, differences between the species of human ESCs and ESCs of this non-human primate might be a useful model to further evaluate the exact mode of action of these compounds.
Collapse
Affiliation(s)
- Ulf Diekmann
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany; Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | | | | | |
Collapse
|
32
|
Vogt EJ, Meglicki M, Hartung KI, Borsuk E, Behr R. Importance of the pluripotency factor LIN28 in the mammalian nucleolus during early embryonic development. Development 2013; 139:4514-23. [PMID: 23172912 PMCID: PMC3912245 DOI: 10.1242/dev.083279] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The maternal nucleolus is required for proper activation of the embryonic genome (EGA) and early embryonic development. Nucleologenesis is characterized by the transformation of a nucleolar precursor body (NPB) to a mature nucleolus during preimplantation development. However, the function of NPBs and the involved molecular factors are unknown. We uncover a novel role for the pluripotency factor LIN28, the biological significance of which was previously demonstrated in the reprogramming of human somatic cells to induced pluripotent stem (iPS) cells. Here, we show that LIN28 accumulates at the NPB and the mature nucleolus in mouse preimplantation embryos and embryonic stem cells (ESCs), where it colocalizes with the nucleolar marker B23 (nucleophosmin 1). LIN28 has nucleolar localization in non-human primate (NHP) preimplantation embryos, but is cytoplasmic in NHP ESCs. Lin28 transcripts show a striking decline before mouse EGA, whereas LIN28 protein localizes to NPBs at the time of EGA. Following knockdown with a Lin28 morpholino, the majority of embryos arrest between the 2- and 4-cell stages and never develop to morula or blastocyst. Lin28 morpholino-injected embryos arrested at the 2-cell stage were not enriched with nucleophosmin at presumptive NPB sites, indicating that functional NPBs were not assembled. Based on these results, we propose that LIN28 is an essential factor of nucleologenesis during early embryonic development.
Collapse
Affiliation(s)
- Edgar J Vogt
- German Primate Center, Stem Cell Biology Unit, Kellnerweg 4, 37077 Goettingen, Germany.
| | | | | | | | | |
Collapse
|
33
|
Warthemann R, Eildermann K, Debowski K, Behr R. False-positive antibody signals for the pluripotency factor OCT4A (POU5F1) in testis-derived cells may lead to erroneous data and misinterpretations. Mol Hum Reprod 2012; 18:605-12. [PMID: 22933709 PMCID: PMC3497886 DOI: 10.1093/molehr/gas032] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Octamer-binding protein 4 (OCT4) is a key player in pluripotent embryonic stem (ES) cells and is essential for the generation of induced pluripotent stem cells. Recently, several reports indicated the spontaneous recovery of pluripotency in cultured adult human testis-derived cells. This was evidenced also by the detection of OCT4 using antibodies. However, the soundness of some data was recently put into question. During our attempts to derive pluripotent cells from the common marmoset monkey (Callithrix jacchus) testis, we obtained inconsistent data which prompted us to analyze deeper the characteristics of three independent OCT4 antibodies that were used in numerous published studies that received greatest attention. All antibodies detected OCT4 by immunofluorescence (IF) in a marmoset monkey ES cell line. Two of the three OCT4 antibodies also gave robust nuclear signals in testis-derived cells. However, the latter cells expressed no OCT4 mRNA as revealed by quantitative RT-PCR and turned out to be mesenchymal cells. When tested in western blot analyses, all antibodies detected heterologously expressed marmoset monkey OCT4 protein. But, importantly, those antibodies that resulted in non-specific signals in IF also showed additional non-specific bands in western blots. In summary, some commercially available OCT4 antibodies result in false-positive signals which may provoke erroneous conclusions when used in studies aiming at the generation of pluripotent cells in vitro. We conclude that (i) antibodies must be carefully characterized before use to prevent misleading observations and (ii) OCT4 expression must be monitored by a second antibody-independent method.
Collapse
Affiliation(s)
- R Warthemann
- Stem Cell Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, D-37077 Göttingen, Germany
| | | | | | | |
Collapse
|
34
|
Aeckerle N, Eildermann K, Drummer C, Ehmcke J, Schweyer S, Lerchl A, Bergmann M, Kliesch S, Gromoll J, Schlatt S, Behr R. The pluripotency factor LIN28 in monkey and human testes: a marker for spermatogonial stem cells? Mol Hum Reprod 2012; 18:477-88. [PMID: 22689537 PMCID: PMC3457707 DOI: 10.1093/molehr/gas025] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Mammalian spermatogenesis is maintained by spermatogonial stem cells (SSCs). However, since evidentiary assays and unequivocal markers are still missing in non-human primates (NHPs) and man, the identity of primate SSCs is unknown. In contrast, in mice, germ cell transplantation studies have functionally demonstrated the presence of SSCs. LIN28 is an RNA-binding pluripotent stem cell factor, which is also strongly expressed in undifferentiated mouse spermatogonia. By contrast, two recent reports indicated that LIN28 is completely absent from adult human testes. Here, we analyzed LIN28 expression in marmoset monkey (Callithrix jacchus) and human testes during development and adulthood and compared it with that in mice. In the marmoset, LIN28 was strongly expressed in migratory primordial germ cells and gonocytes. Strikingly, we found a rare LIN28-positive subpopulation of spermatogonia also in adult marmoset testis. This was corroborated by western blotting and quantitative RT–PCR. Importantly, in contrast to previous publications, we found LIN28-positive spermatogonia also in normal adult human and additional adult NHP testes. Some seasonal breeders exhibit a degenerated (involuted) germinal epithelium consisting only of Sertoli cells and SSCs during their non-breeding season. The latter re-initiate spermatogenesis prior to the next breeding-season. Fully involuted testes from a seasonal hamster and NHP (Lemur catta) exhibited numerous LIN28-positive spermatogonia, indicating an SSC identity of the labeled cells. We conclude that LIN28 is differentially expressed in mouse and NHP spermatogonia and might be a marker for a rare SSC population in NHPs and man. Further characterization of the LIN28-positive population is required.
Collapse
Affiliation(s)
- N Aeckerle
- Stem Cell Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, D-37077 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Eildermann K, Aeckerle N, Debowski K, Godmann M, Christiansen H, Heistermann M, Schweyer S, Bergmann M, Kliesch S, Gromoll J, Ehmcke J, Schlatt S, Behr R. Developmental expression of the pluripotency factor sal-like protein 4 in the monkey, human and mouse testis: restriction to premeiotic germ cells. Cells Tissues Organs 2012; 196:206-20. [PMID: 22572102 DOI: 10.1159/000335031] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2011] [Indexed: 01/09/2023] Open
Abstract
SALL4 (sal-like protein 4) is a pluripotency transcription factor, which is highly expressed in embryonic stem (ES) cells and which is essential for mouse preimplantation development. In adult mouse organs, Sall4 mRNA is highly expressed in the testis and ovary, while there is only little or no expression in other organs. There is also a high expression of SALL4 in human testicular germ cell tumors. However, there is as yet no detailed analysis of SALL4 expression during mammalian testicular development. We analyzed SALL4 expression in ES cells, preimplantation embryos, and the developing and adult testis of a nonhuman primate (NHP) species, the common marmoset monkey (Callithrix jacchus). Immunofluorescence revealed SALL4 in the nuclei of marmoset ES cells and preimplantation embryos. Marmoset SALL4 isoform analysis in ES cells and newborn and adult testis by RT- PCR and Western blotting showed two different isoforms, SALL4-A and SALL4-B. Immunohistochemistry localized this transcription factor to the nuclei of primordial germ cells and most gonocytes in the prenatal and early postnatal marmoset testis. In the pubertal and adult testis SALL4 was present in undifferentiated spermatogonia. In the developing and adult human and mouse testis SALL4 expression mimicked the pattern in the marmoset. Adult testes from additional NHP species, the treeshrew, the cat and the dog also exhibited SALL4 in undifferentiated spermatogonia, indicating a conserved expression in the mammalian testis. Taking into account the importance of SALL4 for mouse development, we conclude that SALL4 may play an important role during mammalian germ cell development and is involved in the regulation of spermatogonial proliferation in the adult testis.
Collapse
Affiliation(s)
- K Eildermann
- Stem Cell Biology Unit, German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Sweeney C, Ward J, Vallender EJ. Naturally occurring, physiologically normal, primate chimeras. CHIMERISM 2012; 3:43-4. [PMID: 22627807 DOI: 10.4161/chim.20729] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Callitrichids, South American primates including marmosets and tamarins, have evolved a unique physiology. Twins share a placenta during gestation and exchange stem cells, resulting in naturally occurring chimeric adults. Our study used a quantitative PCR-based assay to address whether this chimerism was restricted to blood and other cells of the hematopoietic lineage or whether it extended to other somatic tissues. These studies help to characterize species that have adapted evolutionarily to pervasive chimerism, with every individual healthy and unperturbed. This experiment of evolution offers insight into transplantation and histocompatibility, reproductive biology and behavior, and innate and adaptive immunity.
Collapse
Affiliation(s)
- Carolyn Sweeney
- New England Primate Research Center, Harvard Medical School, Southborough, MA, USA
| | | | | |
Collapse
|
37
|
Eildermann K, Gromoll J, Behr R. Misleading and reliable markers to differentiate between primate testis-derived multipotent stromal cells and spermatogonia in culture. Hum Reprod 2012; 27:1754-67. [PMID: 22442249 PMCID: PMC3357197 DOI: 10.1093/humrep/des091] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Several studies have reported the generation of spermatogonia-derived pluripotent stem cells from human testes. The initial aim of the present study was the derivation of equivalent stem cells from an established and experimentally accessible non-human primate model, the common marmoset monkey (Callithrix jacchus). However, an essential prerequisite in the absence of transgenic reporters in primates and man is the availability of validated endogenous markers for the identification of specific cell types in vitro. METHODS AND RESULTS We cultured marmoset testicular cells in a similar way to that described for human testis-derived pluripotent cells and set out to characterize these cultures under different conditions and in differentiation assays applying established marker panels. Importantly, the cells emerged as testicular multipotent stromal cells (TMSCs) instead of (pluripotent) germ cell-derived cells. TMSCs expressed many markers such as GFR-α, GPR125, THY-1 (CD90), ITGA6, SSEA4 and TRA-1-81, which were considered as spermatogonia specific and were previously used for the enrichment or characterization of spermatogonia. Proliferation of TMSCs was highly dependent on basic fibroblast growth factor, a growth factor routinely present in germ cell culture media. As reliable markers for the distinction between spermatogonia and TMSCs, we established VASA, in combination with the spermatogonia-expressed factors, MAGEA4, PLZF and SALL4. CONCLUSIONS Marmoset monkey TMSCs and spermatogonia exhibit an overlap of markers, which may cause erroneous interpretations of experiments with testis-derived stem cells in vitro. We provide a marker panel for the unequivocal identification of spermatogonia providing a better basis for future studies on primate, including human, testis-derived stem cells.
Collapse
Affiliation(s)
- K Eildermann
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, Göttingen, Germany
| | | | | |
Collapse
|
38
|
Derivation of neural progenitors and retinal pigment epithelium from common marmoset and human pluripotent stem cells. Stem Cells Int 2012; 2012:417865. [PMID: 22550507 PMCID: PMC3328338 DOI: 10.1155/2012/417865] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 11/28/2011] [Indexed: 12/23/2022] Open
Abstract
Embryonic and induced pluripotent stem cells (IPSCs) derived from mammalian species are valuable tools for modeling human disease, including retinal degenerative eye diseases that result in visual loss. Restoration of vision has focused on transplantation of neural progenitor cells (NPCs) and retinal pigmented epithelium (RPE) to the retina. Here we used transgenic common marmoset (Callithrix jacchus) and human pluripotent stem cells carrying the enhanced green fluorescent protein (eGFP) reporter as a model system for retinal differentiation. Using suspension and subsequent adherent differentiation cultures, we observed spontaneous in vitro differentiation that included NPCs and cells with pigment granules characteristic of differentiated RPE. Retinal cells derived from human and common marmoset pluripotent stem cells provide potentially unlimited cell sources for testing safety and immune compatibility following autologous or allogeneic transplantation using nonhuman primates in early translational applications.
Collapse
|
39
|
Plentz RR, Palagani V, Wiedemann A, Diekmann U, Glage S, Naujok O, Jörns A, Müller T. Islet microarchitecture and glucose transporter expression of the pancreas of the marmoset monkey display similarities to the human. Islets 2012; 4:123-9. [PMID: 22627676 DOI: 10.4161/isl.19254] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The common marmoset New World monkey (Callithrix jacchus), is a primate model with great potential for scientific research, including research on diabetes. However, in opposite to Rhesus and Java monkeys (Macaca mulatta and Macaca fascicularis) little is known about the marmosets islet microarchitecture, glucose transporter and pancreatic marker gene expression. In this work we analyze differences and similarities in size, shape, cellular composition and intra-islet topography between the common marmoset and the human endocrine pancreas. Different sized, circular and a-circular shaped islets of the common marmoset and human display α-cells in the whole islet organ leading to a ribbon-like islet type. The number of islets was significantly higher in the common marmoset compared with humans. However, the area of insulin-producing cells was significantly higher in the human pancreas. Intra-islet distribution pattern of δ- and β-cells was similar in both species. The morphology of the exocrine pancreas regarding acinar and ductal cells was quite similar as confirmed by ultrastructural analysis. Additionally the ultrastructure of secretory granules from α-, δ- and β-cells of human and non-human primate pancreas showed the same characteristics. Molecular analysis showed the presence of endocrine pancreatic marker genes like PMCA2, NCX1, SUR1, KIR6.2, MAFA, NGN3 and PDX1 also expressed in the human. For the first time we could show presence of Glut 5 and 9 transporters in addition to the low abundance transporter Glut2 and the highly expressed Glut1 glucose transporter. We propose that Callithrix jacchus displays a new animal model for diabetes research and regenerative medicine.
Collapse
Affiliation(s)
- Ruben R Plentz
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Lin ZYC, Imamura M, Sano C, Nakajima R, Suzuki T, Yamadera R, Takehara Y, Okano HJ, Sasaki E, Okano H. Molecular signatures to define spermatogenic cells in common marmoset (Callithrix jacchus). Reproduction 2012; 143:597-609. [PMID: 22323619 DOI: 10.1530/rep-11-0215] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Germ cell development is a fundamental process required to produce offspring. The developmental program of spermatogenesis has been assumed to be similar among mammals. However, recent studies have revealed differences in the molecular properties of primate germ cells compared with the well-characterized mouse germ cells. This may prevent simple application of rodent insights into higher primates. Therefore, thorough investigation of primate germ cells is necessary, as this may lead to the development of more appropriate animal models. The aim of this study is to define molecular signatures of spermatogenic cells in the common marmoset, Callithrix jacchus. Interestingly, NANOG, PRDM1, DPPA3 (STELLA), IFITM3, and ZP1 transcripts, but no POU5F1 (OCT4), were detected in adult marmoset testis. Conversely, mouse testis expressed Pou5f1 but not Nanog, Prdm1, Dppa3, Ifitm3, and Zp1. Other previously described mouse germ cell markers were conserved in marmoset and mouse testes. Intriguingly, marmoset spermatogenic cells underwent dynamic protein expression in a developmental stage-specific manner; DDX4 (VASA) protein was present in gonocytes, diminished in spermatogonial cells, and reexpressed in spermatocytes. To investigate epigenetic differences between adult marmoset and mice, DNA methylation analyses identified unique epigenetic profiles to marmoset and mice. Marmoset NANOG and POU5F1 promoters in spermatogenic cells exhibited a methylation status opposite to that in mice, while the DDX4 and LEFTY1 loci, as well as imprinted genes, displayed an evolutionarily conserved methylation pattern. Marmosets have great advantages as models for human reproductive biology and are also valuable as experimental nonhuman primates; thus, the current study provides an important platform for primate reproductive biology, including possible applications to humans.
Collapse
Affiliation(s)
- Zachary Yu-Ching Lin
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Takabayashi S, Katoh H. Sex identification using the ZFX and ZFY genes in common marmosets (Callithrix jacchus). Exp Anim 2011; 60:417-20. [PMID: 21791881 DOI: 10.1538/expanim.60.417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
We investigated sex determination via the ZFX and ZFY genes using PCR-RFLP in the common marmoset. We designed a novel primer set to detect ZFX and ZFY. A 483-bp band from the ZFX gene and a 471-bp band from the ZFY gene were amplified. Sequencing data of the products amplified from ZFX and ZFY showed the recognition sites of two restriction enzymes, DdeI and MseI, respectively. After digestion of the products using each enzyme, we found that the band patterns between females and males were different. PCR-based sex identification might provide a tool for further breeding studies and experimental embryological studies using marmosets.
Collapse
Affiliation(s)
- Shuji Takabayashi
- Experimental Animals Institute, Hamamatsu University School of Medicine. 1–20–1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | | |
Collapse
|
42
|
Radtke S, Horn PA. Cells, Niche, Fate: Meeting Report on the 6th International Meeting of the Stem Cell Network North Rhine Westphalia. Cell Reprogram 2011; 13:381-4. [DOI: 10.1089/cell.2011.0039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Stefan Radtke
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
43
|
Cryopreservation and quality control of mouse embryonic feeder cells. Cryobiology 2011; 63:104-10. [PMID: 21810414 DOI: 10.1016/j.cryobiol.2011.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 07/07/2011] [Accepted: 07/08/2011] [Indexed: 11/23/2022]
Abstract
Stem cell research is a highly promising and rapidly progressing field inside regenerative medicine. Embryonic stem cells (ESCs), reprogrammed "induced pluripotent" cells (iPS), or lately protein induced pluripotent cells (piPS) share one inevitable factor: mouse embryonic feeder cells (MEFs), which are commonly used for ESC long term culture procedures and colony regeneration. These MEFs originate from different mouse strains, are inactivated by different methods and are differently cryopreserved. Incomprehensibly, there are to date no established quality control parameters for MEFs to insure consistency of ESC experiments and culture. Hence, in this work, we developed a bench-top quality control for embryonic feeder cells. According to our findings, MEFs should be inactivated by irradiation (30Gy) and cryopreserved with optimal 10% DMSO at 1K/min freezing velocity. Thawed cells should be free of mycoplasma and should have above 85 ± 13.1% viability. Values for the metabolic activity should be above 150 ± 10.5% and for the combined gene expression of selected marker genes above 225 ± 43.8% compared to non-irradiated, cryopreserved controls. Cells matching these criteria can be utilized for at least 12 days for ESC culture without detaching from the culture dish or disruption of the cell layer.
Collapse
|
44
|
Bernemann I, Mueller T, Blasczyk R, Glasmacher B, Hofmann N. Colonization of collagen scaffolds by adipocytes derived from mesenchymal stem cells of the common marmoset monkey. Biochem Biophys Res Commun 2011; 411:317-22. [DOI: 10.1016/j.bbrc.2011.06.134] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 06/21/2011] [Indexed: 10/18/2022]
|
45
|
Wianny F, Bourillot PY, Dehay C. Embryonic stem cells in non-human primates: An overview of neural differentiation potential. Differentiation 2011; 81:142-52. [PMID: 21296479 DOI: 10.1016/j.diff.2011.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/18/2010] [Accepted: 01/11/2011] [Indexed: 12/11/2022]
Abstract
Non-human primate (NHP) embryonic stem (ES) cells show unlimited proliferative capacities and a great potential to generate multiple cell lineages. These properties make them an ideal resource both for investigating early developmental processes and for assessing their therapeutic potential in numerous models of degenerative diseases. They share the same markers and the same properties with human ES cells, and thus provide an invaluable transitional model that can be used to address the safety issues related to the clinical use of human ES cells. Here, we review the available information on the derivation and the specific features of monkey ES cells. We comment on the capacity of primate ES cells to differentiate into neural lineages and the current protocols to generate self-renewing neural stem cells. We also highlight the signalling pathways involved in the maintenance of these neural cell types. Finally, we discuss the potential of monkey ES cells for neuronal differentiation.
Collapse
Affiliation(s)
- Florence Wianny
- Inserm, U846, Stem Cell and Brain Research Institute, 18 Avenue Doyen Lépine, 69500 Bron, France.
| | | | | |
Collapse
|
46
|
Shiozawa S, Kawai K, Okada Y, Tomioka I, Maeda T, Kanda A, Shinohara H, Suemizu H, James Okano H, Sotomaru Y, Sasaki E, Okano H. Gene targeting and subsequent site-specific transgenesis at the β-actin (ACTB) locus in common marmoset embryonic stem cells. Stem Cells Dev 2011; 20:1587-99. [PMID: 21126169 DOI: 10.1089/scd.2010.0351] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nonhuman primate embryonic stem (ES) cells have vast promise for preclinical studies. Genetic modification in nonhuman primate ES cells is an essential technique for maximizing the potential of these cells. The common marmoset (Callithrix jacchus), a nonhuman primate, is expected to be a useful transgenic model for preclinical studies. However, genetic modification in common marmoset ES (cmES) cells has not yet been adequately developed. To establish efficient and stable genetic modifications in cmES cells, we inserted the enhanced green fluorescent protein (EGFP) gene with heterotypic lox sites into the β-actin (ACTB) locus of the cmES cells using gene targeting. The resulting knock-in ES cells expressed EGFP ubiquitously under the control of the endogenous ACTB promoter. Using inserted heterotypic lox sites, we demonstrated Cre recombinase-mediated cassette exchange (RMCE) and successfully established a monomeric red fluorescent protein (mRFP) knock-in cmES cell line. Further, a herpes simplex virus-thymidine kinase (HSV-tk) knock-in cmES cell line was established using RMCE. The growth of tumor cells originating from the cell line was significantly suppressed by the administration of ganciclovir. Therefore, the HSV-tk/ganciclovir system is promising as a safeguard for stem cell therapy. The stable and ubiquitous expression of EGFP before RMCE enables cell fate to be tracked when the cells are transplanted into an animal. Moreover, the creation of a transgene acceptor locus for site-specific transgenesis will be a powerful tool, similar to the ROSA26 locus in mice.
Collapse
Affiliation(s)
- Seiji Shiozawa
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chang TC, Liu YG, Eddy CA, Jacoby ES, Binkley PA, Brzyski RG, Schenken RS. Derivation and characterization of novel nonhuman primate embryonic stem cell lines from in vitro-fertilized baboon preimplantation embryos. Stem Cells Dev 2010; 20:1053-62. [PMID: 20874104 DOI: 10.1089/scd.2010.0372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The development of nonhuman primate (NHP) embryonic stem cell (ESC) models holds great promise for cell-mediated treatment of debilitating diseases and to address numerous unanswered questions regarding the therapeutic efficacy of ESCs while supplanting ethical considerations involved with human studies. Here we report successful establishment and characterization of 3 novel baboon (Papio cynocephalus) ESC lines from the inner cell mass of intracytoplasmic sperm injection-derived blastocysts. Embryos were cultured in an improved baboon embryo in vitro culture protocol. The inner cell mass of blastocyst was laser-dissected and plated on mouse embryonic fibroblast feeder cell monolayer in the NHP ESC culture medium. Three cell lines with characteristic ESC morphology have been cultured through an extended period (>14 months), with 2 male cell lines (UT-1 and -2) and 1 female cell line (UT-3) displaying normal baboon karyotypes. Reverse transcription-polymerase chain reaction analysis confirmed that all 3 lines express primate ESC pluripotency markers, including OCT-4, NANOG, SOX-2, TERT, TDGF, LEFTYA, and REX-1. All 3 lines demonstrated positive immunocytochemical staining for OCT-4, stage-specific embryonic antigen-3, stage-specific embryonic antigen-4, TRA-1-60, and TRA-1-81. Baboon ESCs injected into NOD/SCID mice formed teratomas with all 3 germ layers. In addition, embryoid body-like spherical structures were derived and initial outgrowth was observed when embedded into extracellular matrix Matrigel. The ESC lines established in this NHP model have the potential to extend our knowledge in the fields of developmental biology, regenerative medicine, and future applications, including preclinical safety assessment of in vivo stem cell therapy.
Collapse
Affiliation(s)
- Tien-Cheng Chang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Müller T, Hupfeld T, Roessler J, Simoni M, Gromoll J, Behr R. Molecular cloning and functional characterization of endogenous recombinant common marmoset monkey (Callithrix jacchus) follicle-stimulating hormone. J Med Primatol 2010; 40:111-9. [DOI: 10.1111/j.1600-0684.2010.00453.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
49
|
Albert S, Ehmcke J, Wistuba J, Eildermann K, Behr R, Schlatt S, Gromoll J. Germ cell dynamics in the testis of the postnatal common marmoset monkey (Callithrix jacchus). Reproduction 2010; 140:733-42. [DOI: 10.1530/rep-10-0235] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The seminiferous epithelium in the nonhuman primate Callithrix jacchus is similarly organized to man. This monkey has therefore been used as a preclinical model for spermatogenesis and testicular stem cell physiology. However, little is known about the developmental dynamics of germ cells in the postnatal primate testis. In this study, we analyzed testes of newborn, 8-week-old, and adult marmosets employing immunohistochemistry using pluripotent stem cell and germ cell markers DDX4 (VASA), POU5F1 (OCT3/4), and TFAP2C (AP-2γ). Stereological and morphometric techniques were applied for quantitative analysis of germ cell populations and testicular histological changes. Quantitative RT-PCR (qRT-PCR) of testicular mRNA was applied using 16 marker genes establishing the corresponding profiles during postnatal testicular development. Testis size increased during the first 8 weeks of life with the main driver being longitudinal outgrowth of seminiferous cords. The number of DDX4-positive cells per testis doubled between birth and 8 weeks of age whereas TFAP2C- and POU5F1-positive cells remained unchanged. This increase in DDX4-expressing cells indicates dynamic growth of the differentiated A-spermatogonial population. The presence of cells expressing POU5F1 and TFAP2C after 8 weeks reveals the persistence of less differentiated germ cells. The mRNA and protein profiles determined by qRT-PCR and western blot in newborn, 8-week-old, and adult marmosets corroborated the immunohistochemical findings. In conclusion, we demonstrated the presence of distinct spermatogonial subpopulations in the primate testis exhibiting different dynamics during early testicular development. Our study demonstrates the suitability of the marmoset testis as a model for human testicular development.
Collapse
|
50
|
Kong W, Li S, Lorenz HP. Germ plasm-like Dot cells maintain their wound regenerative function after in vitro expansion. Clin Exp Pharmacol Physiol 2010; 37:e136-44. [PMID: 20409081 DOI: 10.1111/j.1440-1681.2010.05343.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Wounds in fetal skin heal without scarring; however, the mechanism for this is unknown. We have identified a novel group of protein and nucleotides-positive particles in fetal and adult mouse blood and in human blood, and termed them 'Dot cells'. Freshly isolated Dot cells regenerate wounds with less scarring and can be cultured without feeder layers. 2. Because the morphology of Dot cells has never been described, in the present study we describe the specific characterizations of Dot cells, including their growth pattern in vitro, and their expressions of stem cell markers using fluorescent cell sorting analyses and immunofluorescent histology. Our data indicates that cultured Dot cells express stem cell surface markers and embryonic stem cell transcription markers, such as Oct4, Nanog and Sox-2. In addition, Dot cells express VASA, the germ plasm specific marker. 3. To confirm whether Dot cells maintain their wound regenerative activity after in vitro expansion, in vitro cultured Dot cells were transplanted to wounded mice. Dot cells from albino mice maintain their wound regenerative activities after intravenous transplantation to black-background diabetic mice. In addition, Dot cells regenerate both the epithelial and dermal cells in the wounds of wild-type mice. The regenerated hair follicles, smooth muscle and dermal tissues express transiently to VASA. 4. Our data demonstrate that Dot cells are newly identified organisms located in the blood and bone marrow of mammals. They express germ cell, embryonic stem cell and adult stem cell markers. Dot cells maintain their regenerative function after in vitro expansion.
Collapse
Affiliation(s)
- Wuyi Kong
- Hagey laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA.
| | | | | |
Collapse
|