1
|
Ghasemi N, Azizi H, Qorbanee A, Skutella T. From unipotency to pluripotency: deciphering protein networks and signaling pathways in the generation of embryonic stem-like cells from murine spermatogonial stem cells. BMC Genomics 2025; 26:426. [PMID: 40307702 PMCID: PMC12042637 DOI: 10.1186/s12864-025-11612-y] [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: 10/19/2024] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
With the significant challenges in using human embryonic stem cells (ESCs) for research and clinical applications, there is a growing impetus to seek alternative pluripotent cell sources. Embryonic stem-like (ES-like) cells emerge as a promising avenue in this pursuit. Our research demonstrates the potential for deriving ES-like cells from spermatogonial stem cells (SSCs) in a time-dependent manner under defined culture conditions. To better understand this process, we investigated the gene expression dynamics and underlying pathways associated with ES-like cell generation from SSCs. A deeper understanding of the signaling pathways underlying this biological process can lead us to refine protocols for ES-like cell generation, which could catalyze the development of more efficient and expedited methodologies inspired by the derivation pathway for future research in regenerative medicine. To identify differentially expressed genes (DEGs), we analyzed publicly available microarray data from murine cells obtained from the Gene Expression Omnibus (GEO). This analysis enabled the prediction of protein-protein interactions (PPIs), which were subsequently used for pathway enrichment analysis to identify biologically relevant pathways. Complementing these computational findings, we conducted in vitro experiments, including Fluidigm qPCR and immunostaining. These experiments serve as validation for our microarray data and the DEGs identified, providing reassurance about the reliability of our research. Among the identified enriched pathways in our investigation are the Toll-like receptor (TLR), GDNF/RET, interleukins (ILs), FGF/FGFR, and SMAD signaling pathway, along with the activation of NIMA kinases. Additionally, miR-410-3p, miRNA let-7e, Miat, and Xist are among some of the predicted non-coding RNAs.
Collapse
Affiliation(s)
- Nima Ghasemi
- Department of Applied Biotechnology and System Biology, College of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Department of Stem Cells and Cancer, College of Biotechnology, Amol University of Special Modern Technologies, P.O. Box 49767, Amol, Iran.
| | - Ali Qorbanee
- Department of Surgery, Faculty of General of Medicine, Koya University, Koya, Kurdistan Region FR, KOY45, Iraq
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Im Neuenheimer Feld 307, Heidelberg, 69120, Germany
| |
Collapse
|
2
|
Roshan MM, Azizi H, Sojoudi K. A novel investigation of NANOG and POU5F1 associations in the pluripotent characterization of ES-like and epiblast cells. Sci Rep 2024; 14:24092. [PMID: 39406879 PMCID: PMC11480366 DOI: 10.1038/s41598-024-75529-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The transcription factors NANOG and POU5F1 (OCT4) play crucial roles in maintaining pluripotency in embryonic stem (ES) cells. While their functions have been well-studied, the specific interactions between NANOG and POU5F1 and their combined effects on pluripotency in ES-like and Epiblast cells remain less understood. Understanding these associations is vital for refining pluripotent stem cell characterization and advancing regenerative medicine. In this matter, we investigated the associations between NANOG and POU5F1 in maintaining pluripotency in ES-like and Epiblast cells and how these interactions contribute to the distinct pluripotent states of these cells. In the present paper, we examined the pattern of NANOG expression by the immunocytochemical method in embryonic stem-like (ES-like) cells and compared it with its expression pattern in embryonic stem cells (ESCs). Similarly, we examined the expression pattern of POU5F1 in ES-like cells, ESCs, and epiblast cells and compared the expression pattern of these two genes with each other. On the other hand, using Fluidigm Biomark system analysis, we compared the amount of NANOG mRNA in these three cell lines and differentiated and undifferentiated Spermatogonial stem cells in several passages. Microscopic observations indicated the cytoplasmic expression of NANOG in the considered cells; moreover, they showed a similar expression pattern of NANOG with POU5F1 in the experimented cells. It has also been suggested that the more limited the cell's pluripotency, the lower the expression of these two genes. However, the decrease in NANOG expression is less than that of POU5F1. Fluidigm real-time RT-PCR analysis also confirmed these results. During the experimental process, protein-protein (PPI) network analysis shows a significant association of NANOG with other stem cell proteins, such as POU5F1. Our findings reveal distinct yet overlapping roles of NANOG and POU5F1 in maintaining pluripotency in ES-like and Epiblast cells. The differential binding patterns and functional interactions between these factors underscore the complexity of pluripotency regulation in different stem cell states. This study provides new insights into the molecular mechanisms governing pluripotency and highlights potential targets for enhancing stem cell-based therapies.
Collapse
Affiliation(s)
- Mehdi Mehdinezhad Roshan
- Faculty of Biotechnology, Amol University of Special Modern Technologies, P.O.Box: 46168-49767, Amol, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, P.O.Box: 46168-49767, Amol, Iran.
| | - Kiana Sojoudi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, P.O.Box: 46168-49767, Amol, Iran
| |
Collapse
|
3
|
Ghasemi N, Azizi H, Skutella T. Unraveling the Significance of Nanog in the Generation of Embryonic Stem-like Cells from Spermatogonia Stem Cells: A Combined In Silico Analysis and In Vitro Experimental Approach. Int J Mol Sci 2024; 25:4833. [PMID: 38732061 PMCID: PMC11084519 DOI: 10.3390/ijms25094833] [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: 02/22/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 05/13/2024] Open
Abstract
Embryonic stem-like cells (ES-like cells) are promising for medical research and clinical applications. Traditional methods involve "Yamanaka" transcription (OSKM) to derive these cells from somatic cells in vitro. Recently, a novel approach has emerged, obtaining ES-like cells from spermatogonia stem cells (SSCs) in a time-related process without adding artificial additives to cell cultures, like transcription factors or small molecules such as pten or p53 inhibitors. This study aims to investigate the role of the Nanog in the conversion of SSCs to pluripotent stem cells through both in silico analysis and in vitro experiments. We used bioinformatic methods and microarray data to find significant genes connected to this derivation path, to construct PPI networks, using enrichment analysis, and to construct miRNA-lncRNA networks, as well as in vitro experiments, immunostaining, and Fluidigm qPCR analysis to connect the dots of Nanog significance. We concluded that Nanog is one of the most crucial differentially expressed genes during SSC conversion, collaborating with critical regulators such as Sox2, Dazl, Pou5f1, Dnmt3, and Cdh1. This intricate protein network positions Nanog as a pivotal factor in pathway enrichment for generating ES-like cells, including Wnt signaling, focal adhesion, and PI3K-Akt-mTOR signaling. Nanog expression is presumed to play a vital role in deriving ES-like cells from SSCs in vitro. Finding its pivotal role in this path illuminates future research and clinical applications.
Collapse
Affiliation(s)
- Nima Ghasemi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol 49767, Iran;
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol 49767, Iran;
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany;
| |
Collapse
|
4
|
Indu S, Devi AN, Sahadevan M, Sengottaiyan J, Basu A, K SR, Kumar PG. Expression profiling of stemness markers in testicular germline stem cells from neonatal and adult Swiss albino mice during their transdifferentiation in vitro. Stem Cell Res Ther 2024; 15:93. [PMID: 38561834 PMCID: PMC10985951 DOI: 10.1186/s13287-024-03701-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Spermatogonial stem cells (SSCs) were considered to be stem cells with limited potencies due to their existence in adult organisms. However, the production of spermatogonial stem cell colonies with broader differentiation capabilities in primary germ cell cultures from mice of select genetic backgrounds (C57BL6/Tg14, ddY, FVB and 129/Ola) indicated that SSCs from these strains were pluripotent. METHODS We established primary cultures of SSCs from neonatal and adult Swiss 3T3 Albino mice. Stemness of SSC colonies were evaluated by performing real-time PCR and immunofluorescence analysis for a panel of chosen stemness markers. Differentiation potentials of SSCs were examined by attempting the generation of embryoid bodies and evaluating the expression of ectodermal, mesodermal and endodermal markers using immunofluorescence and real-time PCR analysis. RESULTS Spermatogonial stem cells from neonatal and mature mice testes colonised in vitro and formed compact spermatogonial stem cell colonies in culture. The presence of stem cell markers ALPL, ITGA6 and CD9 indicated stemness in these colonies. The differentiation potential of these SSC colonies was demonstrated by their transformation into embryoid bodies upon withdrawal of growth factors from the culture medium. SSC colonies and embryoid bodies formed were evaluated using immunofluorescence and real-time PCR analysis. Embryoid body like structures derived from both neonatal and adult mouse testis were quite similar in terms of the expression of germ layer markers. CONCLUSION These results strongly suggest that SSC-derived EB-like structures could be used for further differentiation into cells of interest in cell-based therapeutics.
Collapse
Affiliation(s)
- Sivankutty Indu
- Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, 695 014, Kerala, India
| | - Anandavally N Devi
- Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, 695 014, Kerala, India
| | - Mahitha Sahadevan
- Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, 695 014, Kerala, India
| | - Jeeva Sengottaiyan
- Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, 695 014, Kerala, India
- Department of Biotechnology, University of Kerala, Karyavattom Campus, Thiruvananthapuram, 695581, Kerala, India
| | - Asmita Basu
- Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, 695 014, Kerala, India
- Department of Biotechnology, University of Kerala, Karyavattom Campus, Thiruvananthapuram, 695581, Kerala, India
| | - Shabith Raj K
- Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, 695 014, Kerala, India
- Department of Biotechnology, University of Kerala, Karyavattom Campus, Thiruvananthapuram, 695581, Kerala, India
| | - Pradeep G Kumar
- Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Poojappura, Thiruvananthapuram, 695 014, Kerala, India.
- Department of Biotechnology, University of Kerala, Karyavattom Campus, Thiruvananthapuram, 695581, Kerala, India.
| |
Collapse
|
5
|
Hashemi Karoii D, Azizi H. OCT4 protein and gene expression analysis in the differentiation of spermatogonia stem cells into neurons by immunohistochemistry, immunocytochemistry, and bioinformatics analysis. Stem Cell Rev Rep 2023:10.1007/s12015-023-10548-8. [PMID: 37119454 DOI: 10.1007/s12015-023-10548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Spermatogonia Stem Cells (SSCs) are potential candidates for reprogramming and regeneration. Recent studies have revealed that differentiated cells can be reverted to pluripotent by overexpressing a set of pluripotent transcription factors. OCT4 (encoded by pou5f1), a POU transcription factor family member, is essential to the potential that controls pluripotency, and it is widely expressed in pluripotent stem cells, although it decreased or suppressed after differentiation. METHODS In this investigated research, we examined the OCT4 expression during the differentiation of SSCs into neurons (involving four stages in the following order: SSCs in vivo and in-vitro, embryonic Stem Cell-like (ES-like), Embryonic Bodies (EBs), and finally Neurons) by Immunocytochemistry (ICC), Immunohistochemistry (IMH), and Fluidigm Real-Time polymerase chain reaction. In addition, we use some databases like STRING to predict protein-protein interaction and enrichment analysis. RESULTS We evaluated the expression of OCT4 in this process, and we observed that it is expressed in SSCs, ES-like, and EBs during the differentiation of spermatogonia stem cells into adult neurons. We show that by adding RA to EBs, the expression of OCT4 is reduced and is not expressed in the neuron cells. We observed that the expression of OCT4 is linked and interacts with the differentiation of spermatogonia stem cells into neuron cells, and it has been shown to be biologically functional, like stem cell maintenance and somatic cell reprogramming. CONCLUSION Our findings can help us better understand the process of differentiation of spermatogonia stem cells into neurons, and it can be effective in finding new and more efficient treatments for neurogenesis and repair of neurons. We examined the OCT4 expression during the differentiation of SSCs into neurons (involving four stages in the following order: SSCs in vivo and in-vitro, embryonic Stem Cell-like (ES-like), Embryonic Bodies (EBs), and finally Neurons) by Immunocytochemistry (ICC), Immunohistochemistry (IMH), and Fluidigm Real-Time polymerase chain reaction. In addition, we use some databases like STRING to predict protein-protein interaction and enrichment analysis. We evaluated the expression of OCT4 in this process, and we observed that it is expressed in SSCs, ES-like, and EBs during the differentiation of spermatogonia stem cells into adult neurons. We show that by adding RA to EBs, the expression of OCT4 is reduced and is not expressed in the neuron cells. We observed that the expression of OCT4 is linked and interacts with the differentiation of spermatogonia stem cells into neuron cells, and it has been shown to be biologically functional, like stem cell maintenance and somatic cell reprogramming.
Collapse
Affiliation(s)
- Danial Hashemi Karoii
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran.
| |
Collapse
|
6
|
Martin-Inaraja M, Ferreira M, Taelman J, Eguizabal C, Chuva De Sousa Lopes SM. Improving In Vitro Culture of Human Male Fetal Germ Cells. Cells 2021; 10:cells10082033. [PMID: 34440801 PMCID: PMC8393746 DOI: 10.3390/cells10082033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Male human fetal germ cells (hFGCs) give rise to spermatogonial stem cells (SSCs), which are the adult precursors of the male gametes. Human SSCs are a promising (autologous) source of cells for male fertility preservation; however, in contrast to mouse SSCs, we are still unable to culture them in the long term. Here, we investigated the effect of two different culture media and four substrates (laminin, gelatin, vitronectin and matrigel) in the culture of dissociated second trimester testes, enriched for hFGCs. After 6 days in culture, we quantified the presence of POU5F1 and DDX4 expressing hFGCs. We observed a pronounced difference in hFGC number in different substrates. The combination of gelatin-coated substrate and medium containing GDNF, LIF, FGF2 and EGF resulted in the highest percentage of hFGCs (10% of the total gonadal cells) after 6 days of culture. However, the vitronectin-coated substrate resulted in a comparable percentage of hFGCs regardless of the media used (3.3% of total cells in Zhou-medium and 4.8% of total cells in Shinohara-medium). We provide evidence that not only the choices of culture medium but also choices of the adequate substrate are crucial for optimizing culture protocols for male hFGCs. Optimizing culture conditions in order to improve the expansion of hFGCs will benefit the development of gametogenesis assays in vitro.
Collapse
Affiliation(s)
- Myriam Martin-Inaraja
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, 48960 Galdakao, Spain; (M.M.-I.); (C.E.)
- Biocruces Bizkaia Health Research Institute, Cell Therapy, Stem Cells and Tissues Group, 48903 Barakaldo, Spain
| | - Monica Ferreira
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (M.F.); (J.T.)
| | - Jasin Taelman
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (M.F.); (J.T.)
| | - Cristina Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, 48960 Galdakao, Spain; (M.M.-I.); (C.E.)
- Biocruces Bizkaia Health Research Institute, Cell Therapy, Stem Cells and Tissues Group, 48903 Barakaldo, Spain
| | - Susana M. Chuva De Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (M.F.); (J.T.)
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +31-71-526-9350
| |
Collapse
|
7
|
Application of platelet-rich plasma (PRP) improves self-renewal of human spermatogonial stem cells in two-dimensional and three-dimensional culture systems. Acta Histochem 2020; 122:151627. [PMID: 33002788 DOI: 10.1016/j.acthis.2020.151627] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/17/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
Spermatogonial stem cells (SSCs) are very sensitive to chemotherapy and radiotherapy, so male infertility is a great challenge for prepubertal cancer survivors. Cryoconservation of testicular cells before cancer treatment can preserve SSCs from treatment side effects. Different two-dimensional (2D) and three-dimensional (3D) culture systems of SSCs have been used in many species as a useful technique to in vitro spermatogenesis. We evaluated the proliferation of SSCs in 2D and 3D culture systems of platelet-rich plasma (PRP). testicular cells of four brain-dead patients cultivated in 2D pre-culture system, characterization of SSCs performed by RT-PCR, flow cytometry, immunocytochemistry and their functionality assessed by xenotransplantation to azoospermia mice. PRP prepared and dosimetry carried out to determine the optimized dose of PRP. After preparation of PRP scaffold, cytotoxic and histological evaluation performed and SSCs cultivated into three groups: control, 2D culture by optimized dose of PRP and PRP scaffold. The diameter and number of colonies measured and relative expression of GFRa1 and c-KIT evaluated by real-time PCR. Results indicated the expression of PLZF, VASA, OCT4, GFRa1 and vimentin in colonies after 2D pre-culture, xenotransplantation demonstrated proliferated SSCs have proper functionality to homing in mouse testes. The relative expression of c-KIT showed a significant increase as compared to the control group (*: p < 0.05) in PRP- 2D group, expression of GFRa1 and c-KIT in PRP scaffold group revealed a significant increase as compared to other groups (***: p < 0.001). The number and diameter of colonies in the PRP-2D group showed a considerable increase (p < 0.01) as compared to the control group. In PRP- scaffold group, a significant increase (p < 0.01) was seen only in the number of colonies related to the control group. Our results suggested that PRP scaffold can reconstruct a suitable structure to the in vitro proliferation of SSCs.
Collapse
|
8
|
Mariniello K, Ruiz-Babot G, McGaugh EC, Nicholson JG, Gualtieri A, Gaston-Massuet C, Nostro MC, Guasti L. Stem Cells, Self-Renewal, and Lineage Commitment in the Endocrine System. Front Endocrinol (Lausanne) 2019; 10:772. [PMID: 31781041 PMCID: PMC6856655 DOI: 10.3389/fendo.2019.00772] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
The endocrine system coordinates a wide array of body functions mainly through secretion of hormones and their actions on target tissues. Over the last decades, a collective effort between developmental biologists, geneticists, and stem cell biologists has generated a wealth of knowledge related to the contribution of stem/progenitor cells to both organogenesis and self-renewal of endocrine organs. This review provides an up-to-date and comprehensive overview of the role of tissue stem cells in the development and self-renewal of endocrine organs. Pathways governing crucial steps in both development and stemness maintenance, and that are known to be frequently altered in a wide array of endocrine disorders, including cancer, are also described. Crucially, this plethora of information is being channeled into the development of potential new cell-based treatment modalities for endocrine-related illnesses, some of which have made it through clinical trials.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Emily C. McGaugh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James G. Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
9
|
Suen HC, Qian Y, Liao J, Luk CS, Lee WT, Ng JKW, Chan TTH, Hou HW, Li I, Li K, Chan WY, Feng B, Gao L, Jiang X, Liu YH, Rudd JA, Hobbs R, Qi H, Ng TK, Mak HK, Leung KS, Lee TL. Transplantation of Retinal Ganglion Cells Derived from Male Germline Stem Cell as a Potential Treatment to Glaucoma. Stem Cells Dev 2019; 28:1365-1375. [PMID: 31580778 DOI: 10.1089/scd.2019.0060] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Glaucoma is characterized by retinal ganglion cell (RGC) degeneration and is the second leading cause of blindness worldwide. However, current treatments such as eye drop or surgery have limitations and do not target the loss of RGC. Regenerative therapy using embryonic stem cells (ESCs) holds a promising option, but ethical concern hinders clinical applications on human subjects. In this study, we employed spermatogonial stem cells (SSCs) as an alternative source of ESCs for cell-based regenerative therapy in mouse glaucoma model. We generated functional RGCs from SSCs with a two-step protocol without applying viral transfection or chemical induction. SSCs were first dedifferentiated to embryonic stem-like cells (SSC-ESCs) that resemble ESCs in morphology, gene expression signatures, and stem cell properties. The SSC-ESCs then differentiated toward retinal lineages. We showed SSC-ESC-derived retinal cells expressed RGC-specific marker Brn3b and functioned as bona fide RGCs. To allow in vivo RGC tracing, Brn3b-EGFP reporter SSC-ESCs were generated and the derived RGCs were subsequently transplanted into the retina of glaucoma mouse models by intravitreal injection. We demonstrated that the transplanted RGCs could survive in host retina for at least 10 days after transplantation. SSC-ESC-derived RGCs can thus potentially be a novel alternative to replace the damaged RGCs in glaucomatous retina.
Collapse
Affiliation(s)
- Hoi Ching Suen
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yan Qian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jinyue Liao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Chun Shui Luk
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wing Tung Lee
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Judy Kin Wing Ng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Thomas Ting Hei Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hei Wan Hou
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ingrid Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Kit Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wai-Yee Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Bo Feng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Lin Gao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiaohua Jiang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yuen Hang Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - John A Rudd
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Robin Hobbs
- Aust Regenerative Medicine Institute, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Huayu Qi
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Tsz Kin Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Heather Kayew Mak
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Kai Shun Leung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Tin-Lap Lee
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
10
|
Borzouie Z, Naghibzadeh M, Talebi AR, Pourrajab F, Jebali A, Nikukar H, Molla Hoseini H, Khoradmehr A, Khoradmehr A, Sadeghian-Nodoushan F, Aflatoonian B, Hekmatimoghaddam S. Development of An Artificial Male Germ Cell Niche Using Electrospun Poly Vinyl Alcohol/Human Serum Albumin/Gelatin Fibers. CELL JOURNAL 2019; 21:300-306. [PMID: 31210436 PMCID: PMC6582427 DOI: 10.22074/cellj.2019.6120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 09/29/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Recent achievements in stem cell biotechnology, nanotechnology and tissue engineering have led to development of novel approaches in regenerative medicine. Azoospermia is one of the challenging disorders of the reproductive system. Several efforts were made for isolation and culture of testis-derived stem cells to treat male infertility. However, tissue engineering is the best approach to mimic the three dimensional microenvironment of the testis in vitro. We investigated whether human testis-derived cells (hTCs) obtained by testicular sperm extraction (TESE) can be cultured on a homemade scaffold composed of electrospun nanofibers of homogeneous poly (vinyl alcohol)/human serum albumin/gelatin (PVA/HSA/gelatin). MATERIALS AND METHODS In this experimental lab study, human TCs underwent two steps of enzymatic cell isolation and five culture passages. Nanofibrous scaffolds were characterized by scanning electron microscopy (SEM) and Fouriertransform infrared spectroscopy (FTIR). Attachment of cells onto the scaffold was shown by hematoxylin and eosin (H and E) staining and SEM. Cell viability study using MTT [3-(4, 5-dimethyl-2-thiazolyl) -2, 5-diphenyl -2H- tetrazolium bromide] assay was performed on days 7 and 14. RESULTS Visualization by H and E staining and SEM indicated that hTCs were seeded on the scaffold. MTT test showed that the PVA/HSA/gelatin scaffold is not toxic for hTCs. CONCLUSION It seems that this PVA/HSA/gelatin scaffold is supportive for growth of hTCs.
Collapse
Affiliation(s)
- Zahra Borzouie
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Majid Naghibzadeh
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Reza Talebi
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Pourrajab
- Department of Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Jebali
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Habib Nikukar
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Arezoo Khoradmehr
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Arezoo Khoradmehr
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Sadeghian-Nodoushan
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Behrouz Aflatoonian
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. Electronic Address:
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyedhossein Hekmatimoghaddam
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.Electronic Adress:
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
11
|
Dong L, Kristensen SG, Hildorf S, Gul M, Clasen-Linde E, Fedder J, Hoffmann ER, Cortes D, Thorup J, Andersen CY. Propagation of Spermatogonial Stem Cell-Like Cells From Infant Boys. Front Physiol 2019; 10:1155. [PMID: 31607938 PMCID: PMC6761273 DOI: 10.3389/fphys.2019.01155] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/28/2019] [Indexed: 12/22/2022] Open
Abstract
Background Gonadotoxic treatment of malignant diseases as well as some non-malignant conditions such as cryptorchidism in young boys may result in infertility and failure to father children later in life. As a fertility preserving strategy, several centers collect testicular biopsies to cryopreserve spermatogonial stem cells (SSCs) world-wide. One of the most promising therapeutic strategies is to transplant SSCs back into the seminiferous tubules to initiate endogenous spermatogenesis. However, to obtain sufficient numbers of SSC to warrant transplantation, in vitro propagation of cells is needed together with proper validation of their stem cell identity. Materials and Methods A minute amount of testicular biopsies (between 5 mg and 10 mg) were processed by mechanical and enzymatic digestion. SSCs were enriched by differential plating method in StemPro-34 medium supplemented with several growth factors. SSC-like cell clusters (SSCLCs) were passaged five times. SSCLCs were identified by immunohistochemical and immunofluorescence staining, using protein expression patterns in testis biopsies as reference. Quantitative polymerase chain reaction analysis of SSC markers LIN-28 homolog A (LIN28A), G antigen 1 (GAGE1), promyelocytic leukemia zinc finger protein (PLZF), integrin alpha 6 (ITGA6), ubiquitin carboxy-terminal hydrolase L1 (UCHL1) and integrin beta 1 (ITGB1) were also used to validate the SSC-like cell identity. Results Proliferation of SSCLCs was achieved. The presence of SSCs in SSCLCs was confirmed by positive immunostaining of LIN28, UCHL1 and quantitative polymerase chain reaction for LIN28A, UCHL1, PLZF, ITGA6, and ITGB1, respectively. Conclusion This study has demonstrated that SSCs from infant boys possess the capacity for in vitro proliferation and advance a fertility preservation strategy for pre-pubertal boys who may otherwise lose their fertility.
Collapse
Affiliation(s)
- Lihua Dong
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Simone Hildorf
- Department of Pediatric Surgery, Copenhagen University Hospital, Copenhagen, Denmark
| | - Murat Gul
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Urology, Aksaray University School of Medicine, Aksaray, Turkey
| | - Erik Clasen-Linde
- Department of Pathology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jens Fedder
- Centre of Andrology and Fertility Clinic, Department D, Odense University Hospital, Odense C, Denmark.,Research Unit of Human Reproduction, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Eva R Hoffmann
- Center for Chromosome Stability, Department of Molecular and Cellular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dina Cortes
- Department of Pediatrics, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jorgen Thorup
- Department of Pediatric Surgery, Copenhagen University Hospital, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Kubota H, Brinster RL. Spermatogonial stem cells. Biol Reprod 2019; 99:52-74. [PMID: 29617903 DOI: 10.1093/biolre/ioy077] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/29/2018] [Indexed: 12/19/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are the most primitive spermatogonia in the testis and have an essential role to maintain highly productive spermatogenesis by self-renewal and continuous generation of daughter spermatogonia that differentiate into spermatozoa, transmitting genetic information to the next generation. Since the 1950s, many experimental methods, including histology, immunostaining, whole-mount analyses, and pulse-chase labeling, had been used in attempts to identify SSCs, but without success. In 1994, a spermatogonial transplantation method was reported that established a quantitative functional assay to identify SSCs by evaluating their ability to both self-renew and differentiate to spermatozoa. The system was originally developed using mice and subsequently extended to nonrodents, including domestic animals and humans. Availability of the functional assay for SSCs has made it possible to develop culture systems for their ex vivo expansion, which dramatically advanced germ cell biology and allowed medical and agricultural applications. In coming years, SSCs will be increasingly used to understand their regulation, as well as in germline modification, including gene correction, enhancement of male fertility, and conversion of somatic cells to biologically competent male germline cells.
Collapse
Affiliation(s)
- Hiroshi Kubota
- Laboratory of Cell and Molecular Biology, Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Ralph L Brinster
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Davidoff MS. The Pluripotent Microvascular Pericytes Are the Adult Stem Cells Even in the Testis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:235-267. [PMID: 30937872 DOI: 10.1007/978-3-030-11093-2_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pericytes of the testis are part of the omnipresent population of pericytes in the vertebrate body and are the only true pluripotent adult stem cells able to produce structures typical for the tree primitive germ layers: ectoderm, mesoderm, and endoderm. They originate very early in the embryogenesis from the pluripotent epiblast. The pericytes become disseminated through the whole vertebrate organism by the growing and differentiating blood vessels where they remain in specialized periendothelial vascular niches as resting pluripotent adult stem cells for tissue generation, maintenance, repair, and regeneration. The pericytes are also the ancestors of the perivascular multipotent stromal cells (MSCs). The variable appearance of the pericytes and their progeny reflects the plasticity under the influence of their own epigenetic and the local environmental factors of the host organ. In the testis the pericytes are the ancestors of the neuroendocrine Leydig cells. After activation the pericytes start to proliferate, migrate, and build transit-amplifying cells that transdifferentiate into multipotent stromal cells. These represent progenitors for a number of different cell types in an organ. Finally, it becomes evident that the pericytes are a brilliant achievement of the biological nature aiming to supply every organ with an omnipresent population of pluripotent adult stem cells. Their fascinating features are prerequisites for future therapy concepts supporting cell systems of organs.
Collapse
Affiliation(s)
- Michail S Davidoff
- University Medical Center Hamburg-Eppendorf, Hamburg Museum of Medical History, Hamburg, Germany.
| |
Collapse
|
14
|
De Chiara L, Famulari ES, Fagoonee S, van Daalen SKM, Buttiglieri S, Revelli A, Tolosano E, Silengo L, van Pelt AMM, Altruda F. Characterization of Human Mesenchymal Stem Cells Isolated from the Testis. Stem Cells Int 2018; 2018:4910304. [PMID: 30250488 PMCID: PMC6140008 DOI: 10.1155/2018/4910304] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 01/04/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells hold great promise for regenerative medicine as they can be easily isolated from different sources such as adipose tissue, bone marrow, and umbilical cord blood. Spontaneously arising pluripotent stem cells can be obtained in culture from murine spermatogonial stem cells (SSCs), while the pluripotency of the human counterpart remains a matter of debate. Recent gene expression profiling studies have demonstrated that embryonic stem cell- (ESC-) like cells obtained from the human testis are indeed closer to mesenchymal stem cells (MSCs) than to pluripotent stem cells. Here, we confirm that colonies derived from human testicular cultures, with our isolation protocol, are of mesenchymal origin and do not arise from spermatogonial stem cells (SSCs). The testis, thus, provides an important and accessible source of MSCs (tMSCs) that can be potentially used for nephrotoxicity testing in vitro. We further demonstrate, for the first time, that tMSCs are able to secrete microvesicles that could possibly be applied to the treatment of various chronic diseases, such as those affecting the kidney.
Collapse
Affiliation(s)
- Letizia De Chiara
- Centro di Eccellenza DeNothe, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Elvira Smeralda Famulari
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Sharmila Fagoonee
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
- The Institute of Biostructure and Bioimaging (CNR) c/o Molecular Biotechnology Center, Turin, Italy
| | - Saskia K. M. van Daalen
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Stefano Buttiglieri
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Alberto Revelli
- Obstetrics and Gynecology 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Corso Spezia 60, 10126 Turin, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Lorenzo Silengo
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| | - Ans M. M. van Pelt
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, Netherlands
| | - Fiorella Altruda
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy
| |
Collapse
|
15
|
Bai Y, Zhu C, Feng M, Wei H, Li L, Tian X, Zhao Z, Liu S, Ma N, Zhang X, Shi R, Fu C, Wu Z, Zhang S. Previously claimed male germline stem cells from porcine testis are actually progenitor Leydig cells. Stem Cell Res Ther 2018; 9:200. [PMID: 30021628 PMCID: PMC6052628 DOI: 10.1186/s13287-018-0931-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/27/2018] [Accepted: 06/14/2018] [Indexed: 11/14/2022] Open
Abstract
Background Male germline stem cells (mGSCs) offer great promise in regenerative medicine and animal breeding due to their capacity to maintain self-renewal and to transmit genetic information to the next generation following spermatogenesis. Human testis-derived embryonic stem cell-like cells have been shown to possess potential of mesenchymal progenitors, but there remains confusion about the characteristics and origin of porcine testis-derived stem cells. Methods Porcine testis-derived stem cells were obtained from primary testicular cultures of 5-day old piglets, and selectively expanded using culture conditions for long-term culture and induction differentiation. The stem cell properties of porcine testis-derived stem cells were subsequently assessed by determining the expression of pluripotency-associated markers, alkaline phosphatase (AP) activity, and capacity for sperm and multilineage differentiation in vitro. The gene expression profile was compared via microarray analysis. Results We identified two different types of testis-derived stem cells (termed as C1 and C2 here) during porcine testicular cell culture. The gene expression microarray analysis showed that the transcriptome profile of C1 and C2 differed significantly from each other. The C1 appeared to be morphologically similar to the previously described mouse mGSCs, expressed pluripotency- and germ cell-associated markers, maintained the paternal imprinted pattern of H19, displayed alkaline phosphatase activity, and could differentiate into sperm. Together, these data suggest that C1 represent the porcine mGSC population. Conversely, the C2 appeared similar to the previously described porcine mGSCs with three-dimensional morphology, abundantly expressed Leydig cell lineage and mesenchymal cell-specific markers, and could differentiate into testosterone-producing Leydig cells, suggesting that they are progenitor Leydig cells (PLCs). Conclusion Collectively, we have established the expected characteristics and markers of authentic porcine mGSCs (C1). We found for the first time that, the C2, equivalent to previously claimed porcine mGSCs, are actually progenitor Leydig cells (PLCs). These findings provide new insights into the discrepancies among previous reports and future identification and analyses of testis-derived stem cells. Electronic supplementary material The online version of this article (10.1186/s13287-018-0931-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yinshan Bai
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China.,School of Life Science and Engineering, Foshan University, Foshan, 528231, China
| | - Cui Zhu
- School of Life Science and Engineering, Foshan University, Foshan, 528231, China
| | - Meiying Feng
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Hengxi Wei
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Li Li
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Xiuchun Tian
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, 1390 Storrs Road, Storrs, CT, 06269, USA
| | - Zhihong Zhao
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Shanshan Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ningfang Ma
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xianwei Zhang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China
| | - Ruyi Shi
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Cell Biology and Genetics, Shanxi Medical University, Taiyuan, 030001, China
| | - Chao Fu
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Cell Biology and Genetics, Shanxi Medical University, Taiyuan, 030001, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China.
| | - Shouquan Zhang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, 510642, China.
| |
Collapse
|
16
|
Mahapatra S, Martin D, Gallicano GI. Re-Defining Stem Cell-Cardiomyocyte Interactions: Focusing on the Paracrine Effector Approach. J Stem Cells Regen Med 2018. [PMID: 30018469 PMCID: PMC6043659 DOI: 10.46582/jsrm.1401003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cell research for treating or curing ischemic heart disease has, till date, culminated in three basic approaches: the use of induced pluripotent stem cell (iPSC) technology; reprogramming cardiac fibroblasts; and cardiovascular progenitor cell regeneration. As each approach has been shown to have its advantages and disadvantages, exploiting the advantages while minimizing the disadvantages has been a challenge. Using human germline pluripotent stem cells (hgPSCs) along with a modified version of a relatively novel cell-expansion culture methodology to induce quick, indefinite expansion of normally slow growing hgPSCs, it was possible to emphasize the advantages of all three approaches. We consistently found that unipotent germline stem cells, when removed from their niche and cultured in the correct medium, expressed endogenously, pluripotency genes, which induced them to become hgPSCs. These cells are then capable of producing cell types from all three germ layers. Upon differentiation into cardiac lineages, our data consistently showed that they not only expressed cardiac genes, but also expressed cardiac-promoting paracrine factors. Taking these data a step further, we found that hgPSC-derived cardiac cells could integrate into cardiac tissue in vivo. Note, while the work presented here was based on testes-derived hgPSCs, data from other laboratories have shown that ovaries contain very similar types of stem cells that can give rise to hgPSCs. As a result, hgPSCs should be considered a viable option for eventual use in patients, male or female, with ischemic heart disease
Collapse
Affiliation(s)
- Samiksha Mahapatra
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, 3900 Reservoir Rd, Washington, DC, USA
| | - Dianna Martin
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, 3900 Reservoir Rd, Washington, DC, USA
| | - G Ian Gallicano
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, 3900 Reservoir Rd, Washington, DC, USA
| |
Collapse
|
17
|
Satar NA, Fakiruddin KS, Lim MN, Mok PL, Zakaria N, Fakharuzi NA, Abd Rahman AZ, Zakaria Z, Yahaya BH, Baharuddin P. Novel triple‑positive markers identified in human non‑small cell lung cancer cell line with chemotherapy-resistant and putative cancer stem cell characteristics. Oncol Rep 2018; 40:669-681. [PMID: 29845263 PMCID: PMC6072294 DOI: 10.3892/or.2018.6461] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/03/2018] [Indexed: 02/06/2023] Open
Abstract
Through the specific identification and direct targeting of cancer stem cells (CSCs), it is believed that a better treatment efficacy of cancer may be achieved. Hence, the present study aimed to identify a CSC subpopulation from adenocarcinoma cells (A549) as a model of non-small cell lung cancer (NSCLC). Initially, we sorted two subpopulations known as the triple-positive (EpCAM+/CD166+/CD44+) and triple-negative (EpCAM−/CD166−/CD44−) subpopulation using fluorescence-activated cell sorting (FACS). Sorted cells were subsequently evaluated for proliferation and chemotherapy-resistance using a viability assay and were further characterized for their clonal heterogeneity, self-renewal characteristics, cellular migration, alkaline dehydrogenase (ALDH) activity and the expression of stemness-related genes. According to our findings the triple-positive subpopulation revealed significantly higher (P<0.01) proliferation activity, exhibited better clonogenicity, was mostly comprised of holoclones and had markedly bigger (P<0.001) spheroid formation indicating a better self-renewal capacity. A relatively higher resistance to both 5-fluouracil and cisplatin with 80% expression of ALDH was observed in the triple-positive subpopulation, compared to only 67% detected in the triple-negative subpopulation indicated that high ALDH activity contributed to greater chemotherapy-resistance characteristics. Higher percentage of migrated cells was observed in the triple-positive subpopulation with 56% cellular migration being detected, compared to only 19% in the triple-negative subpopulation on day 2. This was similarly observed on day 3 in the triple-positive subpopulation with 36% higher cellular migration compared to the triple-negative subpopulation. Consistently, elevated levels of the stem cell genes such as REX1 and SSEA4 were also found in the triple-positive subpopulation indicating that the subpopulation displayed a strong characteristic of pluripotency. In conclusion, our study revealed that the triple-positive subpopulation demonstrated similar characteristics to CSCs compared to the triple-negative subpopulation. It also confirmed the feasibility of using the triple-positive (EpCAM+/CD166+/CD44+) marker as a novel candidate marker that may lead to the development of novel therapies targeting CSCs of NSCLC.
Collapse
Affiliation(s)
- Nazilah Abdul Satar
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, 13200 Penang, Malaysia
| | - Kamal Shaik Fakiruddin
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), 50588 Kuala Lumpur, Malaysia
| | - Moon Nian Lim
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), 50588 Kuala Lumpur, Malaysia
| | - Pooi Ling Mok
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor
| | - Norashikin Zakaria
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, 13200 Penang, Malaysia
| | - Noor Atiqah Fakharuzi
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), 50588 Kuala Lumpur, Malaysia
| | - Ahmad Zuhairi Abd Rahman
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), 50588 Kuala Lumpur, Malaysia
| | - Zubaidah Zakaria
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), 50588 Kuala Lumpur, Malaysia
| | - Badrul Hisham Yahaya
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, 13200 Penang, Malaysia
| | - Puteri Baharuddin
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), 50588 Kuala Lumpur, Malaysia
| |
Collapse
|
18
|
Zhang P, Qin Y, Zheng Y, Zeng W. Phospholipase D Family Member 6 Is a Surface Marker for Enrichment of Undifferentiated Spermatogonia in Prepubertal Boars. Stem Cells Dev 2017; 27:55-64. [PMID: 29113556 DOI: 10.1089/scd.2017.0140] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Spermatogonial stem cells (SSCs) have a functional ability to maintain self-renewal and sustain production of spermatozoa throughout the reproductive lifespan of a male. Studies on SSCs can thus better the understandings of spermatogenesis and unravel the mechanisms for self-renewal and differentiation of male germline stem cells. However, the rarity of SSCs in the testis and the lack of reliable surface markers obstruct the related study and further application of SSCs. This is especially the case in livestock animals. In this study, we identified that phospholipase D family member 6 (PLD6) is a surface marker for undifferentiated spermatogonia in boar testes. By magnetic-activated cell sorting, PLD6+ cell fraction comprises 84.45% ± 0.35% of undifferentiated spermatogonia (marked by PLZF). Xenotransplantation of PLD6+ cells into the recipient mouse testis revealed a ninefold increase of donor cell-derived colony formation compared with that in the unselected cell group, indicating the significant enrichment of SSCs. Furthermore, based on the sorted PLD6+ cells with a high SSC content, we established a feeder-free culture system that could maintain porcine undifferentiated spermatogonia for 4 weeks in vitro with the expression of typical markers throughout the culture period. In conclusion, this study demonstrates that PLD6 is a surface marker of undifferentiated spermatogonia in testes of prepubertal boars and could be utilized to unprecedentedly enrich porcine undifferentiated spermatogonia. These data provide the basis for future studies on the refinement of germ cell culture and manipulation of porcine undifferentiated spermatogonia.
Collapse
Affiliation(s)
- Pengfei Zhang
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Yuwei Qin
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| | - Yi Zheng
- 2 Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Centre, University of Amsterdam , Amsterdam, the Netherlands
| | - Wenxian Zeng
- 1 College of Animal Science and Technology, Northwest A&F University , Shaanxi, China
| |
Collapse
|
19
|
Nazm Bojnordi M. The applications and recovery outcome of spermatogonia stem cells in regenerative medicine. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2017. [DOI: 10.1016/j.mefs.2017.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
20
|
Conrad S, Azizi H, Skutella T. Single-Cell Expression Profiling and Proteomics of Primordial Germ Cells, Spermatogonial Stem Cells, Adult Germ Stem Cells, and Oocytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1083:77-87. [DOI: 10.1007/5584_2017_117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Li B, Zhuang M, Wu C, Niu B, Zhang Z, Li X, Wei Z, Li G, Hua J. Bovine male germline stem-like cells cultured in serum- and feeder-free medium. Cytotechnology 2016; 68:2145-2157. [PMID: 26883918 PMCID: PMC5023554 DOI: 10.1007/s10616-015-9933-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/14/2015] [Indexed: 12/19/2022] Open
Abstract
Male germline stem cells (mGSCs) presented in male testis are responsible for spermatogenesis during their whole life. However, little information can be found on the culture of bovine mGSCs, and the current culture system needs to be improved. In this study, we compared the effects of several commercial serum-free media and different extra-cellular matrix on the enrichment and cultivation of mGSCs. To find out the best culture condition, the biological characteristics of the cultured cells were evaluated by morphological observation, RT-PCR and immunofluorescent staining. According to the cells' condition in different experiment groups, we found out an efficient cultivation system for bovine mGSCs derived from neonate testis. In this serum- and feeder-free medium, the cultured cells maintained the typical morphology, and expressed specific surface markers of both pluripotent ES cells and mGSCs, including SSEA-1, CD49f, C-MYC, PLZF, GFRα1, LIN28, NANOG, Oct4 and SOX2 in commercial human ESCs medium PeproGrow-hESC + BIO (6-bromoindirubin-3'-oxime). Embryoid bodies, derived from the bovine mGSCs, and were formed by ganging drop culture. The retinoic acid induced bovine mGSCs were positive for Stra8, SCP3, DZAL, EMA1 and VASA, and resembled spermatid cells morphologically. Thus, we found an efficient bovine mGSCs-cultivation system, which is lack in serum and feeder.
Collapse
Affiliation(s)
- Bo Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Mengru Zhuang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chongyang Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Bowen Niu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhou Zhang
- Department of Reproduction Centre, Shaanxi Provine Women and Children Hospital, Xi'an, 710000, Shaanxi Province, China
| | - Xin Li
- Department of Clinic Medicine, Bengbu Medicine University, Bengbu, Anhui, China
| | - Zhuying Wei
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Guangpeng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China.
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
22
|
Differentiation of Spermatogonia Stem Cells into Functional Mature Neurons Characterized with Differential Gene Expression. Mol Neurobiol 2016; 54:5676-5682. [PMID: 27644129 DOI: 10.1007/s12035-016-0097-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 09/01/2016] [Indexed: 01/02/2023]
Abstract
Transplantation of embryonic stem cells (ESCs) is a promising therapeutic approach for the treatment of neurodegenerative diseases. However, ESCs are not usable clinically due to immunological and ethical limitations. The identification of an alternative safe cell source opens novel options via autologous transplantation in neuro-regeneration circumventing these problems. Here, we examined the neurogenic capacity of embryonic stem-like cells (ES-like cells) derived from the testis using neural growth factor inducers and utilized them to generate functional mature neurons. The neuronal differentiation of ES-like cells is induced in three stages. Stage 1 is related to embryoid body (EB) formation. To induce neuroprogenitor cells, EBs were cultured in the presence of retinoic acid, N2 supplement and fibroblast growth factor followed by culturing in a neurobasal medium containing B27, N2 supplements for additional 10 days, to allow the maturation and development of neuronal progenitor cells. The neurogenic differentiation was confirmed by immunostaining for markers of mature neurons. The differentiated neurons were positive for Tuj1 and Tau1. Real-time PCR dates indicated the expression of Nestin and Neuro D (neuroprogenitor markers) in induced cells at the second stage of the differentiation protocol. The differentiated mature neurons exhibited the specific neuron markers Map2 and β-tubulin. The functional maturity of neurons was confirmed by an electrophysiological analysis of passive and active neural membrane properties. These findings indicated a differentiation capacity of ES-like cells derived from the testis to functionally mature neurons, which proposes them as a novel cell source for neuroregenerative medicine.
Collapse
|
23
|
Tlapakova T, Nguyen TMX, Vegrichtova M, Sidova M, Strnadova K, Blahova M, Krylov V. Identification and characterization of Xenopus tropicalis common progenitors of Sertoli and peritubular myoid cell lineages. Biol Open 2016; 5:1275-82. [PMID: 27464670 PMCID: PMC5051652 DOI: 10.1242/bio.019265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The origin of somatic cell lineages during testicular development is controversial in mammals. Employing basal amphibian tetrapod Xenopus tropicalis we established a cell culture derived from testes of juvenile male. Expression analysis showed transcription of some pluripotency genes and Sertoli cell, peritubular myoid cell and mesenchymal cell markers. Transcription of germline-specific genes was downregulated. Immunocytochemistry revealed that a majority of cells express vimentin and co-express Sox9 and smooth muscle α-actin (Sma), indicating the existence of a common progenitor of Sertoli and peritubular myoid cell lineages. Microinjection of transgenic, red fluorescent protein (RFP)-positive somatic testicular cells into the peritoneal cavity of X. tropicalis tadpoles resulted in cell deposits in heart, pronephros and intestine, and later in a strong proliferation and formation of cell-to-cell net growing through the tadpole body. Immunohistochemistry analysis of transplanted tadpoles showed a strong expression of vimentin in RFP-positive cells. No co-localization of Sox9 and Sma signals was observed during the first three weeks indicating their dedifferentiation to migratory-active mesenchymal cells recently described in human testicular biopsies. Summary: We identified cells co-expressing differentiation markers of Sertoli and peritubular myoid cell lineages in X. tropicalis through the establishment and characterization of cell culture derived from juvenile testis.
Collapse
Affiliation(s)
- Tereza Tlapakova
- Charles University in Prague, Faculty of Science, Vinicna 7, Prague 2 128 44, Czech Republic
| | - Thi Minh Xuan Nguyen
- Charles University in Prague, Faculty of Science, Vinicna 7, Prague 2 128 44, Czech Republic
| | - Marketa Vegrichtova
- Charles University in Prague, Faculty of Science, Vinicna 7, Prague 2 128 44, Czech Republic
| | - Monika Sidova
- Charles University in Prague, Faculty of Science, Vinicna 7, Prague 2 128 44, Czech Republic Laboratory of Gene Expression, Institute of Biotechnology, Academy of Sciences of the Czech Republic, Videnska 1083, Prague 4 142 20, Czech Republic
| | - Karolina Strnadova
- Charles University in Prague, Faculty of Science, Vinicna 7, Prague 2 128 44, Czech Republic
| | - Monika Blahova
- Charles University in Prague, Faculty of Science, Vinicna 7, Prague 2 128 44, Czech Republic
| | - Vladimir Krylov
- Charles University in Prague, Faculty of Science, Vinicna 7, Prague 2 128 44, Czech Republic
| |
Collapse
|
24
|
Bhartiya D, Shaikh A, Anand S, Patel H, Kapoor S, Sriraman K, Parte S, Unni S. Endogenous, very small embryonic-like stem cells: critical review, therapeutic potential and a look ahead. Hum Reprod Update 2016; 23:41-76. [PMID: 27614362 DOI: 10.1093/humupd/dmw030] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/27/2016] [Accepted: 08/04/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Both pluripotent very small embryonic-like stem cells (VSELs) and induced pluripotent stem (iPS) cells were reported in 2006. In 2012, a Nobel Prize was awarded for iPS technology whereas even today the very existence of VSELs is not well accepted. The underlying reason is that VSELs exist in low numbers, remain dormant under homeostatic conditions, are very small in size and do not pellet down at 250-280g. The VSELs maintain life-long tissue homeostasis, serve as a backup pool for adult stem cells and are mobilized under stress conditions. An imbalance in VSELs function (uncontrolled proliferation) may result in cancer. SEARCH METHODS The electronic database 'Medline/Pubmed' was systematically searched with the subject heading term 'very small embryonic-like stem cells'. OBJECTIVE AND RATIONALE The most primitive stem cells that undergo asymmetric cell divisions to self-renew and give rise to progenitors still remain elusive in the hematopoietic system and testes, while the presence of stem cells in ovary is still being debated. We propose to review the available literature on VSELs, the methods of their isolation and characterization, their ontogeny, how they compare with embryonic stem (ES) cells, primordial germ cells (PGCs) and iPS cells, and their role in maintaining tissue homeostasis. The review includes a look ahead on how VSELs will result in paradigm shifts in basic reproductive biology. OUTCOMES Adult tissue-specific stem cells including hematopoietic, spermatogonial, ovarian and mesenchymal stem cells have good proliferation potential and are indeed committed progenitors (with cytoplasmic OCT-4), which arise by asymmetric cell divisions of pluripotent VSELs (with nuclear OCT-4). VSELs are the most primitive stem cells and postulated to be an overlapping population with the PGCs. Rather than migrating only to the gonads, PGCs migrate and survive in various adult body organs throughout life as VSELs. VSELs express both pluripotent and PGC-specific markers and are epigenetically and developmentally more mature compared with ES cells obtained from the inner cell mass of a blastocyst-stage embryo. As a result, VSELs readily differentiate into three embryonic germ layers and spontaneously give rise to both sperm and oocytes in vitro. Like PGCs, VSELs do not divide readily in culture, nor produce teratoma or integrate in the developing embryo. But this property of being relatively quiescent allows endogenous VSELs to survive various kinds of toxic insults. VSELs that survive oncotherapy can be targeted to induce endogenous regeneration of non-functional gonads. Transplanting healthy niche (mesenchymal) cells have resulted in improved gonadal function and live births. WIDER IMPLICATIONS Being quiescent, VSELs possibly do not accumulate genomic (nuclear or mitochondrial) mutations and thus may be ideal endogenous, pluripotent stem cell candidates for regenerative and reproductive medicine. The presence of VSELs in adult gonads and the fact that they survive oncotherapy may obviate the need to bank gonadal tissue for fertility preservation prior to oncotherapy. VSELs and their ability to undergo spermatogenesis/neo-oogenesis in the presence of a healthy niche will help identify newer strategies toward fertility restoration in cancer survivors, delaying menopause and also enabling aged mothers to have better quality eggs.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Ambreen Shaikh
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sandhya Anand
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Hiren Patel
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Sona Kapoor
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India
| | - Kalpana Sriraman
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,The Foundation for Medical Research, 84-A, RG Thadani Marg, Worli, Mumbai 400018, India
| | - Seema Parte
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,Department of Physiology, James Graham Brown Cancer Centre, University of Louisville School of Medicine, 2301 S 3rd St, Louisville, KY 40202, USA
| | - Sreepoorna Unni
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (Indian Council of Medical Research), Jehangir Merwanji Street, Parel, Mumbai 400 012, India.,Inter Disciplinary Studies Department, University College, Zayed University, Academic City, PO Box 19282, Dubai, United Arab Emirates
| |
Collapse
|
25
|
Yamada M, De Chiara L, Seandel M. Spermatogonial Stem Cells: Implications for Genetic Disorders and Prevention. Stem Cells Dev 2016; 25:1483-1494. [PMID: 27596369 PMCID: PMC5035912 DOI: 10.1089/scd.2016.0210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Spermatogonial stem cells (SSCs) propagate mammalian spermatogenesis throughout male reproductive life by continuously self-renewing and differentiating, ultimately, into sperm. SSCs can be cultured for long periods and restore spermatogenesis upon transplantation back into the native microenvironment in vivo. Conventionally, SSC research has been focused mainly on male infertility and, to a lesser extent, on cell reprogramming. With the advent of genome-wide sequencing technology, however, human studies have uncovered a wide range of pathogenic alleles that arise in the male germ line. A subset of de novo point mutations was shown to originate in SSCs and cause congenital disorders in children. This review describes both monogenic diseases (eg, Apert syndrome) and complex disorders that are either known or suspected to be driven by mutations in SSCs. We propose that SSC culture is a suitable model for studying the origin and mechanisms of these diseases. Lastly, we discuss strategies for future clinical implementation of SSC-based technology, from detecting mutation burden by sperm screening to gene correction in vitro.
Collapse
Affiliation(s)
- Makiko Yamada
- Joan and Sanford I Weill Medical College of Cornell University, 12295, Surgery, New York, New York, United States ;
| | - Letizia De Chiara
- Joan and Sanford I Weill Medical College of Cornell University, 12295, Surgery, New York, New York, United States ;
| | - Marco Seandel
- Joan and Sanford I Weill Medical College of Cornell University, 12295, Surgery, New York, New York, United States ;
| |
Collapse
|
26
|
Sadeghian-Nodoushan F, Aflatoonian R, Borzouie Z, Akyash F, Fesahat F, Soleimani M, Aghajanpour S, Moore HD, Aflatoonian B. Pluripotency and differentiation of cells from human testicular sperm extraction: An investigation of cell stemness. Mol Reprod Dev 2016; 83:312-23. [PMID: 27077675 DOI: 10.1002/mrd.22620] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 01/25/2016] [Indexed: 12/22/2022]
Abstract
Human male germ-line stem cells (hmGSCs) and human testis-derived embryonic stem cell-like (htESC-like) cells are claimed to be in vitro pluripotent counterparts of spermatogonial stem cells (SSCs), but the origin and pluripotency of human testis-derived cell cultures are still under debate. The aim of this study was to generate putative pluripotent stem cells in vitro from human testicular sperm-extracted (TESE) samples of infertile men, and to assess their pluripotency and capacity to differentiate. TESE samples were minced, enzymatically disaggregated and dispersed into single-cell or cluster suspensions, and then cultured. Initially, cell clusters resembled those described for hmGSCs and htESC-like cells, and were positive for markers such as OCT4/POU5F1, NANOG, and TRA-2-54. Prolonged propagation of cell clusters expressing pluripotency markers did not thrive; instead, the cells that emerged possessed characteristics of mesenchymal stromal cells (MSCs) such as STRO-1, CD105/EGLN1, CD13/ANPEP, SOX9, vimentin, and fibronectin. KIT, SOX2, and CD44 were not expressed by these MSCs. The multipotential differentiation capacity of these cells was confirmed using Oil Red-O and Alizarin Red staining after induction with specific culture conditions. It is therefore concluded that pluripotent stem cells could not be derived using the conditions previously reported to be successful for TESE samples.
Collapse
Affiliation(s)
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Zahra Borzouie
- Stem Cell Biology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Akyash
- Stem Cell Biology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farzaneh Fesahat
- Stem Cell Biology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mehrdad Soleimani
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samaneh Aghajanpour
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Harry D Moore
- Centre for Stem Cell Biology (CSCB), Department of Biomedical Sciences, The University of Sheffield, Western Bank, Alfred Denny Building, Sheffield, United Kingdom
| | - Behrouz Aflatoonian
- Stem Cell Biology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
27
|
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (ICMR), Mumbai 400 012, Maharashtra, India
| |
Collapse
|
28
|
Chen Z, Li Z, He Z. Plasticity of male germline stem cells and their applications in reproductive and regenerative medicine. Asian J Androl 2016; 17:367-72. [PMID: 25532577 PMCID: PMC4430934 DOI: 10.4103/1008-682x.143739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Spermatogonial stem cells (SSCs), also known as male germline stem cells, are a small subpopulation of type A spermatogonia with the potential of self-renewal to maintain stem cell pool and differentiation into spermatids in mammalian testis. SSCs are previously regarded as the unipotent stem cells since they can only give rise to sperm within the seminiferous tubules. However, this concept has recently been challenged because numerous studies have demonstrated that SSCs cultured with growth factors can acquire pluripotency to become embryonic stem-like cells. The in vivo and in vitro studies from peers and us have clearly revealed that SSCs can directly transdifferentiate into morphologic, phenotypic, and functional cells of other lineages. Direct conversion to the cells of other tissues has important significance for regenerative medicine. SSCs from azoospermia patients could be induced to differentiate into spermatids with fertilization and developmental potentials. As such, SSCs could have significant applications in both reproductive and regenerative medicine due to their unique and great potentials. In this review, we address the important plasticity of SSCs, with focuses on their self-renewal, differentiation, dedifferentiation, transdifferentiation, and translational medicine studies.
Collapse
Affiliation(s)
| | | | - Zuping He
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Cancer, Shanghai 200127; Department of Urology, Shanghai Human Sperm Bank, Shanghai Institute of Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001; Shanghai Key Laboratory of Assisted Reproduction and Reproductive Genetics, Shanghai 200127; Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China
| |
Collapse
|
29
|
Mei XX, Wang J, Wu J. Extrinsic and intrinsic factors controlling spermatogonial stem cell self-renewal and differentiation. Asian J Androl 2016; 17:347-54. [PMID: 25657085 PMCID: PMC4430931 DOI: 10.4103/1008-682x.148080] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Spermatogonial stem cells (SSCs), the stem cells responsible for male fertility, are one of a small number of cells with the abilities of both self-renewal and generation of large numbers of haploid cells. Technology improvements, most importantly, transplantation assays and in vitro culture systems have greatly expanded our understanding of SSC self-renewal and differentiation. Many important molecules crucial for the balance between self-renewal and differentiation have been recently identified although the exact mechanism(s) remain largely undefined. In this review, we give a brief introduction to SSCs, and then focus on extrinsic and intrinsic factors controlling SSCs self-renewal and differentiation.
Collapse
Affiliation(s)
| | | | - Ji Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio X Institutes, Shanghai Jiao Tong University, Shanghai 200240; Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004; Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China
| |
Collapse
|
30
|
von Kopylow K, Schulze W, Salzbrunn A, Spiess AN. Isolation and gene expression analysis of single potential human spermatogonial stem cells. Mol Hum Reprod 2016; 22:229-39. [PMID: 26792870 DOI: 10.1093/molehr/gaw006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/15/2016] [Indexed: 12/18/2022] Open
Abstract
STUDY HYPOTHESIS It is possible to isolate pure populations of single potential human spermatogonial stem cells without somatic contamination for down-stream applications, for example cell culture and gene expression analysis. STUDY FINDING We isolated pure populations of single potential human spermatogonial stem cells (hSSC) without contaminating somatic cells and analyzed gene expression of these cells via single-cell real-time RT-PCR. WHAT IS KNOWN ALREADY The isolation of a pure hSSC fraction could enable clinical applications such as fertility preservation for prepubertal boys and in vitro-spermatogenesis. By utilizing largely nonspecific markers for the isolation of spermatogonia (SPG) and hSSC, previously published cell selection methods are not able to deliver pure target cell populations without contamination by testicular somatic cells. However, uniform cell populations free of somatic cells are necessary to guarantee defined growth conditions in cell culture experiments and to prevent unintended stem cell differentiation. Fibroblast growth factor receptor 3 (FGFR3) is a cell surface protein of human undifferentiated A-type SPG and a promising candidate marker for hSSC. It is exclusively expressed in small, non-proliferating subgroups of this spermatogonial cell type together with the pluripotency-associated protein and spermatogonial nuclear marker undifferentiated embryonic cell transcription factor 1 (UTF1). STUDY DESIGN, SAMPLES/MATERIALS, METHODS We specifically selected the FGFR3-positive spermatogonial subpopulation from two 30 mg biopsies per patient from a total of 37 patients with full spermatogenesis and three patients with meiotic arrest. We then employed cell selection with magnetic beads in combination with a fluorescence-activated cell sorter antibody directed against human FGFR3 to tag and visually identify human FGFR3-positive spermatogonia. Positively selected and bead-labeled cells were subsequently picked with a micromanipulator. Analysis of the isolated cells was carried out by single-cell real-time RT-PCR, real-time RT-PCR, immunocytochemistry and live/dead staining. MAIN RESULTS AND THE ROLE OF CHANCE Single-cell real-time RT-PCR and real-time RT-PCR of pooled cells indicate that bead-labeled single cells express FGFR3 with high heterogeneity at the mRNA level, while bead-unlabeled cells lack FGFR3 mRNA. Furthermore, isolated cells exhibit strong immunocytochemical staining for the stem cell factor UTF1 and are viable. LIMITATIONS, REASONS FOR CAUTION The cell population isolated in this study has to be tested for their potential stem cell characteristics via xenotransplantation. Due to the small amount of the isolated cells, propagation by cell culture will be essential. Other potential hSSC without FGFR3 surface expression will not be captured with the provided experimental design. WIDER IMPLICATIONS OF THE FINDINGS The technical approach as developed in this work could encourage the scientific community to test other established or novel hSSC markers on single SPG that present with potential stem cell-like features. STUDY FUNDING AND COMPETING INTERESTS The project was funded by the DFG Research Unit FOR1041 Germ cell potential (SCH 587/3-2) and DFG grants to K.v.K. (KO 4769/2-1) and A.-N.S. (SP 721/4-1). The authors declare no competing interests.
Collapse
Affiliation(s)
- K von Kopylow
- Department of Andrology, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - W Schulze
- Department of Andrology, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany MVZ Fertility Center Hamburg GmbH, amedes-group, 20095 Hamburg, Germany
| | - A Salzbrunn
- Department of Andrology, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - A-N Spiess
- Department of Andrology, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
31
|
Expression of Genes Related to Germ Cell Lineage and Pluripotency in Single Cells and Colonies of Human Adult Germ Stem Cells. Stem Cells Int 2015; 2016:8582526. [PMID: 26649052 PMCID: PMC4655073 DOI: 10.1155/2016/8582526] [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] [Received: 12/03/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 01/06/2023] Open
Abstract
The aim of this study was to elucidate the molecular status of single human adult germ stem cells (haGSCs) and haGSC colonies, which spontaneously developed from the CD49f MACS and matrix- (collagen−/laminin+ binding-) selected fraction of enriched spermatogonia. Single-cell transcriptional profiling by Fluidigm BioMark system of a long-term cultured haGSCs cluster in comparison to human embryonic stem cells (hESCs) and human fibroblasts (hFibs) revealed that haGSCs showed a characteristic germ- and pluripotency-associated gene expression profile with some similarities to hESCs and with a significant distinction from somatic hFibs. Genome-wide comparisons with microarray analysis confirmed that different haGSC colonies exhibited gene expression heterogeneity with more or less pluripotency. The results of this study confirm that haGSCs are adult stem cells with a specific molecular gene expression profile in vitro, related but not identical to true pluripotent stem cells. Under ES-cell conditions haGSC colonies could be selected and maintained in a partial pluripotent state at the molecular level, which may be related to their cell plasticity and potential to differentiate into cells of all germ layers.
Collapse
|
32
|
Wang X, Chen T, Zhang Y, Li B, Xu Q, Song C. Isolation and Culture of Pig Spermatogonial Stem Cells and Their in Vitro Differentiation into Neuron-Like Cells and Adipocytes. Int J Mol Sci 2015; 16:26333-46. [PMID: 26556335 PMCID: PMC4661817 DOI: 10.3390/ijms161125958] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 10/21/2015] [Accepted: 10/23/2015] [Indexed: 12/14/2022] Open
Abstract
Spermatogonial stem cells (SSCs) renew themselves throughout the life of an organism and also differentiate into sperm in the adult. They are multipopent and therefore, can be induced to differentiate into many cells types in vitro. SSCs from pigs, considered an ideal animal model, are used in studies of male infertility, regenerative medicine, and preparation of transgenic animals. Here, we report on a culture system for porcine SSCs and the differentiation of these cells into neuron-like cells and adipocytes. SSCs and Sertoli cells were isolated from neonatal piglet testis by differential adhesion and SSCs were cultured on a feeder layer of Sertoli cells. Third-generation SSCs were induced to differentiate into neuron-like cells by addition of retinoic acid, β-mercaptoethanol, and 3-isobutyl-1-methylxanthine (IBMX) to the induction media and into adipocytes by the addition of hexadecadrol, insulin, and IBMX to the induction media. The differentiated cells were characterized by biochemical staining, qRT-PCR, and immunocytochemistry. The cells were positive for SSC markers, including alkaline phosphatase and SSC-specific genes, consistent with the cells being undifferentiated. The isolated SSCs survived on the Sertoli cells for 15 generations. Karyotyping confirmed that the chromosomal number of the SSCs were normal for pig (2n = 38, n = 19). Pig SSCs were successfully induced into neuron-like cells eight days after induction and into adipocytes 22 days after induction as determined by biochemical and immunocytochemical staining. qPCR results also support this conclusion. The nervous tissue markers genes, Nestin and β-tubulin, were expressed in the neuron-like cells and the adipocyte marker genes, PPARγ and C/EBPα, were expressed in the adipocytes.
Collapse
Affiliation(s)
- Xiaoyan Wang
- College of Animal Science & Technology, Yangzhou University, Yangzhou 225009, China.
| | - Tingfeng Chen
- College of Animal Science & Technology, Yangzhou University, Yangzhou 225009, China.
| | - Yani Zhang
- College of Animal Science & Technology, Yangzhou University, Yangzhou 225009, China.
| | - Bichun Li
- College of Animal Science & Technology, Yangzhou University, Yangzhou 225009, China.
| | - Qi Xu
- College of Animal Science & Technology, Yangzhou University, Yangzhou 225009, China.
| | - Chengyi Song
- College of Animal Science & Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
33
|
Saito S, Lin YC, Murayama Y, Nakamura Y, Eckner R, Niemann H, Yokoyama KK. Retracted article: In vitro derivation of mammalian germ cells from stem cells and their potential therapeutic application. Cell Mol Life Sci 2015; 72:4545-60. [PMID: 26439925 PMCID: PMC4628088 DOI: 10.1007/s00018-015-2020-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 07/27/2015] [Accepted: 08/07/2015] [Indexed: 01/12/2023]
Abstract
Pluripotent stem cells (PSCs) are a unique type of cells because they
exhibit the characteristics of self-renewal and pluripotency. PSCs may be induced to
differentiate into any cell type, even male and female germ cells, suggesting their
potential as novel cell-based therapeutic treatment for infertility problems.
Spermatogenesis is an intricate biological process that starts from self-renewal of
spermatogonial stem cells (SSCs) and leads to differentiated haploid spermatozoa.
Errors at any stage in spermatogenesis may result in male infertility. During the
past decade, much progress has been made in the derivation of male germ cells from
various types of progenitor stem cells. Currently, there are two main approaches for
the derivation of functional germ cells from PSCs, either the induction of in vitro
differentiation to produce haploid cell products, or combination of in vitro
differentiation and in vivo transplantation. The production of mature and fertile
spermatozoa from stem cells might provide an unlimited source of autologous gametes
for treatment of male infertility. Here, we discuss the current state of the art
regarding the differentiation potential of SSCs, embryonic stem cells, and induced
pluripotent stem cells to produce functional male germ cells. We also discuss the
possible use of livestock-derived PSCs as a novel option for animal reproduction and
infertility treatment.
Collapse
Affiliation(s)
- Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita, Tochigi, 329-1571, Japan. .,SPK Co., Ltd., Aizuwakamatsu, Fukushima, 965-0025, Japan. .,College of Engineering, Nihon University, Koriyama, Fukushima, 963-8642, Japan.
| | - Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaoshiung Medical University, 100 Shin-Chuan 1st Road, Kaohsiung, 807, Taiwan
| | - Yoshinobu Murayama
- College of Engineering, Nihon University, Koriyama, Fukushima, 963-8642, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, 3050074, Japan
| | - Richard Eckner
- Department of Biochemistry and Molecular Biology, Rutgers New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07101, USA
| | - Heiner Niemann
- Institute of Farm Animal Genetics, Friedrich-Löffler-Institut, Mariensee, 31535, Neustadt, Germany.
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine, Center of Stem Cell Research, Center of Environmental Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd, San Ming District, Kaohsiung, 807, Taiwan. .,Faculty of Science and Engineering, Tokushima Bunri University, Sanuki, 763-2193, Japan. .,Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
34
|
Aponte PM. Spermatogonial stem cells: Current biotechnological advances in reproduction and regenerative medicine. World J Stem Cells 2015; 7:669-680. [PMID: 26029339 PMCID: PMC4444608 DOI: 10.4252/wjsc.v7.i4.669] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/13/2015] [Accepted: 04/14/2015] [Indexed: 02/06/2023] Open
Abstract
Spermatogonial stem cells (SSCs) are the germ stem cells of the seminiferous epithelium in the testis. Through the process of spermatogenesis, they produce sperm while concomitantly keeping their cellular pool constant through self-renewal. SSC biology offers important applications for animal reproduction and overcoming human disease through regenerative therapies. To this end, several techniques involving SSCs have been developed and will be covered in this article. SSCs convey genetic information to the next generation, a property that can be exploited for gene targeting. Additionally, SSCs can be induced to become embryonic stem cell-like pluripotent cells in vitro. Updates on SSC transplantation techniques with related applications, such as fertility restoration and preservation of endangered species, are also covered on this article. SSC suspensions can be transplanted to the testis of an animal and this has given the basis for SSC functional assays. This procedure has proven technically demanding in large animals and men. In parallel, testis tissue xenografting, another transplantation technique, was developed and resulted in sperm production in testis explants grafted into ectopical locations in foreign species. Since SSC culture holds a pivotal role in SSC biotechnologies, current advances are overviewed. Finally, spermatogenesis in vitro, already demonstrated in mice, offers great promises to cope with reproductive issues in the farm animal industry and human clinical applications.
Collapse
|
35
|
Sisakhtnezhad S, Bahrami AR, Matin MM, Dehghani H, Momeni-Moghaddam M, Boozarpour S, Farshchian M, Dastpak M. The molecular signature and spermatogenesis potential of newborn chicken spermatogonial stem cells in vitro. In Vitro Cell Dev Biol Anim 2015; 51:415-25. [DOI: 10.1007/s11626-014-9843-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/28/2014] [Indexed: 01/08/2023]
|
36
|
Altman E, Yango P, Moustafa R, Smith JF, Klatsky PC, Tran ND. Characterization of human spermatogonial stem cell markers in fetal, pediatric, and adult testicular tissues. Reproduction 2014; 148:417-27. [PMID: 25030892 PMCID: PMC4599365 DOI: 10.1530/rep-14-0123] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autologous spermatogonial stem cell (SSC) transplantation is a potential therapeutic modality for patients with azoospermia following cancer treatment. For this promise to be realized, definitive membrane markers of prepubertal and adult human SSCs must be characterized in order to permit SSC isolation and subsequent expansion. This study further characterizes the markers of male gonocytes, prespermatogonia, and SSCs in humans. Human fetal, prepubertal, and adult testicular tissues were analyzed by confocal microscopy, fluorescence-activated cell sorting, and qRT-PCR for the expression of unique germ cell membrane markers. During male fetal development, THY1 and KIT (C-Kit) are transient markers of gonocytes but not in prespermatogonia and post-natal SSCs. Although KIT expression is detected in gonocytes, THY1 expression is also detected in the somatic component of the fetal testes in addition to gonocytes. In the third trimester of gestation, THY1 expression shifts exclusively to the somatic cells of the testes where it continues to be detected only in the somatic cells postnatally. In contrast, SSEA4 expression was only detected in the gonocytes, prespermatogonia, SSCs, and Sertoli cells of the fetal and prepubertal testes. After puberty, SSEA4 expression can only be detected in primitive spermatogonia. Thus, although THY1 and KIT are transient markers of gonocytes, SSEA4 is the only common membrane marker of gonocytes, prespermatogonia, and SSCs from fetal through adult human development. This finding is essential for the isolation of prepubertal and adult SSCs, which may someday permit fertility preservation and reversal of azoospermia following cancer treatment.
Collapse
Affiliation(s)
- Eran Altman
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Pamela Yango
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Radwa Moustafa
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - James F Smith
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Peter C Klatsky
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| | - Nam D Tran
- Department of ObstetricsGynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USAHelen Schneider Hospital for WomenRabin Medical Center, Petah Tikva, IsraelDepartment of UrologyUniversity of California, San Francisco, San Francisco, California, USADepartment of Obstetrics and GynecologyAlbert Einstein University, Bronx, New York, USA
| |
Collapse
|
37
|
Nickkholgh B, Mizrak SC, van Daalen SKM, Korver CM, Sadri-Ardekani H, Repping S, van Pelt AMM. Genetic and epigenetic stability of human spermatogonial stem cells during long-term culture. Fertil Steril 2014; 102:1700-7.e1. [PMID: 25256932 DOI: 10.1016/j.fertnstert.2014.08.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 08/08/2014] [Accepted: 08/14/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To determine the genetic and epigenetic stability of human spermatogonial stem cells (SSCs) during long-term culture. DESIGN Experimental basic science study. SETTING Reproductive biology laboratory. PATIENT(S) Cryopreserved human testicular tissue from two prostate cancer patients with normal spermatogenesis. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Testicular cells before and 50 days after culturing were subjected to ITGA6 magnetic-activated cell sorting to enrich for SSCs. Individual spermatogonia were analyzed for aneuploidies with the use of single-cell 24-chromosome screening. Furthermore, the DNA methylation statuses of the paternally imprinted genes H19, H19-DMR (differentially methylated region), and MEG3 and the maternally imprinted genes KCNQ1OT1 and PEG3 were identified by means of bisulfite sequencing. RESULTS(S) Aneuploidy screening showed euploidy with no chromosomal abnormalities in all cultured and most noncultured spermatogonia from both patients. The methylation assays demonstrated demethylation of the paternally imprinted genes H19, H19-DMR, and MEG3 of 11%-28%, 43%-68%, and 18%-26%, respectively, and increased methylation of the maternally imprinted genes PEG 3 and KCNQ1OT of 13%-50% and 30%-38%, respectively, during culture. CONCLUSION(S) In the current culture system for human SSCs propagation, genomic stability is preserved, which is important for future clinical use. Whether the observed changes in methylation status have consequences on functionality of SSCs or health of offspring derived from transplanted SSCs requires further investigation.
Collapse
Affiliation(s)
- Bita Nickkholgh
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - S Canan Mizrak
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Saskia K M van Daalen
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Cindy M Korver
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Hooman Sadri-Ardekani
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sjoerd Repping
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Ans M M van Pelt
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
38
|
Smith JF, Yango P, Altman E, Choudhry S, Poelzl A, Zamah AM, Rosen M, Klatsky PC, Tran ND. Testicular niche required for human spermatogonial stem cell expansion. Stem Cells Transl Med 2014; 3:1043-54. [PMID: 25038247 DOI: 10.5966/sctm.2014-0045] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prepubertal boys treated with high-dose chemotherapy do not have an established means of fertility preservation because no established in vitro technique exists to expand and mature purified spermatogonial stem cells (SSCs) to functional sperm in humans. In this study, we define and characterize the unique testicular cellular niche required for SSC expansion using testicular tissues from men with normal spermatogenesis. Highly purified SSCs and testicular somatic cells were isolated by fluorescence-activated cell sorting using SSEA-4 and THY1 as markers of SSCs and somatic cells. Cells were cultured on various established niches to assess their role in SSC expansion in a defined somatic cellular niche. Of all the niches examined, cells in the SSEA-4 population exclusively bound to adult testicular stromal cells, established colonies, and expanded. Further characterization of these testicular stromal cells revealed distinct mesenchymal markers and the ability to undergo differentiation along the mesenchymal lineage, supporting a testicular multipotent stromal cell origin. In vitro human SSC expansion requires a unique niche provided exclusively by testicular multipotent stromal cells with mesenchymal properties. These findings provide an important foundation for developing methods of inducing SSC growth and maturation in prepubertal testicular tissue, essential to enabling fertility preservation for these boys.
Collapse
Affiliation(s)
- James F Smith
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Pamela Yango
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Eran Altman
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Shweta Choudhry
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Andrea Poelzl
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Alberuni M Zamah
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Mitchell Rosen
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Peter C Klatsky
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| | - Nam D Tran
- Departments of Urology and Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA; Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tiqva, Israel; Department of Obstetrics and Gynecology, Albert Einstein University, Bronx, New York, USA
| |
Collapse
|
39
|
Langenstroth D, Kossack N, Westernströer B, Wistuba J, Behr R, Gromoll J, Schlatt S. Separation of somatic and germ cells is required to establish primate spermatogonial cultures. Hum Reprod 2014; 29:2018-31. [PMID: 24963164 DOI: 10.1093/humrep/deu157] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Can primate spermatogonial cultures be optimized by application of separation steps and well defined culture conditions? SUMMARY ANSWER We identified the cell fraction which provides the best source for primate spermatogonia when prolonged culture is desired. WHAT IS KNOWN ALREADY Man and marmoset show similar characteristics in regard to germ cell development and function. Several protocols for isolation and culture of human testis-derived germline stem cells have been described. Subsequent analysis revealed doubts on the germline origin of these cells and characterized them as mesenchymal stem cells or fibroblasts. Studies using marmosets as preclinical model confirmed that the published isolation protocols did not lead to propagation of germline cells. STUDY DESIGN, SIZE, DURATION Testicular cells derived from nine adult marmoset monkeys (Callithrix jacchus) were cultured for 1, 3, 6 and 11 days and consecutively analyzed for the presence of spermatogonia, differentiating germ cells and testicular somatic cells. PARTICIPANTS/MATERIALS, SETTING, METHODS Testicular tissue of nine adult marmoset monkeys was enzymatically dissociated and subjected to two different cell culture approaches. In the first approach all cells were kept in the same dish (non-separate culture, n = 5). In the second approach the supernatant cells were transferred into a new dish 24 h after seeding and subsequently supernatant and attached cells were cultured separately (separate culture, n = 4). Real-time quantitative PCR and immunofluorescence were used to analyze the expression of reliable germ cell and somatic markers throughout the culture period. Germ cell transplantation assays and subsequent wholemount analyses were performed to functionally evaluate the colonization of spermatogonial cells. MAIN RESULTS AND THE ROLE OF CHANCE This is the first report revealing an efficient isolation and culture of putative marmoset spermatogonial stem cells with colonization ability. Our results indicate that a separation of spermatogonia from testicular somatic cells is a crucial step during cell preparation. We identified the overgrowth of more rapidly expanding somatic cells to be a major problem when establishing spermatogonial cultures. Initiating germ cell cultures from the supernatant and maintaining germ cells in suspension cultures minimized the somatic cell contamination and provided enriched germ cell fractions which displayed after 11 days of culture a significantly higher expression of germ cell markers genes (DDX-4, MAGE A-4; P < 0.05) compared with separately cultured attached cells. Additionally, germ cell transplantation experiments demonstrated a significantly higher absolute number of cells with colonization ability (P < 0.001) in supernatant cells after 11 days of separate culture. LIMITATIONS, REASONS FOR CAUTION This study presents a relevant aspect for the successful setup of spermatogonial cultures but provides limited data regarding the question of whether the long-term maintenance of spermatogonia can be achieved. Transfer of these preclinical data to man may require modifications of the protocol. WIDER IMPLICATIONS OF THE FINDINGS Spermatogonial cultures from rodents have become important and innovative tools for basic and applied research in reproductive biology and veterinary medicine. It is expected that spermatogonia-based strategies will be transformed into clinical applications for the treatment of male infertility. Our data in the marmoset monkey may be highly relevant to establish spermatogonial cultures of human testes. STUDY FUNDING/COMPETING INTERESTS Funding was provided by the DFG-Research Unit FOR 1041 Germ Cell Potential (SCHL394/11-2) and by the Graduate Program Cell Dynamics and Disease (CEDAD) together with the International Max Planck Research School - Molecular Biomedicine (IMPRS-MBM). The authors declare that there is no conflict of interest. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Daniel Langenstroth
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Nina Kossack
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Birgit Westernströer
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Joachim Wistuba
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Rüdiger Behr
- Stem Cell Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
| | - Jörg Gromoll
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| | - Stefan Schlatt
- Institute of Reproduction and Regenerative Biology, Centre of Reproductive Medicine and Andrology, Albert-Schweitzer-Campus 1, Building D11, 48149 Münster, Germany
| |
Collapse
|
40
|
Nickkholgh B, Mizrak SC, Korver CM, van Daalen SKM, Meissner A, Repping S, van Pelt AMM. Enrichment of spermatogonial stem cells from long-term cultured human testicular cells. Fertil Steril 2014; 102:558-565.e5. [PMID: 24864010 DOI: 10.1016/j.fertnstert.2014.04.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 04/13/2014] [Accepted: 04/15/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To evaluate the degree of enrichment of spermatogonial stem cells (SSCs) from human testicular cell cultures by ITGA6+, HLA-/ITGA6+, GPR125+, and HLA-/GPR125+ magnetic-assisted cell sorting (MACS). DESIGN Experimental basic science study. SETTING Reproductive biology laboratory. PATIENT(S) Multiple samples of cryopreserved human testicular cells from two prostate cancer patients with normal spermatogenesis. INTERVENTION(S) Cultured human testicular cells subjected to four sorting strategies based on MACS and xenotransplanted to the testes of mice to determine the enrichment for SSCs. MAIN OUTCOME MEASURE(S) Enrichment for human spermatogonia and SSCs tested by expression analysis of spermatogonial markers ITGA6, GPR125, ZBTB16, UCHL1, and ID4 using quantitative real-time polymerase chain reaction (qPCR) and by xenotransplantation into the testes of mice, respectively. RESULT(S) Compared with the nonsorted cultured testicular cells, only the ITGA6+ and HLA-/GPR125+ sorted cells showed enrichment for ID4. No difference in expression of ZBTB16 and UCHL1 was observed. Xenotransplantation of the sorted cell fractions showed a 7.1-fold enrichment of SSCs with ITGA6+. CONCLUSION(S) Magnetic-assisted cell sorting of cultured human testicular cells using ITGA6 allows for enrichment of SSCs, which aids in further molecular characterization of cultured human SSCs and enhances testicular colonization upon transplantation in future clinical settings.
Collapse
Affiliation(s)
- Bita Nickkholgh
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sefika Canan Mizrak
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Cindy M Korver
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Saskia K M van Daalen
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Andreas Meissner
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sjoerd Repping
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Ans M M van Pelt
- Center for Reproductive Medicine, Women's and Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
41
|
Differential gene expression profiling of enriched human spermatogonia after short- and long-term culture. BIOMED RESEARCH INTERNATIONAL 2014; 2014:138350. [PMID: 24738045 PMCID: PMC3971551 DOI: 10.1155/2014/138350] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/19/2013] [Indexed: 01/15/2023]
Abstract
This study aimed to provide a molecular signature for enriched adult human stem/progenitor spermatogonia during short-term (<2 weeks) and long-term culture (up to more than 14 months) in comparison to human testicular fibroblasts and human embryonic stem cells. Human spermatogonia were isolated by CD49f magnetic activated cell sorting and collagen(-)/laminin(+) matrix binding from primary testis cultures obtained from ten adult men. For transcriptomic analysis, single spermatogonia-like cells were collected based on their morphology and dimensions using a micromanipulation system from the enriched germ cell cultures. Immunocytochemical, RT-PCR and microarray analyses revealed that the analyzed populations of cells were distinct at the molecular level. The germ- and pluripotency-associated genes and genes of differentiation/spermatogenesis pathway were highly expressed in enriched short-term cultured spermatogonia. After long-term culture, a proportion of cells retained and aggravated the "spermatogonial" gene expression profile with the expression of germ and pluripotency-associated genes, while in the majority of long-term cultured cells this molecular profile, typical for the differentiation pathway, was reduced and more genes related to the extracellular matrix production and attachment were expressed. The approach we provide here to study the molecular status of in vitro cultured spermatogonia may be important to optimize the culture conditions and to evaluate the germ cell plasticity in the future.
Collapse
|
42
|
Hou J, Yang S, Yang H, Liu Y, Liu Y, Hai Y, Chen Z, Guo Y, Gong Y, Gao WQ, Li Z, He Z. Generation of male differentiated germ cells from various types of stem cells. Reproduction 2014; 147:R179-88. [PMID: 24534952 DOI: 10.1530/rep-13-0649] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Infertility is a major and largely incurable disease caused by disruption and loss of germ cells. It affects 10-15% of couples, and male factor accounts for half of the cases. To obtain human male germ cells 'especially functional spermatids' is essential for treating male infertility. Currently, much progress has been made on generating male germ cells, including spermatogonia, spermatocytes, and spermatids, from various types of stem cells. These germ cells can also be used in investigation of the pathology of male infertility. In this review, we focused on advances on obtaining male differentiated germ cells from different kinds of stem cells, with an emphasis on the embryonic stem (ES) cells, the induced pluripotent stem (iPS) cells, and spermatogonial stem cells (SSCs). We illustrated the generation of male differentiated germ cells from ES cells, iPS cells and SSCs, and we summarized the phenotype for these stem cells, spermatocytes and spermatids. Moreover, we address the differentiation potentials of ES cells, iPS cells and SSCs. We also highlight the advantages, disadvantages and concerns on derivation of the differentiated male germ cells from several types of stem cells. The ability of generating mature and functional male gametes from stem cells could enable us to understand the precise etiology of male infertility and offer an invaluable source of autologous male gametes for treating male infertility of azoospermia patients.
Collapse
Affiliation(s)
- Jingmei Hou
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Shi Yang
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Hao Yang
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Yang Liu
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Yun Liu
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Yanan Hai
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Zheng Chen
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Ying Guo
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Yuehua Gong
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Zheng Li
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, China
| | - Zuping He
- State Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, ChinaState Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, ChinaState Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, ChinaShanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai 200135, ChinaShanghai Key Laboratory of Reproductive MedicineShanghai 200025, ChinaState Key Laboratory of Oncogenes and Related GenesStem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, ChinaDepartment of UrologyShanghai Human Sperm Bank, Shanghai Institute of Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 845 Linshan Road, Shanghai 200135, Chin
| |
Collapse
|
43
|
Pluripotent Very Small Embryonic-like Stem Cells in Adult Mammalian Gonads. STEM CELL BIOLOGY AND REGENERATIVE MEDICINE 2014. [DOI: 10.1007/978-1-4939-1001-4_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Nazm Bojnordi M, Movahedin M, Tiraihi T, Javan M, Ghasemi Hamidabadi H. Oligoprogenitor Cells Derived from Spermatogonia Stem Cells Improve Remyelination in Demyelination Model. Mol Biotechnol 2013; 56:387-93. [DOI: 10.1007/s12033-013-9722-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
45
|
Chikhovskaya J, van Daalen S, Korver C, Repping S, van Pelt A. Mesenchymal origin of multipotent human testis-derived stem cells in human testicular cell cultures. ACTA ACUST UNITED AC 2013; 20:155-67. [DOI: 10.1093/molehr/gat076] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
46
|
Are Mesenchymal Cells Indeed Pluripotent Stem Cells or Just Stromal Cells? OCT-4 and VSELs Biology Has Led to Better Understanding. Stem Cells Int 2013; 2013:547501. [PMID: 24187558 PMCID: PMC3800663 DOI: 10.1155/2013/547501] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/06/2013] [Accepted: 08/26/2013] [Indexed: 12/24/2022] Open
Abstract
Stem cells have excited researchers because of their potential to regenerate. However, which stem cells will be the best candidate for regenerative medicine remains an enigma. Compared to pluripotent stem cells with associated risks of immune rejection and teratoma formation, adult stem cells especially the mesenchymal stem cells (MSCs) are hyped to be a suitable alternate since they also exhibit pluripotent properties. This review shows that there is a subpopulation of pluripotent very small embryonic-like stem cells (VSELs) among MSCs culture. The two populations differ from each other in expression pattern of OCT-4. VSELs exhibit nuclear OCT-4A, whereas the MSCs have cytoplasmic OCT-4B, similar to our earlier findings in testis and ovary. Pluripotent VSELs with nuclear OCT-4A exist in various adult body organs, and the immediate progenitors express cytoplasmic OCT-4B which is eventually lost as the cell differentiates further. To conclude it is essential to discriminate between nuclear and cytoplasmic OCT-4 expression and also to acknowledge the presence of VSELs.
Collapse
|
47
|
Kossack N, Terwort N, Wistuba J, Ehmcke J, Schlatt S, Schöler H, Kliesch S, Gromoll J. A combined approach facilitates the reliable detection of human spermatogonia in vitro. Hum Reprod 2013; 28:3012-25. [PMID: 24001715 DOI: 10.1093/humrep/det336] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Does a combined approach allow for the unequivocal detection of human germ cells and particularly of spermatogonia in vitro? SUMMARY ANSWER Based on our findings, we conclude that an approach comprising: (i) the detailed characterization of patients and tissue samples prior to the selection of biopsies, (ii) the use of unambiguous markers for the characterization of cultures and (iii) the use of biopsies lacking the germ cell population as a negative control is the prerequisite for the establishment of human germ cell cultures. WHAT IS KNOWN ALREADY The use of non-specific marker genes and the failure to assess the presence of testicular somatic cell types in germ cell cultures may have led to a misinterpretation of results and the erroneous description of germ cells in previous studies. STUDY DESIGN, SIZE, DURATION Testicular biopsies were selected from a pool of 264 consecutively obtained biopsies. Based on the histological diagnosis, biopsies with distinct histological phenotypes were selected (n = 35) to analyze the expression of germ cell and somatic cell markers. For germ cell culture experiments, gonadotrophin levels and clinical data were used as selection criteria resulting in the following two groups: (i) biopsies with qualitatively intact spermatogenesis (n = 4) and (ii) biopsies from Klinefelter syndrome Klinefelter patients lacking the germ cell population (n = 3). PARTICIPANTS/MATERIALS, SETTING, METHODS Quantitative real-time PCR analyses were performed to evaluate the specificity of 18 selected germ cell and 3 somatic marker genes. Cell specificity of individual markers was subsequently validated using immunohistochemistry. Finally, testicular cell cultures were established and were analyzed after 10 days for the expression of germ cell- (UTF1, FGFR3, MAGE A4, DDX4) and somatic cell-specific markers (SMA, VIM, LHCGR) at the RNA and the protein levels. MAIN RESULTS AND THE ROLE OF CHANCE Interestingly, only 9 out of 18 marker genes reflected the presence of germ cells and cell specificity could be validated using immunohistochemistry. Furthermore, VIM, SMA and LHCGR were found to reflect the presence of testicular somatic cells at the RNA and the protein levels. Using this validated marker panel and biopsies lacking the germ cell population (n = 3) as a negative control, we demonstrated that germ cell cultures containing spermatogonia can be established from biopsies with normal spermatogenesis (n = 4) and that these cultures can be maintained for the period of 10 days. However, marker profiling has to be performed at regular time points as the composition of testicular cell types may continuously change under longer term culture conditions. LIMITATIONS, REASONS FOR CAUTION There are significant differences regarding the spermatogonial stem cell (SSC) system and spermatogenesis between rodents and primates. It is therefore possible that marker genes that do not reflect the presence of spermatogonia in the human are specific for spermatogonia in other animal models. WIDER IMPLICATIONS OF THE FINDINGS While some studies have reported that human SSCs can be maintained in vitro and show characteristics of pluripotency, the germ cell origin and the differentiation potential of these cells were subsequently called into question. This study provides critical insights into possible sources for the misinterpretation of results regarding the presence of germ cells in human testicular cell cultures and our findings can therefore help to avoid conflicting reports in the future. STUDY FUNDING/COMPETING INTEREST(S) This project was supported by the Stem Cell Network North Rhine-Westphalia and the Innovative Medical Research of the University of Münster Medical School (Grant KO111014). In addition, it was funded by the DFG-Research Unit FOR 1041 Germ Cell Potential (GR 1547/11-1 and SCHL 394/11-2), the BMBF (01GN0809/10) and the IZKF (CRA 03/09). The authors declare that there is no conflict of interest. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- N Kossack
- Institute for Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Albert-Schweitzer-Campus 1 (D11), Münster 48149, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Harkey MA, Asano A, Zoulas ME, Torok-Storb B, Nagashima J, Travis A. Isolation, genetic manipulation, and transplantation of canine spermatogonial stem cells: progress toward transgenesis through the male germ-line. Reproduction 2013; 146:75-90. [PMID: 23690628 DOI: 10.1530/rep-13-0086] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The dog is recognized as a highly predictive model for preclinical research. Its size, life span, physiology, and genetics more closely match human parameters than do those of the mouse model. Investigations of the genetic basis of disease and of new regenerative treatments have frequently taken advantage of canine models. However, full utility of this model has not been realized because of the lack of easy transgenesis. Blastocyst-mediated transgenic technology developed in mice has been very slow to translate to larger animals, and somatic cell nuclear transfer remains technically challenging, expensive, and low yield. Spermatogonial stem cell (SSC) transplantation, which does not involve manipulation of ova or blastocysts, has proven to be an effective alternative approach for generating transgenic offspring in rodents and in some large animals. Our recent demonstration that canine testis cells can engraft in a host testis, and generate donor-derived sperm, suggests that SSC transplantation may offer a similar avenue to transgenesis in the canine model. Here, we explore the potential of SSC transplantation in dogs as a means of generating canine transgenic models for preclinical models of genetic diseases. Specifically, we i) established markers for identification and tracking canine spermatogonial cells; ii) established methods for enrichment and genetic manipulation of these cells; iii) described their behavior in culture; and iv) demonstrated engraftment of genetically manipulated SSC and production of transgenic sperm. These findings help to set the stage for generation of transgenic canine models via SSC transplantation.
Collapse
Affiliation(s)
- Michael A Harkey
- Clinical Research, Division, Fred Hutchinson Cancer Research Center, Mail Stop D1-100, 1100 Fairview Avenue North, PO Box 19024, Seattle, Washington 98109-1024, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Choi WY, Jeon HG, Chung Y, Lim JJ, Shin DH, Kim JM, Ki BS, Song SH, Choi SJ, Park KH, Shim SH, Moon J, Jung SJ, Kang HM, Park S, Chung HM, Ko JJ, Cha KY, Yoon TK, Kim H, Lee DR. Isolation and characterization of novel, highly proliferative human CD34/CD73-double-positive testis-derived stem cells for cell therapy. Stem Cells Dev 2013; 22:2158-73. [PMID: 23509942 DOI: 10.1089/scd.2012.0385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Human adult stem cells are a readily available multipotent cell source that can be used in regenerative medicine. Despite many advantages, including low tumorigenicity, their rapid senescence and limited plasticity have curtailed their use in cell-based therapies. In this study, we isolated CD34/CD73-double-positive (CD34(+)/CD73(+)) testicular stromal cells (HTSCs) and found that the expression of CD34 was closely related to the cells' stemness and proliferation. The CD34(+)/CD73(+) cells grew in vitro for an extended period of time, yielding a multitude of cells (5.6×10(16) cells) without forming tumors in vivo. They also differentiated into all three germ layer lineages both in vitro and in vivo, produced cartilage more efficiently compared to bone marrow stem cells and, importantly, restored erectile function in a cavernous nerve crush injury rat model. Thus, these HTSCs may represent a promising new autologous cell source for clinical use.
Collapse
Affiliation(s)
- Won Yun Choi
- Fertility Center, CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Anand M, Prasad BV. The computational analysis of human testis transcriptome reveals closer ties to pluripotency. J Hum Reprod Sci 2013; 5:266-73. [PMID: 23531778 PMCID: PMC3604834 DOI: 10.4103/0974-1208.106339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 07/25/2012] [Accepted: 07/28/2012] [Indexed: 11/04/2022] Open
Abstract
AIMS The purpose of this study was to identify the differentially expressed genes (DEG) in human testis and also evaluate the relationship between human testis, human Embryonic Stem Cells (hESC), mouse testis and mouse ESCs (mESC). SETTINGS AND DESIGN It is a prospective analysis designed computationally. METHODS AND MATERIAL The microarray data for human testis, hESCs, mouse testis and mESCs were obtained from NCBI-GEO and analyzed for identification of DEGs. The results were then compared with mouse testis and extended to ESCs. STATISTICAL ANALYSIS USED Data was analyzed in R using various Bioconductor packages. To identify DEGs, 2-fold cut-off and a False Discovery Rate (FDR) below 0.01 criterions was used. RESULTS A total 2868 transcripts (DEGs) were found to be significantly up-regulated and 2011 transcripts significantly down-regulated in human testis compared to other normal tissues. Of the up-regulated transcripts, 232 transcripts were grouped as unclassified i.e. had unknown annotations at the time of analysis. Gene Ontology (GO) based functional annotation of testis specific DEGs indicate that most of the DEGs (~80%) are involved in various metabolic processes. Pathway analysis shows over-representation of Ubiquitin-mediated proteolysis pathway. A core group of 67 transcripts were found to be common among human testis, mouse testis, hESCs and mESCs. CONCLUSIONS Testis seems to be metabolically very active relative to other normal tissues as indicated by functional annotation. The comparison of human and mouse testis shows conserved functions and pathways involved in both species. Large numbers of genes were found conserved between testis and ESCs suggesting very close expression level relationship between reproductive organs and complex phenomenon such as dedifferentiation and reprogramming.
Collapse
Affiliation(s)
- Mt Anand
- IGNOU-/²/T Center of Excellence for Advanced Education and Research, Pune, Maharashtra, India
| | | |
Collapse
|