1
|
Jin C, Yan K, Wang M, Song W, Wang B, Men Y, Niu J, He Y, Zhang Q, Qi J. Dissecting the dynamic cellular transcriptional atlas of adult teleost testis development throughout the annual reproductive cycle. Development 2024; 151:dev202296. [PMID: 38477640 DOI: 10.1242/dev.202296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/09/2024] [Indexed: 03/14/2024]
Abstract
Teleost testis development during the annual cycle involves dramatic changes in cellular compositions and molecular events. In this study, the testicular cells derived from adult black rockfish at distinct stages - regressed, regenerating and differentiating - were meticulously dissected via single-cell transcriptome sequencing. A continuous developmental trajectory of spermatogenic cells, from spermatogonia to spermatids, was delineated, elucidating the molecular events involved in spermatogenesis. Subsequently, the dynamic regulation of gene expression associated with spermatogonia proliferation and differentiation was observed across spermatogonia subgroups and developmental stages. A bioenergetic transition from glycolysis to mitochondrial respiration of spermatogonia during the annual developmental cycle was demonstrated, and a deeper level of heterogeneity and molecular characteristics was revealed by re-clustering analysis. Additionally, the developmental trajectory of Sertoli cells was delineated, alongside the divergence of Leydig cells and macrophages. Moreover, the interaction network between testicular micro-environment somatic cells and spermatogenic cells was established. Overall, our study provides detailed information on both germ and somatic cells within teleost testes during the annual reproductive cycle, which lays the foundation for spermatogenesis regulation and germplasm preservation of endangered species.
Collapse
Affiliation(s)
- Chaofan Jin
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572000, China
| | - Kai Yan
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
| | - Mengya Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572000, China
| | - Weihao Song
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
| | - Bo Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572000, China
| | - Yu Men
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
| | - Jingjing Niu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
| | - Yan He
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572000, China
| | - Quanqi Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572000, China
| | - Jie Qi
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266000, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, 572000, China
| |
Collapse
|
2
|
Dolfini D, Imbriano C, Mantovani R. The role(s) of NF-Y in development and differentiation. Cell Death Differ 2024:10.1038/s41418-024-01388-1. [PMID: 39327506 DOI: 10.1038/s41418-024-01388-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
NF-Y is a conserved sequence-specific trimeric Transcription Factor -TF- binding to the CCAAT element. We review here the role(s) in development, from pre-implantation embryo to terminally differentiated tissues, by rationalizing and commenting on genetic, genomic, epigenetic and biochemical studies. This effort brings to light the impact of NF-YA isoforms on stemness and differentiation, as well as binding to distal vs promoter proximal sites and connections with selected TFs.
Collapse
Affiliation(s)
- Diletta Dolfini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Carol Imbriano
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, Modena, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
3
|
Zhao X, Liu L, Huang Z, Zhu F, Zhang H, Zhou D. PTN from Leydig cells activates SDC2 and modulates human spermatogonial stem cell proliferation and survival via GFRA1. Biol Res 2024; 57:66. [PMID: 39285301 PMCID: PMC11406790 DOI: 10.1186/s40659-024-00546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Spermatogonial stem cells (SSCs) are essential for the maintenance and initiation of male spermatogenesis. Despite the advances in understanding SSC biology in mouse models, the mechanisms underlying human SSC development remain elusive. RESULTS Here, we analyzed the signaling pathways involved in SSC regulation by testicular somatic cells using single-cell sequencing data (GEO datasets: GSE149512 and GSE112013) and identified that Leydig cells communicate with SSCs through pleiotrophin (PTN) and its receptor syndecan-2 (SDC2). Immunofluorescence, STRING prediction, and protein immunoprecipitation assays confirmed the interaction between PTN and SDC2 in spermatogonia, but their co-localization was observed only in approximately 50% of the cells. The knockdown of SDC2 in human SSC lines impaired cell proliferation, DNA synthesis, and the expression of PLZF, a key marker for SSC self-renewal. Transcriptome analysis revealed that SDC2 knockdown downregulated the expression of GFRA1, a crucial factor for SSC proliferation and self-renewal, and inhibited the HIF-1 signaling pathway. Exogenous PTN rescued the proliferation and GFRA1 expression in SDC2 knockdown SSC lines. In addition, we found downregulation of PTN and SDC2 as well as altered localization in non-obstructive azoospermia (NOA) patients, suggesting that downregulation of PTN and SDC2 may be associated with impaired spermatogenesis. CONCLUSIONS Our results uncover a novel mechanism of human SSC regulation by the testicular microenvironment and suggest a potential therapeutic target for male infertility.
Collapse
Affiliation(s)
- Xueheng Zhao
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan, 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
| | - Lvjun Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan, 410000, China
| | - Zenghui Huang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
| | - Fang Zhu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
| | - Huan Zhang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China
| | - Dai Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan, 410000, China.
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410000, China.
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410000, China.
| |
Collapse
|
4
|
Hai L, Maurya VK, DeMayo FJ, Lydon JP. Establishment of Murine Pregnancy Requires the Promyelocytic Leukemia Zinc Finger Transcription Factor. Int J Mol Sci 2024; 25:3451. [PMID: 38542422 PMCID: PMC10970820 DOI: 10.3390/ijms25063451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/02/2024] Open
Abstract
Using an established human primary cell culture model, we previously demonstrated that the promyelocytic leukemia zinc finger (PLZF) transcription factor is a direct target of the progesterone receptor (PGR) and is essential for progestin-dependent decidualization of human endometrial stromal cells (HESCs). These in vitro findings were supported by immunohistochemical analysis of human endometrial tissue biopsies, which showed that the strongest immunoreactivity for endometrial PLZF is detected during the progesterone (P4)-dominant secretory phase of the menstrual cycle. While these human studies provided critical clinical support for the important role of PLZF in P4-dependent HESC decidualization, functional validation in vivo was not possible due to the absence of suitable animal models. To address this deficiency, we recently generated a conditional knockout mouse model in which PLZF is ablated in PGR-positive cells of the mouse (Plzf d/d). The Plzf d/d female was phenotypically analyzed using immunoblotting, real-time PCR, and immunohistochemistry. Reproductive function was tested using the timed natural pregnancy model as well as the artificial decidual response assay. Even though ovarian activity is not affected, female Plzf d/d mice exhibit an infertility phenotype due to an inability of the embryo to implant into the Plzf d/d endometrium. Initial cellular and molecular phenotyping investigations reveal that the Plzf d/d endometrium is unable to develop a transient receptive state, which is reflected at the molecular level by a blunted response to P4 exposure with a concomitant unopposed response to 17-β estradiol. In addition to a defect in P4-dependent receptivity, the Plzf d/d endometrium fails to undergo decidualization in response to an artificial decidual stimulus, providing the in vivo validation for our earlier HESC culture findings. Collectively, our new Plzf d/d mouse model underscores the physiological importance of the PLZF transcription factor not only in endometrial stromal cell decidualization but also uterine receptivity, two uterine cellular processes that are indispensable for the establishment of pregnancy.
Collapse
Affiliation(s)
- Lan Hai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (L.H.); (V.K.M.)
| | - Vineet K. Maurya
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (L.H.); (V.K.M.)
| | - Francesco J. DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (L.H.); (V.K.M.)
| |
Collapse
|
5
|
Pappas MP, Kawakami H, Corcoran D, Chen KQ, Scott EP, Wong J, Gearhart MD, Nishinakamura R, Nakagawa Y, Kawakami Y. Sall4 regulates posterior trunk mesoderm development by promoting mesodermal gene expression and repressing neural genes in the mesoderm. Development 2024; 151:dev202649. [PMID: 38345319 PMCID: PMC10946440 DOI: 10.1242/dev.202649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
The trunk axial skeleton develops from paraxial mesoderm cells. Our recent study demonstrated that conditional knockout of the stem cell factor Sall4 in mice by TCre caused tail truncation and a disorganized axial skeleton posterior to the lumbar level. Based on this phenotype, we hypothesized that, in addition to the previously reported role of Sall4 in neuromesodermal progenitors, Sall4 is involved in the development of the paraxial mesoderm tissue. Analysis of gene expression and SALL4 binding suggests that Sall4 directly or indirectly regulates genes involved in presomitic mesoderm differentiation, somite formation and somite differentiation. Furthermore, ATAC-seq in TCre; Sall4 mutant posterior trunk mesoderm shows that Sall4 knockout reduces chromatin accessibility. We found that Sall4-dependent open chromatin status drives activation and repression of WNT signaling activators and repressors, respectively, to promote WNT signaling. Moreover, footprinting analysis of ATAC-seq data suggests that Sall4-dependent chromatin accessibility facilitates CTCF binding, which contributes to the repression of neural genes within the mesoderm. This study unveils multiple mechanisms by which Sall4 regulates paraxial mesoderm development by directing activation of mesodermal genes and repression of neural genes.
Collapse
Affiliation(s)
- Matthew P. Pappas
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hiroko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dylan Corcoran
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katherine Q. Chen
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Earl Parker Scott
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Julia Wong
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Micah D. Gearhart
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yasushi Nakagawa
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
6
|
Song Y, Zhang X, Desmarais JA, Nagano M. Postnatal development of mouse spermatogonial stem cells as determined by immunophenotype, regenerative capacity, and long-term culture-initiating ability: a model for practical applications. Sci Rep 2024; 14:2299. [PMID: 38280889 PMCID: PMC10821885 DOI: 10.1038/s41598-024-52824-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/24/2024] [Indexed: 01/29/2024] Open
Abstract
Spermatogonial stem cells (SSCs) are the foundation of life-long spermatogenesis. While SSC research has advanced greatly over the past two decades, characterization of SSCs during postnatal development has not been well documented. Using the mouse as a model, in this study, we defined the immunophenotypic profiles of testis cells during the course of postnatal development using multi-parameter flow cytometry with up to five cell-surface antigens. We found that the profiles progress over time in a manner specific to developmental stages. We then isolated multiple cell fractions at different developmental stages using fluorescent-activated cell sorting (FACS) and identified specific cell populations with prominent capacities to regenerate spermatogenesis upon transplantation and to initiate long-term SSC culture. The data indicated that the cell fraction with the highest level of regeneration capacity exhibited the most prominent potential to initiate SSC culture, regardless of age. Interestingly, refinement of cell fractionation using GFRA1 and KIT did not lead to further enrichment of regenerative and culture-initiating stem cells, suggesting that when a high degree of SSC enrichment is achieved, standard markers of SSC self-renewal or commitment may lose their effectiveness to distinguish cells at the stem cell state from committed progenitors. This study provides a significant information resource for future studies and practical applications of mammalian SSCs.
Collapse
Affiliation(s)
- Youngmin Song
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
| | - Xiangfan Zhang
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
| | - Joëlle A Desmarais
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
- JEFO Nutrition Inc, 5020 Avenue Jefo, Saint-Hyachinthe, Quebec, J2R 2E7, Canada
| | - Makoto Nagano
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
7
|
Jokar J, Abdulabbas HT, Alipanah H, Ghasemian A, Ai J, Rahimian N, Mohammadisoleimani E, Najafipour S. Tissue engineering studies in male infertility disorder. HUM FERTIL 2023; 26:1617-1635. [PMID: 37791451 DOI: 10.1080/14647273.2023.2251678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/06/2023] [Indexed: 10/05/2023]
Abstract
Infertility is an important issue among couples worldwide which is caused by a variety of complex diseases. Male infertility is a problem in 7% of all men. In vitro spermatogenesis (IVS) is the experimental approach that has been developed for mimicking seminiferous tubules-like functional structures in vitro. Currently, various researchers are interested in finding and developing a microenvironmental condition or a bioartificial testis applied for fertility restoration via gamete production in vitro. The tissue engineering (TE) has developed new approaches to treat male fertility preservation through development of functional male germ cells. This makes TE a possible future strategy for restoration of male fertility. Although 3D culture systems supply the perception of the effect of cellular interactions in the process of spermatogenesis, formation of a native gradient of autocrine/paracrine factors in 3D culture systems have not been considered. These results collectively suggest that maintaining the microenvironment of testicular cells even in the form of a 3D-culture system is crucial in achieving spermatogenesis ex vivo. It is also possible to engineer the testicular structures using biomaterials to provide a supporting scaffold for somatic and stem cells. The insemination of these cells with GFs is possible for temporally and spatially adjusted release to mimic the microenvironment of the in situ seminiferous epithelium. This review focuses on recent studies and advances in the application of TE strategies to cell-tissue culture on synthetic or natural scaffolds supplemented with growth factors.
Collapse
Affiliation(s)
- Javad Jokar
- Department of Tissue Engineering, Faculty of Medicine, Fasa University of Medical Science, Fasa, Iran
| | | | - Hiva Alipanah
- Department of Physiology, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Jafar Ai
- Tissue Engineering and Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Rahimian
- Department of Biotechnology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Elham Mohammadisoleimani
- Department of Biotechnology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Sohrab Najafipour
- Department of Microbiology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
8
|
Zhao X, Huang Z, Chen Y, Zhou Q, Zhu F, Zhang H, Zhou D. MAGEB2-Mediated Degradation of EGR1 Regulates the Proliferation and Apoptosis of Human Spermatogonial Stem Cell Lines. Stem Cells Int 2023; 2023:3610466. [PMID: 37304127 PMCID: PMC10256451 DOI: 10.1155/2023/3610466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 06/13/2023] Open
Abstract
Spermatogonial stem cells are committed to initiating and maintaining male spermatogenesis, which is the foundation of male fertility. Understanding the mechanisms underlying SSC fate decisions is critical for controlling spermatogenesis and male fertility. However, the key molecules and mechanisms responsible for regulating human SSC development are not clearly understood. Here, we analyzed normal human testis single-cell sequencing data from the GEO dataset (GSE149512 and GSE112013). Melanoma antigen gene B2 (MAGEB2) was found to be predominantly expressed in human SSCs and further validated by immunohistology. Overexpression of MAGEB2 in SSC lines severely weakened cell proliferation and promoted apoptosis. Further, using protein interaction prediction, molecular docking, and immunoprecipitation, we found that MAGEB2 interacted with early growth response protein 1 (EGR1) in SSC lines. Reexpression of EGR1 in MAGEB2 overexpression cells partially rescued decreased cell proliferation. Furthermore, MAGEB2 was shown to be downregulated in specific NOA patients, implying that abnormal expression of MAGEB2 may impair spermatogenesis and male fertility. Our results offer new insights into the functional and regulatory mechanisms in MAGEB2-mediated human SSC line proliferation and apoptosis.
Collapse
Affiliation(s)
- Xueheng Zhao
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Zenghui Huang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Yongzhe Chen
- First Affiliated Hospital of University of South China, Hengyang, Hunan 421000, China
| | - Qianyin Zhou
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Fang Zhu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Huan Zhang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Dai Zhou
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, Hunan 410000, China
- Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
- College of Life Sciences, Hunan Normal University, Changsha, Hunan 410000, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, Hunan 410000, China
| |
Collapse
|
9
|
Kahveci B, Önen S, Akal F, Korkusuz P. Detection of spermatogonial stem/progenitor cells in prepubertal mouse testis with deep learning. J Assist Reprod Genet 2023; 40:1187-1195. [PMID: 36995558 PMCID: PMC10239423 DOI: 10.1007/s10815-023-02784-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
PURPOSE Rapid and easy detection of spermatogonial stem/progenitor cells (SSPCs) is crucial for clinicians dealing with male infertility caused by prepubertal testicular damage. Deep learning (DL) methods may offer visual tools for tracking SSPCs on testicular strips of prepubertal animal models. The purpose of this study is to detect and count the seminiferous tubules and SSPCs in newborn mouse testis sections using a DL method. METHODS Testicular sections of the C57BL/6-type newborn mice were obtained and enumerated. Odd-numbered sections were stained with hematoxylin and eosin (H&E), and even-numbered sections were immune labeled (IL) with SSPC specific marker, SALL4. Seminiferous tubule and SSPC datasets were created using odd-numbered sections. SALL4-labeled sections were used as positive control. The YOLO object detection model based on DL was used to detect seminiferous tubules and stem cells. RESULTS Test scores of the DL model in seminiferous tubules were obtained as 0.98 mAP, 0.93 precision, 0.96 recall, and 0.94 f1-score. The SSPC test scores were obtained as 0.88 mAP, 0.80 precision, 0.93 recall, and 0.82 f1-score. CONCLUSION Seminiferous tubules and SSPCs on prepubertal testicles were detected with a high sensitivity by preventing human-induced errors. Thus, the first step was taken for a system that automates the detection and counting process of these cells in the infertility clinic.
Collapse
Affiliation(s)
- Burak Kahveci
- Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
| | - Selin Önen
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
- Department of Medical Biology, Faculty of Medicine, Atilim University, Ankara, Turkey
| | - Fuat Akal
- Computer Engineering Department, Hacettepe University, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100 Ankara, Turkey
| |
Collapse
|
10
|
Cason C, Lord T. RNA Interference as a Method of Gene Knockdown in Cultured Spermatogonia. Methods Mol Biol 2023; 2656:161-177. [PMID: 37249871 DOI: 10.1007/978-1-0716-3139-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Maintenance and self-renewal of the spermatogonial stem cell (SSC) population in the testis are dictated by the expression of a unique suite of genes. In manipulating gene expression through loss-of-function approaches, we can identify important regulatory mechanisms that dictate spermatogonial fate decisions. One such approach is RNA interference (RNAi), which uses natural cellular responses to small interfering RNAs to decrease levels of a targeted transcript. RNAi is performed in primary cultures of undifferentiated spermatogonia, and can be paired with techniques such as spermatogonial transplantation to assess the functional consequences of downregulated expression of the target gene on stem cell maintenance. This approach provides an alternative or complementary strategy to the generation of knockout mouse lines / cell lines. Here, we describe the methodology of RNAi in undifferentiated spermatogonia, and outline its inherent advantages and disadvantages over other technologies in the study of gene regulation in these cells.
Collapse
Affiliation(s)
- Connor Cason
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW, Australia.
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
11
|
Liu R, Peng Y, Du W, Wu Y, Zhang W, Hu C, Liu M, Liu X, Wu J, Sun J, Zhao X. BMI1 fine-tunes gene repression and activation to safeguard undifferentiated spermatogonia fate. Front Cell Dev Biol 2023; 11:1146849. [PMID: 37169021 PMCID: PMC10164956 DOI: 10.3389/fcell.2023.1146849] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction: Spermatogenesis is sustained by the homeostasis of self-renewal and differentiation of undifferentiated spermatogonia throughout life, which is regulated by transcriptional and posttranscriptional mechanisms. B cell-specific Moloney murine leukemia virus integration site 1 (BMI1), one of spermatogonial stem cell markers, is a member of Polycomb repressive complex 1 (PRC1) and important to spermatogenesis. However, the mechanistic underpinnings of how BMI1 regulates spermatogonia fate remain elusive. Methods: We knocked down BMI1 by siRNA to investigate the role of BMI1 in undifferentiated spermatogonia. Differentially expressed genes were identified by RNA-seq and used for KEGG pathway analysis. We performed ChIP-seq analysis in wild type and BMI1 knockdown cells to explore the underlying molecular mechanisms exerted by BMI1. BMI1-associated alterations in repressive histone modifications were detected via Western blotting and ChIP-seq. Furthermore, we performed mass spectrometry and Co-immunoprecipitation assays to investigate BMI1 co-factors. Finally, we demonstrated the genomic regions occupied by both BMI1 and its co-factor. Results: BMI1 is required for undifferentiated spermatogonia maintenance by both repressing and activating target genes. BMI1 preserves PI3K-Akt signaling pathway for spermatogonia proliferation. Decrease of BMI1 affects the deposition of repressive histone modifications H2AK119ub1 and H3K27me3. BMI also positively regulates H3K27ac deposited genes which are associated with proliferation. Moreover, we demonstrate that BMI1 interacts with Sal-like 4 (SALL4), the transcription factor critical for spermatogonia function, to co-regulate gene expression. Discussion: Overall, our study reveals that BMI1 safeguards undifferentiated spermatogonia fate through multi-functional roles in regulating gene expression programs of undifferentiated spermatogonia.
Collapse
Affiliation(s)
- Ruiqi Liu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yonglin Peng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wenfei Du
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yunqiang Wu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Zhang
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Congxia Hu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Min Liu
- Department of Integrative Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xinhua Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Ji Wu
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Ji Wu, ; Jielin Sun, ; Xiaodong Zhao,
| | - Jielin Sun
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Ji Wu, ; Jielin Sun, ; Xiaodong Zhao,
| | - Xiaodong Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Ji Wu, ; Jielin Sun, ; Xiaodong Zhao,
| |
Collapse
|
12
|
Huang L, Zhang J, Zhang P, Huang X, Yang W, Liu R, Sun Q, Lu Y, Zhang M, Fu Q. Single-cell RNA sequencing uncovers dynamic roadmap and cell-cell communication during buffalo spermatogenesis. iScience 2022; 26:105733. [PMID: 36582818 PMCID: PMC9793287 DOI: 10.1016/j.isci.2022.105733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis carries the task of precise intergenerational transmission of genetic information from the paternal genome and involves complex developmental processes regulated by the testicular microenvironment. Studies performed mainly in mouse models have established the theoretical basis for spermatogenesis, yet the wide interspecies differences preclude direct translation of the findings, and farm animal studies are progressing slowly. More than 32,000 cells from prepubertal (3-month-old) and pubertal (24-month-old) buffalo testes were analyzed by using single-cell RNA sequencing (scRNA-seq), and dynamic gene expression roadmaps of germ and somatic cell development were generated. In addition to identifying the dynamic processes of sequential cell fate transitions, the global cell-cell communication essential to maintain regular spermatogenesis in the buffalo testicular microenvironment was uncovered. The findings provide the theoretical basis for establishing buffalo germline stem cells in vitro or culturing organoids and facilitating the expansion of superior livestock breeding.
Collapse
Affiliation(s)
- Liangfeng Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Junjun Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Pengfei Zhang
- Institute of Medical and Health, Guangxi Academy of Sciences, Nanning 530007, China
| | - Xingchen Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Weihan Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Runfeng Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Qinqiang Sun
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Qiang Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| |
Collapse
|
13
|
Dang H, Martin‐Villalba A, Schiebel E. Centrosome linker protein C-Nap1 maintains stem cells in mouse testes. EMBO Rep 2022; 23:e53805. [PMID: 35599622 PMCID: PMC9253759 DOI: 10.15252/embr.202153805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 04/13/2022] [Accepted: 04/27/2022] [Indexed: 11/27/2022] Open
Abstract
The centrosome linker component C-Nap1 (encoded by CEP250) anchors filaments to centrioles that provide centrosome cohesion by connecting the two centrosomes of an interphase cell into a single microtubule organizing unit. The role of the centrosome linker during development of an animal remains enigmatic. Here, we show that male CEP250-/- mice are sterile because sperm production is abolished. Premature centrosome separation means that germ stem cells in CEP250-/- mice fail to establish an E-cadherin polarity mark and are unable to maintain the older mother centrosome on the basal site of the seminiferous tubules. This failure prompts premature stem cell differentiation in expense of germ stem cell expansion. The concomitant induction of apoptosis triggers the complete depletion of germ stem cells and consequently infertility. Our study reveals a role for centrosome cohesion in asymmetric cell division, stem cell maintenance, and fertility.
Collapse
Affiliation(s)
- Hairuo Dang
- Zentrum für Molekulare Biologie der Universität HeidelbergDeutsches Krebsforschungszentrum‐ZMBH AllianzUniversität HeidelbergHeidelbergGermany
- Heidelberg Biosciences International Graduate School (HBIGS)Universität HeidelbergHeidelbergGermany
| | - Ana Martin‐Villalba
- Deutsches Krebsforschungszentrum‐ZMBH AllianzUniversität HeidelbergHeidelbergGermany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität HeidelbergDeutsches Krebsforschungszentrum‐ZMBH AllianzUniversität HeidelbergHeidelbergGermany
| |
Collapse
|
14
|
Lin H, Cheng K, Kubota H, Lan Y, Riedel SS, Kakiuchi K, Sasaki K, Bernt KM, Bartolomei MS, Luo M, Wang PJ. Histone methyltransferase DOT1L is essential for self-renewal of germline stem cells. Genes Dev 2022; 36:752-763. [PMID: 35738678 PMCID: PMC9296001 DOI: 10.1101/gad.349550.122] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/06/2022] [Indexed: 12/25/2022]
Abstract
Self-renewal of spermatogonial stem cells is vital to lifelong production of male gametes and thus fertility. However, the underlying mechanisms remain enigmatic. Here, we show that DOT1L, the sole H3K79 methyltransferase, is required for spermatogonial stem cell self-renewal. Mice lacking DOT1L fail to maintain spermatogonial stem cells, characterized by a sequential loss of germ cells from spermatogonia to spermatids and ultimately a Sertoli cell only syndrome. Inhibition of DOT1L reduces the stem cell activity after transplantation. DOT1L promotes expression of the fate-determining HoxC transcription factors in spermatogonial stem cells. Furthermore, H3K79me2 accumulates at HoxC9 and HoxC10 genes. Our findings identify an essential function for DOT1L in adult stem cells and provide an epigenetic paradigm for regulation of spermatogonial stem cells.
Collapse
Affiliation(s)
- Huijuan Lin
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei Province 430072, China;,Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Keren Cheng
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Hiroshi Kubota
- Laboratory of Cell and Molecular Biology, Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan
| | - Yemin Lan
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Simone S. Riedel
- Division of Pediatric Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;,Abramson Cancer Center, Philadelphia, Pennsylvania 19104, USA
| | - Kazue Kakiuchi
- Laboratory of Cell and Molecular Biology, Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Aomori 034-8628, Japan
| | - Kotaro Sasaki
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Kathrin M. Bernt
- Division of Pediatric Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;,Abramson Cancer Center, Philadelphia, Pennsylvania 19104, USA
| | - Marisa S. Bartolomei
- Epigenetics Institute, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Mengcheng Luo
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei Province 430072, China
| | - P. Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
15
|
Normal embryonic development and neonatal digit regeneration in mice overexpressing a stem cell factor, Sall4. PLoS One 2022; 17:e0267273. [PMID: 35482646 PMCID: PMC9049339 DOI: 10.1371/journal.pone.0267273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/05/2022] [Indexed: 01/29/2023] Open
Abstract
Sall4 encodes a transcription factor and is known to participate in the pluripotency network of embryonic stem cells. Sall4 expression is known to be high in early stage post-implantation mouse embryos. During early post-gastrulation stages, Sall4 is highly expressed in the tail bud and distal limb buds, where progenitor cells are maintained in an undifferentiated status. The expression of Sall4 is rapidly downregulated during embryonic development. We previously demonstrated that Sall4 is required for limb and posterior axial skeleton development by conditional deletion of Sall4 in the T (Brachyury) lineage. To gain insight into Sall4 functions in embryonic development and postnatal digit regeneration, we genetically overexpressed Sall4 in the mesodermal lineage by the TCre transgene and a novel knockin allele of Rosa26-loxP-stop-loxP-Sall4. In significant contrast to severe defects by Sall4 loss of function reported in previous studies, overexpression of Sall4 resulted in normal morphology and pattern in embryos and neonates. The length of limb long bones showed subtle reduction in Sall4-overexpression mice. It is known that the digit tip of neonatal mice has level-specific regenerative ability after experimental amputation. We observed Sall4 expression in the digit tip by using a sensitive Sall4-LacZ knock-in reporter expression. Sall4 overexpression did not alter the regenerative ability of the terminal phalange that normally regenerates after amputation. Moreover, Sall4 overexpression did not confer regenerative ability to the second phalange that normally does not regenerate after amputation. These genetic experiments show that overexpression of Sall4 does not alter the development of the appendicular and axial skeleton, or neonatal digit regeneration. The results suggest that Sall4 acts as a permissive factor rather than playing an instructive role.
Collapse
|
16
|
Inoue H, Sakurai T, Hasegawa K, Suzuki A, Saga Y. NANOS3 suppresses premature spermatogonial differentiation to expand progenitors and fine-tunes spermatogenesis in mice. Biol Open 2022; 11:274984. [PMID: 35394008 PMCID: PMC9002807 DOI: 10.1242/bio.059146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/25/2022] [Indexed: 12/19/2022] Open
Abstract
In the mouse testis, sperm originate from spermatogonial stem cells (SSCs). SSCs give rise to spermatogonial progenitors, which expand their population until entering the differentiation process that is precisely regulated by a fixed time-scaled program called the seminiferous cycle. Although this expansion process of progenitors is highly important, its regulatory mechanisms remain unclear. NANOS3 is an RNA-binding protein expressed in the progenitor population. We demonstrated that the conditional deletion of Nanos3 at a later embryonic stage results in the reduction of spermatogonial progenitors in the postnatal testis. This reduction was associated with the premature differentiation of progenitors. Furthermore, this premature differentiation caused seminiferous stage disagreement between adjacent spermatogenic cells, which influenced spermatogenic epithelial cycles, leading to disruption of the later differentiation pathway. Our study suggests that NANOS3 plays an important role in timing progenitor expansion to adjust to the proper differentiation timing by blocking the retinoic acid (RA) signaling pathway.
Collapse
Affiliation(s)
- Hiroki Inoue
- Department of Gene Function and Phenomics, Mammalian Development Laboratory, National Institute of Genetics, Mishima, 411-8540Japan
| | - Takayuki Sakurai
- Department of Genetics, School of Life Science, The Graduate University for Advised Studies (SOKENDAI), Mishima, 411-8540Japan
| | - Kazuteru Hasegawa
- Department of Genetics, School of Life Science, The Graduate University for Advised Studies (SOKENDAI), Mishima, 411-8540Japan
| | - Atsushi Suzuki
- Division of Materials Science and Chemical Engineering, Faculty of Engineering, Yokohama National University, Yokohama, Kanagawa, 240-8501Japan
| | - Yumiko Saga
- Department of Gene Function and Phenomics, Mammalian Development Laboratory, National Institute of Genetics, Mishima, 411-8540Japan.,Department of Genetics, School of Life Science, The Graduate University for Advised Studies (SOKENDAI), Mishima, 411-8540Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
17
|
Huang T, Gu W, Liu E, Zhang L, Dong F, He X, Jiao W, Li C, Wang B, Xu G. Screening and Validation of p38 MAPK Involved in Ovarian Development of Brachymystax lenok. Front Vet Sci 2022; 9:752521. [PMID: 35252414 PMCID: PMC8889577 DOI: 10.3389/fvets.2022.752521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/13/2022] [Indexed: 11/17/2022] Open
Abstract
Brachymystax lenok (lenok) is a rare cold-water fish native to China that is of high meat quality. Its wild population has declined sharply in recent years, and therefore, exploring the molecular mechanisms underlying the development and reproduction of lenoks for the purposes of artificial breeding and genetic improvement is necessary. The lenok comparative transcriptome was analyzed by combining single molecule, real-time, and next generation sequencing (NGS) technology. Differentially expressed genes (DEGs) were identified in five tissues (head kidney, spleen, liver, muscle, and gonad) between immature [300 days post-hatching (dph)] and mature [three years post-hatching (ph)] lenoks. In total, 234,124 and 229,008 full-length non-chimeric reads were obtained from the immature and mature sequencing data, respectively. After NGS correction, 61,405 and 59,372 non-redundant transcripts were obtained for the expression level and pathway enrichment analyses, respectively. Compared with the mature group, 719 genes with significantly increased expression and 1,727 genes with significantly decreased expression in all five tissues were found in the immature group. Furthermore, DEGs and pathways involved in the endocrine system and gonadal development were identified, and p38 mitogen-activated protein kinases (MAPKs) were identified as potentially regulating gonadal development in lenok. Inhibiting the activity of p38 MAPKs resulted in abnormal levels of gonadotropin-releasing hormone, follicle-stimulating hormone, and estradiol, and affected follicular development. The full-length transcriptome data obtained in this study may provide a valuable reference for the study of gene function, gene expression, and evolutionary relationships in B. lenok and may illustrate the basic regulatory mechanism of ovarian development in teleosts.
Collapse
Affiliation(s)
- Tianqing Huang
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Wei Gu
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Enhui Liu
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Lanlan Zhang
- Heilongjiang Province General Station of Aquatic Technology Promotion, Harbin, China
| | - Fulin Dong
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Xianchen He
- Heilongjiang Aquatic Animal Resource Conservation Center, Harbin, China
| | - Wenlong Jiao
- Gansu Fisheries Research Institute, Lanzhou, China
| | - Chunyu Li
- Xinjiang Tianyun Organic Agriculture Co., Yili Group, Hohhot, China
| | - Bingqian Wang
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
- *Correspondence: Bingqian Wang
| | - Gefeng Xu
- Key Laboratory of Freshwater Aquatic Biotechnology and Breeding, Ministry of Agriculture and Rural Affairs, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
- Gefeng Xu
| |
Collapse
|
18
|
Huang ZH, Huang C, Ji XR, Zhou WJ, Luo XF, Liu Q, Tang YL, Gong F, Zhu WB. MKK7-mediated phosphorylation of JNKs regulates the proliferation and apoptosis of human spermatogonial stem cells. World J Stem Cells 2021; 13:1797-1812. [PMID: 34909124 PMCID: PMC8641020 DOI: 10.4252/wjsc.v13.i11.1797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/28/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human spermatogonial stem cells (SSCs) are the basis of spermatogenesis. However, little is known about the developmental regulatory mechanisms of SSC due to sample origin and species differences.
AIM To investigates the mechanisms involved in the proliferation of human SSC.
METHODS The expression of mitogen-activated protein kinase kinase 7 (MKK7) in human testis was identified using immunohistochemistry and western blotting (WB). MKK7 was knocked down using small interfering RNA, and cell proliferation and apoptosis were detected by WB, EdU, cell counting kit-8 and fluorescence-activated cell sorting. After bioinformatic analysis, the interaction of MKK7 with c-Jun N-terminal kinases ( JNKs ) was verified by protein co-immunoprecipitation and WB. The phosphorylation of JNKs was inhibited by SP600125, and the phenotypic changes were detected by WB, cell counting kit-8 and fluorescence-activated cell sorting.
RESULTS MKK7 is mainly expressed in human SSCs, and MKK7 knockdown inhibits SSC proliferation and promotes their apoptosis. MKK7 mediated the phosphorylation of JNKs, and after inhibiting the phosphorylation of JNKs, the phenotypic changes of the cells were similar to those after MKK7 downregulation. The expression of MKK7 was significantly downregulated in patients with abnormal spermatogenesis, suggesting that abnormal MKK7 may be associated with spermatogenesis impairment.
CONCLUSION MKK7 regulates the proliferation and apoptosis of human SSC by mediating the phosphorylation of JNKs.
Collapse
Affiliation(s)
- Zeng-Hui Huang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
- Department of Reproductive Center, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Chuan Huang
- Department of Sperm Bank, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Xi-Ren Ji
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Jun Zhou
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Xue-Feng Luo
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Liu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Yu-Lin Tang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Bing Zhu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
19
|
Graniel JV, Bisht K, Friedman A, White J, Perkey E, Vanderbeck A, Moroz A, Carrington LJ, Brandstadter JD, Allen F, Shami AN, Thomas P, Crayton A, Manzor M, Mychalowych A, Chase J, Hammoud SS, Keegan CE, Maillard I, Nandakumar J. Differential impact of a dyskeratosis congenita mutation in TPP1 on mouse hematopoiesis and germline. Life Sci Alliance 2021; 5:5/1/e202101208. [PMID: 34645668 PMCID: PMC8548261 DOI: 10.26508/lsa.202101208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/24/2022] Open
Abstract
A TPP1 mutation known to cause telomere shortening and bone marrow failure in humans recapitulates telomere loss but results in severe germline defects in mice without impacting murine hematopoiesis. Telomerase extends chromosome ends in somatic and germline stem cells to ensure continued proliferation. Mutations in genes critical for telomerase function result in telomeropathies such as dyskeratosis congenita, frequently resulting in spontaneous bone marrow failure. A dyskeratosis congenita mutation in TPP1 (K170∆) that specifically compromises telomerase recruitment to telomeres is a valuable tool to evaluate telomerase-dependent telomere length maintenance in mice. We used CRISPR-Cas9 to generate a mouse knocked in for the equivalent of the TPP1 K170∆ mutation (TPP1 K82∆) and investigated both its hematopoietic and germline compartments in unprecedented detail. TPP1 K82∆ caused progressive telomere erosion with increasing generation number but did not induce steady-state hematopoietic defects. Strikingly, K82∆ caused mouse infertility, consistent with gross morphological defects in the testis and sperm, the appearance of dysfunctional seminiferous tubules, and a decrease in germ cells. Intriguingly, both TPP1 K82∆ mice and previously characterized telomerase knockout mice show no spontaneous bone marrow failure but rather succumb to infertility at steady-state. We speculate that telomere length maintenance contributes differently to the evolutionary fitness of humans and mice.
Collapse
Affiliation(s)
- Jacqueline V Graniel
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.,Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Kamlesh Bisht
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.,Oncology Therapeutic Area, Sanofi, Cambridge, MA, USA
| | - Ann Friedman
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - James White
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.,Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA
| | - Eric Perkey
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA.,Division of Hematology/Oncology, Department of Medicine; Abramson Family Cancer Research Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ashley Vanderbeck
- Division of Hematology/Oncology, Department of Medicine; Abramson Family Cancer Research Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Alina Moroz
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA
| | - Léolène J Carrington
- Division of Hematology/Oncology, Department of Medicine; Abramson Family Cancer Research Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Joshua D Brandstadter
- Division of Hematology/Oncology, Department of Medicine; Abramson Family Cancer Research Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Frederick Allen
- Division of Hematology/Oncology, Department of Medicine; Abramson Family Cancer Research Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Adrienne Niederriter Shami
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI, USA.,Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Peedikayil Thomas
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.,Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA
| | - Aniela Crayton
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA
| | - Mariel Manzor
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA
| | | | - Jennifer Chase
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA.,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Saher S Hammoud
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Catherine E Keegan
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA .,Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Medicine; Abramson Family Cancer Research Institute, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Yang C, Yao C, Ji Z, Zhao L, Chen H, Li P, Tian R, Zhi E, Huang Y, Han X, Hong Y, Zhou Z, Li Z. RNA-binding protein ELAVL2 plays post-transcriptional roles in the regulation of spermatogonia proliferation and apoptosis. Cell Prolif 2021; 54:e13098. [PMID: 34296486 PMCID: PMC8450129 DOI: 10.1111/cpr.13098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022] Open
Abstract
Objectives RNA‐binding proteins (RBPs) play essential post‐transcriptional roles in regulating spermatogonial stem cells (SSCs) maintenance and differentiation. We identified a conserved and SSCs‐enriched RBP ELAVL2 from our single‐cell sequencing data, but its function and mechanism in SSCs were unclear. Materials and methods Expressions of ELAVL2 during human and mouse testis development were validated. Stable C18‐4 and TCam‐2 cell lines with overexpression and knockdown of ELAVL2 were established, which were applied to proliferation and apoptosis analysis. RNA immunoprecipitation and sequencing were used to identify ELAVL2 targets, and regulatory functions of ELAVL2 on target mRNAs were studied. Proteins interacting with ELAVL2 in human and mouse testes were identified using immunoprecipitation and mass spectrometric, which were validated by in vivo and in vitro experiments. Results ELAVL2 was testis‐enriched and preferentially expressed in human and mouse SSCs. ELAVL2 was down‐regulated in NOA patients. ELAVL2 promoted proliferation and inhibited apoptosis of C18‐4 and TCam‐2 cell lines via activating ERK and AKT pathways. ELAVL2 associated with mRNAs encoding essential regulators of SSCs proliferation and survival, and promoted their protein expression at post‐transcriptional level. ELAVL2 interacted with DAZL in vivo and in vitro in both human and mouse testes. Conclusions Taken together, these results indicate that ELAVL2 is a conserved SSCs‐enriched RBP that down‐regulated in NOA, which regulates spermatogonia proliferation and apoptosis by promoting protein expression of targets.
Collapse
Affiliation(s)
- Chao Yang
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chencheng Yao
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyong Ji
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Liangyu Zhao
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huixing Chen
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Li
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruhui Tian
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erlei Zhi
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhua Huang
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Han
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Hong
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zheng Li
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Kawahara T, Kanouchi M, Naniwa Y, Koyago M, Numabe T, Mizutani K, Tanemura K, Hara K. Persistence of undifferentiated spermatogonia in aged Japanese Black cattle. Anim Sci J 2021; 92:e13572. [PMID: 34254411 PMCID: PMC8365669 DOI: 10.1111/asj.13572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/09/2021] [Accepted: 05/14/2021] [Indexed: 12/26/2022]
Abstract
Aging is a major risk factor for spermatogenesis deterioration. However, the influence of age on spermatogenic stem cells and their progenitors in bulls is largely unknown. Here, we report age‐related changes in undifferentiated and differentiating spermatogonia in Japanese Black cattle with nearly constant sperm output, by using spermatogonial markers. The numbers of differentiating spermatogonia and more differentiated spermatogenic cells were significantly decreased in aged bovine testes compared with those in young testes. In contrast, the number of undifferentiated spermatogonia was maintained, and their proliferative activity did not differ significantly between young and aged bovine testes. Although severe calcification was only observed to a small extent in aged testes, fewer Sertoli cells and interstitial fibrosis were observed in noncalcified testicular regions. These results suggest that, even in old bulls with nearly constant sperm output, testicular spermatogenic activity declined whereas undifferentiated spermatogonia numbers were maintained. Thus, we propose that undifferentiated spermatogonia may be resistant to age‐related changes in bovine testes. Because undifferentiated spermatogonia may contain stem cell activity, our findings highlight the potential utility of undifferentiated spermatogonia as an agricultural resource to produce spermatozoa beyond the natural bovine lifetime through transplantation and in vitro spermatogenesis in future animal production.
Collapse
Affiliation(s)
- Terumichi Kawahara
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Miki Kanouchi
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Yousuke Naniwa
- Maebashi Institute of Animal Science, Livestock Improvement Association of Japan, Inc., Maebashi, Gunma, Japan
| | - Masanori Koyago
- Maebashi Institute of Animal Science, Livestock Improvement Association of Japan, Inc., Maebashi, Gunma, Japan
| | - Takashi Numabe
- Miyagi Agricultural Development Corporation, Sendai, Miyagi, Japan
| | - Keishi Mizutani
- Morioka AI Center, Livestock Improvement Association of Japan, Inc., Morioka, Iwate, Japan
| | - Kentaro Tanemura
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Kenshiro Hara
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
22
|
Yu XW, Li TT, Du XM, Shen QY, Zhang MF, Wei YD, Yang DH, Xu WJ, Chen WB, Bai CL, Li XL, Li GP, Li N, Peng S, Liao MZ, Hua JL. Single-cell RNA sequencing reveals atlas of dairy goat testis cells. Zool Res 2021; 42:401-405. [PMID: 34047080 PMCID: PMC8317185 DOI: 10.24272/j.issn.2095-8137.2020.373] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Xiu-Wei Yu
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Tong-Tong Li
- College of Life Science, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Xiao-Min Du
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Qiao-Yan Shen
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Meng-Fei Zhang
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Yu-Dong Wei
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Dong-Hui Yang
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Wen-Jing Xu
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Wen-Bo Chen
- College of Life Science, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Chun-Ling Bai
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Xue-Ling Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Guang-Peng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Na Li
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Sha Peng
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China. E-mail:
| | - Ming-Zhi Liao
- College of Life Science, Northwest A & F University, Yangling, Shaanxi 712100, China. E-mail:
| | - Jin-Lian Hua
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China. E-mail:
| |
Collapse
|
23
|
Khanehzad M, Nourashrafeddin SM, Abolhassani F, Kazemzadeh S, Madadi S, Shiri E, Khanlari P, Khosravizadeh Z, Hedayatpour A. MicroRNA-30a-5p promotes differentiation in neonatal mouse spermatogonial stem cells (SSCs). Reprod Biol Endocrinol 2021; 19:85. [PMID: 34108007 PMCID: PMC8188658 DOI: 10.1186/s12958-021-00758-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The importance of spermatogonial stem cells (SSCs) in spermatogenesis is crucial and intrinsic factors and extrinsic signals mediate fate decisions of SSCs. Among endogenous regulators, microRNAs (miRNAs) play critical role in spermatogenesis. However, the mechanisms which individual miRNAs regulate self- renewal and differentiation of SSCs are unknown. The aim of this study was to investigate effects of miRNA-30a-5p inhibitor on fate determinations of SSCs. METHODS SSCs were isolated from testes of neonate mice (3-6 days old) and their purities were performed by flow cytometry with ID4 and Thy1 markers. Cultured cells were transfected with miRNA- 30a-5p inhibitor. Evaluation of the proliferation (GFRA1, PLZF and ID4) and differentiation (C-Kit & STRA8) markers of SSCs were accomplished by immunocytochemistry and western blot 48 h after transfection. RESULTS Based on the results of flow cytometry with ID4 and Thy1 markers, percentage of purity of SSCs was about 84.3 and 97.4 % respectively. It was found that expression of differentiation markers after transfection was significantly higher in miRNA-30a- 5p inhibitor group compared to other groups. The results of proliferation markers evaluation also showed decrease of GFRA1, PLZF and ID4 protein in SSCs transfected with miRNA-30a-5p inhibitor compared to the other groups. CONCLUSIONS It can be concluded that inhibition of miRNA-30a-5p by overexpression of differentiation markers promotes differentiation of Spermatogonial Stem Cells.
Collapse
Affiliation(s)
- Maryam Khanehzad
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Seyed Mehdi Nourashrafeddin
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, USA
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abolhassani
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Shokoofeh Kazemzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Soheila Madadi
- Department of Anatomy, School of Medicine, Arak University of Medical Science, Arak, Iran
| | - Elham Shiri
- Department of Anatomical Sciences, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parastoo Khanlari
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Zahra Khosravizadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Azim Hedayatpour
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
24
|
A regulatory role for CHD4 in maintenance of the spermatogonial stem cell pool. Stem Cell Reports 2021; 16:1555-1567. [PMID: 33961790 PMCID: PMC8190575 DOI: 10.1016/j.stemcr.2021.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/17/2022] Open
Abstract
Maintenance and self-renewal of the spermatogonial stem cell (SSC) population is the cornerstone of male fertility. Here, we have identified a key role for the nucleosome remodeling protein CHD4 in regulating SSC function. Gene expression analyses revealed that CHD4 expression is highly enriched in the SSC population in the mouse testis. Using spermatogonial transplantation techniques it was established that loss of Chd4 expression significantly impairs SSC regenerative capacity, causing a ∼50% reduction in colonization of recipient testes. An scRNA-seq comparison revealed reduced expression of “self-renewal” genes following Chd4 knockdown, along with increased expression of signature progenitor genes. Co-immunoprecipitation analyses demonstrated that CHD4 regulates gene expression in spermatogonia not only through its traditional association with the remodeling complex NuRD, but also via interaction with the GDNF-responsive transcription factor SALL4. Cumulatively, the results of this study depict a previously unappreciated role for CHD4 in controlling fate decisions in the spermatogonial pool. CHD4 is highly expressed in spermatogonial stem cells in the mouse testis CHD4 expression is required for spermatogonial stem cell maintenance CHD4 interacts with SALL4 and NuRD to activate expression of “self-renewal” genes
Collapse
|
25
|
Wei YD, Du XM, Yang DH, Ma FL, Yu XW, Zhang MF, Li N, Peng S, Liao MZ, Li GP, Bai CL, Liu WS, Hua JL. Dmrt1 regulates the immune response by repressing the TLR4 signaling pathway in goat male germline stem cells. Zool Res 2021; 42:14-27. [PMID: 33420764 PMCID: PMC7840460 DOI: 10.24272/j.issn.2095-8137.2020.186] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Double sex and mab-3-related transcription factor 1 (Dmrt1), which is expressed in goat male germline stem cells (mGSCs) and Sertoli cells, is one of the most conserved transcription factors involved in sex determination. In this study, we highlighted the role of Dmrt1 in balancing the innate immune response in goat mGSCs. Dmrt1 recruited promyelocytic leukemia zinc finger (Plzf), also known as zinc finger and BTB domain-containing protein 16 (Zbtb16), to repress the Toll-like receptor 4 (TLR4)-dependent inflammatory signaling pathway and nuclear factor (NF)-κB. Knockdown of Dmrt1 in seminiferous tubules resulted in widespread degeneration of germ and somatic cells, while the expression of proinflammatory factors were significantly enhanced. We also demonstrated that Dmrt1 stimulated proliferation of mGSCs, but repressed apoptosis caused by the immune response. Thus, Dmrt1 is sufficient to reduce inflammation in the testes, thereby establishing the stability of spermatogenesis and the testicular microenvironment.
Collapse
Affiliation(s)
- Yu-Dong Wei
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Xiao-Min Du
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Dong-Hui Yang
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Fang-Lin Ma
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Xiu-Wei Yu
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Meng-Fei Zhang
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Na Li
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Sha Peng
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China
| | - Ming-Zhi Liao
- College of Life Sciences, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Guang-Peng Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010021, China
| | - Chun-Ling Bai
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010021, China. E-mail:
| | - Wei-Shuai Liu
- Department of Pathology, Yangling Demonstration Zone Hospital, Yangling Shaanxi 712100, China. E-mail:
| | - Jin-Lian Hua
- College of Veterinary Medicine, Northwest A & F University, Shaanxi Centre of Stem Cells Engineering & Technology, Yangling, Shaanxi 712100, China. E-mail:
| |
Collapse
|
26
|
Zeber-Lubecka N, Hennig EE. Genetic Susceptibility to Joint Occurrence of Polycystic Ovary Syndrome and Hashimoto's Thyroiditis: How Far Is Our Understanding? Front Immunol 2021; 12:606620. [PMID: 33746952 PMCID: PMC7968419 DOI: 10.3389/fimmu.2021.606620] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) and Hashimoto’s thyroiditis (HT) are endocrine disorders that commonly occur among young women. A higher prevalence of HT in women with PCOS, relative to healthy individuals, is observed consistently. Combined occurrence of both diseases is associated with a higher risk of severe metabolic and reproductive complications. Genetic factors strongly impact the pathogenesis of both PCOS and HT and several susceptibility loci associated with a higher risk of both disorders have been identified. Furthermore, some candidate gene polymorphisms are thought to be functionally relevant; however, few genetic variants are proposed to be causally associated with the incidence of both disorders together.
Collapse
Affiliation(s)
- Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Ewa E Hennig
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland.,Department of Genetics, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
27
|
Mast4 knockout shows the regulation of spermatogonial stem cell self-renewal via the FGF2/ERM pathway. Cell Death Differ 2020; 28:1441-1454. [PMID: 33219327 DOI: 10.1038/s41418-020-00670-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/22/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis is an important cellular differentiation process that produces the male gametes and remains active throughout the individual's lifespan. Sertoli cell-only syndrome (SCO) refers to the dysfunction of the male reproductive system, including infertility. Accurate self-renewal of spermatogonial stem cells (SSCs) is essential to prevent SCO syndrome. This study investigated the role of microtubule-associated serine/threonine kinase family member 4 (MAST4) in spermatogenesis in mice. MAST4 was localized in Sertoli cells before puberty, providing a somatic niche for spermatogenesis in mice and MAST4 expression shifted to Leydig cells and spermatids throughout puberty. Mast4 knockout (KO) testes were reduced in size compared to wild-type testes, and germ cell depletion associated with an increase in apoptosis and subsequent loss of tubular structure were similar to the SCO phenotype. In addition, MAST4 phosphorylated the Ets-related molecule (ERM), specifically the serine 367 residue. The phosphorylation of ERM ultimately controls the transcription of ERM target genes related to SSC self-renewal. The expression of spermatogenesis-associated proteins was significantly decreased whereas Sertoli cell markers were increased in Mast4 KO testes, which was well-founded by RNA-sequencing analysis. Therefore, MAST4 is associated with the fibroblast growth factor 2 (FGF2)/ERM pathway and this association helps us explore the capacity of SSCs to maintain a vertebrate stem cell niche.
Collapse
|
28
|
Wei Y, Yang D, Du X, Yu X, Zhang M, Tang F, Ma F, Li N, Bai C, Li G, Hua J. Interaction between DMRT1 and PLZF protein regulates self-renewal and proliferation in male germline stem cells. Mol Cell Biochem 2020; 476:1123-1134. [PMID: 33200378 DOI: 10.1007/s11010-020-03977-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/06/2020] [Indexed: 01/04/2023]
Abstract
Double sex and mab-3 related transcription factor 1 (DMRT1) encodes a double sex/mab-3 (DM) domain, which is the most conserved structure that involved in sex determination both in vertebrates and invertebrates. This study revealed important roles of DMRT1 in maintaining self-renewal of male germline stem cells (mGSCs). Our results showed that insufficient expression of DMRT1 in mice testes resulted in decreased number of spermatogonial cells and collapse of testicular niche in vivo. Self-renewal and proliferation of mGSCs were inhibited. Based on the bimolecular fluorescence complementation (BiFC) and co-immunoprecipitation (co-IP) assay, it was finally revealed that the interaction between DMRT1 and promyelocytic leukemia zinc finger (PLZF) protein was essential for maintaining self-renewal of mGSCs. Moreover, BTB domain of PLZF, DM and DMRT1 domain of DMRT1 were indispensable in mGSC, which were responsible for preserving the quantity of germ cells. Our research provided a new scientific basis for studying the mechanism of self-renewal and spermatogenesis in goat mGSCs.
Collapse
Affiliation(s)
- Yudong Wei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Donghui Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Xiaomin Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Xiuwei Yu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Mengfei Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Furong Tang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Fanglin Ma
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China
| | - Chunling Bai
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Guangpeng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, No. 3rd, Taicheng Road, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
29
|
McAninch D, Thomson EP, Thomas PQ. Genome-wide DNA-binding profile of SRY-box transcription factor 3 (SOX3) in mouse testes. Reprod Fertil Dev 2020; 32:1260-1270. [PMID: 33166488 DOI: 10.1071/rd20108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
Spermatogenesis is the male version of gametogenesis, where germ cells are transformed into haploid spermatozoa through a tightly controlled series of mitosis, meiosis and differentiation. This process is reliant on precisely timed changes in gene expression controlled by several different hormonal and transcriptional mechanisms. One important transcription factor is SRY-box transcription factor 3 (SOX3), which is transiently expressed within the uncommitted spermatogonial stem cell population. Sox3-null mouse testes exhibit a block in spermatogenesis, leading to infertility or subfertility. However, the molecular role of SOX3 during spermatogonial differentiation remains poorly understood because the genomic regions targeted by this transcription factor have not been identified. In this study we used chromatin immunoprecipitation sequencing to identify and characterise the endogenous genome-wide binding profile of SOX3 in mouse testes at Postnatal Day 7. We show that neurogenin3 (Neurog3 or Ngn3) is directly targeted by SOX3 in spermatogonial stem cells via a novel testes-specific binding site. We also implicate SOX3, for the first time, in direct regulation of histone gene expression and demonstrate that this function is shared by both neural progenitors and testes, and with another important transcription factor required for spermatogenesis, namely promyelocytic leukaemia zinc-finger (PLZF). Together, these data provide new insights into the function of SOX3 in different stem cell contexts.
Collapse
Affiliation(s)
- Dale McAninch
- School of Biological Sciences and Robinson Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia
| | - Ella P Thomson
- School of Biological Sciences and Robinson Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia
| | - Paul Q Thomas
- School of Biological Sciences and Robinson Research Institute, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia; and Adelaide Medical School, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia; and Precision Medicine Theme, South Australia Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia; and Corresponding author.
| |
Collapse
|
30
|
Histologic Analysis of Testes from Prepubertal Patients Treated with Chemotherapy Associates Impaired Germ Cell Counts with Cumulative Doses of Cyclophosphamide, Ifosfamide, Cytarabine, and Asparaginase. Reprod Sci 2020; 28:603-613. [PMID: 33150486 DOI: 10.1007/s43032-020-00357-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/11/2020] [Indexed: 01/15/2023]
Abstract
Cryopreservation of immature testicular tissue is an experimental strategy for the preservation of fertility in prepubertal boys that will be subjected to a gonadotoxic onset, as is the case of oncologic patients. Therefore, the objective of this study was to assess the impact of chemotherapeutic treatments on the testicular histologic phenotype in prepubertal patients. A total of 56 testicular tissue samples from pediatric patients between 0 and 16 years old (28 with at least one previous chemotherapeutic onset and 28 untreated controls) were histologically analyzed and age-matched compared. At least two 5-μm sections from testis per patient separated by a distance of 100 μm were immunostained for the germ cell marker VASA, the spermatogonial markers UTF1, PLZF, UCHL1, and SALL4, the marker for proliferative cells KI67, and the Sertoli cell marker SOX9. The percentage of tubule cross-sections positive for each marker and the number of positive cells per tubule cross-section were determined and association with the cumulative dose received of each chemotherapeutic drug was statistically assessed. Results indicated that alkylating agents, cyclophosphamide and ifosfamide, but also the antimetabolite cytarabine and asparaginase were associated with a decreased percentage of positive tubules and a lower number of positive cells per tubule for the analyzed markers. Our results provide new evidences of the potential of chemotherapeutic agents previously considered to have low gonadotoxic effects such as cytarabine and asparaginase to trigger a severe testicular phenotype, hampering the potential success of future fertility restoration in experimental programs of fertility preservation in prepubertal boys.
Collapse
|
31
|
Azizi H, Niazi Tabar A, Skutella T, Govahi M. In Vitro and In Vivo Determinations of The Anti-GDNF Family Receptor Alpha 1 Antibody in Mice by Immunochemistry and RT-PCR. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2020; 14:228-233. [PMID: 33098391 PMCID: PMC7604702 DOI: 10.22074/ijfs.2020.6051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 01/11/2020] [Indexed: 12/31/2022]
Abstract
Background The glial cell-derived neurotrophic factor (GDNF) family plays essential roles in the maintenance, growth, regulatory and signalling pathways of spermatogonial stem cells (SSCs). In this study, we analysed the expression of anti-GDNF family receptor alpha 1 antibody (GFRa1) by immunohistochemistry (IHC), immunocytochemistry (ICC), Fluidigm real-time polymerase chain reaction (RT-PCR) and flow cytometry analyses. Materials and Methods In this experiment study, ICC, IHC, Fluidigm RT-PCR and flow cytometry were used to analyse the expression of the germ cell marker GFRa1 in testis tissue and SSC culture. Results IHC analysis showed that there were two groups of GFRa1 positive cells in the seminiferous tubules based on their location and expression shape - a small round punctuated shape on the basal compartment donut shape and a C-shaped expression located between the basal and the luminal compartments of the seminiferous tubules. OCT4 and PLZF positive cells may have similar patterns of expression as the first group. Assessment of the seminiferous tubule sections demonstrated that about 27% of the SSCs were positive for GFRa1. Fluidigm RT-PCR confirmed the significant expression (P<0.001) of GFRa1 in the SSCs compared to testicular stromal cells (TSCs). Flow cytometry analysis demonstrated that about 75% of the isolated SSCs colonies were positive for GFRa1. Conclusion The results indicated that GFRa1 had a specific expression pattern both in vivo and in vitro. This finding could be helpful for understanding the proliferation, maintenance and signalling pathways of SSCs, and differentiation of meiotic and haploid germ cells.
Collapse
Affiliation(s)
- Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran. Electronic Address:
| | - Amirreza Niazi Tabar
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Mostafa Govahi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| |
Collapse
|
32
|
Yang J, Zhao R, Li L, Li G, Yang P, Ma J, Zhao S, Zhao H. Verification of a ZBTB16 variant in polycystic ovary syndrome patients. Reprod Biomed Online 2020; 41:724-728. [PMID: 32773339 PMCID: PMC10730780 DOI: 10.1016/j.rbmo.2020.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/04/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
RESEARCH QUESTION This study investigated whether rs1784692 is a risk factor for polycystic ovary syndrome (PCOS) in Han Chinese women. DESIGN A case-control study was conducted in Han Chinese women, involving 526 PCOS patients and 522 control participants. A TaqMan MGB probe assay was used to genotype the variant rs1784692. Dominant and additive models were employed for genotype-phenotype association analysis in the PCOS and control samples. RESULTS The minor allele C of rs1784692 is protective against PCOS (odds ratio [OR] 0.556, 95% confidence interval [CI] 0.408-0.759, P = 1.83 × 10-4), even after adjustment for body mass index (BMI) and age (ORadj 0.539, 95% CI 0.391-0.743, Padj= 1.62 × 10-4). Genotype-phenotype analysis of the dominant model showed that mean BMI in the CC+CT group was higher than in the TT group in the PCOS group (27.12 ± 5.82 versus 24.57 ± 4.52, P = 1.0 × 10-3), but not in the control groups, indicating that the minor allele C of rs1784692 associates with BMI in women with PCOS. The mean LH (luteinizing hormone) concentration in the CC+CT group was lower than in the TT group in PCOS and control participants (9.33 ± 5.08 versus 10.93 ± 5.91, P = 0.036; 4.39 ± 1.66 versus 4.89 ± 2.07, P = 0.021). Genotype-phenotype analysis of additive model showed that mean BMI in TC group was higher than in the TT group in PCOS patients compared with control participants (27.14 ± 5.81 versus 24.57 ± 4.52, P = 3.06 × 10-3). CONCLUSIONS The SNP rs1784692 in gene ZBTB16 is protective against PCOS but is associated with increased BMI in Han Chinese women with PCOS.
Collapse
Affiliation(s)
- Jie Yang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China
| | - Rusong Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China
| | - Lei Li
- Department of Biological Sciences, University of Notre Dame, Notre Dame IN, USA
| | - Guangyu Li
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Ping Yang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China
| | - Jinlong Ma
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China
| | - Shigang Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China.
| | - Han Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, China.
| |
Collapse
|
33
|
Suzuki S, Diaz VD, Hermann BP. What has single-cell RNA-seq taught us about mammalian spermatogenesis? Biol Reprod 2020; 101:617-634. [PMID: 31077285 DOI: 10.1093/biolre/ioz088] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022] Open
Abstract
Mammalian spermatogenesis is a complex developmental program that transforms mitotic testicular germ cells (spermatogonia) into mature male gametes (sperm) for production of offspring. For decades, it has been known that this several-weeks-long process involves a series of highly ordered and morphologically recognizable cellular changes as spermatogonia proliferate, spermatocytes undertake meiosis, and spermatids develop condensed nuclei, acrosomes, and flagella. Yet, much of the underlying molecular logic driving these processes has remained opaque because conventional characterization strategies often aggregated groups of cells to meet technical requirements or due to limited capability for cell selection. Recently, a cornucopia of single-cell transcriptome studies has begun to lift the veil on the full compendium of gene expression phenotypes and changes underlying spermatogenic development. These datasets have revealed the previously obscured molecular heterogeneity among and between varied spermatogenic cell types and are reinvigorating investigation of testicular biology. This review describes the extent of available single-cell RNA-seq profiles of spermatogenic and testicular somatic cells, how those data were produced and evaluated, their present value for advancing knowledge of spermatogenesis, and their potential future utility at both the benchtop and bedside.
Collapse
Affiliation(s)
- Shinnosuke Suzuki
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Victoria D Diaz
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
34
|
Xie Y, Wei BH, Ni FD, Yang WX. Conversion from spermatogonia to spermatocytes: Extracellular cues and downstream transcription network. Gene 2020; 764:145080. [PMID: 32858178 DOI: 10.1016/j.gene.2020.145080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022]
Abstract
Spermatocyte (spc) formation from spermatogonia (spg) differentiation is the first step of spermatogenesis which produces prodigious spermatozoa for a lifetime. After decades of studies, several factors involved in the functioning of a mouse were discovered both inside and outside spg. Considering the peculiar expression and working pattern of each factor, this review divides the whole conversion of spg to spc into four consecutive development processes with a focus on extracellular cues and downstream transcription network in each one. Potential coordination among Dmrt1, Sohlh1/2 and BMP families mediates Ngn3 upregulation, which marks progenitor spg, with other changes. After that, retinoic acid (RA), as a master regulator, promotes A1 spg formation with its helpers and Sall4. A1-to-B spg transition is under the control of Kitl and impulsive RA signaling together with early and late transcription factors Stra8 and Dmrt6. Finally, RA and its responsive effectors conduct the entry into meiosis. The systematic transcription network from outside to inside still needs research to supplement or settle the controversials in each process. As a step further ahead, this review provides possible drug targets for infertility therapy by cross-linking humans and mouse model.
Collapse
Affiliation(s)
- Yi Xie
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bang-Hong Wei
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fei-Da Ni
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
35
|
Chen KQ, Tahara N, Anderson A, Kawakami H, Kawakami S, Nishinakamura R, Pandolfi PP, Kawakami Y. Development of the Proximal-Anterior Skeletal Elements in the Mouse Hindlimb Is Regulated by a Transcriptional and Signaling Network Controlled by Sall4. Genetics 2020; 215:129-141. [PMID: 32156750 PMCID: PMC7198279 DOI: 10.1534/genetics.120.303069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 03/03/2020] [Indexed: 12/18/2022] Open
Abstract
The vertebrate limb serves as an experimental paradigm to study mechanisms that regulate development of the stereotypical skeletal elements. In this study, we simultaneously inactivated Sall4 using Hoxb6Cre and Plzf in mouse embryos, and found that their combined function regulates development of the proximal-anterior skeletal elements in hindlimbs. The Sall4; Plzf double knockout exhibits severe defects in the femur, tibia, and anterior digits, distinct defects compared to other allelic series of Sall4; Plzf We found that Sall4 regulates Plzf expression prior to hindlimb outgrowth. Further expression analysis indicated that Hox10 genes and GLI3 are severely downregulated in the Sall4; Plzf double knockout hindlimb bud. In contrast, PLZF expression is reduced but detectable in Sall4; Gli3 double knockout limb buds, and SALL4 is expressed in the Plzf; Gli3 double knockout limb buds. These results indicate that Plzf, Gli3, and Hox10 genes downstream of Sall4, regulate femur and tibia development. In the autopod, we show that Sall4 negatively regulates Hedgehog signaling, which allows for development of the most anterior digit. Collectively, our study illustrates genetic systems that regulate development of the proximal-anterior skeletal elements in hindlimbs.
Collapse
Affiliation(s)
| | - Naoyuki Tahara
- Department of Genetics, Cell Biology and Development
- Stem Cell Institute, Minneapolis, Minnesota 55455, and
- Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota 55455
| | | | - Hiroko Kawakami
- Department of Genetics, Cell Biology and Development
- Stem Cell Institute, Minneapolis, Minnesota 55455, and
- Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota 55455
| | - Sho Kawakami
- Department of Genetics, Cell Biology and Development
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan 860-0811
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development
- Stem Cell Institute, Minneapolis, Minnesota 55455, and
- Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
36
|
Zhu H, Zheng L, Wang L, Tang F, Arisha AH, Zhou H, Hua J. p53 inhibits the proliferation of male germline stem cells from dairy goat cultured on poly-L-lysine. Reprod Domest Anim 2020; 55:405-417. [PMID: 31985843 DOI: 10.1111/rda.13645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 12/13/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022]
Abstract
Male germline stem cells (mGSCs) can transmit genetic materials to the next generation and dedifferentiate into pluripotent stem cells. However, in livestock, mGSC lines are difficult to establish, because of the factors that affect their isolation and culture. The extracellular matrix serves as a substrate for attachment and affects the fate of these stem cells. Poly-L-lysine (PL), an extracellular matrix of choice, inhibits and/or kills cancer cells, and promotes the attachment of stem cells in culture. However, how it affects the characteristics and potentials of these stem cells in culture needs to be elucidated. Here, we isolated, enriched and cultured dairy goat mGSCs on five types of extracellular matrices. To explore the best extracellular matrix to use for culturing them, the characteristics and proliferation ability of the cells were determined. Results showed that the cells shared several characteristics with previously reported mGSCs, including the poor effect of PL on their proliferative and colony-forming abilities. Further examination showed upregulation of p53 expression in these cells, which could be inhibiting their proliferation. When a p53 inhibitor was included in the culture medium, it was confirmed to be responsible for the inhibition of proliferation in mGSCs. Optimal concentration of the inhibitor in the culture of these cells was 5 µM. Furthermore, addition of the p53 inhibitor increased the expression of the markers of self-renewal and cell cycle in goat mGSCs. In summary, suppressing p53 is beneficial for the proliferation of dairy goat mGSCs, cultured on PL.
Collapse
Affiliation(s)
- Haijing Zhu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China.,Shaanxi Province Engineering and Technology Research Center of Cashmere Goat, Research Center of Life Science in Yulin University, Yulin, China
| | - Liming Zheng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Long Wang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Furong Tang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Ahmed H Arisha
- Department of physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Hongchao Zhou
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
37
|
Chang WF, Xu J, Lin TY, Hsu J, Hsieh-Li HM, Hwu YM, Liu JL, Lu CH, Sung LY. Survival Motor Neuron Protein Participates in Mouse Germ Cell Development and Spermatogonium Maintenance. Int J Mol Sci 2020; 21:ijms21030794. [PMID: 31991812 PMCID: PMC7037566 DOI: 10.3390/ijms21030794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/13/2022] Open
Abstract
The defective human survival motor neuron 1 (SMN1) gene leads to spinal muscular atrophy (SMA), the most common genetic cause of infant mortality. We previously reported that loss of SMN results in rapid differentiation of Drosophila germline stem cells and mouse embryonic stem cells (ESCs), indicating that SMN also plays important roles in germ cell development and stem cell biology. Here, we show that in healthy mice, SMN is highly expressed in the gonadal tissues, prepubertal spermatogonia, and adult spermatocytes, whereas low SMN expression is found in differentiated spermatid and sperm. In SMA-like mice, the growth of testis tissues is retarded, accompanied with gamete development abnormalities and loss of the spermatogonia-specific marker. Consistently, knockdown of Smn1 in spermatogonial stem cells (SSCs) leads to a compromised regeneration capacity in vitro and in vivo in transplantation experiments. In SMA-like mice, apoptosis and accumulation of the R-loop structure were significantly elevated, indicating that SMN plays a critical role in the survival of male germ cells. The present work demonstrates that SMN, in addition to its critical roles in neuronal development, participates in mouse germ cell and spermatogonium maintenance.
Collapse
Affiliation(s)
- Wei-Fang Chang
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan; (W.-F.C.); (T.-Y.L.); (J.H.)
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA;
| | - Tzu-Ying Lin
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan; (W.-F.C.); (T.-Y.L.); (J.H.)
| | - Jing Hsu
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan; (W.-F.C.); (T.-Y.L.); (J.H.)
| | - Hsiu-Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan;
| | - Yuh-Ming Hwu
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei 10449, Taiwan;
- Department of Obstetrics and Gynecology, Mackay Medical College, New Taipei City 252, Taiwan
- Department of Obstetrics and Gynecology, Mackay Junior College of Medicine, Nursing, and Management, Taipei 11260, Taiwan
| | - Ji-Long Liu
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 2JD, UK;
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Chung-Hao Lu
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei 10449, Taiwan;
- Correspondence: (C.-H.L.); (L.-Y.S.)
| | - Li-Ying Sung
- Institute of Biotechnology, National Taiwan University, Taipei 106, Taiwan; (W.-F.C.); (T.-Y.L.); (J.H.)
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
- Animal Resource Center, National Taiwan University, Taipei 106, Taiwan
- Correspondence: (C.-H.L.); (L.-Y.S.)
| |
Collapse
|
38
|
Wu X, Yang Y, Zhong C, Guo Y, Li S, Lin H, Liu X. Transcriptome profiling of laser-captured germ cells and functional characterization of zbtb40 during 17alpha-methyltestosterone-induced spermatogenesis in orange-spotted grouper (Epinephelus coioides). BMC Genomics 2020; 21:73. [PMID: 31973692 PMCID: PMC6979330 DOI: 10.1186/s12864-020-6477-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Spermatogenesis is an intricate process regulated by a finely organized network. The orange-spotted grouper (Epinephelus coioides) is a protogynous hermaphroditic fish, but the regulatory mechanism of its spermatogenesis is not well-understood. In the present study, transcriptome sequencing of the male germ cells isolated from orange-spotted grouper was performed to explore the molecular mechanism underlying spermatogenesis. RESULTS In this study, the orange-spotted grouper was induced to change sex from female to male by 17alpha-methyltestosterone (MT) implantation. During the spermatogenesis, male germ cells (spermatogonia, spermatocytes, spermatids, and spermatozoa) were isolated by laser capture microdissection. Transcriptomic analysis for the isolated cells was performed. A total of 244,984,338 clean reads were generated from four cDNA libraries. Real-time PCR results of 13 genes related to sex differentiation and hormone metabolism indicated that transcriptome data are reliable. RNA-seq data showed that the female-related genes and genes involved in hormone metabolism were highly expressed in spermatogonia and spermatozoa, suggesting that these genes participate in the spermatogenesis. Interestingly, the expression of zbtb family genes showed significantly changes in the RNA-seq data, and their expression patterns were further examined during spermatogenesis. The analysis of cellular localization of Eczbtb40 and the co-localization of Eczbtb40 and Eccyp17a1 in different gonadal stages suggested that Eczbtb40 might interact with Eccyp17a1 during spermatogenesis. CONCLUSIONS Our study, for the first time, investigated the transcriptome of the male germ cells from orange-spotted grouper, and identified functional genes, GO terms, and KEGG pathways involved in spermatogenesis. Furthermore, Eczbtb40 was first characterized and its role during spermatogenesis was predicted. These data will contribute to future studies on the molecular mechanism of spermatogenesis in teleosts.
Collapse
Affiliation(s)
- Xi Wu
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
| | - Yang Yang
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
| | - Chaoyue Zhong
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
| | - Yin Guo
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
| | - Shuisheng Li
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
- Southern Laboratory of Ocean Science and Engineering, Zhuhai, 519000 People’s Republic of China
| |
Collapse
|
39
|
Hermann BP, Cheng K, Singh A, Roa-De La Cruz L, Mutoji KN, Chen IC, Gildersleeve H, Lehle JD, Mayo M, Westernströer B, Law NC, Oatley MJ, Velte EK, Niedenberger BA, Fritze D, Silber S, Geyer CB, Oatley JM, McCarrey JR. The Mammalian Spermatogenesis Single-Cell Transcriptome, from Spermatogonial Stem Cells to Spermatids. Cell Rep 2019; 25:1650-1667.e8. [PMID: 30404016 PMCID: PMC6384825 DOI: 10.1016/j.celrep.2018.10.026] [Citation(s) in RCA: 342] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 08/15/2018] [Accepted: 10/03/2018] [Indexed: 12/16/2022] Open
Abstract
Spermatogenesis is a complex and dynamic cellular differentiation process critical to male reproduction and sustained by spermatogonial stem cells (SSCs). Although patterns of gene expression have been described for aggregates of certain spermatogenic cell types, the full continuum of gene expression patterns underlying ongoing spermatogenesis in steady state was previously unclear. Here, we catalog single-cell transcriptomes for >62,000 individual spermatogenic cells from immature (postnatal day 6) and adult male mice and adult men. This allowed us to resolve SSC and progenitor spermatogonia, elucidate the full range of gene expression changes during male meiosis and spermiogenesis, and derive unique gene expression signatures for multiple mouse and human spermatogenic cell types and/or subtypes. These transcriptome datasets provide an information-rich resource for studies of SSCs, male meiosis, testicular cancer, male infertility, or contraceptive development, as well as a gene expression roadmap to be emulated in efforts to achieve spermatogenesis in vitro.
Collapse
Affiliation(s)
- Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA; Genomics Core, University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Keren Cheng
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Anukriti Singh
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Lorena Roa-De La Cruz
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Kazadi N Mutoji
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - I-Chung Chen
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Heidi Gildersleeve
- Genomics Core, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Jake D Lehle
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Max Mayo
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Birgit Westernströer
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Nathan C Law
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA
| | - Melissa J Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA
| | - Ellen K Velte
- Department of Anatomy & Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Bryan A Niedenberger
- Department of Anatomy & Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Danielle Fritze
- The UT Transplant Center, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Sherman Silber
- The Infertility Center of St. Louis, Chesterfield, MO 63017, USA
| | - Christopher B Geyer
- Department of Anatomy & Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Jon M Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA.
| |
Collapse
|
40
|
Qi L, Li J, Le W, Zhang J. Low-dose ionizing irradiation triggers apoptosis of undifferentiated spermatogonia in vivo and in vitro. Transl Androl Urol 2019; 8:591-600. [PMID: 32038955 DOI: 10.21037/tau.2019.10.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background The present study aimed to investigate the mechanism of low-dose ionizing radiation (IR) induced apoptosis of undifferentiated spermatogonia in vivo and in vitro. Methods Following 50 mGy IR, testicular tissues were collected from the adult DBA/2 mice at 1, 2 and 24 h; mice in the control group received pseudo-irradiation. Immunofluorescence (IF) staining and TUNEL were performed to assess DNA damage and apoptosis, respectively, in the irradiated testicular tissues. Furthermore, the spermatogonia were also irradiated in vitro, and the expression of apoptosis-related proteins was detected by Western blotting. TUNEL and flow cytometry were applied to assess cell apoptosis. Results γH2AX (a marker of DNA damage) was up-regulated in the seminiferous tubules at 1 and 2 h after IR, but it was reduced following the DNA repair. This was consistent with the finding that apoptosis of germline cells was present in the seminiferous tubules after IR, especially at 1 h (IF and TUNEL). Apoptosis was also present in the PLZF(+) spermatogonia, particularly at 1 h after IR. Apoptotic cells decreased with the increase in DNA repair time after IR. Moreover, the caspase-3 protein was expressed in the undifferentiated spermatogonia following IR. The expression of caspase-3, P53, Ku70 and DNA-PKcs in the cultured spermatogonia was also up-regulated following IR in vitro, but their expression decreased gradually over time after IR, which was supported by the findings from flow cytometry, and the apoptosis of spermatogonia peaked at 24 h post IR. Conclusions IR may induce the apoptosis of spermatogonia at early stage in vivo, but the apoptosis of spermatogonia secondary to IR occurs at a relatively later time point (24 h) in vitro mainly. The apoptosis of spermatogonia is improved over time after IR.
Collapse
Affiliation(s)
- Lixin Qi
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jiaxuan Li
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Wei Le
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jinfu Zhang
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200050, China
| |
Collapse
|
41
|
Wang Z, Xu X, Li JL, Palmer C, Maric D, Dean J. Sertoli cell-only phenotype and scRNA-seq define PRAMEF12 as a factor essential for spermatogenesis in mice. Nat Commun 2019; 10:5196. [PMID: 31729367 PMCID: PMC6858368 DOI: 10.1038/s41467-019-13193-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/22/2019] [Indexed: 01/14/2023] Open
Abstract
Spermatogonial stem cells (SSCs) have the dual capacity to self-renew and differentiate into progenitor spermatogonia that develop into mature spermatozoa. Here, we document that preferentially expressed antigen of melanoma family member 12 (PRAMEF12) plays a key role in maintenance of the spermatogenic lineage. In male mice, genetic ablation of Pramef12 arrests spermatogenesis and results in sterility which can be rescued by transgenic expression of Pramef12. Pramef12 deficiency globally decreases expression of spermatogenic-related genes, and single-cell transcriptional analysis of post-natal male germline cells identifies four spermatogonial states. In the absence of Pramef12 expression, there are fewer spermatogonial stem cells which exhibit lower expression of SSC maintenance-related genes and are defective in their ability to differentiate. The disruption of the first wave of spermatogenesis in juvenile mice results in agametic seminiferous tubules. These observations mimic a Sertoli cell-only syndrome in humans and may have translational implications for reproductive medicine.
Collapse
Affiliation(s)
- Zhengpin Wang
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Xiaojiang Xu
- Integrative Bioinformatics, NIEHS, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics, NIEHS, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Cameron Palmer
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dragan Maric
- NINDS Flow Cytometry Core Facility, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
42
|
La HM, Hobbs RM. Mechanisms regulating mammalian spermatogenesis and fertility recovery following germ cell depletion. Cell Mol Life Sci 2019; 76:4071-4102. [PMID: 31254043 PMCID: PMC11105665 DOI: 10.1007/s00018-019-03201-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/07/2019] [Accepted: 06/19/2019] [Indexed: 12/19/2022]
Abstract
Mammalian spermatogenesis is a highly complex multi-step process sustained by a population of mitotic germ cells with self-renewal potential known as spermatogonial stem cells (SSCs). The maintenance and regulation of SSC function are strictly dependent on a supportive niche that is composed of multiple cell types. A detailed appreciation of the molecular mechanisms underpinning SSC activity and fate is of fundamental importance for spermatogenesis and male fertility. However, different models of SSC identity and spermatogonial hierarchy have been proposed and recent studies indicate that cell populations supporting steady-state germline maintenance and regeneration following damage are distinct. Importantly, dynamic changes in niche properties may underlie the fate plasticity of spermatogonia evident during testis regeneration. While formation of spermatogenic colonies in germ-cell-depleted testis upon transplantation is a standard assay for SSCs, differentiation-primed spermatogonial fractions have transplantation potential and this assay provides readout of regenerative rather than steady-state stem cell capacity. The characterisation of spermatogonial populations with regenerative capacity is essential for the development of clinical applications aimed at restoring fertility in individuals following germline depletion by genotoxic treatments. This review will discuss regulatory mechanisms of SSCs in homeostatic and regenerative testis and the conservation of these mechanisms between rodent models and man.
Collapse
Affiliation(s)
- Hue M La
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Robin M Hobbs
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, 3800, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
43
|
Differential Regulation of TLE3 in Sertoli Cells of the Testes during Postnatal Development. Cells 2019; 8:cells8101156. [PMID: 31569653 PMCID: PMC6848928 DOI: 10.3390/cells8101156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/31/2022] Open
Abstract
Spermatogenesis is a process by which haploid cells differentiate from germ cells in the seminiferous tubules of the testes. TLE3, a transcriptional co-regulator that interacts with DNA-binding factors, plays a role in the development of somatic cells. However, no studies have shown its role during germ cell development in the testes. Here, we examined TLE3 expression in the testes during spermatogenesis. TLE3 was highly expressed in mouse testes and was dynamically regulated in different cell types of the seminiferous tubules, spermatogonia, spermatids, and Sertoli cells, but not in the spermatocytes. Interestingly, TLE3 was not detected in Sertoli cells on postnatal day 7 (P7) but was expressed from P10 onward. The microarray analysis showed that the expression of numerous genes changed upon TLE3 knockdown in a Sertoli cell line TM4. These include 1597 up-regulated genes and 1452 down-regulated genes in TLE3-knockdown TM4 cells. Ingenuity Pathway Analysis (IPA) showed that three factors were up-regulated and two genes were down-regulated upon TLE3 knockdown in TM4 cells. The abnormal expression of the three factors is associated with cellular malfunctions such as abnormal differentiation and Sertoli cell formation. Thus, TLE3 is differentially expressed in Sertoli cells and plays a crucial role in regulating cell-specific genes involved in the differentiation and formation of Sertoli cells during testicular development.
Collapse
|
44
|
Song W, Shi X, Xia Q, Yuan M, Liu J, Hao K, Qian Y, Zhao X, Zou K. PLZF suppresses differentiation of mouse spermatogonial progenitor cells via binding of differentiation associated genes. J Cell Physiol 2019; 235:3033-3042. [PMID: 31541472 DOI: 10.1002/jcp.29208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/03/2019] [Indexed: 11/06/2022]
Abstract
Promyelocytic leukaemia zinc finger (PLZF) is a key factor in inhibiting differentiation of spermatogonial progenitor cells (SPCs), but the underlying mechanisms are still largely unknown. In this study, the regulation of PLZF on Kit, Stra8, Sohlh2, and Dmrt1 (SPCs differentiation related genes) was investigated. We found some PLZF potential binding sites existed in the promoters of Kit, Stra8, Sohlh2, and Dmrt1. Additionally, the expressions of KIT, STRA8, SOHLH2, and DMRT1 were upregulated when PLZF was knockdown in SPCs. Furthermore, chromatin immunoprecipitation quantitative polymerase chain reaction revealed PLZF directly bound to the promoters of Kit, Stra8, Sohlh2, and Dmrt1. Besides, dual luciferase assay verified PLZF repressed those gene expressions. Collectively, our finding indicate that PLZF binds to the promoter regions of Kit, Stra8, Sohlh2, and Dmrt1 to regulate SPCs differentiation, which facilitate us to further understand the regulatory mechanism of PLZF in SPCs fates.
Collapse
Affiliation(s)
- Weixiang Song
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xinglong Shi
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Xia
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Min Yuan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jiaxi Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kunying Hao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yinjuan Qian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xiaodong Zhao
- Shanghai Center for Systems Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Kang Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
45
|
Koubi M, Poplineau M, Vernerey J, N'Guyen L, Tiberi G, Garciaz S, El-Kaoutari A, Maqbool MA, Andrau JC, Guillouf C, Saurin AJ, Duprez E. Regulation of the positive transcriptional effect of PLZF through a non-canonical EZH2 activity. Nucleic Acids Res 2019; 46:3339-3350. [PMID: 29425303 PMCID: PMC5909434 DOI: 10.1093/nar/gky080] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 11/13/2022] Open
Abstract
The transcription factor PLZF (promyelocytic leukemia zinc finger protein) acts as an epigenetic regulator balancing self-renewal and differentiation of hematopoietic cells through binding to various chromatin-modifying factors. First described as a transcriptional repressor, PLZF is also associated with active transcription, although the molecular bases underlying the differences are unknown. Here, we reveal that in a hematopoietic cell line, PLZF is predominantly associated with transcribed genes. Additionally, we identify a new association between PLZF and the histone methyltransferase, EZH2 at the genomic level. We find that co-occupancy of PLZF and EZH2 on chromatin at PLZF target genes is not associated with SUZ12 or trimethylated lysine 27 of histone H3 (H3K27me3) but with the active histone mark H3K4me3 and active transcription. Removal of EZH2 leads to an increase of PLZF binding and increased gene expression. Our results suggest a new role of EZH2 in restricting PLZF positive transcriptional activity independently of its canonical PRC2 activity.
Collapse
Affiliation(s)
- Myriam Koubi
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Mathilde Poplineau
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Julien Vernerey
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Lia N'Guyen
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Guillaume Tiberi
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Sylvain Garciaz
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Abdessamad El-Kaoutari
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| | - Muhammad A Maqbool
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, Cedex 5, France
| | - Jean-Christophe Andrau
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, 34293 Montpellier, Cedex 5, France
| | - Christel Guillouf
- Gustave Roussy, Université Paris-Saclay, Inserm U1170, CNRS Villejuif, France
| | - Andrew J Saurin
- Aix Marseille Université, CNRS, IBDM, UMR 7288, 13288 Marseille, Cedex 9, France
| | - Estelle Duprez
- Epigenetic Factors in Normal and Malignant Hematopoiesis, Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, 13273 Marseille Cedex 9, France
| |
Collapse
|
46
|
Clotaire DZJ, Wei Y, Yu X, Ousman T, Hua J. Functions of promyelocytic leukaemia zinc finger (Plzf) in male germline stem cell development and differentiation. Reprod Fertil Dev 2019; 31:1315-1320. [PMID: 31009592 DOI: 10.1071/rd18252] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 02/16/2019] [Indexed: 01/12/2023] Open
Abstract
Promyelocytic leukaemia zinc finger (Plzf), also known as zinc finger and BTB domain containing 16 (ZBTB16) or zinc-finger protein 145 (ZFP145), is a critical zinc finger protein of male germline stem cells (mGSCs). Multiple lines of evidence indicate that Plzf has a central role in the development, differentiation and maintenance of many stem cells, including mGSCs, and Plzf has been validated as an essential transcription factor for mammalian testis development and spermatogenesis. This review summarises current literature focusing on the significance of Plzf in maintaining and regulating self-renewal and differentiation of mGSCs, especially goat mGSCs. The review summarises evidence of the specificity of Plzf expression in germ cell development stage, the known functions of Plzf and the microRNA-mediated mechanisms that control Plzf expression in mGSCs.
Collapse
Affiliation(s)
- Daguia Zambe John Clotaire
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China; and Laboratoire des sciences Agronomiques et Biologiques pour le Développement (LASBAD), Faculty of Science, University of Bangui, Bangui, 999111, Central Africa
| | - Yudong Wei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiuwei Yu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Tamgue Ousman
- Department of Biochemistry, University of Douala, Douala, 999108, Cameroon
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China; and Corresponding author
| |
Collapse
|
47
|
DDX5 plays essential transcriptional and post-transcriptional roles in the maintenance and function of spermatogonia. Nat Commun 2019; 10:2278. [PMID: 31123254 PMCID: PMC6533336 DOI: 10.1038/s41467-019-09972-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/02/2019] [Indexed: 02/07/2023] Open
Abstract
Mammalian spermatogenesis is sustained by mitotic germ cells with self-renewal potential known as undifferentiated spermatogonia. Maintenance of undifferentiated spermatogonia and spermatogenesis is dependent on tightly co-ordinated transcriptional and post-transcriptional mechanisms. The RNA helicase DDX5 is expressed by spermatogonia but roles in spermatogenesis are unexplored. Using an inducible knockout mouse model, we characterise an essential role for DDX5 in spermatogonial maintenance and show that Ddx5 is indispensable for male fertility. We demonstrate that DDX5 regulates appropriate splicing of key genes necessary for spermatogenesis. Moreover, DDX5 regulates expression of cell cycle genes in undifferentiated spermatogonia post-transcriptionally and is required for cell proliferation and survival. DDX5 can also act as a transcriptional co-activator and we demonstrate that DDX5 interacts with PLZF, a transcription factor required for germline maintenance, to co-regulate select target genes. Combined, our data reveal a critical multifunctional role for DDX5 in regulating gene expression programmes and activity of undifferentiated spermatogonia. Sustained sperm production is dependent on activity of undifferentiated spermatogonia. Here, the authors demonstrate an essential role for RNA helicase DDX5 in maintenance of spermatogonia in adults through control of gene transcription plus RNA processing and export.
Collapse
|
48
|
Grive KJ, Hu Y, Shu E, Grimson A, Elemento O, Grenier JK, Cohen PE. Dynamic transcriptome profiles within spermatogonial and spermatocyte populations during postnatal testis maturation revealed by single-cell sequencing. PLoS Genet 2019; 15:e1007810. [PMID: 30893341 PMCID: PMC6443194 DOI: 10.1371/journal.pgen.1007810] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 04/01/2019] [Accepted: 02/18/2019] [Indexed: 12/22/2022] Open
Abstract
Spermatogenesis is the process by which male gametes are formed from a self-renewing population of spermatogonial stem cells (SSCs) residing in the testis. SSCs represent less than 1% of the total testicular cell population in adults, but must achieve a stable balance between self-renewal and differentiation. Once differentiation has occurred, the newly formed and highly proliferative spermatogonia must then enter the meiotic program in which DNA content is doubled, then halved twice to create haploid gametes. While much is known about the critical cellular processes that take place during the specialized cell division that is meiosis, much less is known about how the spermatocytes in the "first-wave" in juveniles compare to those that contribute to long-term, "steady-state" spermatogenesis in adults. Given the strictly-defined developmental process of spermatogenesis, this study explored the transcriptional profiles of developmental cell stages during testis maturation. Using a combination of comprehensive germ cell sampling with high-resolution, single-cell-mRNA-sequencing, we have generated a reference dataset of germ cell gene expression. We show that discrete developmental stages of spermatogenesis possess significant differences in the transcriptional profiles from neonates compared to juveniles and adults. Importantly, these gene expression dynamics are also reflected at the protein level in their respective cell types. We also show differential utilization of many biological pathways with age in both spermatogonia and spermatocytes, demonstrating significantly different underlying gene regulatory programs in these cell types over the course of testis development and spermatogenic waves. This dataset represents the first unbiased sampling of spermatogonia and spermatocytes during testis maturation, at high-resolution, single-cell depth. Not only does this analysis reveal previously unknown transcriptional dynamics of a highly transitional cell population, it has also begun to reveal critical differences in biological pathway utilization in developing spermatogonia and spermatocytes, including response to DNA damage and double-strand breaks.
Collapse
Affiliation(s)
- Kathryn J. Grive
- Center for Reproductive Genomics, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| | - Yang Hu
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States of America
| | - Eileen Shu
- Center for Reproductive Genomics, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| | - Andrew Grimson
- Center for Reproductive Genomics, Cornell University, Ithaca, NY, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States of America
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States of America
| | - Jennifer K. Grenier
- Center for Reproductive Genomics, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| | - Paula E. Cohen
- Center for Reproductive Genomics, Cornell University, Ithaca, NY, United States of America
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States of America
| |
Collapse
|
49
|
Large-scale genome-wide meta-analysis of polycystic ovary syndrome suggests shared genetic architecture for different diagnosis criteria. PLoS Genet 2018; 14:e1007813. [PMID: 30566500 PMCID: PMC6300389 DOI: 10.1371/journal.pgen.1007813] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/06/2018] [Indexed: 11/19/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a disorder characterized by hyperandrogenism, ovulatory dysfunction and polycystic ovarian morphology. Affected women frequently have metabolic disturbances including insulin resistance and dysregulation of glucose homeostasis. PCOS is diagnosed with two different sets of diagnostic criteria, resulting in a phenotypic spectrum of PCOS cases. The genetic similarities between cases diagnosed based on the two criteria have been largely unknown. Previous studies in Chinese and European subjects have identified 16 loci associated with risk of PCOS. We report a fixed-effect, inverse-weighted-variance meta-analysis from 10,074 PCOS cases and 103,164 controls of European ancestry and characterisation of PCOS related traits. We identified 3 novel loci (near PLGRKT, ZBTB16 and MAPRE1), and provide replication of 11 previously reported loci. Only one locus differed significantly in its association by diagnostic criteria; otherwise the genetic architecture was similar between PCOS diagnosed by self-report and PCOS diagnosed by NIH or non-NIH Rotterdam criteria across common variants at 13 loci. Identified variants were associated with hyperandrogenism, gonadotropin regulation and testosterone levels in affected women. Linkage disequilibrium score regression analysis revealed genetic correlations with obesity, fasting insulin, type 2 diabetes, lipid levels and coronary artery disease, indicating shared genetic architecture between metabolic traits and PCOS. Mendelian randomization analyses suggested variants associated with body mass index, fasting insulin, menopause timing, depression and male-pattern balding play a causal role in PCOS. The data thus demonstrate 3 novel loci associated with PCOS and similar genetic architecture for all diagnostic criteria. The data also provide the first genetic evidence for a male phenotype for PCOS and a causal link to depression, a previously hypothesized comorbid disease. Thus, the genetics provide a comprehensive view of PCOS that encompasses multiple diagnostic criteria, gender, reproductive potential and mental health. We performed an international meta-analysis of genome-wide association studies combining over 10,000,000 genetic markers in more than 10,000 European women with polycystic ovary syndrome (PCOS) and 100,000 controls. We found three new risk variants associated with PCOS. Our data demonstrate that the genetic architecture does not differ based on the diagnostic criteria used for PCOS. We also demonstrate a genetic pathway shared with male pattern baldness, representing the first evidence for shared disease biology in men, and shared genetics with depression, previously postulated based only on observational studies.
Collapse
|
50
|
PAK1 Promotes the Proliferation and Inhibits Apoptosis of Human Spermatogonial Stem Cells via PDK1/KDR/ZNF367 and ERK1/2 and AKT Pathways. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:769-786. [PMID: 30141410 PMCID: PMC6111072 DOI: 10.1016/j.omtn.2018.06.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 01/15/2023]
Abstract
Spermatogonial stem cells (SSCs) have significant applications in reproductive and regenerative medicine. However, nothing is known about genes in mediating human SSCs. Here we have explored for the first time the function and mechanism of P21-activated kinase 1 (PAK1) in regulating the proliferation and apoptosis of the human SSC line. PAK1 level was upregulated by epidermal growth factor (EGF), but not glial cell line-derived neurotrophic factor (GDNF) or fibroblast growth factor 2 (FGF2). PAK1 promoted proliferation and DNA synthesis of the human SSC line, whereas PAK1 suppressed its apoptosis in vitro and in vivo. RNA sequencing identified that PDK1, ZNF367, and KDR levels were downregulated by PAK1 knockdown. Immunoprecipitation and Western blots demonstrated that PAK1 interacted with PDK1. PDK1 and KDR levels were decreased by ZNF367-small interfering RNAs (siRNAs). The proliferation of the human SSC line was reduced by PDK1-, KDR-, and ZNF367-siRNAs, whereas its apoptosis was enhanced by these siRNAs. The levels of phos-ERK1/2, phos-AKT, and cyclin A were decreased by PAK1-siRNAs. Tissue arrays showed that PAK1 level was low in non-obstructive azoospermia patients. Collectively, PAK1 was identified as the first molecule that controls proliferation and apoptosis of the human SSC line through PDK1/KDR/ZNF367 and the ERK1/2 and AKT pathways. This study provides data on novel gene regulation and networks underlying the fate of human SSCs, and it offers new molecular targets for human SSCs in translational medicine.
Collapse
|