101
|
Kubo N, Toh H, Shirane K, Shirakawa T, Kobayashi H, Sato T, Sone H, Sato Y, Tomizawa SI, Tsurusaki Y, Shibata H, Saitsu H, Suzuki Y, Matsumoto N, Suyama M, Kono T, Ohbo K, Sasaki H. DNA methylation and gene expression dynamics during spermatogonial stem cell differentiation in the early postnatal mouse testis. BMC Genomics 2015; 16:624. [PMID: 26290333 PMCID: PMC4546090 DOI: 10.1186/s12864-015-1833-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/07/2015] [Indexed: 12/18/2022] Open
Abstract
Background In the male germline, neonatal prospermatogonia give rise to spermatogonia, which include stem cell population (undifferentiated spermatogonia) that supports continuous spermatogenesis in adults. Although the levels of DNA methyltransferases change dynamically in the neonatal and early postnatal male germ cells, detailed genome-wide DNA methylation profiles of these cells during the stem cell formation and differentiation have not been reported. Results To understand the regulation of spermatogonial stem cell formation and differentiation, we examined the DNA methylation and gene expression dynamics of male mouse germ cells at the critical stages: neonatal prospermatogonia, and early postntal (day 7) undifferentiated and differentiating spermatogonia. We found large partially methylated domains similar to those found in cancer cells and placenta in all these germ cells, and high levels of non-CG methylation and 5-hydroxymethylcytosines in neonatal prospermatogonia. Although the global CG methylation levels were stable in early postnatal male germ cells, and despite the reported scarcity of differential methylation in the adult spermatogonial stem cells, we identified many regions showing stage-specific differential methylation in and around genes important for stem cell function and spermatogenesis. These regions contained binding sites for specific transcription factors including the SOX family members. Conclusions Our findings show a distinctive and dynamic regulation of DNA methylation during spermatogonial stem cell formation and differentiation in the neonatal and early postnatal testes. Furthermore, we revealed a unique accumulation and distribution of non-CG methylation and 5hmC marks in neonatal prospermatogonia. These findings contrast with the reported scarcity of differential methylation in adult spermatogonial stem cell differentiation and represent a unique phase of male germ cell development. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1833-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naoki Kubo
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.,Research Institute for Disease of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hidehiro Toh
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kenjiro Shirane
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takayuki Shirakawa
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, 236-0004, Japan
| | - Hisato Kobayashi
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Tetsuya Sato
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hidetoshi Sone
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, 236-0004, Japan
| | - Yasuyuki Sato
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, 236-0004, Japan
| | - Shin-ichi Tomizawa
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, 236-0004, Japan
| | - Yoshinori Tsurusaki
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Hiroki Shibata
- Division of Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hirotomo Saitsu
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwanoha 5-1-5, Kashiwa, Chiba, 277-8568, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Tomohiro Kono
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo, 156-8502, Japan.,Department of BioScience, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Kazuyuki Ohbo
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, 236-0004, Japan
| | - Hiroyuki Sasaki
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
102
|
Song W, Mu H, Wu J, Liao M, Zhu H, Zheng L, He X, Niu B, Zhai Y, Bai C, Lei A, Li G, Hua J. miR-544 Regulates Dairy Goat Male Germline Stem Cell Self-Renewal via Targeting PLZF. J Cell Biochem 2015; 116:2155-65. [DOI: 10.1002/jcb.25172] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
Affiliation(s)
- Wencong Song
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Hailong Mu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Jiang Wu
- College of Agriculture; Guangdong Ocean University; Zhanjiang 524088 China
| | - Mingzhi Liao
- College of Life Science; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Haijing Zhu
- College of Life Science; Yulin College, Yulin University; 719000 China
| | - Liming Zheng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Xin He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Bowen Niu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Yuanxin Zhai
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Chunling Bai
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education; Inner Mongolia University; Hohhot 010021 China
| | - Anmin Lei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 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 & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| |
Collapse
|
103
|
Linhartová Z, Saito T, Kašpar V, Rodina M, Prášková E, Hagihara S, Pšenička M. Sterilization of sterlet Acipenser ruthenus by using knockdown agent, antisense morpholino oligonucleotide, against dead end gene. Theriogenology 2015; 84:1246-1255.e1. [PMID: 26248520 DOI: 10.1016/j.theriogenology.2015.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/03/2015] [Accepted: 07/03/2015] [Indexed: 11/29/2022]
Abstract
Sturgeons (chondrostean, acipenseridae) are ancient fish species, widely known for their caviar. Nowadays, most of them are critically endangered. The sterlet (Acipenser ruthenus) is a common Eurasian sturgeon species with a small body size and the fastest reproductive cycle among sturgeons. Such species can be used as a host for surrogate production; application is of value for recovery of critically endangered and huge sturgeon species with an extremely long reproductive cycle. One prerequisite for production of the donor's gametes only is to have a sterile host. Commonly used sterilization techniques in fishes such as triploidization or hybridization do not guarantee sterility in sturgeon. Alternatively, sterilization can be achieved by using a temporary germ cell exclusion-specific gene by a knockdown agent, the antisense morpholino oligonucleotide (MO). The targeted gene for the MO is the dead end gene (dnd) which is a vertebrate-specific gene encoding a RNA-binding protein which is crucial for migration and survival of primordial germ cells (PGCs). For this purpose, a dnd homologue of Russian sturgeon (Agdnd), resulting in the same sequence in the start codon region with isolated fragments of sterlet dnd (Ardnd), was used. Reverse transcription polymerase chain reaction confirmed tissue-specific expression of Ardnd only in the gonads of both sexes. Dnd-MO for depletion of PGCs together with fluorescein isothiocyanate (FITC)-biotin-dextran for PGCs labeling was injected into the vegetal region of one- to four-cell-stage sterlet embryos. In the control groups, only FITC was injected to validate the injection method and labeling of PGCs. After optimization of MO concentration together with volume injection, 250-μM MO was applied for sterilization of sturgeon embryos. Primordial germ cells were detected under a fluorescent stereomicroscope in the genital ridge of the FITC-labeled control group only, whereas no PGCs were present in the body cavities of morphants at 21 days after fertilization. Moreover, the body cavities of MO-treated and nontreated fish were examined by histology and in situ hybridization, showing gonads which had no germ cells in morphants at various stages (60, 150, and 210 days after fertilization). Taken together, these results report the first known and functional method of sturgeon sterilization.
Collapse
Affiliation(s)
- Zuzana Linhartová
- Research Institute of Fish Culture and Hydrobiology, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic.
| | - Taiju Saito
- Research Institute of Fish Culture and Hydrobiology, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Vojtěch Kašpar
- Research Institute of Fish Culture and Hydrobiology, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Marek Rodina
- Research Institute of Fish Culture and Hydrobiology, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Eva Prášková
- Research Institute of Fish Culture and Hydrobiology, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| | - Seishi Hagihara
- Division of Marine Life Science, Graduate School of Fisheries Sciences, Hokkaido University, Hakodate, Hokkaido, Japan
| | - Martin Pšenička
- Research Institute of Fish Culture and Hydrobiology, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, Vodňany, Czech Republic
| |
Collapse
|
104
|
A Niche for GFRα1-Positive Spermatogonia in the Terminal Segments of the Seminiferous Tubules in Hamster Testes. Stem Cells 2015; 33:2811-24. [DOI: 10.1002/stem.2065] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/20/2015] [Indexed: 01/15/2023]
|
105
|
Wu J, Liao M, Zhu H, Kang K, Mu H, Song W, Niu Z, He X, Bai C, Li G, Li X, Hua J. CD49f-positive testicular cells in Saanen dairy goat were identified as spermatogonia-like cells by miRNA profiling analysis. J Cell Biochem 2015; 115:1712-23. [PMID: 24817091 DOI: 10.1002/jcb.24835] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 04/29/2014] [Accepted: 05/08/2014] [Indexed: 11/07/2022]
Abstract
miRNAs, a type of small RNA, play critical roles in mammalian spermatogenesis. Spermatogonia are the foundation of spermatogenesis and are valuable for the study of spermatogenesis. However, the expression profiling of the miRNAs in spermatogonia of dairy goats remains unclear. CD49f has been one of the surface markers used for spermatogonia enrichment by magnetic activated cell sorting (MACS). Therefore, we used a CD49f microbead antibody to purify CD49f-positive and -negative cells of dairy goat testicular cells by MACS and then analysed the miRNA expression in these cells in depth using Illumina sequencing technology. The results of miRNA expression profiling in purified CD49f-positive and -negative testicular cells showed that 933 miRNAs were upregulated in CD49f-positive cells and 916 miRNAs were upregulated in CD49f-negative cells with a twofold increase, respectively; several miRNAs and marker genes specific for spermatogonial stem cells (SSCs) in testis had a higher expression level in CD49f-positive testicular cells, including miR-221, miR-23a, miR-29b, miR-24, miR-29a, miR-199b, miR-199a, miR-27a, and miR-21 and CD90, Gfra1, and Plzf. The bioinformatics analysis of differently expressed miRNAs indicated that the target genes of these miRNAs in CD49f-positive cells were involved in cell-cycle biological processes and the cell-cycle KEGG pathway. In conclusion, our comparative miRNAome data provide useful miRNA profiling data of dairy goat spermatogonia cells and suggest that CD49f could be used to enrich dairy goat spermatogonia-like cells, including SSCs.
Collapse
Affiliation(s)
- Jiang Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Affiliation(s)
- F Kent Hamra
- Department of Pharmacology, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
107
|
Production of transgenic spermatozoa by lentiviral transduction and transplantation of porcine spermatogonial stem cells. Tissue Eng Regen Med 2014. [DOI: 10.1007/s13770-014-0078-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
108
|
Reuter K, Schlatt S, Ehmcke J, Wistuba J. Fact or fiction: In vitro spermatogenesis. SPERMATOGENESIS 2014; 2:245-252. [PMID: 23248765 PMCID: PMC3521746 DOI: 10.4161/spmg.21983] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Many previous studies have aimed at spermatogenesis of male murine germ cells in vitro, but no efficient system has been established yet that covers the entire process of mammalian spermatogenesis in a culture dish permanently. In this review, we report on the requirements of spermatogenesis and the current state of different culture methods using testicular tissue fragments, single cell suspensions or three-dimensional culture environments.
Collapse
Affiliation(s)
- Karin Reuter
- Institute of Reproductive and Regenerative Biology; Centre of Reproductive Medicine and Andrology; University of Münster; Münster, Germany
| | | | | | | |
Collapse
|
109
|
Dominguez AA, Chiang HR, Sukhwani M, Orwig KE, Reijo Pera RA. Human germ cell formation in xenotransplants of induced pluripotent stem cells carrying X chromosome aneuploidies. Sci Rep 2014; 4:6432. [PMID: 25242416 PMCID: PMC4170197 DOI: 10.1038/srep06432] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/16/2014] [Indexed: 12/21/2022] Open
Abstract
Turner syndrome is caused by complete or partial loss of the second sex chromosome and is characterized by spontaneous fetal loss in >90% of conceptions. Survivors possess an array of somatic and germline clinical characteristics. Induced pluripotent stem cells (iPSCs) offer an opportunity for insight into genetic requirements of the X chromosome linked to Turner syndrome. We derived iPSCs from Turner syndrome and control individuals and examined germ cell development as a function of X chromosome composition. We demonstrate that two X chromosomes are not necessary for reprogramming or maintenance of pluripotency and that there are minimal differences in gene expression, at the single cell level, linked to X chromosome aneuploidies. Formation of germ cells, as assessed in vivo through a murine xenotransplantation model, indicated that undifferentiated iPSCs, independent of X chromosome composition, are capable of forming germ-cell-like cells (GCLCs) in vivo. In combination with clinical data regarding infertility in women with X chromosome aneuploidies, results suggest that two intact X chromosomes are not required for human germ cell formation, qualitatively or quantitatively, but rather are likely to be required for maintenance of human germ cells to adulthood.
Collapse
Affiliation(s)
- Antonia A Dominguez
- 1] Department of Genetics; Department of Obstetrics and Gynecology; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA, USA [2]
| | - H Rosaria Chiang
- Department of Genetics; Department of Obstetrics and Gynecology; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA, USA
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Renee A Reijo Pera
- 1] Department of Genetics; Department of Obstetrics and Gynecology; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA, USA [2]
| |
Collapse
|
110
|
Yuan J, Zhang D, Wang L, Liu M, Mao J, Yin Y, Ye X, Liu N, Han J, Gao Y, Cheng T, Keefe DL, Liu L. No evidence for neo-oogenesis may link to ovarian senescence in adult monkey. Stem Cells 2014; 31:2538-50. [PMID: 23897655 DOI: 10.1002/stem.1480] [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] [Received: 01/30/2013] [Revised: 06/18/2013] [Accepted: 07/05/2013] [Indexed: 12/15/2022]
Abstract
Female germline or oogonial stem cells transiently residing in fetal ovaries are analogous to the spermatogonial stem cells or germline stem cells (GSCs) in adult testes where GSCs and meiosis continuously renew. Oocytes can be generated in vitro from embryonic stem cells and induced pluripotent stem cells, but the existence of GSCs and neo-oogenesis in adult mammalian ovaries is less clear. Preliminary findings of GSCs and neo-oogenesis in mice and humans have not been consistently reproducible. Monkeys provide the most relevant model of human ovarian biology. We searched for GSCs and neo-meiosis in ovaries of adult monkeys at various ages, and compared them with GSCs from adult monkey testis, which are characterized by cytoplasmic staining for the germ cell marker DAZL and nuclear expression of the proliferative markers PCNA and KI67, and pluripotency-associated genes LIN28 and SOX2, and lack of nuclear LAMIN A, a marker for cell differentiation. Early meiocytes undergo homologous pairing at prophase I distinguished by synaptonemal complex lateral filaments with telomere perinuclear distribution. By exhaustive searching using comprehensive experimental approaches, we show that proliferative GSCs and neo-meiocytes by these specific criteria were undetectable in adult mouse and monkey ovaries. However, we found proliferative nongermline somatic stem cells that do not express LAMIN A and germ cell markers in the adult ovaries, notably in the cortex and granulosa cells of growing follicles. These data support the paradigm that adult ovaries do not undergo germ cell renewal, which may contribute significantly to ovarian senescence that occurs with age.
Collapse
Affiliation(s)
- Jihong Yuan
- State Key Laboratory of Medicinal Chemical Biology, The 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics and College of Life Sciences, Nankai University, Tianjin, China; Key Laboratory of Ministry of Health on Hormones and Development, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Aloisio GM, Nakada Y, Saatcioglu HD, Peña CG, Baker MD, Tarnawa ED, Mukherjee J, Manjunath H, Bugde A, Sengupta AL, Amatruda JF, Cuevas I, Hamra FK, Castrillon DH. PAX7 expression defines germline stem cells in the adult testis. J Clin Invest 2014; 124:3929-44. [PMID: 25133429 PMCID: PMC4153705 DOI: 10.1172/jci75943] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/01/2014] [Indexed: 12/22/2022] Open
Abstract
Spermatogenesis is a complex, multistep process that maintains male fertility and is sustained by rare germline stem cells. Spermatogenic progression begins with spermatogonia, populations of which express distinct markers. The identity of the spermatogonial stem cell population in the undisturbed testis is controversial due to a lack of reliable and specific markers. Here we identified the transcription factor PAX7 as a specific marker of a rare subpopulation of A(single) spermatogonia in mice. PAX7+ cells were present in the testis at birth. Compared with the adult testis, PAX7+ cells constituted a much higher percentage of neonatal germ cells. Lineage tracing in healthy adult mice revealed that PAX7+ spermatogonia self-maintained and produced expanding clones that gave rise to mature spermatozoa. Interestingly, in mice subjected to chemotherapy and radiotherapy, both of which damage the vast majority of germ cells and can result in sterility, PAX7+ spermatogonia selectively survived, and their subsequent expansion contributed to the recovery of spermatogenesis. Finally, PAX7+ spermatogonia were present in the testes of a diverse set of mammals. Our data indicate that the PAX7+ subset of A(single) spermatogonia functions as robust testis stem cells that maintain fertility in normal spermatogenesis in healthy mice and mediate recovery after severe germline injury, such as occurs after cancer therapy.
Collapse
Affiliation(s)
- Gina M. Aloisio
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Yuji Nakada
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Hatice D. Saatcioglu
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher G. Peña
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Michael D. Baker
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Edward D. Tarnawa
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jishnu Mukherjee
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Hema Manjunath
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Abhijit Bugde
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Anita L. Sengupta
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - James F. Amatruda
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ileana Cuevas
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - F. Kent Hamra
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Diego H. Castrillon
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
112
|
Kim JH, Sharma N, Kim SW, Sodhi SS, Ghosh M, Kim NE, Mongre RK, Oh SJ, Jeong DK. Establishment of a pheasant (Phasianus colchicus) spermatogonial stem cell line for the production of interspecies germ line chimeras. ELECTRON J BIOTECHN 2014. [DOI: 10.1016/j.ejbt.2014.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
113
|
Ginsberg JP, Li Y, Carlson CA, Gracia CR, Hobbie WL, Miller VA, Mulhall J, Shnorhavorian M, Brinster RL, Kolon TF. Testicular tissue cryopreservation in prepubertal male children: an analysis of parental decision-making. Pediatr Blood Cancer 2014; 61:1673-8. [PMID: 24777742 PMCID: PMC4676076 DOI: 10.1002/pbc.25078] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/01/2014] [Indexed: 01/14/2023]
Abstract
BACKGROUND Infertility is an unfortunate treatment-related consequence for some pediatric malignancies as well as some non-malignant conditions treated with stem cell transplant. Unlike pubertal males, prepubertal males cannot produce semen for cryopreservation. This manuscript reports on the acceptability and safety of a multi-institutional protocol for offering testicular tissue cryopreservation to families of prepubertal male children at highest risk for infertility. Data on decision influences, decision-making control, and emotional state when considering this option are described. PROCEDURE Prepubertal males facing gonadotoxic therapy were offered testicular cryopreservation. Post-biopsy, patients were followed for acute side effects. In addition, parents and patients were asked to complete questionnaires, whether or not they chose to cryopreserve tissue. RESULTS Seventy-four prepubertal male children were approached. Fifty-seven families (77%) consented to the testicular biopsy; 48 of 57 underwent the procedure. There was one post-operative side effect. Parents who agreed to testicular cryopreservation and those that did not felt in control of this decision. Parents who consented to the biopsy and refusers were not deterred by the experimental nature of the protocol. An important decision-making influence was the risk of the biopsy. CONCLUSION Biopsy and cryopreservation of testicular tissue from prepubertal male children was performed successfully and safely at three institutions. Parents faced with this option at diagnosis can make an informed decision and weigh carefully the risks and benefits. Although asked to make a decision soon after they were given a difficult diagnosis, parents uniformly felt in control of this decision.
Collapse
Affiliation(s)
- Jill P Ginsberg
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Shi X, Li L, Ostrovidov S, Shu Y, Khademhosseini A, Wu H. Stretchable and micropatterned membrane for osteogenic differentation of stem cells. ACS APPLIED MATERIALS & INTERFACES 2014; 6:11915-23. [PMID: 24977302 DOI: 10.1021/am5029236] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Stem cells have emerged as potentially useful cells for regenerative medicine applications. To fully harness this potential, it is important to develop in vitro cell culture platforms with spatially regulated mechanical, chemical, and biological cues to induce the differentiation of stem cells. In this study, a cell culture platform was constructed that used polydopamine (PDA)-coated parafilm. The modified parafilm supports cell attachment and proliferation. In addition, because of the superb plasticity and ductility of the parafilm, it can be easily micropatterned to regulate the spatial arrangements of cells, and can exert different mechanical tensions. Specifically, we constructed a PDA-coated parafilm with grooved micropatterns to induce the osteogenic differentiation of stem cells. Adipose-derived mesenchymal stem cells that were cultured on the PDA-coated parafilm exhibited significantly higher osteogenic commitment in response to mechanical and spatial cues compared to the ones without stretch. Our findings may open new opportunities for inducing osteogenesis of stem cells in vitro using the platform that combines mechanical and spatial cues.
Collapse
Affiliation(s)
- Xuetao Shi
- WPI-Advanced Institute for Materials Research, Tohoku University , Sendai 980-8578, Japan
| | | | | | | | | | | |
Collapse
|
115
|
Chan F, Oatley MJ, Kaucher AV, Yang QE, Bieberich CJ, Shashikant CS, Oatley JM. Functional and molecular features of the Id4+ germline stem cell population in mouse testes. Genes Dev 2014; 28:1351-62. [PMID: 24939937 PMCID: PMC4066404 DOI: 10.1101/gad.240465.114] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Chan et al. generated transgenic mice in which spermatogonial stem cells expressed an Id4-Gfp transgene. Id4-Gfp+ cells exist primarily as a subset of the type Asingle pool and eventually comprise ∼2% of the undifferentiated spermatogonial population in adulthood. RNA sequencing analysis revealed genes whose expression is unique for the Id4-Gfp+/stem cell and Id4-Gfp−/progenitor fractions. These findings provide evidence that stem cells exist as a rare subset of the Asingle pool and reveal transcriptome features distinguishing stem cell and progenitor states within the mammalian male germline. The maintenance of cycling cell lineages relies on undifferentiated subpopulations consisting of stem and progenitor pools. Features that delineate these cell types are undefined for many lineages, including spermatogenesis, which is supported by an undifferentiated spermatogonial population. Here, we generated a transgenic mouse line in which spermatogonial stem cells are marked by expression of an inhibitor of differentiation 4 (Id4)-green fluorescent protein (Gfp) transgene. We found that Id4-Gfp+ cells exist primarily as a subset of the type Asingle pool, and their frequency is greatest in neonatal development and then decreases in proportion during establishment of the spermatogenic lineage, eventually comprising ∼2% of the undifferentiated spermatogonial population in adulthood. RNA sequencing analysis revealed that expression of 11 and 25 genes is unique for the Id4-Gfp+/stem cell and Id4-Gfp−/progenitor fractions, respectively. Collectively, these findings provide the first definitive evidence that stem cells exist as a rare subset of the Asingle pool and reveal transcriptome features distinguishing stem cell and progenitor states within the mammalian male germline.
Collapse
Affiliation(s)
- Frieda Chan
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| | - Melissa J Oatley
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| | - Amy V Kaucher
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| | - Qi-En Yang
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| | - Charles J Bieberich
- Department of Biological Sciences, University of Maryland at Baltimore County, Baltimore, Maryland 21250, USA
| | - Cooduvalli S Shashikant
- Department of Animal Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Jon M Oatley
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington 99164, USA
| |
Collapse
|
116
|
Spermatogonial stem cell enrichment using simple grafting of testis and in vitro cultivation. Sci Rep 2014; 4:5923. [PMID: 25080919 PMCID: PMC4118148 DOI: 10.1038/srep05923] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 07/15/2014] [Indexed: 01/15/2023] Open
Abstract
Enrichment of spermatogonial stem cells (SSCs) from the mammalian adult testis faces several limitations owing to their relatively low numbers among many types of advanced germ cells and somatic cells. The aim of the present study was to improve the isolation efficiency of SSCs using a simple tissue grafting method to eliminate the existing advanced germ cells. Sliced testis parenchyma obtained from adult ICR or EGFP-expressing transgenic mice were grafted heterotropically under the dorsal skin of nude mice. The most advanced germ cells disappeared in the grafted tissues after 2–4 weeks. Grafted tissues were dissociated enzymatically and plated in culture dishes. During in vitro culture, significantly more SSCs were obtained from the grafted testes than from non-grafted controls, and the isolated SSCs had proliferative potential and were successfully maintained. Additionally, EGFP-expressing SSCs derived from graft parenchyma were transplanted into bulsufan-treated recipient mice testes. Finally, we obtained EGFP-expressing pups after in vitro fertilization using spermatozoa derived from transplanted SSCs. These results suggest that subcutaneous grafting of testis parenchyma and the subsequent culture methods provide a simple and efficient isolation method to enrich for SSCs in adult testis without specific cell sorting methods and may be useful tools for clinical applications.
Collapse
|
117
|
Park MH, Park JE, Kim MS, Lee KY, Park HJ, Yun JI, Choi JH, Lee ES, Lee ST. Development of a high-yield technique to isolate spermatogonial stem cells from porcine testes. J Assist Reprod Genet 2014; 31:983-91. [PMID: 24938360 DOI: 10.1007/s10815-014-0271-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 06/01/2014] [Indexed: 01/15/2023] Open
Abstract
PURPOSE To date, the methods available for isolating spermatogonial stem cells (SSCs) from porcine testicular cells have a low efficiency of cell separating. Therefore, we tried to develop a novel isolation technique with a high-yield cell separating ability to isolate SSCs from porcine testes. METHODS We confirmed the presence of SSCs by measuring alkaline phosphatase (AP) activity and SSC-specific gene expression in neonatal porcine testis-derived testicular cells. Subsequently, the isolation of SSCs from testicular cells was performed using different techniques as follows: differential plating (DP), double DP, Petri dish plating post-DP, magnetic-activated cell sorting (MACS), and MACS post-DP. Positive AP staining was used to assess and compare the isolation efficiency of each method. RESULTS Petri dish plating post-DP resulted in the highest isolation efficiency. The putative SSCs isolated using this method was then further characterized by analyzing the expression of SSC-specific genes and -related proteins, and germ cell-specific genes. OCT4, NANOG, EPCAM, THY1, and UCHL1 were expressed transcriptionally, and OCT4, NANOG, SOX2, TRA-1-60, TRA-1-81, and PLZF were expressed translationally in 86 % of the isolated SSCs. In contrast, no difference was observed in the percentage of cells expressing luteinizing hormone receptor (LHR), a Leydig cell-specific protein, or GATA4, a Sertoli cell-specific protein, between SSCs and negative control cells. In addition, transcriptional expression of VASA, a primordial germ cell-specific marker, and DAZL, a premeiotic germ cell-specific marker, wasn't and was detected, respectively. CONCLUSIONS We successfully developed a novel high-yield technique to isolate SSCs from porcine testes to facilitate future porcine SSC-related research.
Collapse
Affiliation(s)
- Min Hee Park
- Department of Animal Life Science, Kangwon National University, Chuncheon, 200-701, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Valli H, Sukhwani M, Dovey SL, Peters KA, Donohue J, Castro CA, Chu T, Marshall GR, Orwig KE. Fluorescence- and magnetic-activated cell sorting strategies to isolate and enrich human spermatogonial stem cells. Fertil Steril 2014; 102:566-580.e7. [PMID: 24890267 DOI: 10.1016/j.fertnstert.2014.04.036] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/23/2014] [Accepted: 04/23/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine the molecular characteristics of human spermatogonia and optimize methods to enrich spermatogonial stem cells (SSCs). DESIGN Laboratory study using human tissues. SETTING Research institute. PATIENT(S) Healthy adult human testicular tissue. INTERVENTION(S) Human testicular tissue was fixed or digested with enzymes to produce a cell suspension. Human testis cells were fractionated by fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS). MAIN OUTCOME MEASURE(S) Immunostaining for selected markers, human-to-nude mouse xenotransplantation assay. RESULT(S) Immunohistochemistry costaining revealed the relative expression patterns of SALL4, UTF1, ZBTB16, UCHL1, and ENO2 in human undifferentiated spermatogonia as well as the extent of overlap with the differentiation marker KIT. Whole mount analyses revealed that human undifferentiated spermatogonia (UCHL1+) were typically arranged in clones of one to four cells whereas differentiated spermatogonia (KIT+) were typically arranged in clones of eight or more cells. The ratio of undifferentiated-to-differentiated spermatogonia is greater in humans than in rodents. The SSC colonizing activity was enriched in the THY1dim and ITGA6+ fractions of human testes sorted by FACS. ITGA6 was effective for sorting human SSCs by MACS; THY1 and EPCAM were not. CONCLUSION(S) Human spermatogonial differentiation correlates with increased clone size and onset of KIT expression, similar to rodents. The undifferentiated-to-differentiated developmental dynamics in human spermatogonia is different than rodents. THY1, ITGA6, and EPCAM can be used to enrich human SSC colonizing activity by FACS, but only ITGA6 is amenable to high throughput sorting by MACS.
Collapse
Affiliation(s)
- Hanna Valli
- Department of Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Meena Sukhwani
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Serena L Dovey
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Karen A Peters
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Julia Donohue
- Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Carlos A Castro
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Tianjiao Chu
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Gary R Marshall
- Department of Natural Sciences, Chatham University, Pittsburgh, Pennsylvania
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Magee-Womens Research Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
119
|
van den Driesche S, Sharpe RM, Saunders PT, Mitchell RT. Regulation of the germ stem cell niche as the foundation for adult spermatogenesis: A role for miRNAs? Semin Cell Dev Biol 2014; 29:76-83. [DOI: 10.1016/j.semcdb.2014.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/01/2014] [Indexed: 12/19/2022]
|
120
|
Majhi SK, Hattori RS, Rahman SM, Strüssmann CA. Surrogate production of eggs and sperm by intrapapillary transplantation of germ cells in cytoablated adult fish. PLoS One 2014; 9:e95294. [PMID: 24748387 PMCID: PMC3991631 DOI: 10.1371/journal.pone.0095294] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 03/25/2014] [Indexed: 01/15/2023] Open
Abstract
Germ cell transplantation (GCT) is a promising assisted reproductive technology for the conservation and propagation of endangered and valuable genetic resources. In teleost fish, GCT in adult gonads has been achieved only in male recipients, limiting greatly the usefulness of this technique in situations where both sexes need equal and timely attention for conservation and/or propagation. Here we describe a simplified GCT approach that ultimately leads to production of donor-derived eggs and sperm in considerably short time. Donor germ cells isolated from young pejerrey Odontesthes bonariensis (Atherinopsidae) were transplanted non-surgically through the genital papilla into the sexually mature gonads of Patagonian pejerrey O. hatcheri recipients whose gonads have been depleted of endogenous GCs by heat (26°C) and chemical treatment (four doses of Busulfan at 30 mg/kg and 40 mg/kg for females and males, respectively). Transplanted spermatogonial and oogonial cells were able to recolonize the recipients' gonads and produce functional donor origin eggs and sperm within 7 months from the GCT. We confirmed the presence of donor-derived gametes by PCR in 17% and 5% of the surrogate O. hatcheri fathers and mothers, respectively. The crosses between surrogate fathers and O. bonariensis mothers yielded 12.6-39.7% pure O. bonariensis and that between a surrogate mother and an O. bonariensis father yielded 52.2% pure O. bonariensis offspring. Our findings confirm that transplantation of germ cells into sexually competent adult fish by non-surgical methods allows the production of functional donor-derived eggs and sperm in a considerably short time. The methods described here could play a vital role in conservation and rapid propagation of endangered fish genetic resources.
Collapse
Affiliation(s)
- Sullip Kumar Majhi
- Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Minato, Tokyo, Japan
- Division of Molecular Biology & Biotechnology, National Bureau of Fish Genetic Resources, Dilkhusa, Lucknow, India
- * E-mail:
| | - Ricardo Shohei Hattori
- Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Minato, Tokyo, Japan
| | - Sheikh Mustafizur Rahman
- Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Minato, Tokyo, Japan
| | - Carlos Augusto Strüssmann
- Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Minato, Tokyo, Japan
| |
Collapse
|
121
|
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
|
122
|
Zheng Y, Zhang Y, Qu R, He Y, Tian X, Zeng W. Spermatogonial stem cells from domestic animals: progress and prospects. Reproduction 2014; 147:R65-74. [PMID: 24357661 DOI: 10.1530/rep-13-0466] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Spermatogenesis, an elaborate and male-specific process in adult testes by which a number of spermatozoa are produced constantly for male fertility, relies on spermatogonial stem cells (SSCs). As a sub-population of undifferentiated spermatogonia, SSCs are capable of both self-renewal (to maintain sufficient quantities) and differentiation into mature spermatozoa. SSCs are able to convert to pluripotent stem cells during in vitro culture, thus they could function as substitutes for human embryonic stem cells without ethical issues. In addition, this process does not require exogenous transcription factors necessary to produce induced-pluripotent stem cells from somatic cells. Moreover, combining genetic engineering with germ cell transplantation would greatly facilitate the generation of transgenic animals. Since germ cell transplantation into infertile recipient testes was first established in 1994, in vivo and in vitro study and manipulation of SSCs in rodent testes have been progressing at a staggering rate. By contrast, their counterparts in domestic animals, despite the failure to reach a comparable level, still burgeoned and showed striking advances. This review outlines the recent progressions of characterization, isolation, in vitro propagation, and transplantation of spermatogonia/SSCs from domestic animals, thereby shedding light on future exploration of these cells with high value, as well as contributing to the development of reproductive technology for large animals.
Collapse
Affiliation(s)
- Yi Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | | | | | | | | |
Collapse
|
123
|
Yeh JR, Nagano MC. Spermatogonial stem cell biomarkers: improved outcomes of spermatogonial transplantation in male fertility restoration? Expert Rev Mol Diagn 2014; 9:109-14. [DOI: 10.1586/14737159.9.2.109] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
124
|
Moreira PN, Montoliu L. Intracytoplasmic sperm injection (ICSI)-mediated transgenesis in mice. Methods Mol Biol 2014; 1194:141-156. [PMID: 25064101 DOI: 10.1007/978-1-4939-1215-5_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Over the years many well-described techniques for the introduction of transgene DNA into host organisms have been used, including pronuclear injection, in vitro fertilization-mediated transgenesis, transfection of ES and spermatogenic cells, nuclear transfer of somatic cell nuclei, and lentiviral vectors. The application of these techniques has been limited however either by the time and effort to be executed or by their narrow efficiency with large transgenes. The greatest advantage of intracytoplasmic sperm injection (ICSI)-mediated transgenesis is precisely its ability to stably introduce large DNA molecules into the genome of host organisms with relatively high efficiency, as compared to alternative procedures. In mice, this procedure has been shown to be a reproducible method to generate transgenic offspring with a high efficiency. Recently, it proved also to be a viable method to generate transgenic rats and pigs, and as such, it is foreseen with great interest for the production of transgenic farm animals, where it would constitute an important tool for the production of recombinant proteins and livestock improvement.
Collapse
Affiliation(s)
- Pedro N Moreira
- Mouse Biology Unit, EMBL Monterotondo, Adriano Buzzati-Traverso Campus, Via Ramarini 32, 00015, Monterotondo, Italy,
| | | |
Collapse
|
125
|
Usmani A, Ganguli N, Sarkar H, Dhup S, Batta SR, Vimal M, Ganguli N, Basu S, Nagarajan P, Majumdar SS. A non-surgical approach for male germ cell mediated gene transmission through transgenesis. Sci Rep 2013; 3:3430. [PMID: 24305437 PMCID: PMC3852150 DOI: 10.1038/srep03430] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 11/20/2013] [Indexed: 01/15/2023] Open
Abstract
Microinjection of foreign DNA in male pronucleus by in-vitro embryo manipulation is difficult but remains the method of choice for generating transgenic animals. Other procedures, including retroviral and embryonic stem cell mediated transgenesis are equally complicated and have limitations. Although our previously reported technique of testicular transgenesis circumvented several limitations, it involved many steps, including surgery and hemicastration, which carried risk of infection and impotency. We improved this technique further, into a two step non-surgical electroporation procedure, for making transgenic mice. In this approach, transgene was delivered inside both testes by injection and modified parameters of electroporation were used for in-vivo gene integration in germ cells. Using variety of constructs, germ cell integration of the gene and its transmission in progeny was confirmed by PCR, slot blot and immunohistochemical analysis. This improved technique is efficient, requires substantially less time and can be easily adopted by various biomedical researchers.
Collapse
Affiliation(s)
- Abul Usmani
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| | - Nirmalya Ganguli
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| | - Hironmoy Sarkar
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| | - Suveera Dhup
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| | | | - Manoj Vimal
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| | - Nilanjana Ganguli
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| | - Sayon Basu
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| | - P. Nagarajan
- Small Animal Facility, National Institute of Immunology, New Delhi, India
| | - Subeer S. Majumdar
- Embryo Biotechnology Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
126
|
Chandrashekran A, Sarkar R, Thrasher A, Fraser SE, Dibb N, Casimir C, Winston R, Readhead C. Efficient generation of transgenic mice by lentivirus‐mediated modification of spermatozoa. FASEB J 2013; 28:569-76. [DOI: 10.1096/fj.13-233999] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Anil Chandrashekran
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Rupa Sarkar
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Adrian Thrasher
- Molecular Immunology UnitUniversity College London Institute of Child HealthLondonUK
| | - Scott E. Fraser
- Biological Imaging CenterBeckman InstituteCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Nicholas Dibb
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Colin Casimir
- Department of Natural SciencesSchool of Science and TechnologyMiddlesex UniversityLondonUK
| | - Robert Winston
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Carol Readhead
- Biological Imaging CenterBeckman InstituteCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| |
Collapse
|
127
|
Eslahi N, Hadjighassem MR, Joghataei MT, Mirzapour T, Bakhtiyari M, Shakeri M, Pirhajati V, Shirinbayan P, Koruji M. The effects of poly L-lactic acid nanofiber scaffold on mouse spermatogonial stem cell culture. Int J Nanomedicine 2013; 8:4563-76. [PMID: 24348035 PMCID: PMC3848747 DOI: 10.2147/ijn.s45535] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION A 3D-nanofiber scaffold acts in a similar way to the extracellular matrix (ECM)/basement membrane that enhances the proliferation and self-renewal of stem cells. The goal of the present study was to investigate the effects of a poly L-lactic acid (PLLA) nanofiber scaffold on frozen-thawed neonate mouse spermatogonial stem cells (SSCs) and testis tissues. METHODS The isolated spermatogonial cells were divided into six culture groups: (1) fresh spermatogonial cells, (2) fresh spermatogonial cells seeded onto PLLA, (3) frozen-thawed spermatogonial cells, (4) frozen-thawed spermatogonial cells seeded onto PLLA, (5) spermatogonial cells obtained from frozen-thawed testis tissue, and (6) spermatogonial cells obtained from frozen-thawed testis tissue seeded onto PLLA. Spermatogonial cells and testis fragments were cryopreserved and cultured for 3 weeks. Cluster assay was performed during the culture. The presence of spermatogonial cells in the culture was determined by a reverse transcriptase polymerase chain reaction for spermatogonial markers (Oct4, GFRα-1, PLZF, Mvh(VASA), Itgα6, and Itgβ1), as well as the ultrastructural study of cell clusters and SSCs transplantation to a recipient azoospermic mouse. The significance of the data was analyzed using the repeated measures and analysis of variance. RESULTS The findings indicated that the spermatogonial cells seeded on PLLA significantly increased in vitro spermatogonial cell cluster formations in comparison with the control groups (culture of SSCs not seeded on PLLA) (P≤0.001). The viability rate for the frozen cells after thawing was 63.00% ± 3.56%. This number decreased significantly (40.00% ± 0.82%) in spermatogonial cells obtained from the frozen-thawed testis tissue. Both groups, however, showed in vitro cluster formation. Although the expression of spermatogonial markers was maintained after 3 weeks of culture, there was a significant downregulation for some spermatogonial genes in the experimental groups compared with those of the control groups. Furthermore, transplantation assay and transmission electron microscopy studies suggested the presence of SSCs among the cultured cells. CONCLUSION Although PLLA can increase the in vitro cluster formation of neonate fresh and frozen-thawed spermatogonial cells, it may also cause them to differentiate during cultivation. The study therefore has implications for SSCs proliferation and germ cell differentiation in vitro.
Collapse
Affiliation(s)
- Neda Eslahi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran ; Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Hadjighassem
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran ; Department of Neurosciences, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran ; Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tooba Mirzapour
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mehrdad Bakhtiyari
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran ; Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Malak Shakeri
- Department of Animal Science, Agricultural Campus, University of Tehran, Tehran, Iran
| | - Vahid Pirhajati
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran ; Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Peymaneh Shirinbayan
- Pediatric Neuro-Rehabilitation Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Morteza Koruji
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran ; Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
128
|
Li X, Mao Z, Wu M, Xia J. Rescuing infertility of Pick1 knockout mice by generating testis-specific transgenic mice via testicular infection. Sci Rep 2013; 3:2842. [PMID: 24100262 PMCID: PMC3792414 DOI: 10.1038/srep02842] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 09/02/2013] [Indexed: 12/16/2022] Open
Abstract
PICK1 (protein interacting with C-kinase 1) is a peripheral membrane protein with high expression in brain, testis, pancreas and other neuroendocrine tissues. Male Pick1 knockout mice are completely infertile, with a phenotype resembling the human disease globozoospermia. Since PICK1 is expressed in both testis and neuroendocrine tissues, infertility of Pick1 knockout mice may be due to either impaired neuroendocrine function or abnormal spermatogenesis. To distinguish these two possibilities, we restored PICK1's expression in the testis by seminiferous tubule microinjection of PICK1-containing lentivirus. By examining the testis-specific Pick1 transgenic mice, we found that PICK1's expression in testis rescued the spermatogenic abnormalities and male infertility in Pick1 knockout mice. Our results indicate that the infertility is caused by the lack of PICK1 in the testis rather than in other organs. In addition, we found that seminiferous tubule microinjection of lentivirus has a strong preference to produce testis-specific transgenic mice.
Collapse
Affiliation(s)
- Xiumao Li
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zhuo Mao
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Min Wu
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jun Xia
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
129
|
Shirakawa T, Yaman-Deveci R, Tomizawa SI, Kamizato Y, Nakajima K, Sone H, Sato Y, Sharif J, Yamashita A, Takada-Horisawa Y, Yoshida S, Ura K, Muto M, Koseki H, Suda T, Ohbo K. An epigenetic switch is crucial for spermatogonia to exit the undifferentiated state toward a Kit-positive identity. Development 2013; 140:3565-76. [PMID: 23903187 DOI: 10.1242/dev.094045] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epigenetic modifications influence gene expression and chromatin remodeling. In embryonic pluripotent stem cells, these epigenetic modifications have been extensively characterized; by contrast, the epigenetic events of tissue-specific stem cells are poorly understood. Here, we define a new epigenetic shift that is crucial for differentiation of murine spermatogonia toward meiosis. We have exploited a property of incomplete cytokinesis, which causes male germ cells to form aligned chains of characteristic lengths, as they divide and differentiate. These chains revealed the stage of spermatogenesis, so the epigenetic differences of various stages could be characterized. Single, paired and medium chain-length spermatogonia not expressing Kit (a marker of differentiating spermatogonia) showed no expression of Dnmt3a2 and Dnmt3b (two de novo DNA methyltransferases); they also lacked the transcriptionally repressive histone modification H3K9me2. By contrast, spermatogonia consisting of ~8-16 chained cells with Kit expression dramatically upregulated Dnmt3a2/3b expression and also displayed increased H3K9me2 modification. To explore the function of these epigenetic changes in spermatogonia in vivo, the DNA methylation machinery was destabilized by ectopic Dnmt3b expression or Np95 ablation. Forced Dnmt3b expression induced expression of Kit; whereas ablation of Np95, which is essential for maintaining DNA methylation, interfered with differentiation and viability only after spermatogonia become Kit positive. These data suggest that the epigenetic status of spermatogonia shifts dramatically during the Kit-negative to Kit-positive transition. This shift might serve as a switch that determines whether spermatogonia self-renew or differentiate.
Collapse
Affiliation(s)
- Takayuki Shirakawa
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Mahmoud H. Concise review: Spermatogenesis in an artificial three-dimensional system. Stem Cells 2013; 30:2355-60. [PMID: 22997006 DOI: 10.1002/stem.1238] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Culture of spermatogonial stem cells has been performed under a variety of conditions. Most featured two-dimensional systems, with different types of sera, conditioned media, feeder layers, and growth factors. Some have used three-dimensional (3D) matrices produced from gelatin, collagen, or other material. In spite of their increasingly sophisticated composition, however, complete spermatogenesis in vitro has not yet been achieved. In the seminiferous tubules, spermatogenesis occurs in an environment where cells are embedded in a 3D structure with specific niches regulating each stage of germ cell maturation mediated by hormones and paracrine/autocrine factors. We have recently reported achievement of complete in vitro spermatogenesis of mouse testicular germ cells in a 3D culture system featuring a soft agar matrix. This review discusses the advantages of the 3D culture system for studying the spermatogenic process in its entirety. Also discussed are the steps necessary to expand the applicability of the 3D culture system to human germ cell development and determine the functionality of culture-produced spermatozoa for generating offspring.
Collapse
Affiliation(s)
- Huleihel Mahmoud
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
131
|
Miao X. Recent advances in the development of new transgenic animal technology. Cell Mol Life Sci 2013; 70:815-28. [PMID: 22833168 PMCID: PMC11113483 DOI: 10.1007/s00018-012-1081-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/30/2012] [Accepted: 07/03/2012] [Indexed: 12/14/2022]
Abstract
Transgenic animal technology is one of the fastest growing biotechnology areas. It is used to integrate exogenous genes into the animal genome by genetic engineering technology so that these genes can be inherited and expressed by offspring. The transgenic efficiency and precise control of gene expression are the key limiting factors in the production of transgenic animals. A variety of transgenic technologies are available. Each has its own advantages and disadvantages and needs further study because of unresolved technical and safety issues. Further studies will allow transgenic technology to explore gene function, animal genetic improvement, bioreactors, animal disease models, and organ transplantation. This article reviews the recently developed animal transgenic technologies, including the germ line stem cell-mediated method to improve efficiency, gene targeting to improve accuracy, RNA interference-mediated gene silencing technology, zinc-finger nuclease gene targeting technology and induced pluripotent stem cell technology. These new transgenic techniques can provide a better platform to develop transgenic animals for breeding new animal varieties and promote the development of medical sciences, livestock production, and other fields.
Collapse
Affiliation(s)
- Xiangyang Miao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
132
|
Hidden gems in the niche: a new approach to the study of spermatogonial stem cells. Asian J Androl 2013; 15:214-5. [PMID: 23314657 DOI: 10.1038/aja.2012.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
133
|
|
134
|
Honke K. Biosynthesis and biological function of sulfoglycolipids. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2013; 89:129-138. [PMID: 23574804 PMCID: PMC3669731 DOI: 10.2183/pjab.89.129] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 02/19/2013] [Indexed: 06/02/2023]
Abstract
Sulfation confers negative charge on glycolipids and the attached sulfate group presents a part of determinants for the molecular interactions. Mammalian sulfoglycolipids are comprised of two major members, sulfatide (SO3-3Gal-ceramide) and seminolipid (SO3-3Gal-alkylacylglycerol). Sulfatide is abundant in the myelin sheath and seminolipid is unique to the spermatogenic cells. The carbohydrate moiety of sulfatide and seminolipid is biosynthesized via sequential reactions catalyzed by common enzymes: ceramide galactosyltransferase (CGT) and cerebroside sulfotransferase (CST). To elucidate the biological function of sulfoglycolipids, we have purified CST, cloned the CST gene, and generated CST-knockout mice. CST-null mice completely lack sulfoglycolipids all over the body. CST-null mice manifest some neurological disorders due to myelin dysfunction, an aberrant enhancement of oligodendrocyte terminal differentiation, and an arrest of spermatogenesis. CST-deficiency ameliorates L-selectin-dependent monocyte infiltration in the renal interstitial inflammation, indicating that sulfatide is an endogenous ligand of L-selectin. Studies on the molecular mechanisms underlying the biological events for which sulfoglycolipids are essential are ongoing
Collapse
Affiliation(s)
- Koichi Honke
- Department of Biochemistry and Kochi System Glycobiology Center, Kochi University Medical School, Kochi, Japan.
| |
Collapse
|
135
|
Hermann BP, Sukhwani M, Winkler F, Pascarella JN, Peters KA, Sheng Y, Valli H, Rodriguez M, Ezzelarab M, Dargo G, Peterson K, Masterson K, Ramsey C, Ward T, Lienesch M, Volk A, Cooper DK, Thomson AW, Kiss JE, Penedo MCT, Schatten GP, Mitalipov S, Orwig KE. Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm. Cell Stem Cell 2012; 11:715-26. [PMID: 23122294 PMCID: PMC3580057 DOI: 10.1016/j.stem.2012.07.017] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 06/16/2012] [Accepted: 07/12/2012] [Indexed: 01/12/2023]
Abstract
Spermatogonial stem cells (SSCs) maintain spermatogenesis throughout a man's life and may have application for treating some cases of male infertility, including those caused by chemotherapy before puberty. We performed autologous and allogeneic SSC transplantations into the testes of 18 adult and 5 prepubertal recipient macaques that were rendered infertile with alkylating chemotherapy. After autologous transplant, the donor genotype from lentivirus-marked SSCs was evident in the ejaculated sperm of 9/12 adult and 3/5 prepubertal recipients after they reached maturity. Allogeneic transplant led to donor-recipient chimerism in sperm from 2/6 adult recipients. Ejaculated sperm from one recipient transplanted with allogeneic donor SSCs were injected into 85 rhesus oocytes via intracytoplasmic sperm injection. Eighty-one oocytes were fertilized, producing embryos ranging from four-cell to blastocyst with donor paternal origin confirmed in 7/81 embryos. This demonstration of functional donor spermatogenesis following SSC transplantation in primates is an important milestone for informed clinical translation.
Collapse
Affiliation(s)
- Brian P. Hermann
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Center for Research in Reproductive Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Magee-Womens Research Institute, Pittsburgh, PA 15213
| | | | | | | | | | - Yi Sheng
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Magee-Womens Research Institute, Pittsburgh, PA 15213
| | - Hanna Valli
- Molecular Genetics and Developmental Biology Graduate Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Magee-Womens Research Institute, Pittsburgh, PA 15213
| | | | - Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
| | - Gina Dargo
- Hemapheresis Department, ITxM Diagnostics Pittsburgh, PA 15213
| | - Kim Peterson
- Hemapheresis Department, ITxM Diagnostics Pittsburgh, PA 15213
| | - Keith Masterson
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Cathy Ramsey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Thea Ward
- Veterinary Genetics Laboratory, University of California Davis, CA 95616
| | | | | | - David K. Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
| | - Angus W. Thomson
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
| | - Joseph E. Kiss
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Hemapheresis Department, ITxM Diagnostics Pittsburgh, PA 15213
| | | | - Gerald P. Schatten
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Magee-Womens Research Institute, Pittsburgh, PA 15213
| | - Shoukhrat Mitalipov
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
- Oregon Stem Cell Center , Oregon Health & Science University, Beaverton, OR 97006
- Departments of Obstetrics & Gynecology and Molecular & Medical Genetics, Oregon Health & Science University, Beaverton, OR 97006
| | - Kyle E. Orwig
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Center for Research in Reproductive Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260
- Magee-Womens Research Institute, Pittsburgh, PA 15213
| |
Collapse
|
136
|
Dobrinski I. De novo morphogenesis of functional testis tissue after ectopic transplantation of isolated cells. Organogenesis 2012; 3:79-82. [PMID: 19279705 DOI: 10.4161/org.3.2.4944] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2007] [Accepted: 08/28/2007] [Indexed: 01/15/2023] Open
Abstract
Development of the mammalian testis begins with sex specific differentiation of the bipotential gonad during fetal development, continues after birth with proliferation and differentiation of testicular somatic cells, and culminates at puberty with germ cell differentiation, meiotic divisions and production of sperm that continues throughout the adult life of the male. Recently, it was demonstrated that functional testicular tissue formed de novo when cells isolated from neonatal porcine or rodent testes were grafted ectopically to mouse hosts. The spermatogenic and interstitial compartments of the testis were regenerated form transplanted cells in a cell autonomous fashion and supported the production of functional haploid germ cells. This fascinating ability of testis cells to recreate the necessary structural and cellular associations to support tissue maturation and germ cell differentiation can now be harnessed to study aspects of mammalian spermatogenesis and testicular morphogenesis in an accessible in vivo system.
Collapse
|
137
|
Barakat B, Itman C, Mendis SH, Loveland KL. Activins and inhibins in mammalian testis development: new models, new insights. Mol Cell Endocrinol 2012; 359:66-77. [PMID: 22406273 DOI: 10.1016/j.mce.2012.02.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 02/20/2012] [Accepted: 02/21/2012] [Indexed: 01/15/2023]
Abstract
The discovery of activin and inhibins as modulators of the hypothalamic-pituitary-gonadal axis has set the foundation for understanding their central importance to many facets of development and disease. This review contains an overview of the processes and cell types that are central to testis development and spermatogenesis and then provides an update focussed on information gathered over the past five years to address new concepts about how these proteins function to control testis development in fetal and juvenile life. Current knowledge about the interactive nature of the transforming growth factor-β (TGFβ) superfamily signalling network is applied to recent findings about activins and inhibins in the testis. Information about the regulated synthesis of signalling components and signalling regulators in the testis is integrated with new concepts that demonstrate their functional significance. The importance of activin bioactivity levels or dosage in controlling balanced growth of spermatogonial cells and their niche at different stages of testis development is highlighted.
Collapse
Affiliation(s)
- B Barakat
- Monash Institute of Reproduction and Development, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
138
|
Abstract
This review addresses current understanding of the germline stem cell niche unit in mammalian testes. Spermatogenesis is a classic model of tissue-specific stem cell function relying on self-renewal and differentiation of spermatogonial stem cells (SSCs). These fate decisions are influenced by a niche microenvironment composed of a growth factor milieu that is provided by several testis somatic support cell populations. Investigations over the last two decades have identified key determinants of the SSC niche including cytokines that regulate SSC functions and support cells providing these factors, adhesion molecules that influence SSC homing, and developmental heterogeneity of the niche during postnatal aging. Emerging evidence suggests that Sertoli cells are a key support cell population influencing the formation and function of niches by secreting soluble factors and possibly orchestrating contributions of other support cells. Investigations with mice have shown that niche influence on SSC proliferation differs during early postnatal development and adulthood. Moreover, there is mounting evidence of an age-related decline in niche function, which is likely influenced by systemic factors. Defining the attributes of stem cell niches is key to developing methods to utilize these cells for regenerative medicine. The SSC population and associated niche comprise a valuable model system for study that provides fundamental knowledge about the biology of tissue-specific stem cells and their capacity to sustain homeostasis of regenerating tissue lineages. While the stem cell is essential for maintenance of all self-renewing tissues and has received considerable attention, the role of niche cells is at least as important and may prove to be more receptive to modification in regenerative medicine.
Collapse
Affiliation(s)
- Jon M Oatley
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| | | |
Collapse
|
139
|
Nagai R, Shinomura M, Kishi K, Aiyama Y, Harikae K, Sato T, Kanai-Azuma M, Kurohmaru M, Tsunekawa N, Kanai Y. Dynamics of GFRα1-positive spermatogonia at the early stages of colonization in the recipient testes of W/Wν male mice. Dev Dyn 2012; 241:1374-84. [PMID: 22745058 DOI: 10.1002/dvdy.23824] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The spermatogonial transplantation experiment can be used as an unequivocal detection assay of spermatogenic stem cells (SSCs) in both a qualitative and quantitative manner, based on their regenerative capacity. In this study, the proliferative patterns and kinetics of donor-derived GFRα1-positive spermatogonia containing potential SSCs were examined during early colonization following spermatogonial transplantation. RESULTS Donor-derived GFRα1-positive cells frequently formed several aggregates of A(al(aligned)) /morula-like structures in a single spermatogenic cell patch before and on day 14 post-transplant, indicating a possible involvement in the formation of a stable spermatogenic colony at 21 days post-transplant. The appearance of these A(al) /morula-like aggregates is positively correlated with regional, high-level expression of immunoreactive GDNF signals, a ligand for GFRα1, associated with colony expansion. CONCLUSIONS These data raise the hypothesis that regional GDNF signals regulate the balance between donor-derived A(al) -like cell aggregates and their differentiation in each small patch, which subsequently leads to further selection of survival colonies at later stages.
Collapse
Affiliation(s)
- Ryohei Nagai
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Koruji M, Shahverdi A, Janan A, Piryaei A, Lakpour MR, Gilani Sedighi MA. Proliferation of small number of human spermatogonial stem cells obtained from azoospermic patients. J Assist Reprod Genet 2012; 29:957-67. [PMID: 22735929 DOI: 10.1007/s10815-012-9817-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 05/29/2012] [Indexed: 12/18/2022] Open
Abstract
PURPOSE This study aims to proliferate spermatogonial stem cells (SSCs) and compare the in-vitro effects of laminin and growth factors on the proliferation of adult human SSC. METHODS Isolated testicular cells were cultured in DMEM supplemented with 5 % fetal calf serum (FCS). During the culture, enriched spermatogonial cells were treated with a combination of glial cell line-derived neurotrophic factor (GDNF), basic fibroblast growth factor (bFGF), epidermal growth factor (EGF) and mouse leukemia inhibitory factor (LIF) in the presence or absence of human placental laminin-coated dishes. Cluster assay was performed during culture. Presence of spermatogonia was determined by an ultrastructural study of the cell clusters, reverse transcription polymerase chain reaction (RT-PCR) for spermatogonial markers and xenotransplantation to the testes of busulfan-treated recipient mice. Statistical significance between mean values was determined using statistical ANOVA tests. RESULTS The findings indicated that the addition of GDNF, bFGF, EGF and LIF on laminin-coated dishes significantly increased in-vitro spermatogonial cell cluster formation in comparison with the control group (p ≤ 0.001). The expression of spermatogonial markers was maintained throughout the culture period. Furthermore, a transplantation experiment showed the presence of SSC among the cultured cells. In addition, a transmission electron microscopy (TEM) study suggested the presence of spermatogonial cells of typical morphology among the cluster cells. CONCLUSIONS It can be concluded that human SSCs obtained from non-obstructive azoospermic (NOA) patients had the ability to self-renew in the culture system. This system can be used for the propagation of a small number of these cells from small biopsies.
Collapse
Affiliation(s)
- Morteza Koruji
- Cellular and Molecular Research Center and Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Hemmat Highway, Tehran, Iran.
| | | | | | | | | | | |
Collapse
|
141
|
Li N, Wang T, Han D. Structural, cellular and molecular aspects of immune privilege in the testis. Front Immunol 2012; 3:152. [PMID: 22701457 PMCID: PMC3371599 DOI: 10.3389/fimmu.2012.00152] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 05/23/2012] [Indexed: 11/17/2022] Open
Abstract
The testis presents a special immunological environment, considering its property of immune privilege that tolerates allo- and auto-antigens. Testicular immune privilege was once believed to be mainly based on the sequestration of antigens from the immune system by the blood–testis barrier in the seminiferous epithelium. Substantial evidence supports the view that the combination of physical structure, testicular cells, and cytokines controls immune responses in the testis to preserve the structural and functional integrity of testicular immune privilege. Both systemic immune tolerance and local immunosuppression help maintain the immune privilege status. Constitutive expression of anti-inflammatory factors in testicular cells is critical for local immunosuppression. However, the testis locally generates an efficient innate immune system against pathogens. Disruption of these mechanisms may lead to orchitis and impair fertility. This review article highlights the current understanding of structural, cellular, and molecular mechanisms underlying the unique immune environment of the testis, particularly its immune privilege status.
Collapse
Affiliation(s)
- Nan Li
- Department of Cell Biology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | | | | |
Collapse
|
142
|
Chen CH, Wang CW, Hsu MI, Huang YH, Lai WFT, Tzeng CR. Bioluminescence imaging as a tool to evaluate germ cells in vitro and transplantation in vivo as fertility preservation of prepubertal male mice. Fertil Steril 2012; 97:1192-8. [PMID: 22424616 DOI: 10.1016/j.fertnstert.2012.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 01/31/2012] [Accepted: 02/02/2012] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To determine the feasibility of bioluminescence imaging (BLI) to evaluate the efficiency of germ cell transplantation in vitro and in vivo in fertility preservation for infertile male mice. DESIGN Transgenic mouse model. SETTING University-based teaching hospital. ANIMAL(S) Transgenic mice. INTERVENTION(S) Busulfan was used to induce testicular failure in 3-week-old immature FVB/NJNarl wild-type recipient mice. At 8 weeks of age they received hemizygotic germinal cells from 3-week-old immature male FVB/N-Tg (PolII-luc) Ltc strain transgenic donor mice, transplanted into the seminiferous tubules. MAIN OUTCOME MEASURE(S) Isolated germinal cells were suspended in multiwell plates with the bioluminescent substrate d-luciferin in excess to quantify viable germ cells in vitro. Quantitatively in vivo BLI was applied to demonstrate the efficiency and success of transplantation and BLI of live pups born from wild-type in vivo. RESULT(S) Live birth pup of FVB/N-Tg (PolII-luc) Ltc transgenic mouse were born and imaged by bioluminescence after mating FVB/NJNarl female wild-type and male wild-type infertile recipient 4-6 months after transplantation of germinal cells of FVB/N-Tg (PolII-luc). CONCLUSION(S) The BLI could be applied successfully to this transgenic small animal model. It proved a useful tool for quantifying germ cells in vitro and for assessing the efficacy of germ cell transplantation in vivo.
Collapse
Affiliation(s)
- Chi-Huang Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
143
|
Sato T, Aiyama Y, Ishii-Inagaki M, Hara K, Tsunekawa N, Harikae K, Uemura-Kamata M, Shinomura M, Zhu XB, Maeda S, Kuwahara-Otani S, Kudo A, Kawakami H, Kanai-Azuma M, Fujiwara M, Miyamae Y, Yoshida S, Seki M, Kurohmaru M, Kanai Y. Cyclical and patch-like GDNF distribution along the basal surface of Sertoli cells in mouse and hamster testes. PLoS One 2011; 6:e28367. [PMID: 22174794 PMCID: PMC3235125 DOI: 10.1371/journal.pone.0028367] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Accepted: 11/07/2011] [Indexed: 12/22/2022] Open
Abstract
Background and Aims In mammalian spermatogenesis, glial cell line-derived neurotrophic factor (GDNF) is one of the major Sertoli cell-derived factors which regulates the maintenance of undifferentiated spermatogonia including spermatogonial stem cells (SSCs) through GDNF family receptor α1 (GFRα1). It remains unclear as to when, where and how GDNF molecules are produced and exposed to the GFRα1-positive spermatogonia in vivo. Methodology and Principal Findings Here we show the cyclical and patch-like distribution of immunoreactive GDNF-positive signals and their close co-localization with a subpopulation of GFRα1-positive spermatogonia along the basal surface of Sertoli cells in mice and hamsters. Anti-GDNF section immunostaining revealed that GDNF-positive signals are mainly cytoplasmic and observed specifically in the Sertoli cells in a species-specific as well as a seminiferous cycle- and spermatogenic activity-dependent manner. In contrast to the ubiquitous GDNF signals in mouse testes, high levels of its signals were cyclically observed in hamster testes prior to spermiation. Whole-mount anti-GDNF staining of the seminiferous tubules successfully visualized the cyclical and patch-like extracellular distribution of GDNF-positive granular deposits along the basal surface of Sertoli cells in both species. Double-staining of GDNF and GFRα1 demonstrated the close co-localization of GDNF deposits and a subpopulation of GFRα1-positive spermatogonia. In both species, GFRα1-positive cells showed a slender bipolar shape as well as a tendency for increased cell numbers in the GDNF-enriched area, as compared with those in the GDNF-low/negative area of the seminiferous tubules. Conclusion/Significance Our data provide direct evidence of regionally defined patch-like GDNF-positive signal site in which GFRα1-positive spermatogonia possibly interact with GDNF in the basal compartment of the seminiferous tubules.
Collapse
Affiliation(s)
- Takeshi Sato
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
- Drug Safety Research Labs, Astellas Pharma Inc., Osaka, Japan
| | - Yoshimi Aiyama
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | | | - Kenshiro Hara
- Division of Germ Cell Biology, National Institute for Basic Biology and Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Naoki Tsunekawa
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | - Kyoko Harikae
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | - Mami Uemura-Kamata
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
- Center for Experimental Animal, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mai Shinomura
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | - Xiao Bo Zhu
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
| | - Seishi Maeda
- Division of Cell Biology, Department of Anatomy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Sachi Kuwahara-Otani
- Division of Cell Biology, Department of Anatomy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Akihiko Kudo
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Japan
| | - Hayato Kawakami
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Japan
| | - Masami Kanai-Azuma
- Center for Experimental Animal, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michio Fujiwara
- Drug Safety Research Labs, Astellas Pharma Inc., Osaka, Japan
| | - Yoichi Miyamae
- Drug Safety Research Labs, Astellas Pharma Inc., Osaka, Japan
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology and Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Makoto Seki
- Division of Cell Biology, Department of Anatomy, Hyogo College of Medicine, Nishinomiya, Japan
| | | | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
144
|
Hermann BP, Sukhwani M, Salati J, Sheng Y, Chu T, Orwig KE. Separating spermatogonia from cancer cells in contaminated prepubertal primate testis cell suspensions. Hum Reprod 2011; 26:3222-31. [PMID: 22016413 DOI: 10.1093/humrep/der343] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Chemotherapy and radiation treatments for cancer and other diseases can cause male infertility. There are currently no options to preserve the fertility of prepubertal boys who are not yet making sperm. Cryopreservation of spermatogonial stem cells (SSCs, obtained via testicular biopsy) followed by autologous transplantation back into the testes at a later date may restore fertility in these patients. However, this approach carries an inherent risk of reintroducing cancer. METHODS To address this aspect of SSC transplantation safety, prepubertal non-human primate testis cell suspensions were inoculated with MOLT4 T-lymphoblastic leukemia cells and subsequently sorted for cell surface markers CD90 (THY-1) and CD45. RESULTS Cancer cells segregated to the CD90-/CD45+ fraction and produced tumors in nude mice. Nearly all sorted DEAD box polypeptide 4-positive (VASA+) spermatogonia segregated to the CD90+/CD45- fraction. In a preliminary experiment, a purity check of the sorted putative stem cell fraction (CD90+/CD45-) revealed 0.1% contamination with cancer cells, which was sufficient to produce tumors in nude mice. We hypothesized that the contamination resulted from mis-sorting due to cell clumping and employed singlet discrimination (SD) in four subsequent experiments. Purity checks revealed no cancer cell contamination in the CD90+/CD45- fraction from three of the four SD replicates and these fractions produced no tumors when transplanted into nude mouse testes. CONCLUSIONS We conclude that spermatogonia can be separated from contaminating malignant cells by fluorescence-activated cell sorting prior to SSC transplantation and that post-sorting purity checks are required to confirm elimination of malignant cells.
Collapse
Affiliation(s)
- Brian P Hermann
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | | | | | | | | | | |
Collapse
|
145
|
Ding LJ, Yan GJ, Ge QY, Yu F, Zhao X, Diao ZY, Wang ZQ, Yang ZZ, Sun HX, Hu YL. FSH acts on the proliferation of type A spermatogonia via Nur77 that increases GDNF expression in the Sertoli cells. FEBS Lett 2011; 585:2437-2444. [PMID: 21726557 DOI: 10.1016/j.febslet.2011.06.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 06/06/2011] [Accepted: 06/11/2011] [Indexed: 12/11/2022]
Abstract
The molecular mechanism responsible for the regulation of GDNF in Sertoli cells remains largely unknown. In the present study, FSH induced the expression of Nur77 and GDNF in mouse testis tissue and human fetal Sertoli cells. Moreover, FSH increased the number of A spermatogonia co-cultured with Sertoli cells. In the additional assays, Nur77 was observed to directly regulate GDNF transcription. Furthermore, overexpression of Nur77 and siRNA-mediated knockdown of Nur77 affected levels of GDNF mRNA and protein in primary human fetal Sertoli cells. These results indicate that FSH-induced Nur77 regulates the expression of GDNF in Sertoli cells to stimulate the proliferation of A spermatogonia in vitro.
Collapse
Affiliation(s)
- Li-Jun Ding
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing City, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Zhang S, Sun J, Pan S, Zhu H, Wang L, Hu Y, Wang J, Wang F, Cao H, Yan X, Hua J. Retinol (vitamin A) maintains self-renewal of pluripotent male germline stem cells (mGSCs) from adult mouse testis. J Cell Biochem 2011; 112:1009-21. [PMID: 21308744 DOI: 10.1002/jcb.23029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Studies have shown that male germline stem cells (mGSCs), which are responsible for maintaining spermatogenesis in the male, could be obtained from mouse and human testis. However, the traditional cultural methods were mostly dependent on serum and feeder, and the initial mGSCs were either obtained from neonatal mice or the detailed description of its potency and origin was not provided. Here we reported a novel (retinol (RE) serum-free and feeder-free) system for the successful culture of adult germline stem cells from adult Kunming mice (8-24 weeks) testis. The isolated mGSCs cultured in RE serum-free and feeder-free medium maintained the typical morphology of undifferentiated embryonic stem cells (ESCs), and they proliferated well in RE medium analyzed by proliferation assay, RT-PCR, microarray, and Western blotting. These cells also showed typical properties of ESCs (alkaline phosphatase (AP) positive, expressions of Oct4, Sox2, Nanog, and SSEA1, with the capacity to form teratomas and differentiate into various types of cells within three germ layers). Taken together, we conclude that RE promotes the self-renewal of mGSCs and maintains the pluripotency of mGSCs, the RE serum-free and feeder-free system may be useful for the culture of pluripotent stem cell lines from adult testis tissues, which provides a new resource for tissue engineering and therapy for infertility.
Collapse
Affiliation(s)
- Shanshan Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Reproductive Physiology & Embryo Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, Shaanxi 712100, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Ebata KT, Yeh JR, Zhang X, Nagano MC. Soluble growth factors stimulate spermatogonial stem cell divisions that maintain a stem cell pool and produce progenitors in vitro. Exp Cell Res 2011; 317:1319-29. [DOI: 10.1016/j.yexcr.2011.03.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 03/01/2011] [Accepted: 03/15/2011] [Indexed: 01/15/2023]
|
148
|
|
149
|
Kubota H, Wu X, Goodyear SM, Avarbock MR, Brinster RL. Glial cell line-derived neurotrophic factor and endothelial cells promote self-renewal of rabbit germ cells with spermatogonial stem cell properties. FASEB J 2011; 25:2604-14. [PMID: 21525489 DOI: 10.1096/fj.10-175802] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Previous studies suggest that exogenous factors crucial for spermatogonial stem cell (SSC) self-renewal are conserved among several mammalian species. Since glial cell line-derived neurotrophic factor (GDNF) and fibroblast growth factor 2 (FGF2) are critical for rodent SSC self-renewal, we hypothesized that they might promote self-renewal of nonrodent SSCs. Therefore, we cultured testicular germ cells from prepubertal rabbits in the presence of GDNF and FGF2 and found they proliferated indefinitely as cellular clumps that displayed characteristics previously identified for rodent SSCs. The rabbit germ cells could not be maintained on mouse embryonic fibroblast (STO) feeders that support rodent SSC self-renewal in vitro but were rather supported on mouse yolk sac-derived endothelial cell (C166) feeder layers. Proliferation of rabbit germ cells was dependent on GDNF. Of critical importance was that clump-forming rabbit germ cells colonized seminiferous tubules of immunodeficient mice, proliferated for at least 6 mo, while retaining an SSC phenotype in the testes of recipient mice, indicating that they were rabbit SSCs. This study demonstrates that GDNF is a mitogenic factor promoting self-renewal that is conserved between rodent and rabbit SSCs; with an evolutionary separation of ∼ 60 million years. These findings provide a foundation to study the mechanisms governing SSC self-renewal in nonrodent species.
Collapse
Affiliation(s)
- Hiroshi Kubota
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, 3850 Baltimore Ave., Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
150
|
Kobayashi H, Nagao K, Nakajima K. Stem cell research for male infertility. Reprod Med Biol 2011; 10:171-174. [PMID: 29699091 DOI: 10.1007/s12522-011-0085-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 04/06/2011] [Indexed: 01/01/2023] Open
Abstract
Stem cells have the ability both to differentiate into numerous tissues and to self-renew. Because of these unique properties, stem cells are promising candidates for use in regenerative medicine. Among stem cell types, embryonic stem (ES) cells have been the most studied; however, alternatives such as induced pluripotent stem cells or other adult stem cells are now being established. In this review, we focus on stem cell research that may have applications in treating male infertility. Stem cells with ES-like properties have been generated from adult human testis tissue. We expect that breakthroughs in stem cell research will increase our understanding of male infertility and lead to treatments in the near future.
Collapse
Affiliation(s)
- Hideyuki Kobayashi
- Department of Urology Toho University School of Medicine 6-11-1 Omori-Nishi, Ota-ku 143-8541 Tokyo Japan
| | - Koichi Nagao
- Department of Urology Toho University School of Medicine 6-11-1 Omori-Nishi, Ota-ku 143-8541 Tokyo Japan
| | - Koichi Nakajima
- Department of Urology Toho University School of Medicine 6-11-1 Omori-Nishi, Ota-ku 143-8541 Tokyo Japan
| |
Collapse
|