1
|
Song Y, Duan Y, Luo H, Yun L, Zhang M, Tran NT, Zheng H, Zhou Q, Li S. Establishment of mud crab (Scylla paramamosain) spermatogonial stem cell line: A potential tool for immunological research. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110349. [PMID: 40254085 DOI: 10.1016/j.fsi.2025.110349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Spermatogonial stem cells (SSCs) can differentiate into sperm and are important for studying on genetic information transmission of animals. However, the establishment of the SSC line in crustaceans is still in its infancy. This study aimed to establish a method for the isolation, culture, and identification of SSCs derived from the gonad of a marine crustacean (mud crab, Scylla paramamosain), and evaluate their differentiation ability and potential application in immunological research, in vitro. SSCs showed robust growth, proliferation, and passaging ability (up to 35 passages) in germ cell culture medium. Proteomic analysis showed that the protein expression profile of SSC was closely related to the gonadal tissue. SSCs were found to be able to express male-specific and pluripotent markers, such as CD9, PIWI, DDX4, DAZL, NANOG, SOX2, and EPHA1. Furthermore, SSCs were differentiated into osteoblasts and adipocytes under in vitro induction. Green fluorescent protein (GFP), packaged by lentivirus, was able to be overexpressed in SSCs after infection. In addition, the infection of white spot syndrome virus (WSSV) simulated the expression of inflammation-associated factors, including TRAF6, TNF-α, MyD88, Dorsal, and Relish, and apoptosis-related genes (BAX and Bcl2) in SSCs. Thus, SSCs were initially isolated and characterized from mud crabs for the first time. Our results proved that SSCs can be used in reproduction technology, germplasm conservation, and immunological studies in crustaceans.
Collapse
Affiliation(s)
- Ying Song
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Yanchuang Duan
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Haiqing Luo
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Linying Yun
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Ming Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Ngoc Tuan Tran
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Huaiping Zheng
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China
| | - Qicun Zhou
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Shengkang Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, 515063, China; Institute of Marine Sciences, Shantou University, Shantou, 515063, China.
| |
Collapse
|
2
|
Du L, Cui Y, Chen W, Li C, He Z. Micro-proteomics reveals distinct protein profiles and SPARC/FGF2/CDH1 regulation of human Sertoli cells between Sertoli cell-only syndrome and normal men. Cell Mol Life Sci 2025; 82:146. [PMID: 40192810 PMCID: PMC11977051 DOI: 10.1007/s00018-025-05678-w] [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: 11/08/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
Sertoli cell-only syndrome (SCOS) is one of the most severe non-obstructive azoospermia (NOA) types, since only Sertoli cells with not any male germ cells exist with the seminiferous tubules. As such, it is of particular significance to elucidate molecular mechanisms underlying SCOS for improving the diagnosis and treatment strategies for this disease. Due to the difficulties in obtaining sufficient human testicular tissues and the limited availability of human cells, the traditional proteomics is inadequate for comparing the differences in large scale of protein expression patterns of human Sertoli cells between SCOS and normal men. To solve this issue on the requirement of large amount of cell numbers, we employed micro-proteomics to reveal distinct global protein expression profiles of human Sertoli cells between SCOS and obstructive azoospermia (OA) with normal spermatogenesis utilizing single human Sertoli cells. We found a significant downregulation of proteins involved in cell adhesion pathways in SCOS Sertoli cells, whereas proteins related to apoptosis were markedly upregulated. Interestingly, we identified the lower expression of SPARC (secreted protein acidic and rich in cysteine) and the higher expression of FGF2 (fibroblast growth factor 2) in human Sertoli cells of the SCOS compared to normal men. SPARC silencing led to upregulation of FGF2 in human Sertoli cells, and SPARC may be associated with the occurrence of SCOS and serves as a reliable marker for the diagnosis of this disease. This study thus comprehensively offers the proteomic landscape of human Sertoli cells in the testes of SCOS patients and it sheds a novel insight into the pathogenesis of SCOS.
Collapse
Affiliation(s)
- Li Du
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Wei Chen
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Chunyun Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, China.
- Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, 571199, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Wang L, Ying Y, Li N, Song Y, Zhao L, Sun H, Wang Z, Liu XH, Wang D. Transcriptome analysis of the hypothalamus and testes in Brandt's Vole: new insights into mechanisms of photoperiodic plasticity in postnatal testicular development. Funct Integr Genomics 2025; 25:55. [PMID: 40053130 DOI: 10.1007/s10142-025-01562-9] [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: 12/19/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 05/13/2025]
Abstract
Postnatal gonadal development is regulated by photoperiod via the hypothalamus, especially in seasonal breeding small rodents. However, the precise molecular mechanisms remain unclear. In this study, we conducted a comparative analysis of the transcriptomes of the hypothalamus and testes in 10-week-old male Brandt's voles born under long (LP, 16L:8D) and short photoperiod (SP, 8L:16D) conditions. Results indicate that the SP group exhibited significantly smaller testes with spermatogenesis halted before meiosis, identifying 129 differentially expressed genes (DEGs) in the hypothalamus and 21,673 DEGs in the testes. In the hypothalamus, genes involved in the thyroid hormone and retinoic acid (RA) pathway were notably altered under SP conditions, including decreased Tshb and Cga expression, increased Dio3, and reduced Crabp1 and Lrat, highlighting their key roles in SP signaling. In the testes, downregulated genes were significantly enriched in male reproduction-related GO terms and metabolic KEGG pathways, such as steroid hormone biosynthesis and retinol metabolism. Key genes for testosterone synthesis (e.g. Star, Cyp11a1) and RA synthesis (e.g. Rdh10, Rdh11) were downregulated, while those linked to RA degradation (Cyp26b1) and undifferentiated spermatogonia maintenance (e.g. Gdnf, Gfra1) were upregulated. These findings outline a molecular microenvironment that favors the preservation of undifferentiated spermatogonia over their differentiation from the hypothalamus to the testes. This study firstly provides valuable insights into the transcriptomic basis of SP-inhibited testicular development in Brandt's voles.
Collapse
Affiliation(s)
- Lewen Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yaqi Ying
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Ning Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Ying Song
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Lijuan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hong Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zhenlong Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiao-Hui Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Dawei Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
- Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji, 831100, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
4
|
Pryzhkova MV, Skinner MW, Candelaria JI, Wellard SR, Jordan PW. The use of deidentified organ donor testes for research. Andrology 2025. [PMID: 39912553 DOI: 10.1111/andr.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
Our knowledge of testis development and function mainly comes from research using mammalian model organisms, primarily the mouse. However, there are integral differences between men and other mammalian species regarding cellular composition and expression profiles during fetal and post-natal testis development and in the mature testis. Therefore, to specifically learn more about human testis development and function, there is a need to use human testis tissue for research. Human testicular tissues that have been donated for research have allowed extensive molecular and cytological assessments, as well as single-cell transcriptome and epigenome analyses. These tissues have also been used for the development of cell technologies and in vitro models that aim to improve infertility treatments and diagnostics. Biopsied material taken from patients and designated for research is usually very small in size and is unsuitable for comprehensive studies. On the other hand, research using whole testes obtained from deceased, deidentified donors has become a valuable resource to assess conservation between humans and other organisms and identify human-specific phenomena. This review discusses the acquisition of donated deidentified human testes and their use for basic science research.
Collapse
Affiliation(s)
- Marina V Pryzhkova
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Marnie W Skinner
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Juliana I Candelaria
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Stephen R Wellard
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Philip W Jordan
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
5
|
Heidari B, Shirazi A, Akbari N, Barzegar-Amini M. Identification and Manipulation of Spermatogonial Stem Cells with the Aim of Inducing Spermatogenesis in Vitro. Reprod Sci 2025; 32:278-288. [PMID: 39424678 DOI: 10.1007/s43032-024-01709-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
Assisted reproduction techniques for infertile men with non-obstructive azoospermia require a sufficient number of functional germ cells produced in vitro. Understanding the mechanisms that allow the resumption of spermatogenesis outside the testicular environment is crucial for fertility preservation in these patients. A review of the literature was conducted using databases such as PubMed, Scopus and Web of Science, with keywords including "spermatogonial stem cell," "germ cells," "male factor infertility," and "enrichment and propagation of SSCs in vitro." Currently, two models-"in vivo" and "in vitro"-have been developed for producing haploid germ cells. The "in vivo" models include spermatogonial stem cell transplantation and testicular xenograft techniques. In contrast, the "in vitro" models consist of conventional culture systems, organ culture, and three-dimensional culture systems, all designed to induce spermatogenesis in vitro. These culture systems enable the simulation of the seminiferous epithelium in vitro, which facilitates better regulation of cell-signaling pathways that control the self-renewal and differentiation of SSCs. This review provides up-to-date information on the organization of SSCs, focusing on the identification, proliferation, and differentiation of spermatogonia in vitro.
Collapse
Affiliation(s)
- Banafsheh Heidari
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.
| | - Abolfazl Shirazi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Nazanin Akbari
- Department of Biology, Shahid Beheshti University, Tehran, Iran
- Clinical Research Development Unit, Faculty of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maral Barzegar-Amini
- Clinical Research Development Unit, Faculty of Medicine, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Rehder P, Packeiser EM, Körber H, Goericke-Pesch S. Altered Sertoli Cell Function Contributes to Spermatogenic Arrest in Dogs with Chronic Asymptomatic Orchitis. Int J Mol Sci 2025; 26:1108. [PMID: 39940876 PMCID: PMC11817828 DOI: 10.3390/ijms26031108] [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: 12/17/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Acquired infertility due to chronic asymptomatic orchitis (CAO) is a common finding in male dogs. It is characterized by spermatogenic arrest, a significant reduction in spermatogonia, immune cell infiltration and a disruption of the blood-testis barrier. Sertoli cells are a key factor for spermatogenesis and the testicular micromilieu. We hypothesize altered Sertoli cell function to be involved in the pathogenesis of canine CAO. Consequently, the aim was to gain further insights into the spermatogonial stem cell niche and Sertoli cell function in CAO-affected dogs. Therefore, the testicular expression of the Sertoli cell-derived factors bFGF, GDNF, WNT5A, BMP4, CXCL12 and LDHC were evaluated in 15 CAO testis tissues and 10 normospermic controls by relative quantitative real-time PCR (qPCR). Additionally, the protein expression patterns of bFGF, GDNF and WNT5A were visualized immunohistochemically (IHC). This study revealed an overexpression of bFGF (IHC, p < 0.0001), GDNF (qPCR, p = 0.0036), WNT5A (IHC, p = 0.0066) and CXCL12 (qPCR, p = 0.0003) and a reduction in BMP4 (qPCR, p = 0.0041) and LDHC (qPCR, p = 0.0003) in CAO-affected testis in dogs, clearly confirming impaired Sertoli cell function in canine CAO. Sertoli cell function is essential for spermatogenesis and must be considered for potential therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Sandra Goericke-Pesch
- Reproductive Unit—Clinic for Small Animals, University of Veterinary Medicine Hannover, Foundation, 30559 Hannover, Germany; (P.R.); (E.-M.P.); (H.K.)
| |
Collapse
|
7
|
Sarkar H, Lee E, Lopez-Darwin SL, Kang Y. Deciphering normal and cancer stem cell niches by spatial transcriptomics: opportunities and challenges. Genes Dev 2025; 39:64-85. [PMID: 39496456 PMCID: PMC11789490 DOI: 10.1101/gad.351956.124] [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] [Indexed: 11/06/2024]
Abstract
Cancer stem cells (CSCs) often exhibit stem-like attributes that depend on an intricate stemness-promoting cellular ecosystem within their niche. The interplay between CSCs and their niche has been implicated in tumor heterogeneity and therapeutic resistance. Normal stem cells (NSCs) and CSCs share stemness features and common microenvironmental components, displaying significant phenotypic and functional plasticity. Investigating these properties across diverse organs during normal development and tumorigenesis is of paramount research interest and translational potential. Advancements in next-generation sequencing (NGS), single-cell transcriptomics, and spatial transcriptomics have ushered in a new era in cancer research, providing high-resolution and comprehensive molecular maps of diseased tissues. Various spatial technologies, with their unique ability to measure the location and molecular profile of a cell within tissue, have enabled studies on intratumoral architecture and cellular cross-talk within the specific niches. Moreover, delineation of spatial patterns for niche-specific properties such as hypoxia, glucose deprivation, and other microenvironmental remodeling are revealed through multilevel spatial sequencing. This tremendous progress in technology has also been paired with the advent of computational tools to mitigate technology-specific bottlenecks. Here we discuss how different spatial technologies are used to identify NSCs and CSCs, as well as their associated niches. Additionally, by exploring related public data sets, we review the current challenges in characterizing such niches, which are often hindered by technological limitations, and the computational solutions used to address them.
Collapse
Affiliation(s)
- Hirak Sarkar
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, New Jersey 08544, USA
- Department of Computer Science, Princeton, New Jersey 08544, USA
| | - Eunmi Lee
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Sereno L Lopez-Darwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA;
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, New Jersey 08544, USA
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
| |
Collapse
|
8
|
Tomizawa SI, Kuroha K, Ono M, Nakajima K, Ohbo K. A behind-the-scenes role of BDNF in the survival and differentiation of spermatogonia. Asian J Androl 2025; 27:37-43. [PMID: 39177410 PMCID: PMC11784946 DOI: 10.4103/aja202457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 05/17/2024] [Indexed: 08/24/2024] Open
Abstract
ABSTRACT Mouse spermatogenesis entails the maintenance and self-renewal of spermatogonial stem cells (SSCs), which require a complex web-like signaling network transduced by various cytokines. Although brain-derived neurotrophic factor (BDNF) is expressed in Sertoli cells in the testis, and its receptor tropomyosin receptor kinase B (TrkB) is expressed in the spermatogonial population containing SSCs, potential functions of BDNF for spermatogenesis have not been uncovered. Here, we generate BDNF conditional knockout mice and find that BDNF is dispensable for in vivo spermatogenesis and fertility. However, in vitro , we reveal that BDNF -deficient germline stem cells (GSCs) exhibit growth potential not only in the absence of glial cell line-derived neurotrophic factor (GDNF), a master regulator for GSC proliferation, but also in the absence of other factors, including epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and insulin. GSCs grown without these factors are prone to differentiation, yet they maintain expression of promyelocytic leukemia zinc finger ( Plzf ), an undifferentiated spermatogonial marker. Inhibition of phosphoinositide 3-kinase (PI3K), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), and Src pathways all interfere with the growth of BDNF-deficient GSCs. Thus, our findings suggest a role for BDNF in maintaining the undifferentiated state of spermatogonia, particularly in situations where there is a shortage of growth factors.
Collapse
Affiliation(s)
- Shin-ichi Tomizawa
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Kazushige Kuroha
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Michio Ono
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Kuniko Nakajima
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Kazuyuki Ohbo
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
9
|
Kim SH, Shin SH, Kim SM, Jung SE, Shin BJ, Ahn JS, Lim KT, Kim DH, Lee K, Ryu BY. Bisphenol Analogs Downregulate the Self-Renewal Potential of Spermatogonial Stem Cells. World J Mens Health 2025; 43:154-165. [PMID: 38606862 PMCID: PMC11704178 DOI: 10.5534/wjmh.230166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/10/2023] [Accepted: 12/28/2023] [Indexed: 04/13/2024] Open
Abstract
PURPOSE In this study, we investigated the effect of bisphenol-A (BPA) and its major analogs, bisphenol-F (BPF), and bisphenol-S (BPS), on spermatogonial stem cells (SSCs) populations using in vitro SSC culture and in vivo transplantation models. MATERIALS AND METHODS SSCs enriched from 6- to 8-day-old C57BL/6-eGFP⁺ male mice testes were treated with varying concentrations of bisphenols for 7 days to examine bisphenol-derived cytotoxicity and changes in SSC characteristics. We utilized flow cytometry, immunocytochemistry, real-time quantitative reverse transcription-PCR, and western blot analysis. The functional alteration of SSCs was further investigated by examining donor SSC-derived spermatogenesis evaluation through in vivo transplantation and subsequent testis analysis. RESULTS BPF exhibited a similar inhibitory effect on SSCs as BPA, demonstrating a significant decrease in SSC survival, inhibition of proliferation, and induction of apoptosis. On the other hand, while BPS was comparatively weaker than BPA and BPF, it still showed significant SSC cytotoxicity. Importantly, SSCs exposed to BPA, BPF, and BPS exhibited a significant reduction in donor SSC-derived germ cell colonies per total number of cultured cells, indicating that, like BPA, BPF, and BPS can induce a comparable reduction in functional SSCs in the recipient animals. However, the progress of spermatogenesis, as evidenced by histochemistry and the expressions of PCNA and SSC specific markers, collectively indicates that BPA, BPF, and BPS may not adversely affect the spermatogenesis. CONCLUSIONS Our findings indicate that the major BPA substitutes, BPF and BPS, have significant cytotoxic effects on SSCs, similar to BPA. These effects may lead to a reduction in the functional self-renewal stem cell population and potential impacts on male fertility.
Collapse
Affiliation(s)
- Seo-Hee Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Korea
| | - Seung Hee Shin
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Korea
| | - Seok-Man Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Korea
| | - Sang-Eun Jung
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Korea
| | - Beom-Jin Shin
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Korea
| | - Jin Seop Ahn
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Korea
| | - Kyoung Taek Lim
- Department of Urology, Maria Fertility Hospital, Seoul, Korea
| | - Dong-Hwan Kim
- Department of Animal Sciences, The Ohio State University, Columbus, OH, USA
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH, USA
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Korea.
| |
Collapse
|
10
|
Zheng T, Fok EKL. The Biology and Regulation of Spermatogonial Stem Cells in the Niche. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:333-354. [PMID: 40301263 DOI: 10.1007/978-3-031-82990-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Spermatogenesis, the process responsible for the daily production of millions of sperm, originates from spermatogonial stem cells (SSCs). Dysregulation of spermatogenesis is a major contributing factor to male infertility. Additionally, cryopreservation of SSCs followed by transplantation is a viable approach to restore spermatogenesis after sterilizing treatments such as chemotherapy and radiotherapy for cancer treatment. Therefore, investigating the biology and regulatory mechanisms involved in maintaining SSCs will provide valuable insights into the etiology of male fertility disorders and inform clinical strategies for fertility preservation and restoration. In this chapter, we will review the origin of SSCs, their biological and functional properties, and the various types of cells that contribute to the SSC niche. Additionally, we will discuss the regulation of SSC self-renewal and differentiation by niche factors, cell-cell and cell-extracellular matrix interactions, intrinsic gene regulation, and emerging intercellular communication mechanisms.
Collapse
Affiliation(s)
- Tingting Zheng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Ellis Kin Lam Fok
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China.
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR, China.
- Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Chengdu, PR, China.
| |
Collapse
|
11
|
Li SY, Kumar S, Gu X, DeFalco T. Testicular immunity. Mol Aspects Med 2024; 100:101323. [PMID: 39591799 PMCID: PMC11624985 DOI: 10.1016/j.mam.2024.101323] [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: 04/02/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024]
Abstract
The testis is a unique environment where immune responses are suppressed to allow the development of sperm that possess autoimmunogenic antigens. There are several contributors responsible for testicular immune privilege, including the blood-testis barrier, testicular immune cells, immunomodulation by Sertoli cells, and high levels of steroid hormones. Despite multiple mechanisms in place to regulate the testicular immune environment, pathogens that disrupt testicular immunity can lead to long-term effects such as infertility. If testicular immunity is disturbed, autoimmune reactions can also occur, leading to aberrant immune cell infiltration and subsequent attack of autoimmunogenic germ cells. Here we discuss cellular and molecular factors underlying testicular immunity and how testicular infection or autoimmunity compromise immune privilege. We also describe infections and autoimmune diseases that impact the testis. Further research into testicular immunity will reveal how male fertility is maintained and will help update therapeutic strategies for infertility and other testicular disorders.
Collapse
Affiliation(s)
- Shu-Yun Li
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sudeep Kumar
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
12
|
Zhang Z, Miao J, Wang H, Ali I, Nguyen D, Chen W, Wang Y. Accelerated mitochondrial dynamics promote spermatogonial differentiation. Stem Cell Reports 2024; 19:1548-1563. [PMID: 39393359 PMCID: PMC11589200 DOI: 10.1016/j.stemcr.2024.09.006] [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: 03/23/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024] Open
Abstract
At different stages of spermatogenesis, germ cell mitochondria differ remarkably in morphology, architecture, and functions. However, it remains elusive how mitochondria change their features during spermatogonial differentiation, which in turn impacts spermatogonial stem cell fate decision. In this study, we observed that mitochondrial fusion and fission were both upregulated during spermatogonial differentiation. As a result, the mitochondrial morphology remained unaltered. Enhanced mitochondrial fusion and fission promoted spermatogonial differentiation, while the deficiency in DRP1-mediated fission led to a stage-specific blockage of spermatogenesis at differentiating spermatogonia. Our data further revealed that increased expression of pro-fusion factor MFN1 upregulated mitochondrial metabolism, whereas DRP1 specifically regulated mitochondrial permeability transition pore opening in differentiating spermatogonia. Taken together, our findings unveil how proper spermatogonial differentiation is precisely controlled by concurrently accelerated and properly balanced mitochondrial fusion and fission in a germ cell stage-specific manner, thereby providing critical insights about mitochondrial contribution to stem cell fate decision.
Collapse
Affiliation(s)
- Zhaoran Zhang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Junru Miao
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Hanben Wang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Izza Ali
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Duong Nguyen
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Wei Chen
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Yuan Wang
- Department of Animal Sciences, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
13
|
Bhardwaj JK, Siwach A, Sachdeva D, Sachdeva SN. Revisiting cadmium-induced toxicity in the male reproductive system: an update. Arch Toxicol 2024; 98:3619-3639. [PMID: 39317800 DOI: 10.1007/s00204-024-03871-7] [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: 08/01/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Heavy metals like cadmium (Cd) are one of the main environmental pollutants, with no biological role in the human body. Cd has been well-documented to have disastrous effects on both plants and animals. It is known to accumulate in kidneys, lungs, liver, and testes and is thought to affect these organs' function over time, which is linked to a very long biological half-life and a very poor rate of elimination. According to recent researches, the testes are extremely vulnerable to cadmium. The disruption of the blood-testis barrier, seminiferous tubules, Sertoli cells, and Leydig cells caused by cadmium leads to the loss of sperm through various mechanisms, such as oxidative stress, spermatogenic cell death, testicular swelling, dysfunction in androgen-producing cells, interference with gene regulation, disruption of ionic homeostasis, and damage to the vascular endothelium. Additionally, through epigenetic control, cadmium disrupts the function of germ cells and somatic cells, resulting in infertile or subfertile males. A full grasp of the mechanisms underlying testicular toxicity caused by Cd is very important to develop suitable strategies to ameliorate male fertility. Therefore, this review article outlines cadmium's impact on growth and functions of the testicles, reviews therapeutic approaches and protective mechanisms, considers recent research findings, and identifies future research directions.
Collapse
Affiliation(s)
- Jitender Kumar Bhardwaj
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, 136119, India.
| | - Anshu Siwach
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Drishty Sachdeva
- Reproductive Physiology Laboratory, Department of Zoology, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Som Nath Sachdeva
- Department of Civil Engineering, National Institute of Technology, Kurukshetra and Kurukshetra University, Kurukshetra, Haryana, India
| |
Collapse
|
14
|
Damyanova KB, Nixon B, Johnston SD, Gambini A, Benitez PP, Lord T. Spermatogonial stem cell technologies: applications from human medicine to wildlife conservation†. Biol Reprod 2024; 111:757-779. [PMID: 38993049 PMCID: PMC11473898 DOI: 10.1093/biolre/ioae109] [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: 05/01/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024] Open
Abstract
Spermatogonial stem cell (SSC) technologies that are currently under clinical development to reverse human infertility hold the potential to be adapted and applied for the conservation of endangered and vulnerable wildlife species. The biobanking of testis tissue containing SSCs from wildlife species, aligned with that occurring in pediatric human patients, could facilitate strategies to improve the genetic diversity and fitness of endangered populations. Approaches to utilize these SSCs could include spermatogonial transplantation or testis tissue grafting into a donor animal of the same or a closely related species, or in vitro spermatogenesis paired with assisted reproduction approaches. The primary roadblock to progress in this field is a lack of fundamental knowledge of SSC biology in non-model species. Herein, we review the current understanding of molecular mechanisms controlling SSC function in laboratory rodents and humans, and given our particular interest in the conservation of Australian marsupials, use a subset of these species as a case-study to demonstrate gaps-in-knowledge that are common to wildlife. Additionally, we review progress in the development and application of SSC technologies in fertility clinics and consider the translation potential of these techniques for species conservation pipelines.
Collapse
Affiliation(s)
- Katerina B Damyanova
- Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Brett Nixon
- Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Stephen D Johnston
- School of Environment, The University of Queensland, Gatton, QLD 4343, Australia
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Andrés Gambini
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
- School of Agriculture and Food Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Patricio P Benitez
- School of Agriculture and Food Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Tessa Lord
- Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
15
|
Lord T, Oatley JM. Spermatogenic Stem Cells: Core Biology, Defining Features, and Utilities. Mol Reprod Dev 2024; 91:e23777. [PMID: 39392153 DOI: 10.1002/mrd.23777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
The actions of spermatogenic stem cells (SSCs) provide the foundation for continual spermatogenesis and regeneration of the cognate lineage following cytotoxic insult or transplantation. Several decades of research with rodent models have yielded knowledge about the core biology, morphological features, and molecular profiles of mammalian SSCs. Translation of these discoveries to utilities for human fertility preservation, improving animal agriculture, and wildlife conservation are actively being pursued. Here, we provide overviews of these aspects covering both historical and current states of understanding.
Collapse
Affiliation(s)
- Tessa Lord
- Discipline of Biological Sciences, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales, Australia
- Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jon M Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
16
|
Li J, Zhang SX, Wang DL, Qi JJ, Bai CY, Sun H, Sun BX, Liang S. Thyroxine regulates pig Sertoli cell line proliferation and maturation through the IKK/NFκB and p38 MAPK signaling pathways. Theriogenology 2024; 227:1-8. [PMID: 38981313 DOI: 10.1016/j.theriogenology.2024.06.031] [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: 11/20/2023] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
The aim of this study was to investigate the signaling pathways involved in the proliferation and differentiation of pig Sertoli cells (SCs) mediated by thyroid hormone (T3) to provide a theoretical and practical basis for enhancing pig semen production. The effects of different concentrations of T3 on the proliferation of pig SCs were evaluated using the CCK8 assay. The impact of T3 on the proliferation and differentiation of pig SCs was further examined using RNA-seq, qPCR, and Western Blotting techniques. Additionally, the involvement of the p38 MAPK and NFκB pathways in mediating the effects of T3 on SCs proliferation and differentiation was investigated. Our findings revealed a strong correlation between the dosage of T3 and the inhibition of pig SCs proliferation and promotion of maturation. T3 regulated the activation state of the NFκB signaling pathway by upregulating IKKα, downregulating IKKβ, and promoting IκB phosphorylation. Furthermore, T3 facilitated SCs maturation by upregulating AR and FSHR expression while downregulating KRT-18. In conclusion, T3 inhibits pig SCs proliferation and promote pig SCs maturation through the IKK/NFκB and p38 MAPK pathways. These findings provide valuable insights into the mechanisms by which T3 influences the proliferation and maturation of pig SCs.
Collapse
Affiliation(s)
- Jing Li
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China
| | - Shao-Xuan Zhang
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China
| | - Da-Li Wang
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China
| | - Jia-Jia Qi
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China
| | - Chun-Yan Bai
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China
| | - Hao Sun
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China
| | - Bo-Xing Sun
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China.
| | - Shuang Liang
- Department of Animals Sciences, College of Animal Sciences, Jilin University, Changchun, China.
| |
Collapse
|
17
|
Liu W, Du L, Li J, He Y, Tang M. Microenvironment of spermatogonial stem cells: a key factor in the regulation of spermatogenesis. Stem Cell Res Ther 2024; 15:294. [PMID: 39256786 PMCID: PMC11389459 DOI: 10.1186/s13287-024-03893-z] [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: 05/10/2024] [Accepted: 08/25/2024] [Indexed: 09/12/2024] Open
Abstract
Spermatogonial stem cells (SSCs) play a crucial role in the male reproductive system, responsible for maintaining continuous spermatogenesis. The microenvironment or niche of SSCs is a key factor in regulating their self-renewal, differentiation and spermatogenesis. This microenvironment consists of multiple cell types, extracellular matrix, growth factors, hormones and other molecular signals that interact to form a complex regulatory network. This review aims to provide an overview of the main components of the SSCs microenvironment, explore how they regulate the fate decisions of SSCs, and discuss the potential impact of microenvironmental abnormalities on male reproductive health.
Collapse
Affiliation(s)
- Wei Liu
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Li Du
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Junjun Li
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China
| | - Yan He
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China.
| | - Mengjie Tang
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China.
| |
Collapse
|
18
|
Wang Y, Hu S, Han C. A Simple and Efficient Procedure for Developing Mouse Germline Stem Cell Lines with Gene Knock-in via CRISPR/Cas9 Technology. Curr Protoc 2024; 4:e70002. [PMID: 39264143 DOI: 10.1002/cpz1.70002] [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] [Indexed: 09/13/2024]
Abstract
Cultured mammalian spermatogonial stem cells (SSCs), also known as germline stem cells (GSCs), hold great promise for applications such as fertility preservation, gene therapy, and animal breeding, particularly in conjunction with accurate gene editing. Although the in vitro development of mouse GSC (mGSC) lines, and gene-targeting procedures for such lines, were initially established about two decades ago, it remains challenging for beginners to efficiently accomplish these tasks, partly because mGSCs proliferate more slowly and are more resistant to lipid-mediated gene transfection than pluripotent stem cells (PSCs). Meanwhile, methods for mGSC culture and gene editing have been evolving constantly to become simpler and more efficient. Here, we describe how to develop mGSC lines from small mouse testis samples and how to carry out gene knock-in in these cells using CRISPR/Cas9 technology, detailing three basic protocols that constitute a streamlined procedure. Using these simple and efficient procedures, site-specific knock-in mGSC lines can be obtained in 3 months. We hope that these protocols will help researchers use genetically modified GSCs to explore scientific questions of interest and to accumulate experience for application to GSC research in other mammalian species. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Establishment of mouse GSCs lines from small testicular samples Basic Protocol 2: Preparation of plasmids for gene knock-in using the CRISPR/Cas9 system Basic Protocol 3: Establishment of gene knock-in mGSC lines by electroporation gene delivery.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Shuaitao Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Chunsheng Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
19
|
Zhang YW, Wu SX, Wang GW, Wan RD, Yang QE. Single-cell analysis identifies critical regulators of spermatogonial development and differentiation in cattle-yak bulls. J Dairy Sci 2024; 107:7317-7336. [PMID: 38642661 DOI: 10.3168/jds.2023-24442] [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: 11/22/2023] [Accepted: 03/11/2024] [Indexed: 04/22/2024]
Abstract
Spermatogenesis is a continuous process in which functional sperm are produced through a series of mitotic and meiotic divisions and morphological changes in germ cells. The aberrant development and fate transitions of spermatogenic cells cause hybrid sterility in mammals. Cattle-yak, a hybrid animal between taurine cattle (Bos taurus) and yak (Bos grunniens), exhibits male-specific sterility due to spermatogenic failure. In the present study, we performed single-cell RNA sequencing analysis to identify differences in testicular cell composition and the developmental trajectory of spermatogenic cells between yak and cattle-yak. The composition and molecular signatures of spermatogonial subtypes were dramatically different between these 2 animals, and the expression of genes associated with stem cell maintenance, cell differentiation and meiotic entry was altered in cattle-yak, indicating the impairment of undifferentiated spermatogonial fate decisions. Cell communication analysis revealed that signaling within different spermatogenic cell subpopulations was weakened, and progenitor spermatogonia were unable to or delayed receiving and sending signals for transformation to the next stage in cattle-yak. Simultaneously, the communication between niche cells and germ cells was also abnormal. Collectively, we obtained the expression profiles of transcriptome signatures of different germ cells and testicular somatic cell populations at the single-cell level and identified critical regulators of spermatogonial differentiation and meiosis in yak and sterile cattle-yak. The findings of this study shed light on the genetic mechanisms that lead to hybrid sterility and speciation in bovid species.
Collapse
Affiliation(s)
- Yi-Wen Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shi-Xin Wu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guo-Wen Wang
- Qinghai Academy of Animal Husbandry and Veterinary Sciences, Xining, Qinghai 810016, China
| | - Rui-Dong Wan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi-En Yang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China; Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810001, China.
| |
Collapse
|
20
|
Ibtisham F, Tang S, Song Y, Wanze W, Xiao M, Honaramooz A, An L. Optimal isolation, culture, and in vitro propagation of spermatogonial stem cells in Huaixiang chicken. Reprod Domest Anim 2024; 59:e14661. [PMID: 38979950 DOI: 10.1111/rda.14661] [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: 03/25/2024] [Revised: 05/15/2024] [Accepted: 06/18/2024] [Indexed: 07/10/2024]
Abstract
Spermatogonial stem cells (SSCs) comprise the foundation of spermatogenesis and hence have great potential for fertility preservation of rare or endangered species and the development of transgenic animals and birds. Yet, developing optimal conditions for the isolation, culture, and maintenance of SSCs in vitro remains challenging, especially for chicken. The objectives of this study were to (1) find the optimal age for SSC isolation in Huaixiang chicken, (2) develop efficient protocols for the isolation, (3) enrichment, and (4) culture of isolated SSCs. In the present study, we first compared the efficiency of SSC isolation using 11 different age groups (8-79 days of age) of Huaixiang chicken. We found that the testes of 21-day-old chicken yielded the highest cell viability. Next, we compared two different enzymatic combinations for isolating SSCs and found that 0.125% trypsin and 0.02 g/L EDTA supported the highest number and viability of SSCs. This was followed by investigating optimal conditions for the enrichment of SSCs, where we observed that differential plating had the highest enrichment efficiency compared to the Percoll gradient and magnetic-activated cell sorting methods. Lastly, to find the optimal culture conditions of SSCs, we compared adding different concentrations of foetal bovine serum (FBS; 2%, 5%, 7%, and 10%) and different concentrations of GDNF, bFGF, or LIF (5, 10, 20, or 30 ng/mL). We found that a combination of 2% FBS and individual growth factors, including GDNF (20 ng/mL), bFGF (30 ng/mL), or LIF (5 ng/mL), best supported the proliferation and colony formation of SSCs. In conclusion, SSCs can be optimally isolated through enzymatic digestion from testes of 21-day-old chicken, followed by enrichment using differential plating. Furthermore, adding 2% FBS and optimized concentrations of GFNF, bFGF, or LIF in the culture promotes the proliferation of chicken SSCs.
Collapse
Affiliation(s)
- Fahar Ibtisham
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Shuyan Tang
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yiping Song
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Wang Wanze
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Mei Xiao
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Ali Honaramooz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Lilong An
- Agricultural College, Guangdong Ocean University, Zhanjiang, Guangdong, China
| |
Collapse
|
21
|
Kanatsu-Shinohara M, Yamamoto T, Morimoto H, Liu T, Shinohara T. Spermatogonial stem cells in the 129 inbred strain exhibit unique requirements for self-renewal. Development 2024; 151:dev202553. [PMID: 38934417 DOI: 10.1242/dev.202553] [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: 11/20/2023] [Accepted: 05/19/2024] [Indexed: 06/28/2024]
Abstract
Spermatogonial stem cells (SSCs) undergo self-renewal division to sustain spermatogenesis. Although it is possible to derive SSC cultures in most mouse strains, SSCs from a 129 background never proliferate under the same culture conditions, suggesting they have distinct self-renewal requirements. Here, we established long-term culture conditions for SSCs from mice of the 129 background (129 mice). An analysis of 129 testes showed significant reduction of GDNF and CXCL12, whereas FGF2, INHBA and INHBB were higher than in testes of C57BL/6 mice. An analysis of undifferentiated spermatogonia in 129 mice showed higher expression of Chrna4, which encodes an acetylcholine (Ach) receptor component. By supplementing medium with INHBA and Ach, SSC cultures were derived from 129 mice. Following lentivirus transduction for marking donor cells, transplanted cells re-initiated spermatogenesis in infertile mouse testes and produced transgenic offspring. These results suggest that the requirements of SSC self-renewal in mice are diverse, which has important implications for understanding self-renewal mechanisms in various animal species.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo 100-0004, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroko Morimoto
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Tianjiao Liu
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
22
|
Chen WB, Zhang MF, Yang F, Hua JL. Applications of single-cell RNA sequencing in spermatogenesis and molecular evolution. Zool Res 2024; 45:575-585. [PMID: 38766742 PMCID: PMC11188606 DOI: 10.24272/j.issn.2095-8137.2024.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/08/2024] [Indexed: 05/22/2024] Open
Abstract
Spermatogenic cell heterogeneity is determined by the complex process of spermatogenesis differentiation. However, effectively revealing the regulatory mechanisms underlying mammalian spermatogenic cell development and differentiation via traditional methods is difficult. Advances in technology have led to the emergence of many single-cell transcriptome sequencing protocols, which have partially addressed these challenges. In this review, we detail the principles of 10x Genomics technology and summarize the methods for downstream analysis of single-cell transcriptome sequencing data. Furthermore, we explore the role of single-cell transcriptome sequencing in revealing the heterogeneity of testicular ecological niche cells, delineating the establishment and disruption of testicular immune homeostasis during human spermatogenesis, investigating abnormal spermatogenesis in humans, and, ultimately, elucidating the molecular evolution of mammalian spermatogenesis.
Collapse
Affiliation(s)
- Wen-Bo Chen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Meng-Fei Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Fan Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
- Key Laboratory of Livestock Biology, Northwest A & F University, Yangling, Shaanxi 712100, China
| | - Jin-Lian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A & F University, Yangling, Shaanxi 712100, China
- Key Laboratory of Livestock Biology, Northwest A & F University, Yangling, Shaanxi 712100, China. E-mail:
| |
Collapse
|
23
|
Gao J, Xu Z, Song W, Huang J, Liu W, He Z, He L. USP11 regulates proliferation and apoptosis of human spermatogonial stem cells via HOXC5-mediated canonical WNT/β-catenin signaling pathway. Cell Mol Life Sci 2024; 81:211. [PMID: 38722330 PMCID: PMC11082041 DOI: 10.1007/s00018-024-05248-6] [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: 01/02/2024] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 05/12/2024]
Abstract
Spermatogonial stem cells (SSCs) are capable of transmitting genetic information to the next generations and they are the initial cells for spermatogenesis. Nevertheless, it remains largely unknown about key genes and signaling pathways that regulate fate determinations of human SSCs and male infertility. In this study, we explored the expression, function, and mechanism of USP11 in controlling the proliferation and apoptosis of human SSCs as well as the association between its abnormality and azoospermia. We found that USP11 was predominantly expressed in human SSCs as shown by database analysis and immunohistochemistry. USP11 silencing led to decreases in proliferation and DNA synthesis and an enhancement in apoptosis of human SSCs. RNA-sequencing identified HOXC5 as a target of USP11 in human SSCs. Double immunofluorescence, Co-immunoprecipitation (Co-IP), and molecular docking demonstrated an interaction between USP11 and HOXC5 in human SSCs. HOXC5 knockdown suppressed the growth of human SSCs and increased apoptosis via the classical WNT/β-catenin pathway. In contrast, HOXC5 overexpression reversed the effect of proliferation and apoptosis induced by USP11 silencing. Significantly, lower levels of USP11 expression were observed in the testicular tissues of patients with spermatogenic disorders. Collectively, these results implicate that USP11 regulates the fate decisions of human SSCs through the HOXC5/WNT/β-catenin pathway. This study thus provides novel insights into understanding molecular mechanisms underlying human spermatogenesis and the etiology of azoospermia and it offers new targets for gene therapy of male infertility.
Collapse
Affiliation(s)
- Jun Gao
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhipeng Xu
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Weijie Song
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiwei Huang
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University, Changsha, Hunan, 410013, China.
| | - Leye He
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
24
|
Yang L, Liao J, Huang H, Lee TL, Qi H. Stage-specific regulation of undifferentiated spermatogonia by AKT1S1-mediated AKT-mTORC1 signaling during mouse spermatogenesis. Dev Biol 2024; 509:11-27. [PMID: 38311163 DOI: 10.1016/j.ydbio.2024.02.002] [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: 06/29/2023] [Revised: 11/03/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Undifferentiated spermatogonia are composed of a heterogeneous cell population including spermatogonial stem cells (SSCs). Molecular mechanisms underlying the regulation of various spermatogonial cohorts during their self-renewal and differentiation are largely unclear. Here we show that AKT1S1, an AKT substrate and inhibitor of mTORC1, regulates the homeostasis of undifferentiated spermatogonia. Although deletion of Akt1s1 in mouse appears not grossly affecting steady-state spermatogenesis and male mice are fertile, the subset of differentiation-primed OCT4+ spermatogonia decreased significantly, whereas self-renewing GFRα1+ and proliferating PLZF+ spermatogonia were sustained. Both neonatal prospermatogonia and the first wave spermatogenesis were greatly reduced in Akt1s1-/- mice. Further analyses suggest that OCT4+ spermatogonia in Akt1s1-/- mice possess altered PI3K/AKT-mTORC1 signaling, gene expression and carbohydrate metabolism, leading to their functionally compromised developmental potential. Collectively, these results revealed an important role of AKT1S1 in mediating the stage-specific signals that regulate the self-renewal and differentiation of spermatogonia during mouse spermatogenesis.
Collapse
Affiliation(s)
- Lele Yang
- Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jinyue Liao
- GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Hongying Huang
- The Experimental Animal Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Tin Lap Lee
- GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Huayu Qi
- Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
25
|
Zhang Y, Li X, Gao L, Dong X, Xue J, Zhao M, Xie J, Niyaz A, Ren L, Zhou X. The role of Sertoli cells-secreted factors in different stages of germ cells development in mice exposed to BDE-209. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123775. [PMID: 38503350 DOI: 10.1016/j.envpol.2024.123775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 03/10/2024] [Indexed: 03/21/2024]
Abstract
Decabromodiphenyl ether (BDE-209), a frequently used brominated flame retardant, readily enters the environment and is difficult to degrade with bioaccumulation. BDE-209 could cause male reproductive toxicity, but the regulatory functions of Sertoli cells-secreted factors remain uncertain. In present study, male mice were treated with 75 mg/kg BDE-209 and then stopped exposure for 50 days. Exogenous Glial cell line-derived neurotrophic factor (GDNF), a Sertoli cell-secreted factor, was injected into testes of mice treated with BDE-209 for 50 days to explore the role of GDNF in BDE-209-induced reproductive toxicity. The mouse spermatogonia cell line GC-1 spg was used in vitro to further verify regulatory effects of Sertoli cells-secreted factors on meiotic initiation. The results showed that BDE-209 inhibited expressions of the self-renewal pathway GFRα-1/RAS/ERK1/2 in spermatogonial stem cells (SSCs), and reduced expressions of spermatogonia proliferation-related pathway NRG3/ERBB4 and meiosis initiation factor Stra8. Furthermore, BDE-209 decreased the levels of both GDNF and retinoic acid (RA) secreted by Sertoli cells in testes. Importantly, the alterations of above indicators induced by BDE-209 did not recover after 50-day recovery period. After exogenous GDNF injection, the decreased expression of GFRα-1/RAS/ERK in SSCs was reversed. However, the level of RA and expressions of NRG3/ERBB4/Stra8 were not restored. The in vitro experimental results showed that exogenous RA reversed the reductions in NRG3/ERBB4/Stra8 and ameliorated inhibition of GC-1 spg cells proliferation induced by BDE-209. These results suggested that Sertoli cells-secreted factors play roles in regulating various stages of germ cell development. Specifically, BDE-209 affected the self-renewal of SSCs by decreasing GDNF secretion resulting in the inhibition of GFRα-1/RAS/ERK pathway; BDE-209 hindered the proliferation of spermatogonia and initiation of meiosis by inhibiting the secretion of RA and preventing RA from binding to RARα, resulting in the suppression of NRG3/ERBB4/Stra8 pathway. As a consequence, spermatogenesis was compromised, leading to persistent male reproductive toxicity.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xiangyang Li
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Leqiang Gao
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xiaomin Dong
- Experimental Center for Basic Medical Teaching, Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jinglong Xue
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Moxuan Zhao
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Junhong Xie
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Aliekram Niyaz
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Lihua Ren
- School of Nursing, Peking University, Beijing, 100191, China
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
26
|
Tian H, Wang X, Li X, Song W, Mi J, Zou K. Regulation of spermatogonial stem cell differentiation by Sertoli cells-derived exosomes through paracrine and autocrine signaling. J Cell Physiol 2024; 239:e31202. [PMID: 38291718 DOI: 10.1002/jcp.31202] [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: 10/07/2023] [Revised: 12/28/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
In the orchestrated environment of the testicular niche, the equilibrium between self-renewal and differentiation of spermatogonial stem cells (SSCs) is meticulously maintained, ensuring a stable stem cell reserve and robust spermatogenesis. Within this milieu, extracellular vesicles, specifically exosomes, have emerged as critical conveyors of intercellular communication. Despite their recognized significance, the implications of testicular exosomes in modulating SSC fate remain incompletely characterized. Given the fundamental support and regulatory influence of Sertoli cells (SCs) on SSCs, we were compelled to explore the role of SC-derived exosomes (SC-EXOs) in the SSC-testicular niche. Our investigation hinged on the hypothesis that SC-EXOs, secreted by SCs from the testes of 5-day-old mice-a developmental juncture marking the onset of SSC differentiation-participate in the regulation of this process. We discovered that exposure to SC-EXOs resulted in an upsurge of PLZF, MVH, and STRA8 expression in SSC cultures, concomitant with a diminution of ID4 and GFRA1 levels. Intriguingly, obstructing exosomal communication in a SC-SSC coculture system with the exosome inhibitor GW4869 attenuated SSC differentiation, suggesting that SC-EXOs may modulate this process via paracrine signaling. Further scrutiny revealed the presence of miR-493-5p within SC-EXOs, which suppresses Gdnf mRNA in SCs to indirectly restrain SSC differentiation through the modulation of GDNF expression-an indication of autocrine regulation. Collectively, our findings illuminate the complex regulatory schema by which SC-EXOs affect SSC differentiation, offering novel perspectives and laying the groundwork for future preclinical and clinical investigations.
Collapse
Affiliation(s)
- Hairui Tian
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Xingju Wang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Xiaoxiao Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Weixiang Song
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Jiaqi Mi
- Department of Cancer Biology, Cancer Center and Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
27
|
Hu M, Yeh YH, Maezawa S, Nakagawa T, Yoshida S, Namekawa S. PRC1 directs PRC2-H3K27me3 deposition to shield adult spermatogonial stem cells from differentiation. Nucleic Acids Res 2024; 52:2306-2322. [PMID: 38142439 PMCID: PMC10954450 DOI: 10.1093/nar/gkad1203] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/16/2023] [Accepted: 12/11/2023] [Indexed: 12/26/2023] Open
Abstract
Spermatogonial stem cells functionality reside in the slow-cycling and heterogeneous undifferentiated spermatogonia cell population. This pool of cells supports lifelong fertility in adult males by balancing self-renewal and differentiation to produce haploid gametes. However, the molecular mechanisms underpinning long-term stemness of undifferentiated spermatogonia during adulthood remain unclear. Here, we discover that an epigenetic regulator, Polycomb repressive complex 1 (PRC1), shields adult undifferentiated spermatogonia from differentiation, maintains slow cycling, and directs commitment to differentiation during steady-state spermatogenesis in adults. We show that PRC2-mediated H3K27me3 is an epigenetic hallmark of adult undifferentiated spermatogonia. Indeed, spermatogonial differentiation is accompanied by a global loss of H3K27me3. Disruption of PRC1 impairs global H3K27me3 deposition, leading to precocious spermatogonial differentiation. Therefore, PRC1 directs PRC2-H3K27me3 deposition to maintain the self-renewing state of undifferentiated spermatogonia. Importantly, in contrast to its role in other tissue stem cells, PRC1 negatively regulates the cell cycle to maintain slow cycling of undifferentiated spermatogonia. Our findings have implications for how epigenetic regulators can be tuned to regulate the stem cell potential, cell cycle and differentiation to ensure lifelong fertility in adult males.
Collapse
Affiliation(s)
- Mengwen Hu
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA 95616, USA
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yu-Han Yeh
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA 95616, USA
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - So Maezawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Faculty of Science and Technology, Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba 281-8510, Japan
| | - Toshinori Nakagawa
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
- Course for Basic Biology, The Graduate Institute for Advanced Studies, SOKENDAI, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
- Course for Basic Biology, The Graduate Institute for Advanced Studies, SOKENDAI, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Satoshi H Namekawa
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA 95616, USA
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
28
|
Ding X, Li L, Gao J, Yi D, Schimenti JC. Scalable and Efficient Generation of Mouse Primordial Germ Cell-like Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580543. [PMID: 38405756 PMCID: PMC10888945 DOI: 10.1101/2024.02.15.580543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Primordial germ cells (PGCs) are the founder cells of the germline. The ability to generate PGC-like cells (PGCLCs) from pluripotent stem cells has advanced our knowledge of gametogenesis and holds promise for developing infertility treatments. However, generating an ample supply of PGCLCs for demanding applications such as high-throughput genetic screens has been a limitation. Here, we demonstrated that simultaneous overexpressing 4 transcriptional factors - Nanog and three PGC master regulators Prdm1, Prdm14 and Tfap2c - in suspended mouse epiblast like cells (EpiLCs) and formative embryonic stem cells (ESCs) results in efficient and cost-effective production of PGCLCs. The overexpression of Nanog enhances the PGC regulatory network and suppresses differentiation of somatic lineages, enabling a significant improvement in the efficiency of PGCLC production. Transcriptomic analysis reveals that differentiated PGCLCs exhibit similarities to in vivo PGCs and are more advanced compared to cytokine-induced PGCLCs. These differentiated PGCLCs could be sustained over prolonged periods of culture and could differentiate into spermatogonia-like cells in vitro. Importantly, the ability to produce PGCLCs at scale, without using costly cytokines, enables biochemical and functional genomic screens to dissect mechanisms of germ cell development and infertility.
Collapse
Affiliation(s)
- Xinbao Ding
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853
| | - Liangdao Li
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853
| | - Jingyi Gao
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853
| | - Dain Yi
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853
| | - John C Schimenti
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853
| |
Collapse
|
29
|
Jorban A, Lunenfeld E, Huleihel M. Effect of Temperature on the Development of Stages of Spermatogenesis and the Functionality of Sertoli Cells In Vitro. Int J Mol Sci 2024; 25:2160. [PMID: 38396838 PMCID: PMC10889116 DOI: 10.3390/ijms25042160] [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: 12/26/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Spermatogenesis is the process of proliferation and differentiation of spermatogonial cells to meiotic and post-meiotic stages and sperm generation. Normal spermatogenesis occurs in vivo at 34 °C to 35 °C, and high temperatures are known to cause male infertility. The aim of the present study was to examine the effect of temperature (35 °C compared to 37 °C) on the viability/apoptosis of developed cells, on the development of different stages of spermatogenesis in 3D in vitro culture conditions, and the functionality of Sertoli cells under these conditions. We used isolated cells from seminiferous tubules of sexually immature mice. The cells were cultured in methylcellulose (as a three-dimensional (3D) in vitro culture system) and incubated in a CO2 incubator at 35 °C or 37 °C. After two to six weeks, the developed cells and organoids were collected and examined for cell viability and apoptosis markers. The development of different stages of spermatogenesis was evaluated by immunofluorescence staining or qPCR analysis using specific antibodies or primers, respectively, for cells at each stage. Factors that indicate the functionality of Sertoli cells were assessed by qPCR analysis. The developed organoids were examined by a confocal microscope. Our results show that the percentages and/or the expression levels of the developed pre-meiotic, meiotic, and post-meiotic cells were significantly higher at 35 °C compared to those at 37 °C, including the expression levels of the androgen receptor, the FSH receptor, transferrin, the androgen-binding protein (ABP), and the glial-derived nerve growth factor (GDNF) which were similarly significantly higher at 35 °C than at 37 °C. The percentages of apoptotic cells (according to acridine orange staining) and the expression levels of BAX, FAS, and CASPAS 3 were significantly higher in cultures incubated at 37 °C compared to those incubated at 35 °C. These findings support the in vivo results regarding the negative effect of high temperatures on the process of spermatogenesis and suggest a possible effect of high temperatures on the viability/apoptosis of spermatogenic cells. In addition, increasing the temperature in vitro also impaired the functionality of Sertoli cells. These findings may deepen our understanding of the mechanisms behind optimal conditions for normal spermatogenesis in vivo and in vitro.
Collapse
Affiliation(s)
- Areej Jorban
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Eitan Lunenfeld
- Adelson School of Medicine, Ariel University, Ariel 4076414, Israel;
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| |
Collapse
|
30
|
Yi S, Wang W, Su L, Meng L, Li Y, Tan C, Liu Q, Zhang H, Fan L, Lu G, Hu L, Du J, Lin G, Tan YQ, Tu C, Zhang Q. Deleterious variants in X-linked RHOXF1 cause male infertility with oligo- and azoospermia. Mol Hum Reprod 2024; 30:gaae002. [PMID: 38258527 DOI: 10.1093/molehr/gaae002] [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: 06/08/2023] [Revised: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
Oligozoospermia and azoospermia are two common phenotypes of male infertility characterized by massive sperm defects owing to failure of spermatogenesis. The deleterious impact of candidate variants with male infertility is to be explored. In our study, we identified three hemizygous missense variants (c.388G>A: p.V130M, c.272C>T: p.A91V, and c.467C>T: p.A156V) and one hemizygous nonsense variant (c.478C>T: p.R160X) in the Rhox homeobox family member 1 gene (RHOXF1) in four unrelated cases from a cohort of 1201 infertile Chinese men with oligo- and azoospermia using whole-exome sequencing and Sanger sequencing. RHOXF1 was absent in the testicular biopsy of one patient (c.388G>A: p.V130M) whose histological analysis showed a phenotype of Sertoli cell-only syndrome. In vitro experiments indicated that RHOXF1 mutations significantly reduced the content of RHOXF1 protein in HEK293T cells. Specifically, the p.V130M, p.A156V, and p.R160X mutants of RHOXF1 also led to increased RHOXF1 accumulation in cytoplasmic particles. Luciferase assays revealed that p.V130M and p.R160X mutants may disrupt downstream spermatogenesis by perturbing the regulation of doublesex and mab-3 related transcription factor 1 (DMRT1) promoter activity. Furthermore, ICSI treatment could be beneficial in the context of oligozoospermia caused by RHOXF1 mutations. In conclusion, our findings collectively identified mutated RHOXF1 to be a disease-causing X-linked gene in human oligo- and azoospermia.
Collapse
Affiliation(s)
- Sibing Yi
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Weili Wang
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Center for Biology Post-Doctoral studies, College of Life Science, Hunan Normal University, Changsha, China
| | - Lilan Su
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Lanlan Meng
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Yong Li
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Chen Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qiang Liu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Department of Hepatobiliary Surgery, Hunan Cancer Hospital and the Affiliated Cancer of Xiangya School of Medicine, Central South University, Changsha, China
| | - Huan Zhang
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Liqing Fan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, China
| | - Guangxiu Lu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, China
| | - Liang Hu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Center for Biology Post-Doctoral studies, College of Life Science, Hunan Normal University, Changsha, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, China
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Center for Biology Post-Doctoral studies, College of Life Science, Hunan Normal University, Changsha, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Center for Biology Post-Doctoral studies, College of Life Science, Hunan Normal University, Changsha, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, China
| | - Chaofeng Tu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Qianjun Zhang
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
- Center for Biology Post-Doctoral studies, College of Life Science, Hunan Normal University, Changsha, China
- Key Laboratory of Stem Cell and Reproduction Engineering, Ministry of Health, Changsha, China
| |
Collapse
|
31
|
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
|
32
|
van Maaren J, Alves LF, van Wely M, van Pelt AMM, Mulder CL. Favorable culture conditions for spermatogonial propagation in human and non-human primate primary testicular cell cultures: a systematic review and meta-analysis. Front Cell Dev Biol 2024; 11:1330830. [PMID: 38259514 PMCID: PMC10800969 DOI: 10.3389/fcell.2023.1330830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction: Autologous transplantation of spermatogonial stem cells (SSCs) isolated from cryopreserved testicular biopsies obtained before oncological treatment could restore fertility in male childhood cancer survivors. There is a clear necessity for in vitro propagation of the limited SSCs from the testicular biopsy prior to transplantation due to limited numbers of spermatogonia in a cryopreserved testicular biopsy. Still, there is no consensus regarding their optimal culture method. Methods: We performed a systematic review and meta-analysis of studies reporting primary testicular cell cultures of human and non-human primate origin through use of Pubmed, EMBASE, and Web of Science core collection databases. Of 760 records, we included 42 articles for qualitative and quantitative analysis. To quantify in vitro spermatogonial propagation, spermatogonial colony doubling time (CDT) was calculated, which measures the increase in the number of spermatogonial colonies over time. A generalized linear mixed model analysis was used to assess the statistical effect of various culture conditions on CDT. Results: Our analysis indicates decreased CDTs, indicating faster spermatogonial propagation in cultures with a low culture temperature (32°C); with use of non-cellular matrices; use of StemPro-34 medium instead of DMEM; use of Knockout Serum Replacement; and when omitting additional growth factors in the culture medium. Discussion: The use of various methods and markers to detect the presence of spermatogonia within the reported cultures could result in detection bias, thereby potentially influencing comparability between studies. However, through use of CDT in the quantitative analysis this bias was reduced. Our results provide insight into critical culture conditions to further optimize human spermatogonial propagation in vitro, and effectively propagate and utilize these cells in a future fertility restoration therapy and restore hope of biological fatherhood for childhood cancer survivors.
Collapse
Affiliation(s)
- Jillis van Maaren
- Reproductive Biology Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Luis F. Alves
- Reproductive Biology Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Madelon van Wely
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Centre for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ans M. M. van Pelt
- Reproductive Biology Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Callista L. Mulder
- Reproductive Biology Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
33
|
López-Jiménez P, Berenguer I, Pérez-Moreno I, de Aledo JG, Parra MT, Page J, Gómez R. The Organotypic Culture of Mouse Seminiferous Tubules as a Reliable Methodology for the Study of Meiosis In Vitro. Methods Mol Biol 2024; 2818:147-160. [PMID: 39126472 DOI: 10.1007/978-1-0716-3906-1_9] [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] [Indexed: 08/12/2024]
Abstract
Male mouse meiosis has been traditionally studied using descriptive methods like histological sections and spreading or squashing techniques, which allow the observation of fixed meiocytes in either wildtype or genetically modified mice. For these studies, the sacrifice of the males and the extraction of the testicles are required to obtain the material of study. Other functional in vivo studies include the administration of intravenous or intraperitoneal drugs, or the exposure to mutagenic agents or generators of DNA damage, in order to study their impact on meiosis progression. However, in these studies, the exposure times or drug concentration are important limitations to consider when acknowledging animal welfare. Recently, several approaches have been proposed to offer alternative methodologies that allow the in vitro study of spermatocytes with a considerable reduction in the use of animals. Here we revisit and validate an optimal technique of organotypic culture of fragments of seminiferous tubules for meiotic studies. This technique is a trustable methodology to develop functional studies that preserve the histological configuration of the seminiferous tubule, aim homogeneity of the procedures (the use of the same animal for different study conditions), and allow procedures that would compromise the animal welfare. Therefore, this methodology is highly recommendable for the study of meiosis and spermatogenesis, while it supports the principle of 3R's for animal research.
Collapse
Affiliation(s)
- Pablo López-Jiménez
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Meiosis group, MRC Laboratory of Medical Sciences, London, UK
| | - Inés Berenguer
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CBMSO), Madrid, Spain
| | - Irene Pérez-Moreno
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | | | - María Teresa Parra
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Jesús Page
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| | - Rocío Gómez
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| |
Collapse
|
34
|
Uju CN, Unniappan S. Growth factors and female reproduction in vertebrates. Mol Cell Endocrinol 2024; 579:112091. [PMID: 37863469 DOI: 10.1016/j.mce.2023.112091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Female reproductive efficiency is influenced by the outcomes of various processes, including folliculogenesis, apoptosis, response to gonadotropin signaling, oocyte maturation, and ovulation. The role of hormones in regulating these processes and other reproductive activities has been well established. It is becoming increasingly evident that in addition to well-characterized hormones, growth factors play vital roles in regulating some of these reproductive activities. Growth factors and their receptors are widely distributed in vertebrate ovaries at different stages of ovarian development, indicating their involvement in intraovarian reproductive functions. In the ovary, cell surface receptors allow growth factors to regulate intraovarian reproductive activities. Understanding these actions in the reproductive axis would provide a tool to target growth factors and/or their receptors to yield desirable reproductive outcomes. These include enrichment of in vitro maturation and fertilization culture media, and management of infertility. This review discusses some widely characterized growth factors belonging to the TGF, EGF, IGF, FGF, and BDNF family of peptides and their role in female reproduction in vertebrates, with a focus on mammals.
Collapse
Affiliation(s)
- Chinelo N Uju
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5B4, Canada
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5B4, Canada.
| |
Collapse
|
35
|
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
|
36
|
Li C, Bi R, Wang L, Ma YH, Yao YG, Zheng P. Characterization of long-term ex vivo expansion of tree shrew spermatogonial stem cells. Zool Res 2023; 44:1080-1094. [PMID: 37914523 PMCID: PMC10802108 DOI: 10.24272/j.issn.2095-8137.2023.317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/30/2023] [Indexed: 11/03/2023] Open
Abstract
Tree shrews ( Tupaia belangeri chinensis) share a close relationship to primates and have been widely used in biomedical research. We previously established a spermatogonial stem cell (SSC)-based gene editing platform to generate transgenic tree shrews. However, the influences of long-term expansion on tree shrew SSC spermatogenesis potential remain unclear. Here, we examined the in vivo spermatogenesis potential of tree shrew SSCs cultured across different passages. We found that SSCs lost spermatogenesis ability after long-term expansion (>50 passages), as indicated by the failure to colonize the seminiferous epithelium and generate donor spermatogonia (SPG)-derived spermatocytes or spermatids marking spermatogenesis. RNA sequencing (RNA-seq) analysis of undifferentiated SPGs across different passages revealed significant gene expression changes after sub-culturing primary SPG lines for more than 40 passages on feeder layers. Specifically, DNA damage response and repair genes (e.g., MRE11, SMC3, BLM, and GEN1) were down-regulated, whereas genes associated with mitochondrial function (e.g., NDUFA9, NDUFA8, NDUFA13, and NDUFB8) were up-regulated after expansion. The DNA damage accumulation and mitochondrial dysfunction were experimentally validated in high-passage cells. Supplementation with nicotinamide adenine dinucleotide (NAD +) precursor nicotinamide riboside (NR) exhibited beneficial effects by reducing DNA damage accumulation and mitochondrial dysfunction in SPG elicited by long-term culture. Our research presents a comprehensive analysis of the genetic and physiological attributes critical for the sustained expansion of undifferentiated SSCs in tree shrews and proposes an effective strategy for extended in vitro maintenance.
Collapse
Affiliation(s)
- Cong Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Lin Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Yu-Hua Ma
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China. E-mail:
| | - Ping Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650204, China. E-mail:
| |
Collapse
|
37
|
Luo Y, Yamada M, N’Tumba-Byn T, Asif H, Gao M, Hu Y, Marangoni P, Liu Y, Evans T, Rafii S, Klein OD, Voss HU, Hadjantonakis AK, Elemento O, Martin LA, Seandel M. SPRY4-dependent ERK negative feedback demarcates functional adult stem cells in the male mouse germline†. Biol Reprod 2023; 109:533-551. [PMID: 37552049 PMCID: PMC10577279 DOI: 10.1093/biolre/ioad089] [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: 02/15/2023] [Revised: 06/28/2023] [Accepted: 08/05/2023] [Indexed: 08/09/2023] Open
Abstract
Niche-derived growth factors support self-renewal of mouse spermatogonial stem and progenitor cells through ERK MAPK signaling and other pathways. At the same time, dysregulated growth factor-dependent signaling has been associated with loss of stem cell activity and aberrant differentiation. We hypothesized that growth factor signaling through the ERK MAPK pathway in spermatogonial stem cells is tightly regulated within a narrow range through distinct intracellular negative feedback regulators. Evaluation of candidate extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK)-responsive genes known to dampen downstream signaling revealed robust induction of specific negative feedback regulators, including Spry4, in cultured mouse spermatogonial stem cells in response to glial cell line-derived neurotrophic factor or fibroblast growth factor 2. Undifferentiated spermatogonia in vivo exhibited high levels of Spry4 mRNA. Quantitative single-cell analysis of ERK MAPK signaling in spermatogonial stem cell cultures revealed both dynamic signaling patterns in response to growth factors and disruption of such effects when Spry4 was ablated, due to dysregulation of ERK MAPK downstream of RAS. Whereas negative feedback regulator expression decreased during differentiation, loss of Spry4 shifted cell fate toward early differentiation with concomitant loss of stem cell activity. Finally, a mouse Spry4 reporter line revealed that the adult spermatogonial stem cell population in vivo is demarcated by strong Spry4 promoter activity. Collectively, our data suggest that negative feedback-dependent regulation of ERK MAPK is critical for preservation of spermatogonial stem cell fate within the mammalian testis.
Collapse
Affiliation(s)
- Yanyun Luo
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Makiko Yamada
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | | | - Hana Asif
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Meng Gao
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Yang Hu
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Pauline Marangoni
- Program in Craniofacial Biology, Department of Orofacial Sciences, University of California, San Francisco, CA, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Ying Liu
- Division of Regenerative Medicine, Department of Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Department of Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ophir D Klein
- Program in Craniofacial Biology, Department of Orofacial Sciences, University of California, San Francisco, CA, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Henning U Voss
- College of Human Ecology, Cornell University, Ithaca, NY, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Laura A Martin
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Marco Seandel
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
38
|
Liao X, Tao B, Zhang X, Chen L, Chen J, Song Y, Hu W. Loss of gdnfa disrupts spermiogenesis and male courtship behavior in zebrafish. Mol Cell Endocrinol 2023; 576:112010. [PMID: 37419437 DOI: 10.1016/j.mce.2023.112010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/19/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Spermatogenesis is essential for establishment and maintenance of reproduction in male vertebrates. Spermatogenesis, which is mainly regulated by the combined action of hormones, growth factors, and epigenetic factors, is highly conserved. Glial cell line-derived neurotrophic factor (GDNF) is a member of the transforming growth factor-β superfamily. In this study, global gdnfa knockout and Tg (gdnfa: mcherry) transgenic zebrafish lines were generated. Loss of gdnfa resulted in disorganized testes, decreased gonadosomatic index, and low percentage of mature spermatozoa. In the Tg (gdnfa: mcherry) zebrafish line, we found that gdnfa was expressed in Leydig cells. The mutation in gdnfa significantly decreased Leydig cell marker gene expression and androgen secretion in Leydig cells. In addition, courtship behavior was disrupted in the male mutants. We present in vivo data showing that global knockout of gdnfa disrupts spermiogenesis and male courtship behavior in zebrafish. The first viable vertebrate model with a global gdnfa knockout may be valuable for studying the role of GDNF in animal reproduction.
Collapse
Affiliation(s)
- Xianyao Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Binbin Tao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China.
| | - Xiya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lu Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ji Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Yanlong Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; Guangdong Laboratory for Lingnan Modem Agriculture, China; Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
39
|
Oback B, Cossey DA. Chimaeras, complementation, and controlling the male germline. Trends Biotechnol 2023; 41:1237-1247. [PMID: 37173191 DOI: 10.1016/j.tibtech.2023.03.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 05/15/2023]
Abstract
Animal breeding drives genetic progress mainly through the male germline. This process is slow to respond to rapidly mounting environmental pressures that threaten sustainable food security from animal protein production. New approaches promise to accelerate breeding by producing chimaeras, which comprise sterile host and fertile donor genotypes, to exclusively transmit elite male germlines. Following gene editing to generate sterile host cells, the missing germline can be restored by transplanting either: (i) spermatogonial stem cells (SSCs) into the testis; or (ii) embryonic stem cells (ESCs) into early embryos. Here we compare these alternative germline complementation strategies and their impact on agribiotechnology and species conservation. We propose a novel breeding platform that integrates embryo-based complementation with genomic selection, multiplication, and gene modification.
Collapse
Affiliation(s)
- Björn Oback
- AgResearch, Ruakura Research Centre, Hamilton, New Zealand; School of Sciences, University of Waikato, Hamilton, New Zealand; School of Medical Sciences, University of Auckland, Auckland, New Zealand.
| | - Daniel A Cossey
- AgResearch, Ruakura Research Centre, Hamilton, New Zealand; School of Sciences, University of Waikato, Hamilton, New Zealand
| |
Collapse
|
40
|
Salem M, Khadivi F, Javanbakht P, Mojaverrostami S, Abbasi M, Feizollahi N, Abbasi Y, Heidarian E, Rezaei Yazdi F. Advances of three-dimensional (3D) culture systems for in vitro spermatogenesis. Stem Cell Res Ther 2023; 14:262. [PMID: 37735437 PMCID: PMC10512562 DOI: 10.1186/s13287-023-03466-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
The loss of germ cells and spermatogenic failure in non-obstructive azoospermia are believed to be the main causes of male infertility. Laboratory studies have used in vitro testicular models and different 3-dimensional (3D) culture systems for preservation, proliferation and differentiation of spermatogonial stem cells (SSCs) in recent decades. The establishment of testis-like structures would facilitate the study of drug and toxicity screening, pathological mechanisms and in vitro differentiation of SSCs which resulted in possible treatment of male infertility. The different culture systems using cellular aggregation with self-assembling capability, the use of different natural and synthetic biomaterials and various methods for scaffold fabrication provided a suitable 3D niche for testicular cells development. Recently, 3D culture models have noticeably used in research for their architectural and functional similarities to native microenvironment. In this review article, we briefly investigated the recent 3D culture systems that provided a suitable platform for male fertility preservation through organ culture of testis fragments, proliferation and differentiation of SSCs.
Collapse
Affiliation(s)
- Maryam Salem
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Farnaz Khadivi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Parinaz Javanbakht
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Narjes Feizollahi
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Yasaman Abbasi
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Heidarian
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Farzane Rezaei Yazdi
- Department of Anatomy, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
41
|
Ibtisham F, Cham TC, Fayaz MA, Honaramooz A. Effects of Growth Factors on In Vitro Culture of Neonatal Piglet Testicular Tissue Fragments. Cells 2023; 12:2234. [PMID: 37759457 PMCID: PMC10526381 DOI: 10.3390/cells12182234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
In vitro spermatogenesis (IVS) has important applications including fertility preservation of prepubertal cancer patients; however, thus far, IVS has only been achieved using mouse models. To study the effects of growth factors on the maintenance of testicular tissue integrity, germ cell numbers, and potential induction of IVS using a porcine model, we cultured small testicular fragments (~2 mg) from 1-wk-old piglets under six different media conditions (DMEM + 10%KSR alone or supplemented with GDNF, bFGF, SCF, EGF, or a combination of all) for 8 weeks. Overall, tissues supplemented with GDNF and bFGF had the greatest seminiferous tubule integrity and least number of apoptotic cells. GDNF-supplemented tissues had the greatest number of gonocytes per tubule, followed by bFGF-supplemented tissues. There was evidence of gradual Sertoli cell maturation in all groups. Moreover, histological examination and the expression of c-KIT (a marker of differentiating spermatogonia and spermatocytes) and STRA8 (a marker of the pre/meiotic stage germ cells) confirmed the induction of IVS in all groups. However, GDNF- and bFGF-supplemented tissue cultures had greater numbers of seminiferous tubules with spermatocytes compared to other groups. In conclusion, overall, GDNF and bFGF supplementation better maintained the tissue integrity and gonocyte numbers and induced IVS in cultured testicular tissues.
Collapse
Affiliation(s)
| | | | | | - Ali Honaramooz
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada; (F.I.); (T.-C.C.); (M.A.F.)
| |
Collapse
|
42
|
Cao Y, Wang J, Li X, Liu B, Li C, Sun Y, Zou K. Gastrodin protects porcine sertoli cells from zearalenone-induced abnormal secretion of glial cell line-derived neurotrophic factor through the NOTCH signaling pathway. Reprod Biol 2023; 23:100781. [PMID: 37285694 DOI: 10.1016/j.repbio.2023.100781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/25/2023] [Accepted: 05/28/2023] [Indexed: 06/09/2023]
Abstract
Zearalenone (ZEA) is a prevalent mycotoxin found in moldy diets and is associated with reproductive dysfunction. However, the molecular underpinning of ZEA in impairment of spermatogenesis remains largely unknown. To unveil the toxic mechanism of ZEA, we established a co-culture model using porcine Sertoli cells and porcine spermatogonial stem cells (pSSCs) to investigate the impact of ZEA on these cell types and their associated signaling pathways. Our findings showed that low concentration of ZEA inhibited cell apoptosis, while high concentration induced cell apoptosis. Furthermore, the expression levels of Wilms' tumor 1 (WT1), proliferating cell nuclear antigen (PCNA) and glial cell line-derived neurotrophic factor (GDNF) were significantly decreased in ZEA treatment group, while concurrently upregulating the transcriptional levels of the NOTCH signaling pathway target genes HES1 and HEY1. The addition of the NOTCH signaling pathway inhibitor DAPT (GSI-IX) alleviated the damage to porcine Sertoli cells caused by ZEA. Gastrodin (GAS) significantly increased the expression levels of WT1, PCNA and GDNF, and inhibited the transcription of HES1 and HEY1. GAS also efficiently restored the decreased expression levels of DDX4, PCNA and PGP9.5 in co-cultured pSSCs suggesting its potential in ameliorating the damage caused by ZEA to Sertoli cells and pSSCs. In conclusion, the present study demonstrates that ZEA disrupts pSSCs self-renewal by affecting the function of porcine Sertoli cell, and highlights the protective mechanism of GAS through the regulation of the NOTCH signaling pathway. These findings may offer a novel strategy for alleviating ZEA-induced male reproductive dysfunction in animal production.
Collapse
Affiliation(s)
- Yulu Cao
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingjing Wang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxiao Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing 210095, China
| | - Biyun Liu
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chongjun Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yijin Sun
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
43
|
Voigt AL, de Lima e Martins Lara N, Dobrinski I. Comparing the adult and pre-pubertal testis: Metabolic transitions and the change in the spermatogonial stem cell metabolic microenvironment. Andrology 2023; 11:1132-1146. [PMID: 36690000 PMCID: PMC10363251 DOI: 10.1111/andr.13397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND Survivors of childhood cancer often suffer from infertility. While sperm cryopreservation is not feasible before puberty, the patient's own spermatogonial stem cells could serve as a germ cell reservoir, enabling these patients to father their own children in adulthood through the isolation, in vitro expansion, and subsequent transplantation of spermatogonial stem cells. However, this approach requires large numbers of stem cells, and methods for successfully propagating spermatogonial stem cells in the laboratory are yet to be established for higher mammals and humans. The improvement of spermatogonial stem cell culture requires deeper understanding of their metabolic requirements and the mechanisms that regulate metabolic homeostasis. AIM This review gives a summary on our knowledge of spermatogonial stem cell metabolism during maintenance and differentiation and highlights the potential influence of Sertoli cell and stem cell niche maturation on spermatogonial stem cell metabolic requirements during development. RESULTS AND CONCLUSIONS Fetal human spermatogonial stem cell precursors, or gonocytes, migrate into the seminiferous cords and supposedly mature to adult stem cells within the first year of human development. However, the spermatogonial stem cell niche does not fully differentiate until puberty, when Sertoli cells dramatically rearrange the architecture and microenvironment within the seminiferous epithelium. Consequently, pre-pubertal and adult spermatogonial stem cells experience two distinct niche environments potentially affecting spermatogonial stem cell metabolism and maturation. Indeed, the metabolic requirements of mouse primordial germ cells and pig gonocytes are distinct from their adult counterparts, and novel single-cell RNA sequencing analysis of human and porcine spermatogonial stem cells during development confirms this metabolic transition. Knowledge of the metabolic requirements and their changes and regulation during spermatogonial stem cell maturation is necessary to implement laboratory-based techniques and enable clinical use of spermatogonial stem cells. Based on the advancement in our understanding of germline metabolism circuits and maturation events of niche cells within the testis, we propose a new definition of spermatogonial stem cell maturation and its amendment in the light of metabolic change.
Collapse
Affiliation(s)
- Anna Laura Voigt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; and Faculty of Veterinary Medicine, University of Calgary, AB, Canada
| | - Nathalia de Lima e Martins Lara
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; and Faculty of Veterinary Medicine, University of Calgary, AB, Canada
| | - Ina Dobrinski
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine; and Faculty of Veterinary Medicine, University of Calgary, AB, Canada
| |
Collapse
|
44
|
Shakeel M, Yoon M. Effects of insulin-like growth factor-1 on the proliferation and apoptosis of stallion testicular cells under normal and heat stress culture conditions. Anim Reprod Sci 2023; 256:107319. [PMID: 37633109 DOI: 10.1016/j.anireprosci.2023.107319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023]
Abstract
This study investigated the effect of heat stress on stallion testicular cells (TCs) and the effect of insulin-like growth factor (IGF)-1 on TC viability, proliferation, and apoptosis, including different stages of germ cells. TCs were divided into control or treatment groups with 0.01, 0.1, 1, 10, and 100 ng/mL of recombinant human IGF-1 (rhIGF-1) for 24 h at 34 °C and 37 °C. The population and viability were measured before and after treatment. The effects of rhIGF-1 on TC viability, proliferation, and apoptosis were determined using RT-qPCR. Proliferating cell nuclear antigen (PCNA) and marker of proliferation Ki-67 (MKI-67) were used as proliferation markers. Myeloid leukemia-1 (MCL-1) was used as an antiapoptotic marker. BCL2 antagonist/killer-1 (BAK-1) was used as a proapoptotic marker. The relative abundance of mRNA transcript of undifferentiated cell transcription factor 1 (UTF-1), protein gene product 9.5 (PGP9.5), and deleted in azoospermia-like (DAZL), was measured for spermatogenesis progression. TCs treated with 1 ng/mL rhIGF-1 at 34 °C exhibited the highest viability. Significant upregulation of the relative abundance of mRNA transcript of PCNA, MKI-67, and MCL-1 was observed in treated TCs compared with untreated TCs; however, BAK-1 was significantly downregulated in treated TCs. Germ cells treated with 1 ng/mL rhIGF-1 exhibited the highest relative abundance of mRNA transcript of UTF-1 and DAZL, whereas TCs exposed to 0.1 ng/mL showed the highest PGP9.5 level. These data confirm that heat stress in stallions decreases TC viability. These findings may help identify a basal IGF-1 level for TC proliferation and apoptosis during heat stress-induced testicular degeneration in stallions.
Collapse
Affiliation(s)
- Muhammad Shakeel
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea; Department of Clinical Studies, Faculty of Veterinary and Animal Sciences, Pir Mehr Ali Shah, Arid Agriculture University, Rawalpindi 44000, Pakistan
| | - Minjung Yoon
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Republic of Korea; Department of Horse, Companion, and Wild Animal Science, Kyungpook National University, Sangju 37224, Republic of Korea; Research Centre for Horse Industry, Kyungpook National University, Sangju 37224, Republic of Korea.
| |
Collapse
|
45
|
Khaleel B, Lunenfeld E, Kapelushnik J, Huleihel M. Effect of Granulocyte Colony-Stimulating Factor on the Development of Spermatogenesis in the Adulthood of Juvenile AML Mice Model Treated with Cytarabine. Int J Mol Sci 2023; 24:12229. [PMID: 37569605 PMCID: PMC10419160 DOI: 10.3390/ijms241512229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Pediatric acute myeloid leukemia (AML) generally occurs de novo. The treatment of AML includes cytarabine (CYT) and other medications. The granulocyte-colony stimulating factor (GCSF) is used in the clinic in cases of neutropenia after chemotherapies. We show that the administration of GCSF in combination with CYT in AML-diagnosed mice (AML+CYT+GCSF) extended the survival of mice for additional 20 days. However, including GCSF in all treatment modalities does not affect the testis' weight or the histology of the seminiferous tubules (STs). We show that GCSF does not affect normal ST histology from AML-, CYT-, or (AML+CYT)-treated groups compared to the relevant treated group without GCSF 2, 4, and 5 weeks post-injection. However, when comparing the percentages of normal STs between the AML+CYT+GCSF-treated groups and those without GCSF, we observe an increase of 17%-42% in STs at 4 weeks and 5.5 weeks post-injection. Additionally, we show that the injection of GCSF into the normal, AML-alone, or CYT-alone groups, or in combination with AML, significantly decreases the percentage of STs with apoptotic cells compared to the relevant groups without GCSF and to the CT (untreated mice) only 2 weeks post-injection. We also show that injection of GCSF into the CT group increases the examined spermatogonial marker PLZF within 2 weeks post-injection. However, GCSF does not affect the count of meiotic cells (CREM) but decreases the post-meiotic cells (ACROSIN) within 4 weeks post-injection. Furthermore, GCSF not only extends the survival of the AML+CYT-treated group, but it also leads to the generation of sperm (1.2 ± 0.04 × 106/mL) at 5.5 weeks post-injection. In addition, we demonstrate that the injection of GCSF into the CT group increases the RNA expression level of IL-10 but not IL-6 compared to CT 2 weeks post-treatment. However, the injection of GCSF into the AML-treated group reverses the expression levels of both IL-10 and IL-6 to normal levels compared to CT 2 weeks post-injection. Thus, we suggest that the addition of GCSF to the regimen of AML after CYT may assist in the development of future therapeutic strategies to preserve male fertility in AML prepubertal patients.
Collapse
Affiliation(s)
- Bara’ah Khaleel
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Eitan Lunenfeld
- Adelson School of Medicine, Ariel University, Ariel 4076414, Israel;
| | - Joseph Kapelushnik
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
- Department of Pediatric Oncology and Hematology, Soroka Medical Center, Beer-Sheva, and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- The Center of Advanced Research and Education in Reproduction (CARER), Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel;
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| |
Collapse
|
46
|
Whiley PAF, Nathaniel B, Stanton PG, Hobbs RM, Loveland KL. Spermatogonial fate in mice with increased activin A bioactivity and testicular somatic cell tumours. Front Cell Dev Biol 2023; 11:1237273. [PMID: 37564373 PMCID: PMC10409995 DOI: 10.3389/fcell.2023.1237273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
Adult male fertility depends on spermatogonial stem cells (SSCs) which undergo either self-renewal or differentiation in response to microenvironmental signals. Activin A acts on Sertoli and Leydig cells to regulate key aspects of testis development and function throughout life, including steroid production. Recognising that activin A levels are elevated in many pathophysiological conditions, this study investigates effects of this growth factor on the niche that determines spermatogonial fate. Although activin A can promote differentiation of isolated spermatogonia in vitro, its impacts on SSC and spermatogonial function in vivo are unknown. To assess this, we examined testes of Inha KO mice, which feature elevated activin A levels and bioactivity, and develop gonadal stromal cell tumours as adults. The GFRA1+ SSC-enriched population was more abundant and proliferative in Inha KO compared to wildtype controls, suggesting that chronic elevation of activin A promotes a niche which supports SSC self-renewal. Intriguingly, clusters of GFRA1+/EOMES+/LIN28A- cells, resembling a primitive SSC subset, were frequently observed in tubules adjacent to tumour regions. Transcriptional analyses of Inha KO tumours, tubules adjacent to tumours, and tubules distant from tumour regions revealed disrupted gene expression in each KO group increased in parallel with tumour proximity. Modest transcriptional changes were documented in Inha KO tubules with complete spermatogenesis. Importantly, tumours displaying upregulation of activin responsive genes were also enriched for factors that promote SSC self-renewal, including Gdnf, Igf1, and Fgf2, indicating the tumours generate a supportive microenvironment for SSCs. Tumour cells featured some characteristics of adult Sertoli cells but lacked consistent SOX9 expression and exhibited an enhanced steroidogenic phenotype, which could arise from maintenance or acquisition of a fetal cell identity or acquisition of another somatic phenotype. Tumour regions were also heavily infiltrated with endothelial, peritubular myoid and immune cells, which may contribute to adjacent SSC support. Our data show for the first time that chronically elevated activin A affects SSC fate in vivo. The discovery that testis stromal tumours in the Inha KO mouse create a microenvironment that supports SSC self-renewal but not differentiation offers a strategy for identifying pathways that improve spermatogonial propagation in vitro.
Collapse
Affiliation(s)
- Penny A. F. Whiley
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Benedict Nathaniel
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Peter G. Stanton
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Robin M. Hobbs
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Kate L. Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
47
|
Rajachandran S, Zhang X, Cao Q, Caldeira-Brant AL, Zhang X, Song Y, Evans M, Bukulmez O, Grow EJ, Nagano M, Orwig KE, Chen H. Dissecting the spermatogonial stem cell niche using spatial transcriptomics. Cell Rep 2023; 42:112737. [PMID: 37393620 PMCID: PMC10530051 DOI: 10.1016/j.celrep.2023.112737] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/07/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023] Open
Abstract
Spermatogonial stem cells (SSCs) in the testis support the lifelong production of sperm. SSCs reside within specialized microenvironments called "niches," which are essential for SSC self-renewal and differentiation. However, our understanding of the molecular and cellular interactions between SSCs and niches remains incomplete. Here, we combine spatial transcriptomics, computational analyses, and functional assays to systematically dissect the molecular, cellular, and spatial composition of SSC niches. This allows us to spatially map the ligand-receptor (LR) interaction landscape in both mouse and human testes. Our data demonstrate that pleiotrophin regulates mouse SSC functions through syndecan receptors. We also identify ephrin-A1 as a potential niche factor that influences human SSC functions. Furthermore, we show that the spatial re-distribution of inflammation-related LR interactions underlies diabetes-induced testicular injury. Together, our study demonstrates a systems approach to dissect the complex organization of the stem cell microenvironment in health and disease.
Collapse
Affiliation(s)
- Shreya Rajachandran
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xin Zhang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qiqi Cao
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andre L Caldeira-Brant
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiangfan Zhang
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Youngmin Song
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Melanie Evans
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Orhan Bukulmez
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward J Grow
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Makoto Nagano
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Haiqi Chen
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
48
|
Giassetti MI, Miao D, Law NC, Oatley MJ, Park J, Robinson LD, Maddison LA, Bernhardt ML, Oatley JM. ARRDC5 expression is conserved in mammalian testes and required for normal sperm morphogenesis. Nat Commun 2023; 14:2111. [PMID: 37069147 PMCID: PMC10110545 DOI: 10.1038/s41467-023-37735-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/28/2023] [Indexed: 04/19/2023] Open
Abstract
In sexual reproduction, sperm contribute half the genomic material required for creation of offspring yet core molecular mechanisms essential for their formation are undefined. Here, the α-arrestin molecule arrestin-domain containing 5 (ARRDC5) is identified as an essential regulator of mammalian spermatogenesis. Multispecies testicular tissue transcriptome profiling indicates that expression of Arrdc5 is testis enriched, if not specific, in mice, pigs, cattle, and humans. Knockout of Arrdc5 in mice leads to male specific sterility due to production of low numbers of sperm that are immotile and malformed. Spermiogenesis, the final phase of spermatogenesis when round spermatids transform to spermatozoa, is defective in testes of Arrdc5 deficient mice. Also, epididymal sperm in Arrdc5 knockouts are unable to capacitate and fertilize oocytes. These findings establish ARRDC5 as an essential regulator of mammalian spermatogenesis. Considering the role of arrestin molecules as modulators of cellular signaling and ubiquitination, ARRDC5 is a potential male contraceptive target.
Collapse
Affiliation(s)
- Mariana I Giassetti
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Deqiang Miao
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Nathan C Law
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Melissa J Oatley
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Julie Park
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - LeeLa D Robinson
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Lisette A Maddison
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| | - Miranda L Bernhardt
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| | - Jon M Oatley
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA.
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| |
Collapse
|
49
|
Li C, Yao Z, Ma L, Song X, Wang W, Wan C, Ren S, Chen D, Zheng Y, Zhu YT, Chang G, Wu S, Miao K, Luo F, Zhao XY. Lovastatin promotes the self-renewal of murine and primate spermatogonial stem cells. Stem Cell Reports 2023; 18:969-984. [PMID: 37044069 PMCID: PMC10147841 DOI: 10.1016/j.stemcr.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 04/14/2023] Open
Abstract
The spermatogonial stem cell (SSC) niche is critical for SSC maintenance and subsequent spermatogenesis. Numerous reproductive hazards impair the SSC niche, thereby resulting in aberrant SSC self-renewal and male infertility. However, promising agents targeting the impaired SSC niche to promote SSC self-renewal are still limited. Here, we screen out and assess the effects of Lovastatin on the self-renewal of mouse SSCs (mSSCs). Mechanistically, Lovastatin promotes the self-renewal of mSSCs and inhibits its inflammation and apoptosis through the regulation of isoprenoid intermediates. Remarkably, treatment by Lovastatin could promote the proliferation of undifferentiated spermatogonia in the male gonadotoxicity model generated by busulfan injection. Of note, we demonstrate that Lovastatin could enhance the proliferation of primate undifferentiated spermatogonia. Collectively, our findings uncover that lovastatin could promote the self-renewal of both murine and primate SSCs and have implications for the treatment of certain types of male infertility using small compounds.
Collapse
Affiliation(s)
- Chaohui Li
- Shunde Hospital of Southern Medical University, Shunde, Guangdong, China; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhaokai Yao
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Linzi Ma
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Reproductive Medicine Center, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuling Song
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen Wang
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Cong Wan
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Shaofang Ren
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Dingyao Chen
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Zheng
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yong-Tong Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Gang Chang
- Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Shihao Wu
- Shunde Hospital of Southern Medical University, Shunde, Guangdong, China
| | - Kai Miao
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau, SAR, China.
| | - Fang Luo
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| | - Xiao-Yang Zhao
- State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China; Sino-America Joint Research Center for Translational Medicine in Developmental Disabilities, Guangzhou, China; Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; National Clinical Research Canter for Kidney Disease, Guangzhou, China; Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, China.
| |
Collapse
|
50
|
Woodward E, Schlingmann K, Tobias J, Turner R. Characterisation of the testicular transcriptome in stallions with age-related testicular degeneration. Equine Vet J 2023; 55:239-252. [PMID: 35569039 DOI: 10.1111/evj.13588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Age-related testicular degeneration can be defined as the progressive deterioration of the testis that typically occurs in middle-aged or older males and that leads to diminished testicular function and subfertility. In the equine breeding industry, genetically valuable males maintain their value as breeding animals well into old age. Because testicular degeneration is common in middle-aged and older stallions, the disease often has a significant negative impact on a stallion's breeding career and leads to economic losses in the horse breeding industry. OBJECTIVE Because testicular degeneration is a tissue autologous disease in the horse, the objective of this study was to use whole-transcriptome sequencing to compare the testicular transcriptomes of normal, fertile stallions to those of stallions affected by age-related testicular degeneration in order to better understand the pathophysiology of the disease. STUDY DESIGN Cross sectional. METHODS Testicular tissue samples from clinical castrations or euthanasia were collected from normal healthy (n = 3) or older subfertile (n = 4) stallions. Samples were processed and sequenced on an Illumina HiSeq™ 2000 Sequencing System. Bioinformatic analysis of the data was performed in R/RStudio, and the transcriptomes were compared between the two groups. Genes were considered to be differentially expressed between healthy and diseased tissue if they demonstrated at least a ±1.5× fold change difference and had a false discovery rate-adjusted P value <0.05. Gene ontology analysis was performed using Ingenuity® IPA. RESULTS Analyses of differential expression of individual genes, as well as computer-based gene ontology analysis, identified upregulation of cytokine-mediated inflammatory pathways in testes from stallions affected with testicular degeneration. This upregulation of inflammation was associated with upregulation of cell survival pathways, inhibition of apoptotic pathways and increases in collagen formation. MAIN LIMITATIONS There are unavoidable confounding factors (e.g. differences in breed, management, environment, age) that could create non disease-related genetic variation between our normal and affected samples. In addition, there are practical limitations to applying computer-based gene ontology analysis to equine samples. Gene ontology software relies on published information (mostly non-equine), and some biological processes (e.g. apoptosis and inflammation) are more commonly studied than others and so are over-represented in the literature and therefore more likely to be identified by computer algorithms. Caution should be taken when interpreting the data, as alterations in gene expression can be the cause of disease processes or can be the result of disease processes. CONCLUSIONS These results suggest that chronic, low-grade inflammation may be involved in the pathophysiology of age-related testicular degeneration in stallions.
Collapse
Affiliation(s)
- Elizabeth Woodward
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Karen Schlingmann
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania, USA
| | - John Tobias
- Penn Genome Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Regina Turner
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania, USA
| |
Collapse
|