1
|
Carosi S, Innocenti F, Monaco L, Laurenzi G, Saracino R, Canipari R, Vicini E. Hormonal Regulation of Urokinase- and Tissue-Type Plasminogen Activator in Mouse Sertoli Cells. Mol Reprod Dev 2025; 92:e70012. [PMID: 39871771 PMCID: PMC11773371 DOI: 10.1002/mrd.70012] [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: 07/19/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 01/29/2025]
Abstract
A role for the plasminogen activator (PA) system has been postulated in mammalian gonads, considering the complex process of morphogenesis these organs undergo during their development. Our results show that mouse Sertoli cells under basal conditions produce both types of PA, tissue-type PA (tPA) and urokinase-type PA (uPA), and hormonal treatments increase the production of both enzymes. The increased enzyme secretion does not correlate with a parallel increase in their mRNAs. However, the proteolytic activity results from a balance between enzyme activity and inhibitors. Hormonal stimulation decreased the expression of the inhibitor PAI-1, suggesting that the increase in proteolytic activity might depend on the decreased production of PAI-1. The expression of the two enzymes and their inhibitor depends on the seminiferous epithelium stage. We observed higher uPA mRNA levels at stages VII-VIII and IX-XII, tPA peaks at stages VII-VIII, and PAI-1 mRNA levels decreased at stages VII-VIII and IX-XII. The testes from mice lacking the uPA gene (uPA-/-) presented statistically smaller sizes and weights. Histological analysis of uPA-/-animals showed tubular morphology defects and atypical residual bodies (RB), suggesting a defect in Sertoli cell phagocytosis. Moreover, we show lower sperm concentration and motility in uPA-/- mice. These data suggested an effective deficiency of testicular development in the absence of uPA.
Collapse
Affiliation(s)
- Sara Carosi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRomeItaly
| | - Federica Innocenti
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRomeItaly
| | - Lucia Monaco
- Department of Physiology and PharmacologySapienza University of RomeRomeItaly
| | - Gaia Laurenzi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRomeItaly
| | - Rossana Saracino
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRomeItaly
| | - Rita Canipari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRomeItaly
| | - Elena Vicini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRomeItaly
| |
Collapse
|
2
|
Havel SL, Griswold MD. Temporal maturation of Sertoli cells during the establishment of the cycle of the seminiferous epithelium†. Biol Reprod 2024; 111:959-974. [PMID: 39077996 PMCID: PMC11473899 DOI: 10.1093/biolre/ioae115] [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: 02/28/2024] [Revised: 06/05/2024] [Accepted: 07/29/2024] [Indexed: 07/31/2024] Open
Abstract
Sertoli cells, omnipresent, somatic cells within the seminiferous tubules of the mammalian testis are essential to male fertility. Sertoli cells maintain the integrity of the testicular microenvironment, regulate hormone synthesis, and of particular importance, synthesize the active derivative of vitamin A, all trans retinoic acid (atRA), which is required for germ cell differentiation and the commitment of male germ cells to meiosis. Stages VIII-IX, when atRA synthesis occurs in the testis, coincide with multiple germ cell development and testicular restructuring events that rely on Sertoli cell gene products to proceed normally. In this study, we have synchronized and captured the mouse testis at four recurrent points of atRA synthesis to observe transcriptomic changes within Sertoli cells as mice age and the Sertoli cells are exposed to increasingly developed germ cell subtypes. This work provides comprehensive, high-resolution characterization of the timing of induction of functional Sertoli cell genes across the first wave of spermatogenesis, and outlines in silico predictions of germ cell derived signaling mechanisms targeting Sertoli cells. We have found that Sertoli cells adapt to their environment, especially to the needs of the germ cell populations present and establish germ-Sertoli cell and Sertoli-Sertoli cell junctions early but gain many of their known immune-regulatory and protein secretory functions in preparation for spermiogenesis and spermiation. Additionally, we have found unique patterns of germ-Sertoli signaling present at each endogenous pulse of atRA, suggesting individual functions of the various germ cells in germ-Sertoli communication.
Collapse
Affiliation(s)
- Shelby L Havel
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | - Michael D Griswold
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
3
|
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
|
4
|
Liu S, Wu J, Zhao X, Yu M, Taniguchi M, Bao H, Kang K. Recent Progress of Induced Spermatogenesis In Vitro. Int J Mol Sci 2024; 25:8524. [PMID: 39126092 PMCID: PMC11313507 DOI: 10.3390/ijms25158524] [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: 05/29/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Sperm, a crucial gamete for reproduction in sexual reproduction, is generated through the proliferation, differentiation, and morphological transformations of spermatogonial stem cells within the specialized microenvironment of the testes. Replicating this environment artificially presents challenges. However, interdisciplinary advancements in physics, materials science, and cell engineering have facilitated the utilization of innovative materials, technologies, and structures for inducing in vitro sperm production. This article offers a comprehensive overview of research progress on inducing in vitro sperm production by categorizing techniques into two major systems based on matrix-based and non-matrix-based approaches, respectively. Detailed discussions are provided for both types of technology systems through comparisons of their similarities and differences, as well as research advancements. The aim is to provide researchers in this field with a comprehensive panoramic view while presenting our own perspectives and prospects.
Collapse
Affiliation(s)
- Siqi Liu
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| | - Jiang Wu
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| | - Xin Zhao
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| | - Meng Yu
- State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
| | - Masayasu Taniguchi
- Department of Veterinary Medicine, Yamaguchi University, Yamaguchi 753-0841, Japan
| | - Huimingda Bao
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| | - Kai Kang
- College of Coastal Agriculture Science, Guangdong Ocean University, Zhanjiang 524088, China; (S.L.); (J.W.)
| |
Collapse
|
5
|
Zhao Y, Deng S, Li C, Cao J, Wu A, Chen M, Ma X, Wu S, Lian Z. The Role of Retinoic Acid in Spermatogenesis and Its Application in Male Reproduction. Cells 2024; 13:1092. [PMID: 38994945 PMCID: PMC11240464 DOI: 10.3390/cells13131092] [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: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Spermatogenesis in mammalian testes is essential for male fertility, ensuring a continuous supply of mature sperm. The testicular microenvironment finely tunes this process, with retinoic acid, an active metabolite of vitamin A, serving a pivotal role. Retinoic acid is critical for various stages, including the differentiation of spermatogonia, meiosis in spermatogenic cells, and the production of mature spermatozoa. Vitamin A deficiency halts spermatogenesis, leading to the degeneration of numerous germ cells, a condition reversible with retinoic acid supplementation. Although retinoic acid can restore fertility in some males with reproductive disorders, it does not work universally. Furthermore, high doses may adversely affect reproduction. The inconsistent outcomes of retinoid treatments in addressing infertility are linked to the incomplete understanding of the molecular mechanisms through which retinoid signaling governs spermatogenesis. In addition to the treatment of male reproductive disorders, the role of retinoic acid in spermatogenesis also provides new ideas for the development of male non-hormone contraceptives. This paper will explore three facets: the synthesis and breakdown of retinoic acid in the testes, its role in spermatogenesis, and its application in male reproduction. Our discussion aims to provide a comprehensive reference for studying the regulatory effects of retinoic acid signaling on spermatogenesis and offer insights into its use in treating male reproductive issues.
Collapse
Affiliation(s)
- Yue Zhao
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Biological Sciences, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.Z.); (M.C.)
| | - Shoulong Deng
- National Center of Technology Innovation for Animal Model, National Health Commission of China (NHC) Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China;
| | - Chongyang Li
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, China;
| | - Jingchao Cao
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Biological Sciences, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.Z.); (M.C.)
| | - Aowu Wu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Biological Sciences, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.Z.); (M.C.)
| | - Mingming Chen
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Biological Sciences, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.Z.); (M.C.)
| | - Xuehai Ma
- Xinjiang Key Laboratory of Mental Development and Learning Science, College of Psychology, Xinjiang Normal University, Urumqi 830017, China
| | - Sen Wu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Biological Sciences, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.Z.); (M.C.)
| | - Zhengxing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Biological Sciences, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.Z.); (M.C.)
| |
Collapse
|
6
|
Ishiguro KI. Mechanisms of meiosis initiation and meiotic prophase progression during spermatogenesis. Mol Aspects Med 2024; 97:101282. [PMID: 38797021 DOI: 10.1016/j.mam.2024.101282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/16/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Meiosis is a critical step for spermatogenesis and oogenesis. Meiosis commences with pre-meiotic S phase that is subsequently followed by meiotic prophase. The meiotic prophase is characterized by the meiosis-specific chromosomal events such as chromosome recombination and homolog synapsis. Meiosis initiator (MEIOSIN) and stimulated by retinoic acid gene 8 (STRA8) initiate meiosis by activating the meiotic genes by installing the meiotic prophase program at pre-meiotic S phase. This review highlights the mechanisms of meiotic initiation and meiotic prophase progression from the point of the gene expression program and its relevance to infertility. Furthermore, upstream pathways that regulate meiotic initiation will be discussed in the context of spermatogenic development, indicating the sexual differences in the mode of meiotic entry.
Collapse
Affiliation(s)
- Kei-Ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
7
|
Hong SH, Castro G, Wang D, Nofsinger R, Kane M, Folias A, Atkins AR, Yu RT, Napoli JL, Sassone-Corsi P, de Rooij DG, Liddle C, Downes M, Evans RM. Targeting nuclear receptor corepressors for reversible male contraception. Proc Natl Acad Sci U S A 2024; 121:e2320129121. [PMID: 38377195 PMCID: PMC10907271 DOI: 10.1073/pnas.2320129121] [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: 11/30/2023] [Accepted: 01/06/2024] [Indexed: 02/22/2024] Open
Abstract
Despite numerous female contraceptive options, nearly half of all pregnancies are unintended. Family planning choices for men are currently limited to unreliable condoms and invasive vasectomies with questionable reversibility. Here, we report the development of an oral contraceptive approach based on transcriptional disruption of cyclical gene expression patterns during spermatogenesis. Spermatogenesis involves a continuous series of self-renewal and differentiation programs of spermatogonial stem cells (SSCs) that is regulated by retinoic acid (RA)-dependent activation of receptors (RARs), which control target gene expression through association with corepressor proteins. We have found that the interaction between RAR and the corepressor silencing mediator of retinoid and thyroid hormone receptors (SMRT) is essential for spermatogenesis. In a genetically engineered mouse model that negates SMRT-RAR binding (SMRTmRID mice), the synchronized, cyclic expression of RAR-dependent genes along the seminiferous tubules is disrupted. Notably, the presence of an RA-resistant SSC population that survives RAR de-repression suggests that the infertility attributed to the loss of SMRT-mediated repression is reversible. Supporting this notion, we show that inhibiting the action of the SMRT complex with chronic, low-dose oral administration of a histone deacetylase inhibitor reversibly blocks spermatogenesis and fertility without affecting libido. This demonstration validates pharmacologic targeting of the SMRT repressor complex for non-hormonal male contraception.
Collapse
Affiliation(s)
- Suk-Hyun Hong
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Glenda Castro
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Dan Wang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Russell Nofsinger
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Maureen Kane
- Department of Nutritional Sciences and Toxicology, The University of California, Berkeley, CA94720
| | - Alexandra Folias
- Department of Nutritional Sciences and Toxicology, The University of California, Berkeley, CA94720
| | - Annette R. Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Joseph L. Napoli
- Department of Nutritional Sciences and Toxicology, The University of California, Berkeley, CA94720
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, U1233 INSERM, University of California, Irvine, CA92697
| | - Dirk G. de Rooij
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CHUtrecht, The Netherlands
| | - Christopher Liddle
- Storr Liver Centre, The Westmead Institute for Medical Research and Sydney Medical School, University of Sydney, Westmead, NSW2145, Australia
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA92037
| |
Collapse
|
8
|
Khalil A, Daradkeh M, Alrabie A, Abo Siam H. Assessment of the In Vivo Reprotoxicity of Isotretinoin in Sprague-Dawley Male Rat. Curr Drug Discov Technol 2024; 21:e160823219865. [PMID: 37587808 DOI: 10.2174/1570163820666230816155855] [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: 03/30/2023] [Revised: 05/30/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Isotretinoin (ISO) belongs to a family of drugs called retinoids. It is the most effective drug prescribed by dermatologists for the treatment of the inflammatory disease, acne vulgaris. A significant barrier to the use of ISO has worries regarding its adverse effect profile. Despite the well-recognized reproductive toxicity and teratogenicity in females, there is no warning related to the use by male patients in the medication prospectus. Current data on the effects on human male fertility is contradictory and inconclusive. OBJECTIVES This study was undertaken to investigate the potential effects of ISO oral doses in the Sprague-Dawley male rat germ cells using the sperm morphology assay. Also, the serum levels of the follicle-stimulating hormone (FSH), luteinizing hormone (LH), and testosterone were measured. METHODS The rat groups were given varying ISO doses via gastric gavage for seven consecutive days. The epididymis sperm specimens were microscopically examined for the following reproductive toxicity parameters: sperm concentration, examined viability, motility, and morphology. The serum FSH, LH, and testosterone levels were measured by using the corresponding enzyme-linked immunosorbent assay (ELISA) kit. The data were analyzed statistically by one-way analysis of variance (ANOVA) followed by the Tukey test at P ≤ 0.05 significance level. RESULTS The results indicated that the drug did not significantly increase the sex hormone levels but notably affected both the sperm quantity and quality. CONCLUSION These observations suggest that ISO was reprotoxic, and future therapies should be further reassessed.
Collapse
Affiliation(s)
- Ahmad Khalil
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid, Jordan
| | - Mai Daradkeh
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid, Jordan
| | - Amneh Alrabie
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid, Jordan
| | - Hasan Abo Siam
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid, Jordan
| |
Collapse
|
9
|
Li B, Zhao X, Jin T, Wu Z, Yang H. Efficient isolation and purification of spermatogonia, spermatocytes, and spermatids from mice, piglets, and adult boars using an optimized STA-PUT method. Theriogenology 2024; 213:97-108. [PMID: 37820498 DOI: 10.1016/j.theriogenology.2023.09.023] [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: 10/04/2022] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Spermatogenesis is a delicate and complex biological process in which spermatogonial stem cells continue to proliferate and differentiate into mature spermatozoa, maintaining sperm production in male mammals throughout the lifetime. To study the molecular mechanism of spermatogenesis, researchers had to isolate different germ cell subpopulations for in vitro culture and characterization. However, due to the existence of several stages of germ cells and a variety of populations of somatic cells in the testis of male mammals, it is a challenge for us to obtain high-purity germ cell subpopulations for further research. Here, we optimized the STA-PUT device and successfully applied it to isolate and purify spermatogonia populations in piglets, and multiple germ cell populations at different developmental stages in testes of adult mice and boars. This work provides a simple platform for germ cell fractionation to facilitate the molecular mechanistic study of animal spermatogenesis in vitro.
Collapse
Affiliation(s)
- Bin Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xin Zhao
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Taili Jin
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
10
|
He M, Liu K, Cao J, Chen Q. An update on the role and potential mechanisms of clock genes regulating spermatogenesis: A systematic review of human and animal experimental studies. Rev Endocr Metab Disord 2023; 24:585-610. [PMID: 36792803 DOI: 10.1007/s11154-022-09783-0] [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] [Accepted: 12/25/2022] [Indexed: 02/17/2023]
Abstract
Circadian clocks can be traced in nearly all life kingdoms, with the male reproductive system no exception. However, our understanding of the circadian clock in spermatogenesis seems to fall behind other scenarios. The present review aims to summarize the current knowledge about the role and especially the potential mechanisms of clock genes in spermatogenesis regulation. Accumulating studies have revealed rhythmic oscillation in semen parameters and some physiological events of spermatogenesis. Disturbing the clock gene expression by genetic mutations or environmental changes will also notably damage spermatogenesis. On the other hand, the mechanisms of spermatogenetic regulation by clock genes remain largely unclear. Some recent studies, although not revealing the entire mechanisms, indeed attempted to shed light on this issue. Emerging clues hinted that gonadal hormones, retinoic acid signaling, homologous recombination, and the chromatoid body might be involved in the regulation of spermatogenesis by clock genes. Then we highlight the challenges and the promising directions for future studies so as to stimulate attention to this critical field which has not gained adequate concern.
Collapse
Affiliation(s)
- Mengchao He
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Kun Liu
- Center for Disease Control and Prevention of Southern Theatre Command, Guangzhou, 510630, China
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
11
|
Kirsanov O, Johnson TA, Niedenberger BA, Malachowski TN, Hale BJ, Chen Q, Lackford B, Wang J, Singh A, Schindler K, Hermann BP, Hu G, Geyer CB. Retinoic acid is dispensable for meiotic initiation but required for spermiogenesis in the mammalian testis. Development 2023; 150:dev201638. [PMID: 37350382 PMCID: PMC10357014 DOI: 10.1242/dev.201638] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
Retinoic acid (RA) is the proposed mammalian 'meiosis inducing substance'. However, evidence for this role comes from studies in the fetal ovary, where germ cell differentiation and meiotic initiation are temporally inseparable. In the postnatal testis, these events are separated by more than 1 week. Exploiting this difference, we discovered that, although RA is required for spermatogonial differentiation, it is dispensable for the subsequent initiation, progression and completion of meiosis. Indeed, in the absence of RA, the meiotic transcriptome program in both differentiating spermatogonia and spermatocytes entering meiosis was largely unaffected. Instead, transcripts encoding factors required during spermiogenesis were aberrant during preleptonema, and the subsequent spermatid morphogenesis program was disrupted such that no sperm were produced. Taken together, these data reveal a RA-independent model for male meiotic initiation.
Collapse
Affiliation(s)
- Oleksandr Kirsanov
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Taylor A. Johnson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Bryan A. Niedenberger
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Taylor N. Malachowski
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Benjamin J. Hale
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
| | - Qing Chen
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Brad Lackford
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Jiajia Wang
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Anukriti Singh
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Brian P. Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Guang Hu
- Epigenetics and Stem Cell Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Christopher B. Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
- East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
12
|
Johnson TA, Niedenberger BA, Kirsanov O, Harrington EV, Malachowski T, Geyer CB. Differential responsiveness of spermatogonia to retinoic acid dictates precocious differentiation but not meiotic entry during steady-state spermatogenesis†. Biol Reprod 2023; 108:822-836. [PMID: 36708226 PMCID: PMC10183363 DOI: 10.1093/biolre/ioad010] [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: 10/24/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 01/29/2023] Open
Abstract
The foundation of mammalian spermatogenesis is provided by undifferentiated spermatogonia, which comprise of spermatogonial stem cells (SSCs) and transit-amplifying progenitors that differentiate in response to retinoic acid (RA) and are committed to enter meiosis. Our laboratory recently reported that the foundational populations of SSCs, undifferentiated progenitors, and differentiating spermatogonia are formed in the neonatal testis in part based on their differential responsiveness to RA. Here, we expand on those findings to define the extent to which RA responsiveness during steady-state spermatogenesis in the adult testis regulates the spermatogonial fate. Our results reveal that both progenitor and differentiating spermatogonia throughout the testis are capable of responding to exogenous RA, but their resulting fates were quite distinct-undifferentiated progenitors precociously differentiated and proceeded into meiosis on a normal timeline, while differentiating spermatogonia were unable to hasten their entry into meiosis. This reveals that the spermatogonia responding to RA must still complete the 8.6 day differentiation program prior to their entry into meiosis. Addition of exogenous RA enriched testes with preleptotene and pachytene spermatocytes one and two seminiferous cycles later, respectively, supporting recent clinical studies reporting increased sperm production and enhanced fertility in subfertile men on long-term RA analog treatment. Collectively, our results reveal that a well-buffered system exists within mammalian testes to regulate spermatogonial RA exposure, that exposed undifferentiated progenitors can precociously differentiate, but must complete a normal-length differentiation program prior to entering meiosis, and that daily RA treatments increased the numbers of advanced germ cells by directing undifferentiated progenitors to continuously differentiate.
Collapse
Affiliation(s)
- Taylor A Johnson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Bryan A Niedenberger
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Oleksandr Kirsanov
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Ellen V Harrington
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Taylor Malachowski
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NCUSA
- East Carolina Diabetes and Obesity Institute at East Carolina University, Greenville, NCUSA
| |
Collapse
|
13
|
Schleif C, Gewiss R, Griswold M. Chromatin Remodeling via Retinoic Acid Action during Murine Spermatogonial Development. Life (Basel) 2023; 13:690. [PMID: 36983846 PMCID: PMC10058303 DOI: 10.3390/life13030690] [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: 02/11/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Spermatogonial differentiation is a process that commits germ cells to the complex process of spermatogenesis. Spermatogonial differentiation is mediated by the action of retinoic acid, which triggers major morphological and transcriptional changes. While these transcriptional changes have been well explored, there has been little effort devoted to epigenetic regulation surrounding spermatogonial development. This study aimed to uncover the timing and dynamics of chromatin organization during spermatogonial development within the context of these transcriptional changes. Using germ cell synchrony and the assay for transposase accessible chromatin and next generation sequencing (ATAC-seq) to isolate subpopulations of developing spermatogonia and identify accessible regions within their genome, we found that 50% of accessible regions in undifferentiated spermatogonia were condensed following retinoic acid action within 18 h. Surprisingly, genes with known functional relevance during spermatogonial development were accessible at all times, indicating that chromatin state does not impact transcription at these sites. While there was an overall decrease in gene accessibility during spermatogonial development, we found that transcriptionally active regions were not predictive of chromatin state.
Collapse
Affiliation(s)
| | | | - Michael Griswold
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
14
|
Shamhari A‘A, Jefferi NES, Abd Hamid Z, Budin SB, Idris MHM, Taib IS. The Role of Promyelocytic Leukemia Zinc Finger (PLZF) and Glial-Derived Neurotrophic Factor Family Receptor Alpha 1 (GFRα1) in the Cryopreservation of Spermatogonia Stem Cells. Int J Mol Sci 2023; 24:ijms24031945. [PMID: 36768269 PMCID: PMC9915902 DOI: 10.3390/ijms24031945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 01/20/2023] Open
Abstract
The cryopreservation of spermatogonia stem cells (SSCs) has been widely used as an alternative treatment for infertility. However, cryopreservation itself induces cryoinjury due to oxidative and osmotic stress, leading to reduction in the survival rate and functionality of SSCs. Glial-derived neurotrophic factor family receptor alpha 1 (GFRα1) and promyelocytic leukemia zinc finger (PLZF) are expressed during the self-renewal and differentiation of SSCs, making them key tools for identifying the functionality of SSCs. To the best of our knowledge, the involvement of GFRα1 and PLZF in determining the functionality of SSCs after cryopreservation with therapeutic intervention is limited. Therefore, the purpose of this review is to determine the role of GFRα1 and PLZF as biomarkers for evaluating the functionality of SSCs in cryopreservation with therapeutic intervention. Therapeutic intervention, such as the use of antioxidants, and enhancement in cryopreservation protocols, such as cell encapsulation, cryoprotectant agents (CPA), and equilibrium of time and temperature increase the expression of GFRα1 and PLZF, resulting in maintaining the functionality of SSCs. In conclusion, GFRα1 and PLZF have the potential as biomarkers in cryopreservation with therapeutic intervention of SSCs to ensure the functionality of the stem cells.
Collapse
Affiliation(s)
- Asma’ ‘Afifah Shamhari
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Nur Erysha Sabrina Jefferi
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Zariyantey Abd Hamid
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Siti Balkis Budin
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
| | - Muhd Hanis Md Idris
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), Puncak Alam Campus, Bandar Puncak Alam 42300, Selangor, Malaysia
| | - Izatus Shima Taib
- Center of Diagnostics, Therapeutics, and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Wilayah Persekutuan, Malaysia
- Correspondence: ; Tel.: +603-928-97608
| |
Collapse
|
15
|
Ciccarelli M, Oatley JM. Perspectives: Approaches for Studying Livestock Spermatogonia. Methods Mol Biol 2023; 2656:325-339. [PMID: 37249879 DOI: 10.1007/978-1-0716-3139-3_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
At present, the knowledge base on characteristics and biology of spermatogonia in livestock is limited in comparison to rodents, yet the importance of studying these cells for comparative species analysis and enhancing reproductive capacity in food animals is high. Previous studies have established that although many core attributes of organ physiology and mechanisms governing essential cellular functions are conserved across eutherians, significant differences exist between mice and higher order mammals. In this chapter, we briefly discuss distinguishing aspects of testicular anatomy and the spermatogenic lineage in livestock and critical considerations for studying spermatogonial stem cell biology in these species.
Collapse
Affiliation(s)
- Michela Ciccarelli
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Jon M Oatley
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
16
|
Griswold M, Hogarth C. Synchronizing spermatogenesis in the mouse. Biol Reprod 2022; 107:1159-1165. [PMID: 35871549 DOI: 10.1093/biolre/ioac148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/28/2022] [Accepted: 07/20/2022] [Indexed: 01/24/2025] Open
Abstract
The formation of spermatozoa starts with a germ-line stem cell creating a pool of progenitor cells or undifferentiated spermatogonia. There is a requirement for these progenitor cells to be stimulated by retinoic acid (RA) to enter differentiation and ultimately form spermatocytes, undergo meiosis, form spermatids, and ultimately spermatozoa. After the stimulation by RA, which occurs at sites in the seminiferous tubules, it takes ~35 days to complete this complex process. As a result, the adult testis contains germ cells in all possible states of differentiation, and the isolation of individual cell types or study of functional aspects of the cycle of the seminiferous epithelium is very difficult. We describe the use of WIN 18 446-an inhibitor of RA synthesis followed by injection of RA as a mechanism for the synchronization of spermatogenesis to one to three stages of the cycle of the seminiferous epithelium. The result is that only one to four germ cell types are prevalent during the first wave of spermatogenesis. In the adult only a predictable few stages of the cycle are present throughout the entire testis enriching the targeted cells or stages of the cycle.
Collapse
Affiliation(s)
- Michael Griswold
- School of Molecular Biosciences, Washington State University, Pullman, WA, USA
| | - Cathryn Hogarth
- Department of Pharmacy and Biomedical Sciences, LaTrobe University, Wodonga, Victoria, Australia
| |
Collapse
|
17
|
R-Loop Formation in Meiosis: Roles in Meiotic Transcription-Associated DNA Damage. EPIGENOMES 2022; 6:epigenomes6030026. [PMID: 36135313 PMCID: PMC9498298 DOI: 10.3390/epigenomes6030026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/24/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Meiosis is specialized cell division during gametogenesis that produces genetically unique gametes via homologous recombination. Meiotic homologous recombination entails repairing programmed 200–300 DNA double-strand breaks generated during the early prophase. To avoid interference between meiotic gene transcription and homologous recombination, mammalian meiosis is thought to employ a strategy of exclusively transcribing meiotic or post-meiotic genes before their use. Recent studies have shown that R-loops, three-stranded DNA/RNA hybrid nucleotide structures formed during transcription, play a crucial role in transcription and genome integrity. Although our knowledge about the function of R-loops during meiosis is limited, recent findings in mouse models have suggested that they play crucial roles in meiosis. Given that defective formation of an R-loop can cause abnormal transcription and transcription-coupled DNA damage, the precise regulatory network of R-loops may be essential in vivo for the faithful progression of mammalian meiosis and gametogenesis.
Collapse
|
18
|
Topping T, Griswold MD. Global Deletion of ALDH1A1 and ALDH1A2 Genes Does Not Affect Viability but Blocks Spermatogenesis. Front Endocrinol (Lausanne) 2022; 13:871225. [PMID: 35574006 PMCID: PMC9097449 DOI: 10.3389/fendo.2022.871225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/15/2022] [Indexed: 12/02/2022] Open
Abstract
The transition of undifferentiated A spermatogonia to differentiated spermatogonia requires the action of retinoic acid (RA). The synthesis of retinoic acid from retinal in the seminiferous epithelium is a result of the action of aldehyde dehydrogenases termed ALDH1A1, ALDH1A2, and ALDH1A3. We used a mouse with a global deletion of the Aldh1a1 gene that is phenotypically normal and the CRE-loxP approach to eliminate Aldh1a2 genes globally and from Sertoli cells and germ cells. The results show that global elimination of Aldh1a1 and Aldh1a2 genes blocks spermatogenesis but does not appear to affect viability. The cell specific elimination of Aldh1a2 gene showed that retinoic acid synthesis by Sertoli cells is required for the initial round of spermatogonial differentiation but that there is no requirement for retinoic acid synthesis by germ cells. In both the global gene deletion and the cell specific gene deletions the maintenance of Aldh1a3 activity could not compensate.
Collapse
Affiliation(s)
| | - Michael D. Griswold
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| |
Collapse
|
19
|
Yabut KCB, Isoherranen N. CRABPs Alter all-trans-Retinoic Acid Metabolism by CYP26A1 via Protein-Protein Interactions. Nutrients 2022; 14:1784. [PMID: 35565751 PMCID: PMC9105409 DOI: 10.3390/nu14091784] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Cellular retinoic acid binding proteins (CRABP1 and CRABP2) bind all-trans-retinoic acid (atRA), the active metabolite of vitamin A, with high affinity. CRABP1 and CRABP2 have been shown to interact with the atRA-clearing cytochrome P450 enzymes CYP26B1 and CYP26C1 and with nuclear retinoic acid receptors (RARs). We hypothesized that CRABP1 and CRABP2 also alter atRA metabolism and clearance by CYP26A1, the third key atRA-metabolizing enzyme in the CYP26 family. Based on stopped-flow experiments, atRA bound CRABP1 and CRABP2 with Kd values of 4.7 nM and 7.6 nM, respectively. The unbound atRA Km values for 4-OH-atRA formation by CYP26A1 were 4.7 ± 0.8 nM with atRA, 6.8 ± 1.7 nM with holo-CRABP1 and 6.1 ± 2.7 nM with holo-CRABP2 as a substrate. In comparison, the apparent kcat value was about 30% lower (0.71 ± 0.07 min-1 for holo-CRABP1 and 0.75 ± 0.09 min-1 for holo-CRABP2) in the presence of CRABPs than with free atRA (1.07 ± 0.08 min-1). In addition, increasing concentrations in apo-CRABPs decreased the 4-OH-atRA formation rates by CYP26A1. Kinetic analyses suggest that apo-CRABP1 and apo-CRABP2 inhibit CYP26A1 (Ki = 0.39 nM and 0.53 nM, respectively) and holo-CRABPs channel atRA for metabolism by CYP26A1. These data suggest that CRABPs play a critical role in modulating atRA metabolism and cellular atRA concentrations.
Collapse
Affiliation(s)
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
20
|
Schleif MC, Havel SL, Griswold MD. Function of Retinoic Acid in Development of Male and Female Gametes. Nutrients 2022; 14:1293. [PMID: 35334951 PMCID: PMC8951023 DOI: 10.3390/nu14061293] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/07/2023] Open
Abstract
Retinoic acid, an active metabolite of vitamin A, is necessary for many developmental processes in mammals. Much of the field of reproduction has looked toward retinoic acid as a key transcriptional regulator and catalyst of differentiation events. This review focuses on the effects of retinoic acid on male and female gamete formation and regulation. Within spermatogenesis, it has been well established that retinoic acid is necessary for the proper formation of the blood-testis barrier, spermatogonial differentiation, spermiation, and assisting in meiotic completion. While many of the roles of retinoic acid in male spermatogenesis are known, investigations into female oogenesis have provided differing results.
Collapse
Affiliation(s)
| | | | - Michael D. Griswold
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, Pullman, WA 99163, USA; (M.C.S.); (S.L.H.)
| |
Collapse
|
21
|
The netrin-1 receptor UNC5C contributes to the homeostasis of undifferentiated spermatogonia in adult mice. Stem Cell Res 2022; 60:102723. [PMID: 35247845 DOI: 10.1016/j.scr.2022.102723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/24/2022] Open
Abstract
In adult testis, the cell mobility is essential for spermatogonia differentiation and is suspected to regulate spermatogonial stem cell fate. Netrin-1 controls cell migration and/or survival according to the cellular context. Its involvement in some self-renewing lineages raises the possibility that Netrin-1 could have a role in spermatogenesis. We show that in addition to Sertoli cells, a fraction of murine undifferentiated spermatogonia express the Netrin-1 receptor UNC5c and that UNC5c contributes to spermatogonia differentiation. Receptor loss in Unc5crcm males leads to the concomitant accumulation of transit-amplifying progenitors and short syncytia of spermatogonia. Without altering cell death rates, the consequences of Unc5c loss worsen with age: the increase in quiescent undifferentiated progenitors associated with a higher spermatogonial stem cell enriched subset leads to the spermatocyte I decline. We demonstrate in vitro that Netrin-1 promotes a guidance effect as it repulses both undifferentiated and differentiating spermatogonia. Finally, we propose that UNC5c triggers undifferentiated spermatogonia adhesion/ migration and that the repulsive activity of Netrin-1 receptors could regulate spermatogonia differentiation, and maintain germ cell homeostasis.
Collapse
|
22
|
Action and Interaction between Retinoic Acid Signaling and Blood–Testis Barrier Function in the Spermatogenesis Cycle. Cells 2022; 11:cells11030352. [PMID: 35159162 PMCID: PMC8834282 DOI: 10.3390/cells11030352] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/20/2021] [Indexed: 02/04/2023] Open
Abstract
Spermatogenesis is a complex process occurring in mammalian testes, and constant sperm production depends on the exact regulation of the microenvironment in the testes. Many studies have indicated the crucial role of blood–testis barrier (BTB) junctions and retinoic acid (RA) signaling in the spermatogenesis process. The BTB consists of junctions between adjacent Sertoli cells, comprised mainly of tight junctions and gap junctions. In vitamin A-deficient mice, halted spermatogenesis could be rebooted by RA or vitamin A administration, indicating that RA is absolutely required for spermatogenesis. Accordingly, this manuscript will review and discuss how RA and the BTB regulate spermatogenesis and the interaction between RA signaling and BTB function.
Collapse
|
23
|
Ruthig VA, Lamb DJ. Updates in Sertoli Cell-Mediated Signaling During Spermatogenesis and Advances in Restoring Sertoli Cell Function. Front Endocrinol (Lausanne) 2022; 13:897196. [PMID: 35600584 PMCID: PMC9114725 DOI: 10.3389/fendo.2022.897196] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/31/2022] [Indexed: 01/16/2023] Open
Abstract
Since their initial description by Enrico Sertoli in 1865, Sertoli cells have continued to enchant testis biologists. Testis size and germ cell carrying capacity are intimately tied to Sertoli cell number and function. One critical Sertoli cell function is signaling from Sertoli cells to germ cells as part of regulation of the spermatogenic cycle. Sertoli cell signals can be endocrine or paracrine in nature. Here we review recent advances in understanding the interplay of Sertoli cell endocrine and paracrine signals that regulate germ cell state. Although these findings have long-term implications for treating male infertility, recent breakthroughs in Sertoli cell transplantation have more immediate implications. We summarize the surge of advances in Sertoli cell ablation and transplantation, both of which are wedded to a growing understanding of the unique Sertoli cell niche in the transitional zone of the testis.
Collapse
Affiliation(s)
- Victor A. Ruthig
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
- Sexual Medicine Lab, Weill Cornell Medicine, New York, NY, United States
| | - Dolores J. Lamb
- Department of Urology, Weill Cornell Medicine, New York, NY, United States
- Center for Reproductive Genomics, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Dolores J. Lamb,
| |
Collapse
|
24
|
Nakagawa T, Jörg DJ, Watanabe H, Mizuno S, Han S, Ikeda T, Omatsu Y, Nishimura K, Fujita M, Takahashi S, Kondoh G, Simons BD, Yoshida S, Nagasawa T. A multistate stem cell dynamics maintains homeostasis in mouse spermatogenesis. Cell Rep 2021; 37:109875. [PMID: 34686326 DOI: 10.1016/j.celrep.2021.109875] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 08/17/2021] [Accepted: 09/29/2021] [Indexed: 01/15/2023] Open
Abstract
In mouse testis, a heterogeneous population of undifferentiated spermatogonia (Aundiff) harbors spermatogenic stem cell (SSC) potential. Although GFRα1+ Aundiff maintains the self-renewing pool in homeostasis, the functional basis of heterogeneity and the implications for their dynamics remain unresolved. Here, through quantitative lineage tracing of SSC subpopulations, we show that an ensemble of heterogeneous states of SSCs supports homeostatic, persistent spermatogenesis. Such heterogeneity is maintained robustly through stochastic interconversion of SSCs between a renewal-biased Plvap+/GFRα1+ state and a differentiation-primed Sox3+/GFRα1+ state. In this framework, stem cell commitment occurs not directly but gradually through entry into licensed but uncommitted states. Further, Plvap+/GFRα1+ cells divide slowly, in synchrony with the seminiferous epithelial cycle, while Sox3+/GFRα1+ cells divide much faster. Such differential cell-cycle dynamics reduces mitotic load, and thereby the potential to acquire harmful de novo mutations of the self-renewing pool, while keeping the SSC density high over the testicular open niche.
Collapse
Affiliation(s)
- 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; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan; Department of Immunobiology and Hematology, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| | - David J Jörg
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, J. J. Thomson Avenue, University of Cambridge, Cambridge CB3 0HE, UK
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Trans-border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Seungmin Han
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 A0W, UK
| | - Tatsuro Ikeda
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Yoshiki Omatsu
- Department of Immunobiology and Hematology, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, World Premier International Immunology Frontier Research Center, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keiko Nishimura
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Miyako Fujita
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center and Trans-border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Benjamin D Simons
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 A0W, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, Wilberforce Road, Cambridge CB3 0WA, UK
| | - 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; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan.
| | - Takashi Nagasawa
- Department of Immunobiology and Hematology, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, World Premier International Immunology Frontier Research Center, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
25
|
Newkirk SJ, An W. UHRF1: a jack of all trades, and a master epigenetic regulator during spermatogenesis. Biol Reprod 2021; 102:1147-1152. [PMID: 32101289 DOI: 10.1093/biolre/ioaa026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 02/26/2020] [Indexed: 01/03/2023] Open
Affiliation(s)
- Simon J Newkirk
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, USA
| | - Wenfeng An
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, USA
| |
Collapse
|
26
|
Gewiss RL, Law NC, Helsel AR, Shelden EA, Griswold MD. Two distinct Sertoli cell states are regulated via germ cell crosstalk. Biol Reprod 2021; 105:1591-1602. [PMID: 34494084 PMCID: PMC8689118 DOI: 10.1093/biolre/ioab160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/30/2021] [Accepted: 08/13/2021] [Indexed: 01/15/2023] Open
Abstract
Sertoli cells are a critical component of the testis environment for their role in maintaining seminiferous tubule structure, establishing the blood-testis barrier, and nourishing maturing germ cells in a specialized niche. This study sought to uncover how Sertoli cells are regulated in the testis environment via germ cell crosstalk in the mouse. We found two major clusters of Sertoli cells as defined by their transcriptomes in Stages VII-VIII of the seminiferous epithelium and a cluster for all other stages. Additionally, we examined transcriptomes of germ cell-deficient testes and found that these existed in a state independent of either of the germ cell-sufficient clusters. Together, we highlight two main transcriptional states of Sertoli cells in an unperturbed testis environment, and a germ cell-deficient environment does not allow normal Sertoli cell transcriptome cycling and results in a state unique from either of those seen in Sertoli cells from a germ cell-sufficient environment.
Collapse
Affiliation(s)
- Rachel L Gewiss
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA.,Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Nathan C Law
- Center for Reproductive Biology, Washington State University, Pullman, Washington, USA.,Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Aileen R Helsel
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA.,Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Eric A Shelden
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA.,Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Michael D Griswold
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA.,Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
27
|
Abstract
Male meiosis is a complex process whereby spermatocytes undergo cell division to form haploid cells. This review focuses on the role of retinoic acid (RA) in meiosis, as well as several processes regulated by RA before cell entry into meiosis that are critical for proper meiotic entry and completion. Here, we discuss RA metabolism in the testis as well as the roles of stimulated by retinoic acid gene 8 (STRA8) and MEIOSIN, which are responsive to RA and are critical for meiosis. We assert that transcriptional regulation in the spermatogonia is critical for successful meiosis.
Collapse
Affiliation(s)
- Rachel L Gewiss
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - M Christine Schleif
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Michael D Griswold
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
28
|
The male germline-specific protein MAPS is indispensable for pachynema progression and fertility. Proc Natl Acad Sci U S A 2021; 118:2025421118. [PMID: 33602822 PMCID: PMC7923350 DOI: 10.1073/pnas.2025421118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Meiosis is a specialized cell division that creates haploid germ cells from diploid progenitors. Through differential RNA expression analyses, we previously identified a number of mouse genes that were dramatically elevated in spermatocytes, relative to their very low expression in spermatogonia and somatic organs. Here, we investigated in detail 1700102P08Rik, one of these genes, and independently conclude that it encodes a male germline-specific protein, in agreement with a recent report. We demonstrated that it is essential for pachynema progression in spermatocytes and named it male pachynema-specific (MAPS) protein. Mice lacking Maps (Maps -/- ) suffered from pachytene arrest and spermatocyte death, leading to male infertility, whereas female fertility was not affected. Interestingly, pubertal Maps -/- spermatocytes were arrested at early pachytene stage, accompanied by defects in DNA double-strand break (DSB) repair, crossover formation, and XY body formation. In contrast, adult Maps -/- spermatocytes only exhibited partially defective crossover but nonetheless were delayed or failed in progression from early to mid- and late pachytene stage, resulting in cell death. Furthermore, we report a significant transcriptional dysregulation in autosomes and XY chromosomes in both pubertal and adult Maps -/- pachytene spermatocytes, including failed meiotic sex chromosome inactivation (MSCI). Further experiments revealed that MAPS overexpression in vitro dramatically decreased the ubiquitination levels of cellular proteins. Conversely, in Maps -/- pachytene cells, protein ubiquitination was dramatically increased, likely contributing to the large-scale disruption in gene expression in pachytene cells. Thus, MAPS is a protein essential for pachynema progression in male mice, possibly in mammals in general.
Collapse
|
29
|
O'Donnell L, Smith LB, Rebourcet D. Sertoli cells as key drivers of testis function. Semin Cell Dev Biol 2021; 121:2-9. [PMID: 34229950 DOI: 10.1016/j.semcdb.2021.06.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/14/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022]
Abstract
Sertoli cells are the orchestrators of spermatogenesis; they support fetal germ cell commitment to the male pathway and are essential for germ cell development, from maintenance of the spermatogonial stem cell niche and spermatogonial populations, through meiosis and spermiogeneis and to the final release of mature spermatids during spermiation. However, Sertoli cells are also emerging as key regulators of other testis somatic cells, including supporting peritubular myoid cell development in the pre-pubertal testis and supporting the function of the testicular vasculature and in contributing to testicular immune privilege. Sertoli cells also have a major role in regulating androgen production within the testis, by specifying interstitial cells to a steroidogenic fate, contributing to androgen production in the fetal testis, and supporting fetal and adult Leydig cell development and function. Here, we provide an overview of the specific roles for Sertoli cells in the testis and highlight how these cells are key drivers of testicular sperm output, and of adult testis size and optimal function of other testicular somatic cells, including the steroidogenic Leydig cells.
Collapse
Affiliation(s)
- Liza O'Donnell
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; Monash University, Clayton 3168, Victoria, Australia.
| | - Lee B Smith
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia; MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Diane Rebourcet
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
30
|
Suzuki S, McCarrey JR, Hermann BP. Differential RA responsiveness among subsets of mouse late progenitor spermatogonia. Reproduction 2021; 161:645-655. [PMID: 33835049 PMCID: PMC8105290 DOI: 10.1530/rep-21-0031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/07/2021] [Indexed: 01/11/2023]
Abstract
Initiation of spermatogonial differentiation in the mouse testis begins with the response to retinoic acid (RA) characterized by activation of KIT and STRA8 expression. In the adult, spermatogonial differentiation is spatiotemporally coordinated by a pulse of RA every 8.6 days that is localized to stages VII-VIII of the seminiferous epithelial cycle. Dogmatically, progenitor spermatogonia that express retinoic acid receptor gamma (RARG) at these stages will differentiate in response to RA, but this has yet to be tested functionally. Previous single-cell RNA-seq data identified phenotypically and functionally distinct subsets of spermatogonial stem cells (SSCs) and progenitor spermatogonia, where late progenitor spermatogonia were defined by expression of RARG and Dppa3. Here, we found late progenitor spermatogonia (RARGhigh KIT-) were further divisible into two subpopulations based on Dppa3 reporter expression (Dppa3-ECFP or Dppa3-EGFP) and were observed across all stages of the seminiferous epithelial cycle. However, nearly all Dppa3+ spermatogonia were differentiating (KIT+) late in the seminiferous epithelial cycle (stages X-XII), while Dppa3- late progenitors remained abundant, suggesting that Dppa3+ and Dppa3- late progenitors differentially responded to RA. Following acute RA treatment (2-4 h), significantly more Dppa3+ late progenitors induced KIT, including at the midpoint of the cycle (stages VI-IX), than Dppa3- late progenitors. Subsequently, single-cell analyses indicated a subset of Dppa3+ late progenitors expressed higher levels of Rxra, which we confirmed by RXRA whole-mount immunostaining. Together, these results indicate RARG alone is insufficient to initiate a spermatogonial response to RA in the adult mouse testis and suggest differential RXRA expression may discriminate responding cells.
Collapse
Affiliation(s)
- Shinnosuke Suzuki
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249 USA
| | - John R. McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249 USA
| | - Brian P. Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas 78249 USA
| |
Collapse
|
31
|
Rodríguez-Casuriaga R, Geisinger A. Contributions of Flow Cytometry to the Molecular Study of Spermatogenesis in Mammals. Int J Mol Sci 2021; 22:1151. [PMID: 33503798 PMCID: PMC7865295 DOI: 10.3390/ijms22031151] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/12/2021] [Accepted: 01/17/2021] [Indexed: 12/18/2022] Open
Abstract
Mammalian testes are very heterogeneous organs, with a high number of different cell types. Testicular heterogeneity, together with the lack of reliable in vitro culture systems of spermatogenic cells, have been an obstacle for the characterization of the molecular bases of the unique events that take place along the different spermatogenic stages. In this context, flow cytometry has become an invaluable tool for the analysis of testicular heterogeneity, and for the purification of stage-specific spermatogenic cell populations, both for basic research and for clinical applications. In this review, we highlight the importance of flow cytometry for the advances on the knowledge of the molecular groundwork of spermatogenesis in mammals. Moreover, we provide examples of different approaches to the study of spermatogenesis that have benefited from flow cytometry, including the characterization of mutant phenotypes, transcriptomics, epigenetic and genome-wide chromatin studies, and the attempts to establish cell culture systems for research and/or clinical aims such as infertility treatment.
Collapse
Affiliation(s)
- Rosana Rodríguez-Casuriaga
- Department of Molecular Biology, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), 11600 Montevideo, Uruguay
| | - Adriana Geisinger
- Department of Molecular Biology, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), 11600 Montevideo, Uruguay
- Biochemistry-Molecular Biology, Facultad de Ciencias, Universidad de la República (UdelaR), 11400 Montevideo, Uruguay
| |
Collapse
|
32
|
Saintilnord WN, Tenlep SYN, Preston JD, Duregon E, DeRouchey JE, Unrine JM, de Cabo R, Pearson KJ, Fondufe-Mittendorf YN. Chronic Exposure to Cadmium Induces Differential Methylation in Mice Spermatozoa. Toxicol Sci 2021; 180:262-276. [PMID: 33483743 PMCID: PMC8041459 DOI: 10.1093/toxsci/kfab002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cadmium exposure is ubiquitous and has been linked to diseases including cancers and reproductive defects. Since cadmium is nonmutagenic, it is thought to exert its gene dysregulatory effects through epigenetic reprogramming. Several studies have implicated germline exposure to cadmium in developmental reprogramming. However, most of these studies have focused on maternal exposure, while the impact on sperm fertility and disease susceptibility has received less attention. In this study, we used reduced representation bisulfite sequencing to comprehensively investigate the impact of chronic cadmium exposure on mouse spermatozoa DNA methylation. Adult male C57BL/J6 mice were provided water with or without cadmium chloride for 9 weeks. Sperm, testes, liver, and kidney tissues were collected at the end of the treatment period. Cadmium exposure was confirmed through gene expression analysis of metallothionein-1 and 2, 2 well-known cadmium-induced genes. Analysis of sperm DNA methylation changes revealed 1788 differentially methylated sites present at regulatory regions in sperm of mice exposed to cadmium compared with vehicle (control) mice. Furthermore, most of these differential methylation changes positively correlated with changes in gene expression at both the transcription initiation stage as well as the splicing levels. Interestingly, the genes targeted by cadmium exposure are involved in several critical developmental processes. Our results present a comprehensive analysis of the sperm methylome in response to chronic cadmium exposure. These data, therefore, highlight a foundational framework to study gene expression patterns that may affect fertility in the exposed individual as well as their offspring, through paternal inheritance.
Collapse
Affiliation(s)
- Wesley N Saintilnord
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536-0509, USA
| | - Sara Y N Tenlep
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky 40536-0509, USA
| | - Joshua D Preston
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky 40536-0509, USA,Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Eleonora Duregon
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 20892, USA
| | - Jason E DeRouchey
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40536-0509, USA
| | - Jason M Unrine
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, Kentucky 40536-0509, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 20892, USA
| | - Kevin J Pearson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky 40536-0509, USA,To whom correspondence should be addressed at Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536-0509, USA. E-mail: ; Department of Molecular and Cellular Biochemistry, University of Kentucky, 800 Rose Street, 273 BBSRB, Lexington, KY 40536-0509, USA. E-mail:
| | - Yvonne N Fondufe-Mittendorf
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536-0509, USA,To whom correspondence should be addressed at Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536-0509, USA. E-mail: ; Department of Molecular and Cellular Biochemistry, University of Kentucky, 800 Rose Street, 273 BBSRB, Lexington, KY 40536-0509, USA. E-mail:
| |
Collapse
|
33
|
Prokai D, Pudasaini A, Kanchwala M, Moehlman AT, Waits AE, Chapman KM, Chaudhary J, Acevedo J, Keller P, Chao X, Carr BR, Hamra FK. Spermatogonial Gene Networks Selectively Couple to Glutathione and Pentose Phosphate Metabolism but Not Cysteine Biosynthesis. iScience 2021; 24:101880. [PMID: 33458605 PMCID: PMC7797946 DOI: 10.1016/j.isci.2020.101880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 11/02/2020] [Accepted: 11/25/2020] [Indexed: 01/15/2023] Open
Abstract
In adult males, spermatogonia maintain lifelong spermatozoa production for oocyte fertilization. To understand spermatogonial metabolism we compared gene profiles in rat spermatogonia to publicly available mouse, monkey, and human spermatogonial gene profiles. Interestingly, rat spermatogonia expressed metabolic control factors Foxa1, Foxa2, and Foxa3. Germline Foxa2 was enriched in Gfra1Hi and Gfra1Low undifferentiated A-single spermatogonia. Foxa2-bound loci in spermatogonial chromatin were overrepresented by conserved stemness genes (Dusp6, Gfra1, Etv5, Rest, Nanos2, Foxp1) that intersect bioinformatically with conserved glutathione/pentose phosphate metabolism genes (Tkt, Gss, Gc l c , Gc l m, Gpx1, Gpx4, Fth), marking elevated spermatogonial GSH:GSSG. Cystine-uptake and intracellular conversion to cysteine typically couple glutathione biosynthesis to pentose phosphate metabolism. Rat spermatogonia, curiously, displayed poor germline stem cell viability in cystine-containing media, and, like primate spermatogonia, exhibited reduced transsulfuration pathway markers. Exogenous cysteine, cysteine-like mercaptans, somatic testis cells, and ferroptosis inhibitors counteracted the cysteine-starvation-induced spermatogonial death and stimulated spermatogonial growth factor activity in vitro.
Collapse
Affiliation(s)
- David Prokai
- Division of Reproductive Endocrinology and Infertility, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashutosh Pudasaini
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- GenomeDesigns Laboratory, LLC, 314 Stonebridge Drive, Richardson, TX 75080, USA
| | - Mohammed Kanchwala
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrew T. Moehlman
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alexandrea E. Waits
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Karen M. Chapman
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jaideep Chaudhary
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jesus Acevedo
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Cecil H. & Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Patrick Keller
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Cecil H. & Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xing Chao
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bruce R. Carr
- Division of Reproductive Endocrinology and Infertility, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - F. Kent Hamra
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Cecil H. & Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Low retinoic acid levels mediate regionalization of the Sertoli valve in the terminal segment of mouse seminiferous tubules. Sci Rep 2021; 11:1110. [PMID: 33441739 PMCID: PMC7806815 DOI: 10.1038/s41598-020-79987-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/15/2020] [Indexed: 01/29/2023] Open
Abstract
In mammalian testes, undifferentiated spermatogonia (Aundiff) undergo differentiation in response to retinoic acid (RA), while their progenitor states are partially maintained by fibroblast growth factors (FGFs). Sertoli valve (SV) is a region located at the terminal end of seminiferous tubule (ST) adjacent to the rete testis (RT), where the high density of Aundiff is constitutively maintained with the absence of active spermatogenesis. However, the molecular and cellular characteristics of SV epithelia still remain unclear. In this study, we first identified the region-specific AKT phosphorylation in the SV Sertoli cells and demonstrated non-cell autonomous specialization of Sertoli cells in the SV region by performing a Sertoli cell ablation/replacement experiment. The expression of Fgf9 was detected in the RT epithelia, while the exogenous administration of FGF9 caused ectopic AKT phosphorylation in the Sertoli cells of convoluted ST. Furthermore, we revealed the SV region-specific expression of Cyp26a1, which encodes an RA-degrading enzyme, and demonstrated that the increased RA levels in the SV region disrupt its pool of Aundiff by inducing their differentiation. Taken together, RT-derived FGFs and low levels of RA signaling contribute to the non-cell-autonomous regionalization of the SV epithelia and its local maintenance of Aundiff in the SV region.
Collapse
|
35
|
Yoshida S. Mouse Spermatogenesis Reflects the Unity and Diversity of Tissue Stem Cell Niche Systems. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036186. [PMID: 32152184 DOI: 10.1101/cshperspect.a036186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mouse spermatogenesis is supported by spermatogenic stem cells (SSCs). SSCs maintain their pool while migrating over an open (or facultative) niche microenvironment of testicular seminiferous tubules, where ligands that support self-renewal are likely distributed widely. This contrasts with the classic picture of closed (or definitive) niches in which stem cells are gathered and the ligands are highly localized. Some of the key properties observed in the dynamics of SSCs in the testicular niche in vivo, which show the flexible and stochastic (probabilistic) fate behaviors, are found to be generic for a wide range of, if not all, tissue stem cells. SSCs also show properties characteristic of an open niche-supported system, such as high motility. Motivated by the properties of SSCs, in this review, I will reconsider the potential unity and diversity of tissue stem cell systems, with an emphasis on the varying degrees of ligand distribution and stem cell motility.
Collapse
Affiliation(s)
- Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences; and Department of Basic Biology, School of Life Science, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
36
|
Florke Gee RR, Chen H, Lee AK, Daly CA, Wilander BA, Fon Tacer K, Potts PR. Emerging roles of the MAGE protein family in stress response pathways. J Biol Chem 2020; 295:16121-16155. [PMID: 32921631 PMCID: PMC7681028 DOI: 10.1074/jbc.rev120.008029] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
The melanoma antigen (MAGE) proteins all contain a MAGE homology domain. MAGE genes are conserved in all eukaryotes and have expanded from a single gene in lower eukaryotes to ∼40 genes in humans and mice. Whereas some MAGEs are ubiquitously expressed in tissues, others are expressed in only germ cells with aberrant reactivation in multiple cancers. Much of the initial research on MAGEs focused on exploiting their antigenicity and restricted expression pattern to target them with cancer immunotherapy. Beyond their potential clinical application and role in tumorigenesis, recent studies have shown that MAGE proteins regulate diverse cellular and developmental pathways, implicating them in many diseases besides cancer, including lung, renal, and neurodevelopmental disorders. At the molecular level, many MAGEs bind to E3 RING ubiquitin ligases and, thus, regulate their substrate specificity, ligase activity, and subcellular localization. On a broader scale, the MAGE genes likely expanded in eutherian mammals to protect the germline from environmental stress and aid in stress adaptation, and this stress tolerance may explain why many cancers aberrantly express MAGEs Here, we present an updated, comprehensive review on the MAGE family that highlights general characteristics, emphasizes recent comparative studies in mice, and describes the diverse functions exerted by individual MAGEs.
Collapse
Affiliation(s)
- Rebecca R Florke Gee
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Helen Chen
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Anna K Lee
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Christina A Daly
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Benjamin A Wilander
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Klementina Fon Tacer
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; School of Veterinary Medicine, Texas Tech University, Amarillo, Texas, USA.
| | - Patrick Ryan Potts
- Cell and Molecular Biology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
37
|
Larose H, Kent T, Ma Q, Shami AN, Harerimana N, Li JZ, Hammoud SS, Handel MA. Regulation of meiotic progression by Sertoli-cell androgen signaling. Mol Biol Cell 2020; 31:2841-2862. [PMID: 33026960 PMCID: PMC7851862 DOI: 10.1091/mbc.e20-05-0334] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Androgen receptor (AR) signaling in Sertoli cells is known to be important for germ-cell progression through meiosis, but the extent to which androgens indirectly regulate specific meiotic stages is not known. Here, we combine synchronization of spermatogenesis, cytological analyses and single-cell RNAseq (scRNAseq) in the Sertoli-cell androgen receptor knockout (SCARKO) mutant and control mice, and demonstrate that SCARKO mutant spermatocytes exhibited normal expression and localization of key protein markers of meiotic prophase events, indicating that initiation of meiotic prophase is not androgen dependent. However, spermatocytes from SCARKO testes failed to acquire competence for the meiotic division phase. ScRNAseq analysis of wild-type and SCARKO mutant testes revealed a molecular transcriptomic block in an early meiotic prophase state (leptotene/zygotene) in mutant germ cells, and identified several misregulated genes in SCARKO Sertoli cells, many of which have been previously implicated in male infertility. Together, our coordinated cytological and scRNAseq analyses identified germ-cell intrinsic and extrinsic genes responsive to Sertoli-cell androgen signaling that promotes cellular states permissive for the meiotic division phase.
Collapse
Affiliation(s)
- Hailey Larose
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109
| | - Travis Kent
- The Jackson Laboratory, Bar Harbor, ME 04609
| | - Qianyi Ma
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | | | | | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109
| | - Saher Sue Hammoud
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109.,Department of Urology, University of Michigan, Ann Arbor, MI 48109
| | | |
Collapse
|
38
|
Suzuki S, Diaz VD, Hermann BP. What has single-cell RNA-seq taught us about mammalian spermatogenesis? Biol Reprod 2020; 101:617-634. [PMID: 31077285 DOI: 10.1093/biolre/ioz088] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022] Open
Abstract
Mammalian spermatogenesis is a complex developmental program that transforms mitotic testicular germ cells (spermatogonia) into mature male gametes (sperm) for production of offspring. For decades, it has been known that this several-weeks-long process involves a series of highly ordered and morphologically recognizable cellular changes as spermatogonia proliferate, spermatocytes undertake meiosis, and spermatids develop condensed nuclei, acrosomes, and flagella. Yet, much of the underlying molecular logic driving these processes has remained opaque because conventional characterization strategies often aggregated groups of cells to meet technical requirements or due to limited capability for cell selection. Recently, a cornucopia of single-cell transcriptome studies has begun to lift the veil on the full compendium of gene expression phenotypes and changes underlying spermatogenic development. These datasets have revealed the previously obscured molecular heterogeneity among and between varied spermatogenic cell types and are reinvigorating investigation of testicular biology. This review describes the extent of available single-cell RNA-seq profiles of spermatogenic and testicular somatic cells, how those data were produced and evaluated, their present value for advancing knowledge of spermatogenesis, and their potential future utility at both the benchtop and bedside.
Collapse
Affiliation(s)
- Shinnosuke Suzuki
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Victoria D Diaz
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
39
|
Xie Y, Wei BH, Ni FD, Yang WX. Conversion from spermatogonia to spermatocytes: Extracellular cues and downstream transcription network. Gene 2020; 764:145080. [PMID: 32858178 DOI: 10.1016/j.gene.2020.145080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022]
Abstract
Spermatocyte (spc) formation from spermatogonia (spg) differentiation is the first step of spermatogenesis which produces prodigious spermatozoa for a lifetime. After decades of studies, several factors involved in the functioning of a mouse were discovered both inside and outside spg. Considering the peculiar expression and working pattern of each factor, this review divides the whole conversion of spg to spc into four consecutive development processes with a focus on extracellular cues and downstream transcription network in each one. Potential coordination among Dmrt1, Sohlh1/2 and BMP families mediates Ngn3 upregulation, which marks progenitor spg, with other changes. After that, retinoic acid (RA), as a master regulator, promotes A1 spg formation with its helpers and Sall4. A1-to-B spg transition is under the control of Kitl and impulsive RA signaling together with early and late transcription factors Stra8 and Dmrt6. Finally, RA and its responsive effectors conduct the entry into meiosis. The systematic transcription network from outside to inside still needs research to supplement or settle the controversials in each process. As a step further ahead, this review provides possible drug targets for infertility therapy by cross-linking humans and mouse model.
Collapse
Affiliation(s)
- Yi Xie
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bang-Hong Wei
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fei-Da Ni
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
40
|
Lord T, Nixon B. Metabolic Changes Accompanying Spermatogonial Stem Cell Differentiation. Dev Cell 2020; 52:399-411. [PMID: 32097651 DOI: 10.1016/j.devcel.2020.01.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/27/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Male fertility is driven by spermatogonial stem cells (SSCs) that self-renew while also giving rise to differentiating spermatogonia. Spermatogonial transitions are accompanied by a shift in gene expression, however, whether equivalent changes in metabolism occur remains unexplored. In this review, we mined recently published scRNA-seq databases from mouse and human testes to compare expression profiles of spermatogonial subsets, focusing on metabolism. Comparisons revealed a conserved upregulation of genes involved in mitochondrial function, biogenesis, and oxidative phosphorylation in differentiating spermatogonia, while gene expression in SSCs reflected a glycolytic cell. Here, we also discuss the relationship between metabolism and the external microenvironment within which spermatogonia reside.
Collapse
Affiliation(s)
- Tessa Lord
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, the University of Newcastle, Callaghan, Newcastle, NSW 2300, Australia; Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, Newcastle, NSW 2305, Australia.
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, the University of Newcastle, Callaghan, Newcastle, NSW 2300, Australia; Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, Newcastle, NSW 2305, Australia
| |
Collapse
|
41
|
Chung SSW, Vizcarra N, Wolgemuth DJ. Filamentous actin disorganization and absence of apical ectoplasmic specialization disassembly during spermiation upon interference with retinoid signaling†. Biol Reprod 2020; 103:378-389. [PMID: 32678439 PMCID: PMC7401411 DOI: 10.1093/biolre/ioaa123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 11/29/2022] Open
Abstract
Spermiation is a multiple-step process involving profound cellular changes in both spermatids and Sertoli cells. We have observed spermiation defects, including abnormalities in spermatid orientation, translocation and release, in mice deficient in the retinoic acid receptor alpha (RARA) and upon treatment with a pan-RAR antagonist. To elucidate the role of retinoid signaling in regulating spermiation, we first characterized the time course of appearance of spermiogenic defects in response to treatment with the pan-RAR antagonist. The results revealed that defects in spermiation are indeed among the earliest abnormalities in spermatogenesis observed upon inhibition of retinoid signaling. Using fluorescent dye-conjugated phalloidin to label the ectoplasmic specialization (ES), we showed for the first time that these defects involved improper formation of filamentous actin (F-actin) bundles in step 8–9 spermatids and a failure of the actin-surrounded spermatids to move apically to the lumen and to disassemble the ES. The aberrant F-actin organization is associated with diminished nectin-3 expression in both RARA-deficient and pan-RAR antagonist-treated testes. An abnormal localization of both tyrosinated and detyrosinated tubulins was also observed during spermatid translocation in the seminiferous epithelium in drug-treated testes. These results highlight a crucial role of RAR receptor-mediated retinoid signaling in regulating microtubules and actin dynamics in the cytoskeleton rearrangements, required for proper spermiation. This is critical to understand in light of ongoing efforts to inhibit retinoid signaling as a novel approach for male contraception and may reveal spermiation components that could also be considered as new targets for male contraception.
Collapse
Affiliation(s)
- Sanny S W Chung
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Nika Vizcarra
- The Institute of Human Nutrition Columbia University Irving Medical Center, New York, NY, USA
| | - Debra J Wolgemuth
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
- The Institute of Human Nutrition Columbia University Irving Medical Center, New York, NY, USA
- The Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Correspondence: Department of Genetics & Development, Columbia University Irving Medical Center , Russ Berrie Pavilion, Room 608, 1150 St. Nicholas Avenue, New York, NY 10032, USA. Tel: (212) 851-4754; E-mail:
| |
Collapse
|
42
|
Yu K, Zhang Y, Zhang BL, Wu HY, Jiang WQ, Wang ST, Han DP, Liu YX, Lian ZX, Deng SL. In-vitro differentiation of early pig spermatogenic cells to haploid germ cells. Mol Hum Reprod 2020; 25:507-518. [PMID: 31328782 DOI: 10.1093/molehr/gaz043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/26/2019] [Indexed: 01/06/2023] Open
Abstract
Spermatogonial stem cells (SSCs) self-renew and contribute genetic information to the next generation. Pig is wildly used as a model animal for understanding reproduction mechanisms of human being. Inducing directional differentiation of porcine SSCs may be an important strategy in exploring the mechanisms of spermatogenesis and developing better treatment methods for male infertility. Here, we established an in-vitro culture model for porcine small seminiferous tubule segments, to induce SSCs to differentiate into single-tail haploid spermatozoa. The culture model subsequently enabled spermatozoa to express the sperm-specific protein acrosin and oocytes to develop to blastocyst stage after round spermatid injection. The addition of retinoic acid (RA) to the differentiation media promoted the efficiency of haploid differentiation. RT-PCR analysis indicated that RA stimulated the expression of Stra8 but reduced the expression of NANOS2 in spermatogonia. Genes involved in post-meiotic development, transition protein 1 (Tnp1) and protamine 1 (Prm1) were upregulated in the presence of RA. The addition of an RA receptor (RAR) inhibitor, BMS439, showed that RA enhanced the expression of cAMP responsive-element binding protein through RAR and promoted the formation of round spermatids. We established an efficient culture system for in-vitro differentiation of pig SSCs. Our study represents a model for human testis disease and toxicology screening. Molecular regulators of SSC differentiation revealed in this study might provide a therapeutic strategy for male infertility.
Collapse
Affiliation(s)
- Kun Yu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, People's Republic of China
| | - Yi Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Chaoyang District, Beijing, People's Republic of China.,Department of Medicine, Panzhihua University, Sichuan, Sichuan, People's Republic of China
| | - Bao-Lu Zhang
- Marine Consulting Center of MNR, Oceanic Counseling Center, Ministry of Natural Resources of the People's Republic of China, Feng-tai District, Beijing, People's Republic of China
| | - Han-Yu Wu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, People's Republic of China
| | - Wu-Qi Jiang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, People's Republic of China
| | - Su-Tian Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Xiangfang District, People's Republic of China
| | - De-Ping Han
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, People's Republic of China
| | - Yi-Xun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, People's Republic of China
| | - Zheng-Xing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Haidian District, Beijing, People's Republic of China
| | - Shou-Long Deng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Chaoyang District, Beijing, People's Republic of China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, People's Republic of China
| |
Collapse
|
43
|
Beedle MT, Stevison F, Zhong G, Topping T, Hogarth C, Isoherranen N, Griswold MD. Sources of all-trans retinal oxidation independent of the aldehyde dehydrogenase 1A isozymes exist in the postnatal testis†. Biol Reprod 2020; 100:547-560. [PMID: 30247516 DOI: 10.1093/biolre/ioy200] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/01/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023] Open
Abstract
Despite the essential role of the active metabolite of vitamin A, all-trans retinoic acid (atRA) in spermatogenesis, the enzymes, and cellular populations responsible for its synthesis in the postnatal testis remain largely unknown. The aldehyde dehydrogenase 1A (ALDH1A) family of enzymes residing within Sertoli cells is responsible for the synthesis of atRA, driving the first round of spermatogenesis. Those studies also revealed that the atRA required to drive subsequent rounds of spermatogenesis is possibly derived from the ALDH1A enzymes residing within the meiotic and post-meiotic germ cells. Three ALDH1A isozymes (ALDH1A1, ALDH1A2, and ALDH1A3) are present in the testis. Although, ALDH1A1 is expressed in adult Sertoli cells and is suggested to contribute to the atRA required for the pre-meiotic transitions, ALDH1A2 is proposed to be the essential isomer involved in testicular atRA biosynthesis. In this report, we first examine the requirement for ALDH1A2 via the generation and analysis of a conditional Aldh1a2 germ cell knockout and a tamoxifen-induced Aldh1a2 knockout model. We then utilized the pan-ALDH1A inhibitor (WIN 18446) to test the collective contribution of the ALDH1A enzymes to atRA biosynthesis following the first round of spermatogenesis. Collectively, our data provide the first in vivo evidence demonstrating that animals severely deficient in ALDH1A2 postnatally proceed normally through spermatogenesis. Our studies with a pan-ALDH1A inhibitor (WIN 18446) also suggest that an alternative source of atRA biosynthesis independent of the ALDH1A enzymes becomes available to maintain atRA levels for several spermatogenic cycles following an initial atRA injection.
Collapse
Affiliation(s)
- My-Thanh Beedle
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Faith Stevison
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Guo Zhong
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Traci Topping
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Cathryn Hogarth
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Michael D Griswold
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
44
|
Saracino R, Capponi C, Di Persio S, Boitani C, Masciarelli S, Fazi F, Fera S, Vicini E. Regulation of
Gdnf
expression by retinoic acid in Sertoli cells. Mol Reprod Dev 2020; 87:419-429. [DOI: 10.1002/mrd.23323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Rossana Saracino
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| | - Chiara Capponi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| | - Sara Di Persio
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| | - Carla Boitani
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| | - Silvia Masciarelli
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| | - Francesco Fazi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| | - Stefania Fera
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| | - Elena Vicini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of HistologySapienza University of RomeRoma Italy
| |
Collapse
|
45
|
Retinoic Acid and Germ Cell Development in the Ovary and Testis. Biomolecules 2019; 9:biom9120775. [PMID: 31771306 PMCID: PMC6995559 DOI: 10.3390/biom9120775] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/23/2019] [Indexed: 02/07/2023] Open
Abstract
Retinoic acid (RA), a derivative of vitamin A, is critical for the production of oocytes and sperm in mammals. These gametes derive from primordial germ cells, which colonize the nascent gonad, and later undertake sexual differentiation to produce oocytes or sperm. During fetal development, germ cells in the ovary initiate meiosis in response to RA, whereas those in the testis do not yet initiate meiosis, as they are insulated from RA, and undergo cell cycle arrest. After birth, male germ cells resume proliferation and undergo a transition to spermatogonia, which are destined to develop into haploid spermatozoa via spermatogenesis. Recent findings indicate that RA levels change periodically in adult testes to direct not only meiotic initiation, but also other key developmental transitions to ensure that spermatogenesis is precisely organized for the prodigious output of sperm. This review focuses on how female and male germ cells develop in the ovary and testis, respectively, and the role of RA in this process.
Collapse
|
46
|
Gewiss R, Topping T, Griswold MD. Cycles, waves, and pulses: Retinoic acid and the organization of spermatogenesis. Andrology 2019; 8:892-897. [PMID: 31670467 PMCID: PMC7496180 DOI: 10.1111/andr.12722] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/16/2019] [Accepted: 10/28/2019] [Indexed: 12/27/2022]
Abstract
Background Spermatogenesis in mammals is organized in a manner that maximizes sperm production. The central aspect of this organization is the cycle of the seminiferous epithelium that is characterized by an asynchronous repeating series of germ cell associations. These cell associations are the result of a fixed point of entry into the cycle at regular short time intervals and the longer time required for cells to fully differentiate and exit the cycle. Objective This review will examine the current information on the action and metabolism of retinoic acid in the testis, the interaction of retinoic acid (RA) with the cycle and the spermatogenic wave, and the mechanisms that can lead to synchronous spermatogenesis. Finally, the unique applications of synchronous spermatogenesis to the study of the cycle and the mass isolation of specific germ cell populations are described. Materials and methods Retinoic acid metabolism and spermatogonial differentiation have been examined by gene deletions, immunocytochemistry, chemical inhibitors, and mass spectrometry. Results, discussion, and conclusion Both the Sertoli cells and the germ cells have the capacity to synthesize retinoic acid from retinol and in the mouse the entry into the cycle of the seminiferous epithelium, and the subsequent conversion of undifferentiated spermatogonia into differentiating spermatogonia is governed by a peak of RA synthesis occurring at stages VIII‐IX of the cycle. Normal asynchronous spermatogenesis can be modified by altering RA levels, and as a result the entire testis will consist of a few closely related stages of the cycle.
Collapse
Affiliation(s)
- Rachel Gewiss
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| | - Traci Topping
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| | - Michael D Griswold
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| |
Collapse
|
47
|
Jauregui EJ, Mitchell D, Garza SM, Topping T, Hogarth CA, Griswold MD. Leydig cell genes change their expression and association with polysomes in a stage-specific manner in the adult mouse testis. Biol Reprod 2019; 98:722-738. [PMID: 29408990 DOI: 10.1093/biolre/ioy031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/30/2018] [Indexed: 11/13/2022] Open
Abstract
Spermatogenesis in mammals occurs in a very highly organized manner within the seminiferous epithelium regulated by different cell types in the testis. Testosterone produced by Leydig cells regulates blood-testis barrier formation, meiosis, spermiogenesis, and spermiation. However, it is unknown whether Leydig cell function changes with the different stages of the seminiferous epithelium. This study utilized the WIN 18,446 and retinoic acid (RA) treatment regime combined with the RiboTag mouse methodology to synchronize male germ cell development and allow for the in vivo mapping of the Leydig cell translatome across the different stages of one cycle of the seminiferous epithelium. Using microarrays analysis, we identified 11 Leydig cell-enriched genes that were expressed in stage-specific manner such as the glucocorticoid synthesis and transport genes, Cyp21a1 and Serpina6. In addition, there were nine Leydig cell transcripts that change their association with polysomes in correlation with the different stages of the spermatogenic cycle including Egr1. Interestingly, the signal intensity of EGR1 and CYP21 varied among Leydig cells in the adult asynchronous testis. However, testosterone levels across the different stages of germ cell development did not cycle. These data show, for the first time, that Leydig cell gene expression changes in a stage-specific manner during the cycle of the seminiferous epithelium and indicate that a heterogeneous Leydig cell population exists in the adult mouse testis.
Collapse
Affiliation(s)
- Estela J Jauregui
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Debra Mitchell
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Savanna M Garza
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Traci Topping
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Cathryn A Hogarth
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Michael D Griswold
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
48
|
Helsel A, Griswold MD. Retinoic acid signaling and the cycle of the seminiferous epithelium. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2019; 6:1-6. [PMID: 32832726 PMCID: PMC7442248 DOI: 10.1016/j.coemr.2019.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Affiliation(s)
- Aileen Helsel
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, WA, 99164-7520, United States
| | - Michael D Griswold
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, WA, 99164-7520, United States
| |
Collapse
|
49
|
Velte EK, Niedenberger BA, Serra ND, Singh A, Roa-DeLaCruz L, Hermann BP, Geyer CB. Differential RA responsiveness directs formation of functionally distinct spermatogonial populations at the initiation of spermatogenesis in the mouse. Development 2019; 146:dev.173088. [PMID: 31023878 DOI: 10.1242/dev.173088] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 04/16/2019] [Indexed: 12/22/2022]
Abstract
In the mammalian testis, sustained spermatogenesis relies on spermatogonial stem cells (SSCs); their progeny either remain as stem cells (self-renewal) or proliferate and differentiate to enter meiosis in response to retinoic acid (RA). Here, we sought to uncover elusive mechanisms regulating a key switch fundamental to spermatogonial fate: the capacity of spermatogonia to respond to RA. Using the developing mouse testis as a model, we found that spermatogonia and precursor prospermatogonia exhibit a heterogeneous capacity to respond to RA with at least two underlying causes. First, progenitor spermatogonia are prevented from responding to RA by catabolic activity of cytochrome P450 family 26 enzymes. Second, a smaller subset of undifferentiated spermatogonia enriched for SSCs exhibit catabolism-independent RA insensitivity. Moreover, for the first time, we observed that precursor prospermatogonia are heterogeneous and comprise subpopulations that exhibit the same differential RA responsiveness found in neonatal spermatogonia. We propose a novel model by which mammalian prospermatogonial and spermatogonial fates are regulated by their intrinsic capacity to respond (or not) to the differentiation signal provided by RA before, and concurrent with, the initiation of spermatogenesis.
Collapse
Affiliation(s)
- Ellen K Velte
- Departments of Anatomy and Cell Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Bryan A Niedenberger
- Departments of Anatomy and Cell Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Nicholas D Serra
- Departments of Anatomy and Cell Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Anukriti Singh
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Lorena Roa-DeLaCruz
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Christopher B Geyer
- Departments of Anatomy and Cell Biology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA .,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
50
|
Faisal I, Cisneros-Montalvo S, Hamer G, Tuominen MM, Laurila PP, Tumiati M, Jauhiainen M, Kotaja N, Toppari J, Mäkelä JA, Kauppi L. Transcription Factor USF1 Is Required for Maintenance of Germline Stem Cells in Male Mice. Endocrinology 2019; 160:1119-1136. [PMID: 30759202 DOI: 10.1210/en.2018-01088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/08/2019] [Indexed: 12/22/2022]
Abstract
A prerequisite for lifelong sperm production is that spermatogonial stem cells (SSCs) balance self-renewal and differentiation, yet factors required for this balance remain largely undefined. Using mouse genetics, we now demonstrate that the ubiquitously expressed transcription factor upstream stimulatory factor (USF)1 is critical for the maintenance of SSCs. We show that USF1 is not only detected in Sertoli cells as previously reported, but also in SSCs. Usf1-deficient mice display progressive spermatogenic decline as a result of age-dependent loss of SSCs. According to our data, the germ cell defect in Usf1-/- mice cannot be attributed to impairment of Sertoli cell development, maturation, or function, but instead is likely due to an inability of SSCs to maintain a quiescent state. SSCs of Usf1-/- mice undergo continuous proliferation, which provides an explanation for their age-dependent depletion. The proliferation-coupled exhaustion of SSCs in turn results in progressive degeneration of the seminiferous epithelium, gradual decrease in sperm production, and testicular atrophy. We conclude that the general transcription factor USF1 is indispensable for the proper maintenance of mammalian spermatogenesis.
Collapse
Affiliation(s)
- Imrul Faisal
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Doctoral Program in Biomedicine, Doctoral School in Health Sciences, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sheyla Cisneros-Montalvo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Turku Doctoral Program of Molecular Medicine, University of Turku, Turku, Finland
| | - Geert Hamer
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Minna M Tuominen
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pirkka-Pekka Laurila
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Manuela Tumiati
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Jauhiainen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Biomedicum, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Noora Kotaja
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Juho-Antti Mäkelä
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland
| | - Liisa Kauppi
- Genome-Scale Biology Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|