1
|
Naro C, Sette C, Geremia R. Purification by STA-PUT Technique of Male Germ Cells from Single Mouse and RNA-Extraction for Transcriptomic Analysis. Methods Mol Biol 2024; 2770:37-52. [PMID: 38351445 DOI: 10.1007/978-1-0716-3698-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Transcriptomic analyses of germ cells at different stages of differentiation have shed light on the transcriptional and post-transcriptional mechanisms regulating gene expression that ensure the correct progression of spermatogenesis and male fertility. In this chapter, we describe a method to isolate meiotic and post-meiotic germ cells, based on gravimetric sedimentation, starting from a testicular germ cell suspension isolated from a single adult mouse. We also describe how to assess the purity and quality of the collected fractions of germ cells and how to optimize the extraction from these samples of RNA for subsequent RNA-sequencing experiment. In our experience, this protocol is suitable for germ cell isolation and transcriptomic analysis for mouse models with spermatogenic defects, overcoming the limits that reduced fertility poses to the obtaining of experimental animals.
Collapse
Affiliation(s)
- Chiara Naro
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Raffaele Geremia
- Department of Biomedicine and Prevention, Section of Human Anatomy, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
2
|
Guo H, Shen X, Hu H, Zhou P, He T, Xia L, Tan D, Zhang X, Zhang Y. Alteration of RNA modification signature in human sperm correlates with sperm motility. Mol Hum Reprod 2022; 28:gaac031. [PMID: 35959987 PMCID: PMC9422301 DOI: 10.1093/molehr/gaac031] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/27/2022] [Indexed: 11/30/2022] Open
Abstract
RNA modifications, which are introduced post-transcriptionally, have recently been assigned pivotal roles in the regulation of spermatogenesis and embryonic development. However, the RNA modification landscape in human sperm is poorly characterized, hampering our understanding about the potential role played by RNA modification in sperm. Through our recently developed high-throughput RNA modification detection platform based on liquid chromatography with tandem mass spectroscopy, we are the first to have characterized the RNA modification signature in human sperm. The RNA modification signature was generated on the basis of 49 samples from participants, including 13 healthy controls, 21 patients with asthenozoospermia (AZS) and 15 patients with teratozoospermia (TZS). In total, we identified 13 types of RNA modification marks on the total RNA in sperm, and 16 types of RNA modification marks on sperm RNA fragments of different sizes. The levels of these RNA modifications on the RNA of patients with AZS or TZS were altered, compared to controls, especially on sperm RNA fragments > 80 nt. A few types of RNA modifications, such as m1G, m5C, m2G and m1A, showed clear co-expression patterns as well as high linear correlations with clinical sperm motility. In conclusion, we characterized the RNA modification signature of human sperm and identified its correlation with sperm motility, providing promising candidates for use in clinical sperm quality assessment and new research insights for exploring the underlying pathological mechanisms in human male infertility syndromes.
Collapse
Affiliation(s)
- Huanping Guo
- Medical Center of Hematology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xipeng Shen
- Medical Center of Hematology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Hua Hu
- Center for Reproductive & Genetic Medical, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Peng Zhou
- Center for Reproductive & Genetic Medical, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Tong He
- Medical Center of Hematology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Lin Xia
- Medical Center of Hematology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Dongmei Tan
- Laboratory Animal Center, Chongqing Medical University, Chongqing, China
| | - Xi Zhang
- Medical Center of Hematology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yunfang Zhang
- Medical Center of Hematology, The Second Affiliated Hospital of Army Medical University, Chongqing, China
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
3
|
Differential Expression and Localization of EHBP1L1 during the First Wave of Rat Spermatogenesis Suggest Its Involvement in Acrosome Biogenesis. Biomedicines 2022; 10:biomedicines10010181. [PMID: 35052860 PMCID: PMC8773523 DOI: 10.3390/biomedicines10010181] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/01/2023] Open
Abstract
The identification and characterization of new proteins involved in spermatogenesis is fundamental, considering that good-quality gametes are basic in ensuring proper reproduction. Here, we further analyzed the temporal and spatial localization during the first spermatogenic wave of rat testis of EHBP1L1, which is involved in vesicular trafficking due to the CH and bMERB domains, which bind to actin and Rab8/10, respectively. Western blot and immunofluorescence analyses showed that EHBP1L1 protein expression started at 21 days post-partum (dpp) concomitantly with the appearance of primary spermatocytes (I SPC). In subsequent stages, EHBP1L1 specifically localized together with actin in the perinuclear cytoplasm close to the acrosomal and Golgian regions of spermatids (SPT) during the different phases of acrosome biogenesis (AB). Moreover, it was completely absent in elongated SPT and in mature spermatozoa, suggesting that its role was completed in previous stages. The combined data, also supported by our previous report demonstrating that EHBP1L1 mRNA was expressed by primary (I) and secondary (II) SPC, lead us to hypothesize its specific role during AB. Although these results are suggestive, further studies are needed to better clarify the underlying molecular mechanisms of AB, with the aim to use EHBP1L1 as a potential new marker for spermatogenesis.
Collapse
|
4
|
Yang C, Yao C, Ji Z, Zhao L, Chen H, Li P, Tian R, Zhi E, Huang Y, Han X, Hong Y, Zhou Z, Li Z. RNA-binding protein ELAVL2 plays post-transcriptional roles in the regulation of spermatogonia proliferation and apoptosis. Cell Prolif 2021; 54:e13098. [PMID: 34296486 PMCID: PMC8450129 DOI: 10.1111/cpr.13098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022] Open
Abstract
Objectives RNA‐binding proteins (RBPs) play essential post‐transcriptional roles in regulating spermatogonial stem cells (SSCs) maintenance and differentiation. We identified a conserved and SSCs‐enriched RBP ELAVL2 from our single‐cell sequencing data, but its function and mechanism in SSCs were unclear. Materials and methods Expressions of ELAVL2 during human and mouse testis development were validated. Stable C18‐4 and TCam‐2 cell lines with overexpression and knockdown of ELAVL2 were established, which were applied to proliferation and apoptosis analysis. RNA immunoprecipitation and sequencing were used to identify ELAVL2 targets, and regulatory functions of ELAVL2 on target mRNAs were studied. Proteins interacting with ELAVL2 in human and mouse testes were identified using immunoprecipitation and mass spectrometric, which were validated by in vivo and in vitro experiments. Results ELAVL2 was testis‐enriched and preferentially expressed in human and mouse SSCs. ELAVL2 was down‐regulated in NOA patients. ELAVL2 promoted proliferation and inhibited apoptosis of C18‐4 and TCam‐2 cell lines via activating ERK and AKT pathways. ELAVL2 associated with mRNAs encoding essential regulators of SSCs proliferation and survival, and promoted their protein expression at post‐transcriptional level. ELAVL2 interacted with DAZL in vivo and in vitro in both human and mouse testes. Conclusions Taken together, these results indicate that ELAVL2 is a conserved SSCs‐enriched RBP that down‐regulated in NOA, which regulates spermatogonia proliferation and apoptosis by promoting protein expression of targets.
Collapse
Affiliation(s)
- Chao Yang
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chencheng Yao
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyong Ji
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Liangyu Zhao
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huixing Chen
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Li
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruhui Tian
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erlei Zhi
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhua Huang
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Han
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Hong
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zheng Li
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Venditti M, Donizetti A, Aniello F, Minucci S. EH domain binding protein 1-like 1 (EHBP1L1), a protein with calponin homology domain, is expressed in the rat testis. ZYGOTE 2020; 28:441-446. [PMID: 32795384 DOI: 10.1017/s0967199420000301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this paper, with the aim to find new genes involved in mammalian spermatogenesis, we isolated, for the first time in the rat testis, a partial cDNA clone that encoded EH domain binding protein 1-like 1 (Ehbp1l1), a protein that has a single calponin homology domain (CH). Bioinformatic analysis showed that EHBP1l1 contains three domains: the N-terminal C2-like, the CH and the C-terminal bivalent Mical/EHBP Rab binding (bMERB) domains, which are evolutionarily conserved in vertebrates. We found that Ehbp1l1 mRNA was expressed in several rat tissues, including the liver, intestine, kidney and also in the testis during its development, with a higher level in testis from 12-month-old animals. Interestingly, in situ hybridization experiments revealed that Ehbp1l1 is specifically expressed by types I and II spermatocytes, this result was validated by RT-PCR performed on total RNA obtained from enriched fractions of different testicular cell types. As EHBP1l1 has been described as linked to vesicular transport to the actin cytoskeleton and as an effector of the small GTPase Rab8, we hypothesized that it could participate both in cytoskeletal remodelling and in the regulation of vesicle sorting from the trans-Golgi network to the apical plasma membrane. Our findings provide a better understand of the molecular mechanisms of the differentiation process of spermatogenesis; Ehbp1l1 may also be used as a new marker of testicular activity.
Collapse
Affiliation(s)
- Massimo Venditti
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate 'F. Bottazzi', Università degli Studi della Campania 'Luigi Vanvitelli' via Costantinopoli, 16-80138 - Napoli, Italy
| | - Aldo Donizetti
- Dipartimento di Biologia, Università di Napoli 'Federico II, via Cinthia', 21-80126 - Napoli, Italy
| | - Francesco Aniello
- Dipartimento di Biologia, Università di Napoli 'Federico II, via Cinthia', 21-80126 - Napoli, Italy
| | - Sergio Minucci
- Dipartimento di Medicina Sperimentale, Sez. Fisiologia Umana e Funzioni Biologiche Integrate 'F. Bottazzi', Università degli Studi della Campania 'Luigi Vanvitelli' via Costantinopoli, 16-80138 - Napoli, Italy
| |
Collapse
|
6
|
Nucleolus structural integrity during the first meiotic prophase in rat spermatocytes. Exp Cell Res 2019; 383:111587. [PMID: 31454492 DOI: 10.1016/j.yexcr.2019.111587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022]
Abstract
A typical nucleolus structure is shaped by three components. A meshwork of fine fibers forming the fibrillar center (FC) is surrounded by densely packed fibers forming the dense fibrillar component (DFC). Meanwhile, wrapping the FC and DFC is the granular component (GC). During the mitotic prophase, the nucleolus undergoes disassembling of its components. On the contrary, throughout the first meiotic prophase that occurs in the cells of the germ line, small nucleoli are assembled into one nucleolus by the end of the prophase. These nucleoli are transcriptionally active, suggesting that they are fully functional. Electron microscopy analysis has suggested that these nucleoli display their three main components but a typical organization has not been observed. Here, by immunolabeling and electron microscopy, we show that the nucleolus has its three main components. The GC is interlaced with the DFC and is not as well defined as previously thought during leptotene and zygotene stage.
Collapse
|
7
|
Carvalho RK, Andersen ML, Mazaro-Costa R. The effects of cannabidiol on male reproductive system: A literature review. J Appl Toxicol 2019; 40:132-150. [PMID: 31313338 DOI: 10.1002/jat.3831] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/25/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023]
Abstract
Cannabidiol (CBD) is one of the most abundant phytocannabinoids present in the plant Cannabis sativa (marijuana). There have been several studies of CBD in the last few decades, mainly focused on its neuroprotective properties, particularly after the identification of the endocannabinoid system and its participation in the central nervous system. On the other hand, the peripheral effects of CBD, particularly on reproductive physiology, were also evidenced. A narrative review was conducted using the PubMed database to identify studies that analyzed the pharmacological effects of CBD on the male reproductive system of vertebrates and invertebrates. Thirty-two citations (in vivo and in vitro) were identified. Among the vertebrates, the studies were carried out with men, monkeys, rats and mice. Studies with invertebrates are centered exclusively on the sea urchin. The CBD treatment periods include mostly acute and subacute evaluations. Exposure to CBD is associated with a reduction in mammalian testis size, the number of germ and Sertoli cells in spermatogenesis, fertilization rates, and plasma concentrations of hypothalamic, pituitary and gonadal hormones. Moreover, chronic doses of CBD have impaired sexual behavior in mice. From the studies identified in this review, it is possible to conclude that CBD has negative effects on the reproductive system of males. However, knowledge is still limited, and additional research is required to elucidate fully the mechanisms of action, as well as the reversibility of CBD effects on the reproductive system.
Collapse
Affiliation(s)
- Renata K Carvalho
- Department of Pharmacology, Laboratory of Physiology and Pharmacology of Reproduction, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Monica L Andersen
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Renata Mazaro-Costa
- Department of Pharmacology, Laboratory of Physiology and Pharmacology of Reproduction, Universidade Federal de Goiás, Goiânia, GO, Brazil
| |
Collapse
|
8
|
|
9
|
Sutherland JM, Siddall NA, Hime GR, McLaughlin EA. RNA binding proteins in spermatogenesis: an in depth focus on the Musashi family. Asian J Androl 2016; 17:529-36. [PMID: 25851660 PMCID: PMC4492041 DOI: 10.4103/1008-682x.151397] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Controlled gene regulation during gamete development is vital for maintaining reproductive potential. During the complex process of mammalian spermatogenesis, male germ cells experience extended periods of the inactive transcription despite heavy translational requirements for continued growth and differentiation. Hence, spermatogenesis is highly reliant on mechanisms of posttranscriptional regulation of gene expression, facilitated by RNA binding proteins (RBPs), which remain abundantly expressed throughout this process. One such group of proteins is the Musashi family, previously identified as critical regulators of testis germ cell development and meiosis in Drosophila, and also shown to be vital to sperm development and reproductive potential in the mouse. This review describes the role and function of RBPs within the scope of male germ cell development, focusing on our recent knowledge of the Musashi proteins in spermatogenesis. The functional mechanisms utilized by RBPs within the cell are outlined in depth, and the significance of sub-cellular localization and stage-specific expression in relation to the mode and impact of posttranscriptional regulation is also highlighted. We emphasize the historical role of the Musashi family of RBPs in stem cell function and cell fate determination, as originally characterized in Drosophila and Xenopus, and conclude with our current understanding of the differential roles and functions of the mammalian Musashi proteins, Musashi-1 and Musashi-2, with a primary focus on our findings in spermatogenesis. This review highlights both the essential contribution of RBPs to posttranscriptional regulation and the importance of the Musashi family as master regulators of male gamete development.
Collapse
Affiliation(s)
| | | | | | - Eileen A McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
10
|
Licatalosi DD. Roles of RNA-binding Proteins and Post-transcriptional Regulation in Driving Male Germ Cell Development in the Mouse. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 907:123-51. [PMID: 27256385 DOI: 10.1007/978-3-319-29073-7_6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue development and homeostasis are dependent on highly regulated gene expression programs in which cell-specific combinations of regulatory factors determine which genes are expressed and the post-transcriptional fate of the resulting RNA transcripts. Post-transcriptional regulation of gene expression by RNA-binding proteins has critical roles in tissue development-allowing individual genes to generate multiple RNA and protein products, and the timing, location, and abundance of protein synthesis to be finely controlled. Extensive post-transcriptional regulation occurs during mammalian gametogenesis, including high levels of alternative mRNA expression, stage-specific expression of mRNA variants, broad translational repression, and stage-specific activation of mRNA translation. In this chapter, an overview of the roles of RNA-binding proteins and the importance of post-transcriptional regulation in male germ cell development in the mouse is presented.
Collapse
Affiliation(s)
- Donny D Licatalosi
- Center for RNA Molecular Biology, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
11
|
Oyama T, Sasagawa S, Takeda S, Hess RA, Lieberman PM, Cheng EH, Hsieh JJ. Cleavage of TFIIA by Taspase1 activates TRF2-specified mammalian male germ cell programs. Dev Cell 2014; 27:188-200. [PMID: 24176642 DOI: 10.1016/j.devcel.2013.09.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 08/12/2013] [Accepted: 09/26/2013] [Indexed: 01/25/2023]
Abstract
The evolution of tissue-specific general transcription factors (GTFs), such as testis-specific TBP-related factor 2 (TRF2), enables the spatiotemporal expression of highly specialized genetic programs. Taspase1 is a protease that cleaves nuclear factors MLL1, MLL2, TFIIAα-β, and ALFα-β (TFIIAτ). Here, we demonstrate that Taspase1-mediated processing of TFIIAα-β drives mammalian spermatogenesis. Both Taspase1(-/-) and noncleavable TFIIAα-βnc/nc testes release immature germ cells with impaired transcription of Transition proteins (Tnp) and Protamines (Prm), exhibiting chromatin compaction defects and recapitulating those observed with TRF2(-/-) testes. Although the unprocessed TFIIA still complexes with TRF2, this complex is impaired in targeting and thus activating Tnp1 and Prm1 promoters. The current study presents a paradigm in which a protease (Taspase1) cleaves a ubiquitously expressed GTF (TFIIA) to enable tissue-specific (testis) transcription, meeting the demand for sophisticated regulation of distinct subsets of genes in higher organisms.
Collapse
Affiliation(s)
- Toshinao Oyama
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Satoru Sasagawa
- Department of Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka 537-8511, Japan
| | - Shugaku Takeda
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Rex A Hess
- Veterinary Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | | | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - James J Hsieh
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
12
|
Teperek M, Miyamoto K. Nuclear reprogramming of sperm and somatic nuclei in eggs and oocytes. Reprod Med Biol 2013; 12:133-149. [PMID: 24273450 PMCID: PMC3824936 DOI: 10.1007/s12522-013-0155-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/18/2013] [Indexed: 10/26/2022] Open
Abstract
Eggs and oocytes have a prominent ability to reprogram sperm nuclei for ensuring embryonic development. The reprogramming activity that eggs/oocytes intrinsically have towards sperm is utilised to reprogram somatic nuclei injected into eggs/oocytes in nuclear transfer (NT) embryos. NT embryos of various species can give rise to cloned animals, demonstrating that eggs/oocytes can confer totipotency even to somatic nuclei. However, many studies indicate that reprogramming of somatic nuclei is not as efficient as that of sperm nuclei. In this review, we explain how and why sperm and somatic nuclei are differentially reprogrammed in eggs/oocytes. Recent studies have shown that sperm chromatin is epigenetically modified to be adequate for early embryonic development, while somatic nuclei do not have such modifications. Moreover, epigenetic memories encoded in sperm chromatin are transgenerationally inherited, implying unique roles of sperm. We also discuss whether somatic nuclei can be artificially modified to acquire sperm-like chromatin states in order to increase the efficiency of nuclear reprogramming.
Collapse
Affiliation(s)
- Marta Teperek
- The Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, CB2 1QN Cambridge, United Kingdom ; Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
13
|
Hussain S, Tuorto F, Menon S, Blanco S, Cox C, Flores JV, Watt S, Kudo NR, Lyko F, Frye M. The mouse cytosine-5 RNA methyltransferase NSun2 is a component of the chromatoid body and required for testis differentiation. Mol Cell Biol 2013; 33:1561-70. [PMID: 23401851 PMCID: PMC3624257 DOI: 10.1128/mcb.01523-12] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 01/30/2013] [Indexed: 11/20/2022] Open
Abstract
Posttranscriptional regulatory mechanisms are crucial for protein synthesis during spermatogenesis and are often organized by the chromatoid body. Here, we identify the RNA methyltransferase NSun2 as a novel component of the chromatoid body and, further, show that NSun2 is essential for germ cell differentiation in the mouse testis. In NSun2-depleted testes, genes encoding Ddx4, Miwi, and Tudor domain-containing (Tdr) proteins are repressed, indicating that RNA-processing and posttranscriptional pathways are impaired. Loss of NSun2 specifically blocked meiotic progression of germ cells into the pachytene stage, as spermatogonial and Sertoli cells were unaffected in knockout mice. We observed the same phenotype when we simultaneously deleted NSun2 and Dnmt2, the only other cytosine-5 RNA methyltransferase characterized to date, indicating that Dnmt2 was not functionally redundant with NSun2 in spermatogonial stem cells or Sertoli cells. Specific NSun2- and Dnmt2-methylated tRNAs decreased in abundance when both methyltransferases were deleted, suggesting that RNA methylation pathways play an essential role in male germ cell differentiation.
Collapse
Affiliation(s)
- Shobbir Hussain
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Francesca Tuorto
- Division of Epigenetics, German Cancer Research Center, Heidelberg, Germany
| | - Suraj Menon
- CR-UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Sandra Blanco
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Claire Cox
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Joana V. Flores
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Stephen Watt
- CR-UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Nobuaki R. Kudo
- IRDB, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Frank Lyko
- Division of Epigenetics, German Cancer Research Center, Heidelberg, Germany
| | - Michaela Frye
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
14
|
Ferrara D, Izzo G, Pariante P, Donizetti A, d'Istria M, Aniello F, Minucci S. Expression of prothymosin alpha in meiotic and post-meiotic germ cells during the first wave of rat spermatogenesis. J Cell Physiol 2010; 224:362-8. [DOI: 10.1002/jcp.22131] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
15
|
Wilkerson DC, Sarge KD. RNA polymerase II interacts with the Hspa1b promoter in mouse epididymal spermatozoa. Reproduction 2009; 137:923-9. [PMID: 19336471 DOI: 10.1530/rep-09-0015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Hspa1b (Hsp70.1) gene is one of the first genes expressed after fertilization, with expression occurring during the minor zygotic genome activation (ZGA) in the absence of stress. This expression can take place in the male pronucleus as early as the one-cell stage of embryogenesis. The importance of HSPA1B for embryonic viability during times of stress is supported by studies showing that depletion of this protein results in a significant reduction in embryos developing to the blastocyte stage. Recently, we have begun addressing the mechanism responsible for allowing expression of Hspa1b during the minor ZGA and found that heat shock transcription factor (HSF) 1 and 2 bind the Hspa1b promoter during late spermatogenesis. In this report, we have extended those studies using western blots and chromatin immunoprecipitation assays and found that RNA polymerase II (Pol II) is present in epididymal spermatozoa and bound to the Hspa1b promoter. These present results, in addition to our previous results, support a model in which the binding of HSF1, HSF2, SP1, and Pol II to the promoter of Hspa1b would allow the rapid formation of a transcription-competent state during the minor ZGA, thereby allowing Hspa1b expression.
Collapse
Affiliation(s)
- Donald C Wilkerson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | | |
Collapse
|
16
|
|
17
|
Zheng J, Xia X, Ding H, Yan A, Hu S, Gong X, Zong S, Zhang Y, Sheng HZ. Erasure of the paternal transcription program during spermiogenesis: the first step in the reprogramming of sperm chromatin for zygotic development. Dev Dyn 2008; 237:1463-76. [PMID: 18386827 DOI: 10.1002/dvdy.21499] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Male germ cells possess a unique epigenetic program and express a male-specific transcription profile. However, when its chromatin is passed onto the zygote, it expresses an transcription/epigenetic program characteristic of the zygote. The mechanism underlying this reprogramming process is not understood at present. In this study, we show that an extensive range of chromatin factors (CFs), including essential transcription factors and regulators, remodeling factors, histone deacetylases, heterochromatin-binding proteins, and topoisomerases, were removed from chromatin during spermiogenesis. This process will erase the paternal epigenetic program to generate a relatively naive chromatin, which is likely to be essential for installation of the zygotic developmental program after fertilization. We have also showed that transcription termination in male germ cells was temporally correlated with CF dissociation. A genome-wide CF dissociation will inevitably disassemble the transcription apparatus and regulatory mechanism and lead to transcription silence. Based on data presented in this and previous studies (Sun et al., Cell Research [2007] 17:117-134), we propose that paternal-zygotic transcription reprogramming begins with a genome-wide CF dissociation to erase the existing transcription program in later stages of spermatogenesis. This will be followed by assembling of the zygotic equivalent after fertilization. The transcription/epigenetic program of the male germ cell is transformed into a zygotic one using an erase-and-rebuild strategy similar to that used in the maternal-zygotic transition. It is also noted that transcription is terminated long after meiosis is completed and before chromatin becomes highly condensed during spermatogenesis. The temporal order of these events suggests that transcription silence does not have to be coupled to meiosis or chromatin condensation.
Collapse
Affiliation(s)
- Junke Zheng
- Center for Developmental Biology, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wilkerson DC, Murphy LA, Sarge KD. Interaction of HSF1 and HSF2 with the Hspa1b promoter in mouse epididymal spermatozoa. Biol Reprod 2008; 79:283-8. [PMID: 18434628 DOI: 10.1095/biolreprod.107.066241] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The Hspa1b gene is one of the first genes expressed after fertilization, with expression observed in the male pronucleus as early as the one-cell stage of embryogenesis. This expression can occur in the absence of stress and is initiated during the minor zygotic genome activation. There is a significant reduction in the number of embryos developing to the blastocyte stage when HSPA1B levels are depleted, which supports the importance of this protein for embryonic viability. However, the mechanism responsible for allowing expression of Hspa1b during the minor zygotic genome activation (ZGA) is unknown. In this report, we investigated the role of HSF1 and HSF2 in bookmarking Hspa1b during late spermatogenesis. Western blot results show that both HSF1 and HSF2 are present in epididymal spermatozoa, and immunofluorescence analysis revealed that some of the HSF1 and HSF2 proteins in these cells overlap the 4',6'-diamidino-2-phenylindole-stained DNA region. Results from chromatin immunoprecipitation assays showed that HSF1, HSF2, and SP1 are bound to the Hspa1b promoter in epididymal spermatozoa. Furthermore, we observed an increase in HSF2 binding to the Hspa1b promoter in late spermatids versus early spermatids, suggesting a likely period during spermatogenesis when transcription factor binding could occur. These results support a model in which the binding of HSF1, HSF2, and SP1 to the promoter of Hspa1b would allow the rapid formation of a transcription-competent state during the minor ZGA, thereby allowing Hspa1b expression.
Collapse
Affiliation(s)
- Donald C Wilkerson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | |
Collapse
|
19
|
Gonzalez-Herrera IG, Prado-Lourenco L, Pileur F, Conte C, Morin A, Cabon F, Prats H, Vagner S, Bayard F, Audigier S, Prats AC. Testosterone regulates FGF-2 expression during testis maturation by an IRES-dependent translational mechanism. FASEB J 2006; 20:476-8. [PMID: 16423876 DOI: 10.1096/fj.04-3314fje] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Spermatogenesis is a complex process involving cell proliferation, differentiation, and apoptosis. Fibroblast growth factor 2 (FGF-2) is involved in testicular function, but its role in spermatogenesis has not been fully documented. The control of FGF-2 expression particularly occurs at the translational level, by an internal ribosome entry site (IRES)-dependent mechanism driving the use of alternative initiation codons. To study IRES activity regulation in vivo, we have developed transgenic mice expressing a bicistronic construct coding for two luciferase genes. Here, we show that the FGF-2 IRES is age-dependently activated in mouse testis, whereas EMCV and c-myc IRESs are not. Real-time PCR confirms that this regulation is translational. By using immunohistological techniques, we demonstrate that FGF-2 IRES stimulation occurs in adult, but not in immature, type-A spermatogonias. This is correlated with activation of endogenous FGF-2 expression in spermatogonia; whereas FGF-2 mRNA transcription is known to decrease in adult testis. Interestingly, the FGF-2 IRES activation is triggered by testosterone and is partially inhibited by siRNA directed against the androgen receptor. Two-dimensional analysis of proteins bound to the FGF-2 mRNA 5'UTR after UV cross-linking reveals that testosterone treatment correlates with the binding of several proteins. These data suggest a paracrine loop where IRES-dependent FGF-2 expression, stimulated by Sertoli cells in response to testosterone produced by Leydig cells, would in turn activate Leydig function and testosterone production. In addition, nuclear FGF-2 isoforms could be involved in an intracrine function of FGF-2 in the start of spermatogenesis, mitosis, or meiosis initiation. This report demonstrates that mRNA translation regulation by an IRES-dependent mechanism participates in a physiological process.
Collapse
MESH Headings
- 5' Untranslated Regions
- Age Factors
- Androgen Receptor Antagonists
- Animals
- Codon
- Fibroblast Growth Factor 2/biosynthesis
- Fibroblast Growth Factor 2/genetics
- Fibroblast Growth Factor 2/physiology
- Genes, Reporter
- Genes, Synthetic
- Leydig Cells/physiology
- Luciferases, Renilla/genetics
- Male
- Meiosis
- Mice
- Mice, Transgenic
- Mitosis
- Paracrine Communication
- Peptide Chain Initiation, Translational/physiology
- Protein Biosynthesis
- Protein Isoforms/physiology
- RNA, Messenger/genetics
- RNA, Messenger/radiation effects
- RNA, Small Interfering/pharmacology
- Receptors, Androgen/genetics
- Recombinant Fusion Proteins/physiology
- Regulatory Sequences, Nucleic Acid
- Ribosomes/metabolism
- Sertoli Cells/physiology
- Spermatogenesis/physiology
- Testis/growth & development
- Testis/metabolism
- Testis/physiology
- Testosterone/metabolism
- Testosterone/pharmacology
- Testosterone/physiology
- Ultraviolet Rays
Collapse
|
20
|
Kimura T, Kosaka J, Nomura T, Yamada T, Miki Y, Takagi K, Kogami T, Sasaki J. Quantification of in situ hybridization signals in rat testes. J Histochem Cytochem 2004; 52:813-20. [PMID: 15150290 DOI: 10.1369/jhc.4a6249.2004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We performed basic research into quantifying in situ hybridization (ISH) signals in rat testis, a suitable organ for the quantification because germ cells undergo synchronized development and show stage-specific gene expression. In this model experiment, rRNA was selected as the hybridizable RNA in paraffin sections. Specimens fixed with Bouin's fixative and hybridized with digoxygenin-labeled probes could easily be analyzed quantitatively through "posterization" of the images. The amount of rRNA hybridized with the probe was greatest in early primary spermatocytes, followed by pachytene primary spermatocytes, then diplotene spermatocytes, and finally by secondary spermatocytes and spermatids. The amounts reached low levels in metaphase, anaphase, and telophase of meiotic division and early step 1 spermatids, and then slightly increased during spermiogenesis. ISH rRNA staining was a useful parameter for evaluation of the quantitative analysis of mRNA and the levels of hybridizable RNA in tissue sections.
Collapse
Affiliation(s)
- Touji Kimura
- Department of Cytology and Histology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Li M, Arimura A. Neuropeptides of the pituitary adenylate cyclase-activating polypeptide/vasoactive intestinal polypeptide/growth hormone-releasing hormone/secretin family in testis. Endocrine 2003. [PMID: 12721498 DOI: 10.1385/endo: 20: 3: 201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mammalian testicular development and the maintenance of spermatogenesis are hormone-dependent processes that are controlled by the pituitary gonadotropins and testosterone. Recent studies have demonstrated the presence of many neuropeptides and their receptors in the testis, suggesting that these peptides operate as local regulators of testicular germ cell development and function. Among these testicular neuropeptides, the peptides that belong to the pituitary adenylate cyclase-activating polypeptide (PACAP) family, particularly growth hormone-releasing hormone and secretin, appear to show some unique common features in terms of intratesticular localization and the time of expression during the spermatogenic cycle. However, their precise physiologic roles and mechanisms of action remain unknown. This review analyzes the available information on the functional interactions among the testicular cells that appear to be mediated by locally produced neuropeptides, with a special emphasis on the peptides of the PACAP family.
Collapse
Affiliation(s)
- Min Li
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
| | | |
Collapse
|
22
|
Li M, Arimura A. Neuropeptides of the pituitary adenylate cyclase-activating polypeptide/vasoactive intestinal polypeptide/growth hormone-releasing hormone/secretin family in testis. Endocrine 2003; 20:201-14. [PMID: 12721498 DOI: 10.1385/endo:20:3:201] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2002] [Revised: 11/25/2002] [Accepted: 12/09/2002] [Indexed: 12/12/2022]
Abstract
Mammalian testicular development and the maintenance of spermatogenesis are hormone-dependent processes that are controlled by the pituitary gonadotropins and testosterone. Recent studies have demonstrated the presence of many neuropeptides and their receptors in the testis, suggesting that these peptides operate as local regulators of testicular germ cell development and function. Among these testicular neuropeptides, the peptides that belong to the pituitary adenylate cyclase-activating polypeptide (PACAP) family, particularly growth hormone-releasing hormone and secretin, appear to show some unique common features in terms of intratesticular localization and the time of expression during the spermatogenic cycle. However, their precise physiologic roles and mechanisms of action remain unknown. This review analyzes the available information on the functional interactions among the testicular cells that appear to be mediated by locally produced neuropeptides, with a special emphasis on the peptides of the PACAP family.
Collapse
Affiliation(s)
- Min Li
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
| | | |
Collapse
|
23
|
Vigodner M, Lewin LM, Shochat L, Mittelman L, Golan R. Meiosis in the golden hamster: a confocal microscopy and flow cytometric analysis. Mol Reprod Dev 2003; 64:86-95. [PMID: 12420303 DOI: 10.1002/mrd.10212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In this study, confocal microscopy and flow-cytometry were utilized to follow meiosis in hamster spermatogenesis. Confocal microscopy was used as an analytical tool to observe spermatocytes inside the tubules following meiotic progression consecutively at defined spermatogenic stages. To study spermatocyte differentiation, the structure of the synaptonemal complex was studied in detail at various stages of hamster spermatogenesis using the antibody against SC3 (the protein of axial/lateral element). The synaptonemal complex was observed from the leptotene stage until the first meiotic division with maximal staining in mid-pachytene spermatocytes, suggesting a role for SC3 at this postrecombinational stage. In addition, 3-dimensional (3D) images of synaptonemal complex were observed, providing information about spatial distribution of the chromosomes within the nuclei of spermatocytes at different stages of meiosis. Changes in spermatocyte sizes and DNA condensation allowed assessment of meiosis by flow cytometry. Changes in chromatin condensation at different stages of hamster meiosis were followed, revealing decondensation from early to late pachytene stages. The analysis also allowed a comparing of chromatin status of mitotic and meiotic chromosomes, confirming the less compact structure of the latter, possibly connected to increased transcriptional activity during meiosis.
Collapse
Affiliation(s)
- M Vigodner
- Department of Clinical Biochemistry, Sackler Medical School, Tel Aviv University, Ramat Aviv, Israel.
| | | | | | | | | |
Collapse
|
24
|
Aguilar-Mahecha A, Hales BF, Robaire B. Chronic cyclophosphamide treatment alters the expression of stress response genes in rat male germ cells. Biol Reprod 2002; 66:1024-32. [PMID: 11906922 DOI: 10.1095/biolreprod66.4.1024] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Increases in the survival rate of men treated with chemotherapeutic drugs and their desire to have children precipitate concerns about the effects of these drugs on germ cells. Azoospermia, oligospermia, and infertility are common outcomes resulting from treatment with cyclophosphamide, an alkylating agent. Exposure of male rats to cyclophosphamide results in dose-dependent and time-specific adverse effects on progeny outcome. Elucidation of the effects of chronic low-dose cyclophosphamide treatment on the expression of stress response genes in male germ cells may provide insight into the mechanisms underlying such adverse effects. Male rats were gavaged with saline or cyclophosphamide (6 mg/kg) for 4-5 wk; pachytene spermatocytes, round spermatids, and elongating spermatids were isolated; RNA was extracted and probed on cDNA arrays containing 216 cDNAs. After saline treatment, 125 stress response genes were expressed in pachytene spermatocytes (57% of genes studied), 122 in round spermatids (56%), and 83 in elongating spermatids (38%). Cyclophosphamide treatment reduced the number of genes detected in all germ cell types. The predominant effect of chronic cyclophosphamide exposure was to decrease the expression level of genes in pachytene spermatocytes (34% of genes studied), round spermatids (29%), and elongating spermatids (4%). In elongating spermatids only, drug treatment increased the expression of 8% of the genes studied. The expression profiles of genes involved in DNA repair, posttranslational modification, and antioxidant defense in male germ cells were altered by chronic cyclophosphamide treatment. We hypothesize that the effects of cyclophosphamide exposure on germ cell gene expression during spermatogenesis may have adverse consequences on male fertility and progeny outcome.
Collapse
Affiliation(s)
- Adriana Aguilar-Mahecha
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montreal, Quebec, Canada H3G 1Y6
| | | | | |
Collapse
|
25
|
Aguilar-Mahecha A, Hales BF, Robaire B. Acute cyclophosphamide exposure has germ cell specific effects on the expression of stress response genes during rat spermatogenesis. Mol Reprod Dev 2001; 60:302-11. [PMID: 11599041 DOI: 10.1002/mrd.1092] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Exposure of male rats to cyclophosphamide, a commonly used anticancer and immunosuppressive drug, has been shown to alter fertility and progeny outcome in a male germ cell phase-specific manner. The effect of toxicant exposure on male germ cells depends in part on the stress response mechanisms present during the different stages of spermatogenesis. To assess how acute cyclophosphamide exposure affects the expression of stress response genes, we examined the expression of 216 genes, using gene expression arrays, in isolated rat spermatogenic cell types (pachytene spermatocytes, round spermatids, and elongating spermatids). Cyclophosphamide exposure affected gene expression in all cell types but most dramatically in round spermatids. Increased transcript levels were observed for 30 genes in round spermatids compared to seven genes in pachytene spermatocytes and two in elongating spermatids. The expression of genes involved in apoptosis, DNA-damage recognition and repair, transcriptional activation, and in the heat shock protein-chaperone response was most affected by cyclophosphamide in round spermatids. Our results demonstrate that cyclophosphamide alters the expression of stress response genes during spermatogenesis in a germ cell-specific manner. The greater response of round spermatids to cyclophosphamide suggests that this cell type may be more susceptible to the damaging effects induced by this drug, possibly due to the chromatin remodeling that is taking place at this stage of spermatogenesis. This observation is consistent with the reported higher level of abnormal progeny outcome seen when the germ cells were first exposed to cyclophosphamide as round spermatids.
Collapse
Affiliation(s)
- A Aguilar-Mahecha
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir-William-Osler, Montréal, Québec, Canada H3G 1Y6
| | | | | |
Collapse
|
26
|
Aguilar-Mahecha A, Hales BF, Robaire B. Expression of stress response genes in germ cells during spermatogenesis. Biol Reprod 2001; 65:119-27. [PMID: 11420231 DOI: 10.1095/biolreprod65.1.119] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
During germ cell development different spermatogenic cell types show remarkable variation in their susceptibility to stressful stimuli. Various cellular mechanisms are triggered in germ cells after exposure to stress, but the expression of only a few of the genes involved in such pathways has been studied during spermatogenesis. In the present study we determined the expression profiles of 216 stress response genes in isolated rat germ cells (pachytene spermatocytes, and round and elongating spermatids) using cDNA atlas arrays. Of the 216 genes studied, 86 were detected in pachytene spermatocytes, 82 in round spermatids, and 52 in elongating spermatids. Fifty percent (48) of the total number of genes detected during spermatogenesis were detected in all three cell types while nearly 25% (25) were expressed exclusively in pachytene spermatocytes and round spermatids; some cell specific transcripts were observed also. The use of the K means clustering method allowed us to group genes by their pattern of expression during spermatogenesis; five specific expression profiles were obtained and analyzed. To determine how stress response genes are regulated throughout spermatogenesis, we examined the expression of genes involved in stress response mechanisms such as heat shock proteins-chaperones, DNA repair, and oxidative stress. Genes belonging to these families were differentially expressed during germ cell development. We suggest that the differential expression of stress response genes during spermatogenesis contributes to the selectivity of the susceptibility of germ cells to stress.
Collapse
Affiliation(s)
- A Aguilar-Mahecha
- Departments of Pharmacology and Therapeutics, and of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada H3G 1Y6
| | | | | |
Collapse
|
27
|
Catalano RD, Hillhouse EW, Vlad M. Developmental expression and characterization of FS39, a testis complementary DNA encoding an intermediate filament-related protein of the sperm fibrous sheath. Biol Reprod 2001; 65:277-87. [PMID: 11420250 DOI: 10.1095/biolreprod65.1.277] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Proteins immunologically related to intermediate filaments have been identified in the sperm fibrous sheath but remain uncharacterized. We isolated and characterized a novel intermediate filament-related protein (FS39) localized to the fibrous sheath of the sperm tail. We used Northern blot analysis to establish that FS39 is transcribed predominantly in the testis of mice >18-20 days old. At this age, spermatogenesis has proceeded to the development of the first round haploid spermatids. In situ hybridization revealed that FS39 mRNA is first detectable in late step 3 spermatids, is at its highest level during steps 9 and 10, and diminishes in steps 13 and 14. Western blot analysis identified a single protein of 39 kDa in mouse and rat testis and epididymis, suggesting the protein is conserved in rodents. Indirect immunofluorescence localized FS39 to the fibrous sheath of the sperm tail, and in testis sections expression was detected from step 13 and step 14 spermatids onward, indicating FS39 is under translational control. Southern blot analysis showed FS39 to be a single copy gene, and hybridization to human genomic DNA suggested that a human equivalent gene is present. These results demonstrate that FS39 is transcribed in testis tissue during the haploid phase of spermatogenesis, is present in mature sperm, and codes for a novel 39-kDa intermediate filament-related protein of the fibrous sheath.
Collapse
Affiliation(s)
- R D Catalano
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, United Kingdom.
| | | | | |
Collapse
|
28
|
Hundscheid RD, Sistermans EA, Thomas CM, Braat DD, Straatman H, Kiemeney LA, Oostra BA, Smits AP. Imprinting effect in premature ovarian failure confined to paternally inherited fragile X premutations. Am J Hum Genet 2000; 66:413-8. [PMID: 10677300 PMCID: PMC1288093 DOI: 10.1086/302774] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Fragile X premutations are considered to be a risk factor for premature ovarian failure (POF), which is usually defined as menopause at age <40 years. Since premutations may be inherited from either the mother or the father, we evaluated the influence of the inheritance pattern on the duration of reproductive life in female carriers. The occurrence of POF and age at menopause in women with a paternally inherited fragile X premutation (PIP) were compared to those in women with a maternally inherited fragile X premutation (MIP). We identified 148 women in whom the parental origin of the premutation could be determined. In 109 of these women we were able to establish whether POF had occurred: 82 women had a PIP, and 27 had a MIP. Twenty-three of the women (28%) with a PIP had POF, versus only 1 (3.7%) with a MIP (two -tailed Fisher's exact test; P=. 007). Kaplan-Meier analysis of all 148 premutations showed that the age at menopause was significantly lower in the women with a PIP than in the woman with a MIP (Breslow test in Kaplan-Meier analysis; P=.003). Our data strongly suggest that, when POF occurs in fragile X premutation carriers, a considerable proportion of the premutations are inherited paternally (parent-of-origin effect). We hypothesize that this may be owing to a paternal genomic imprinting effect.
Collapse
Affiliation(s)
- R D Hundscheid
- Department of Human Genetics, University Hospital Nijmegen, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
The many events of meiotic prophase can now be viewed as a series of specialized incidents that are monitored by meiotic checkpoints, some of which are similar to their mitotic counterparts, and some of which are probably unique to meiosis. This shift in perspective means that meiotic sterility in mammals must be reexamined and viewed as the result of errors subject to meiotic checkpoint controls. Like their mitotic counterparts, the meiotic checkpoints detect defects and halt normal progression until these mistakes can be repaired. Some of these checkpoints utilize mitotic checkpoint proteins, others may involve meiotic-specific proteins, or splice forms. If repair is impossible, the checkpoints then either trigger immediate apoptosis or cause an arrest of meiotic progression followed by eventual cell death. If a sufficient number of spermatocytes are involved, either alternative results in sterility. Identification of these meiotic checkpoints and delineation of the signal transduction cascades involved has only just begun. While yeast, or other model organisms, may provide clues to some of these pathways, others appears to have arisen during vertebrate evolution. The study of mammalian meiosis has entered a new era and the foundations are being laid for a growing understanding of the many problems that may contribute to sterility.
Collapse
Affiliation(s)
- T Ashley
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
30
|
Lassalle B, Ziyyat A, Testart J, Finaz C, Lefèvre A. Flow cytometric method to isolate round spermatids from mouse testis. Hum Reprod 1999; 14:388-94. [PMID: 10099985 DOI: 10.1093/humrep/14.2.388] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The purpose of this study was to isolate pure populations of round spermatids from mouse testis by flow cytometry followed by cell sorting. Cell suspensions from mouse testis were enriched in germ cells by centrifugation on a discontinuous Percoll gradient, then analysed using a FACScalibur flow cytometer measuring the cell size and density. A large and well-delimited population of cells (R1) expected to contain round spermatids was observed on the dot plot diagram. Sorted R1 cells were very homogeneous in size (approximately 11 microns) and displayed the characteristic cytological aspect of round spermatids. Spermatid-specific gene expression was confirmed by reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of R1 cells using primers for protamine 2 gene (PRM2) and SP-10. A positive signal for SP-10 was obtained with a single cell using nested primers. The 5.5 kb transcript of c-kit, which is not expressed in spermatids, was not detected by nested RT-PCR, excluding a contamination with spermatogonia. Our results clearly established that flow cytometry followed by cell sorting allows the isolation of a highly homogeneous population of round spermatids from the testis.
Collapse
Affiliation(s)
- B Lassalle
- Unité Maturation Gamètique et Fécondation, INSERM unité 355 (Maturation gamètique et fécondation), Clamart, France
| | | | | | | | | |
Collapse
|
31
|
Mahoney MG, Tang W, Xiang MM, Moss SB, Gerton GL, Stanley JR, Tseng H. Translocation of the zinc finger protein basonuclin from the mouse germ cell nucleus to the midpiece of the spermatozoon during spermiogenesis. Biol Reprod 1998; 59:388-94. [PMID: 9687312 DOI: 10.1095/biolreprod59.2.388] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Basonuclin was first described as a human keratinocyte zinc finger protein present in the nuclei of proliferative basal keratinocytes in the epidermis. It disappears from keratinocytes that have lost their proliferative ability and have entered terminal differentiation. We now report that basonuclin is present also in the germ cells of the mouse testis and ovary. Immunocytochemical staining detected basonuclin in the nuclei of spermatogonia and spermatocytes at various developmental stages. During spermiogenesis, it relocated from the nucleus to the midpiece of the flagellum of the spermatozoa. In the ovary, basonuclin was found mainly in the nuclei of developing oocytes. The dual presence of basonuclin in differentiated spermatozoa and oocytes suggests that it may play a role in their differentiation and the early development of an embryo.
Collapse
Affiliation(s)
- M G Mahoney
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Schalles U, Shao X, van der Hoorn FA, Oko R. Developmental expression of the 84-kDa ODF sperm protein: localization to both the cortex and medulla of outer dense fibers and to the connecting piece. Dev Biol 1998; 199:250-60. [PMID: 9698445 DOI: 10.1006/dbio.1998.8931] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Outer dense fibers (ODF) are specialized cytoskeletal elements of the mammalian sperm tail which are composed of several prominent proteins. We previously reported the isolation of a cDNA (111-450) encoding a putative 84-kDa ODF protein. Here we demonstrate by independent cDNA isolations and by translational/immunoprecipitation of testicular mRNAs using anti-ODF 84 antibodies that 111-450 cDNA encodes the 84-kDa protein. We then analyzed the testicular expression of the ODF 84 mRNA and protein. Riboprobes generated from the clones recognized four testicular-specific transcripts of 1.6, 2.2, 2.4, and 2.8 kb in both rat and bull of which the immunoprecipitable product of the 2.4-kb mRNA comigrates with ODF 84 protein. Developmental Northerns indicated that the 2.2- and 2.4-kb mRNAs are first transcribed during meiotic prophase while the other two species are first expressed in round spermatids. The levels of all the transcripts steadily increased up to elongated spermatids. Immunocytochemistry revealed that the anti-84 reactive ODF proteins were synthesized and assembled in the cytoplasm of elongated spermatids (steps 9-18) with peak activity occurring in step 16 of spermiogenesis. Immunogold labeling was selective to the assembling ODF and connecting piece of the tail and to granulated bodies of the cytoplasmic lobe. Both the striated collar and capitulum of the connecting piece were immunolabeled as well as the basal plate of the implantation fossa. A combination of pre- and postembedding immunogold labeling provided evidence that the 84-kDa ODF protein is localized to both the cortex and medulla of the ODF in contrast to the sole medullary localization of the major 27-kDa ODF protein. Thus the 84-kDa ODF protein, encoded by the 2.4 transcript, is translationally regulated, packaged after synthesis into granulated bodies, assembled in a proximal to distal direction along the axoneme and may interact by means of leucine zippers specifically with the 27-kDa ODF protein during assembly. Its localization to both the cortex and medulla of the ODF, as opposed to exclusive medullary localization of the 27-kDa ODF protein, and the presence of two leucine zippers, only one of which interacts with the 27-kDa ODF, suggests that it could act as a link between proteins of the two regions of the ODF.
Collapse
Affiliation(s)
- U Schalles
- Department of Anatomy and Cell Biology, Queen's University, Kingston, Ontario, Canada
| | | | | | | |
Collapse
|
33
|
Abstract
The inactive X chromosome differs from the active X in a number of ways; some of these, such as allocyclic replication and altered histone acetylation, are associated with all types of epigenetic silencing, whereas others, such as DNA methylation, are of more restricted use. These features are acquired progressively by the inactive X after onset of initiation. Initiation of X-inactivation is controlled by the X-inactivation center (Xic) and influenced by the X chromosome controlling element (Xce), which causes primary nonrandom X-inactivation. Other examples of nonrandom X-inactivation are also presented in this review. The definition of a major role for Xist, a noncoding RNA, in X-inactivation has enabled investigation of the mechanism leading to establishment of the heterochromatinized X-chromosome and also of the interactions between X-inactivation and imprinting as well as between X-inactivation and developmental processes in the early embryo.
Collapse
Affiliation(s)
- E Heard
- Unité de Génétique Moléculaire Murine, URA CNRS 1968, Institut Pasteur, Paris, France.
| | | | | |
Collapse
|
34
|
Abstract
Pairing between homologous chromosomes is essential for successful meiosis; generally only paired homologs recombine and segregate correctly into haploid germ cells. Homologs also pair in some somatic cells (e.g. in diploid and polytene cells of Drosophila). How homologs find their partners is a mystery. First, I review some explanations of how they might do so; most involve base-pairing (i.e. DNA-DNA) interactions. Then I discuss the remarkable fact that chromosomes only pair when they are transcriptionally active. Finally, I present a general model for pairing based upon the DNA-protein interactions involved in transcription. Each chromosome in the haploid set has a unique array of transcription units strung along its length. Therefore, each chromatin fibre will be folded into a unique array of loops associated with clusters of polymerases and transcription factors; only homologs share similar arrays. As these loops and clusters, or transcription factories, move continually, they make and break contact with others. Correct pairing would be nucleated when a promoter in a loop tethered to one factory binds to a homologous polymerizing site in another factory, before transcription stabilizes the association. This increases the chances that adjacent promoters will bind to their homologs, so that chromosomes eventually become zipped together with their partners. Pairing is then the inevitable consequence of transcription of partially-condensed chromosomes.
Collapse
Affiliation(s)
- P R Cook
- CRC Nuclear Structure and Function Research Group, Sir William Dunn School of Pathology, University of Oxford, UK.
| |
Collapse
|
35
|
Abstract
The goals of this work were to create germ-cell-stage-specific cDNA libraries from mouse spermatogenic cells and to employ a novel two-step genetic screen to identify gene sequences present during the critical meiotic stage of spermatogenesis. Highly enriched germ-cell fractions were prepared from adult and juvenile mouse testes, and purity of these fractions was extensively analyzed by light and electron microscopy. Standard techniques were used to prepare cDNA libraries from populations of mixed leptotene and zygotene (L/Z) spermatocytes, pachytene (P) spermatocytes, and round spermatids. These libraries were analyzed with respect to representation of sequences from ubiquitously expressed genes, and from genes expressed at specific germ-cell stages as well as from genes expressed in testicular somatic cells. For the first step of the screening procedure, testicular cDNA was prepared from mutant mice carrying the T(X;11)38H chromosomal translocation that causes spermatogenic arrest at early meiotic prophase. This mixed cDNA probe was used to screen the libraries from L/Z and P spermatocytes to detect sequences failed to hybridize. The clones identified were characterized for ability to hybridize to various germ-cell-specific cDNAs to verify that they represented sequences present in normal spermatogenic meiotic cells. These clones were then subjected to a second screening with another mutant probe; this time the cDNA probe was from testes of sterile mice bearing the T(X;16)16H chromosomal translocation that causes spermatogenic arrest at late meiotic prophase. This screen identified 27 clones that were not represented in testicular cDNA from T38-bearing mice or from T16-bearing mice. These clones may represent sequences essential for normal completion of the genetic events of meiosis during spermatogenesis. Likewise, the secondary screen identified 19 clones that were not represented in testicular cDNA from T38-bearing mice but were represented in testicular cDNA of T16-bearing mice. These clones are thus gene sequences present in spermatogenic cells during the time from early meiotic prophase to mid-to-late prophase. This strategy represents the first use of genetic aberrations in differential screening to identify genes expressed at specific times during mammalian spermatogenesis.
Collapse
Affiliation(s)
- K A Caldwell
- Department of Zoology, University of Tennessee, Knoxville, USA
| | | | | |
Collapse
|
36
|
Papp S, Robaire B, Hermo L. Immunocytochemical localization of the Ya, Yc, Yb1, and Yb2 subunits of glutathione S-transferases in the testis and epididymis of adult rats. Microsc Res Tech 1995; 30:1-23. [PMID: 7711317 DOI: 10.1002/jemt.1070300102] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Glutathione S-transferases (GSTs) are dimeric proteins that come from a multigene family. They can be grouped into five classes (alpha, mu, pi, sigma, theta) based on the degree of amino acid homology of their subunits. These GST isozymes are able to catalyze the conjugation of glutathione with a wide variety of electrophiles, thereby protecting important cellular constituents from electrophilic attack. In the present study, the distribution of the Ya and Yc subunits from the alpha family, as well as the Yb1 and Yb2 subunits from the mu gene family was examined using immunocytochemistry in the adult rat testis and epididymis. The results of these four GST subunits were also compared with two other subunits, the Yf and Yo proteins, which have already been investigated in our laboratory [Veri et al. (1993), J. Androl., 14:23-44; Veri et al. (in press), J. Androl.]. In the testis, Leydig cells were intensely stained for all six subunits. Within the seminiferous epithelium, Sertoli cells were reactive only for antibodies raised against the Ya, Yb1 and Yf subunits. Among germ cells, all spermatogonia, spermatocytes and step 1-15 spermatids were virtually unreactive for each of the six GSTs. However, moderate to intense staining was seen over steps 16-19 spermatids with the anti-Yo and anti-Ya antibodies, and intense staining over step 19 spermatids with the anti-Yb1 and anti-Yb2 antibodies. In the rete testis, Yf, Yo, Yb1, and Yb2 subunits were intensely reactive over the epithelial cells with weak staining for Yc and no staining for Ya antibodies. Interestingly, in the efferent ducts the Yc, Yb1, and Yf proteins were intensely reactive over ciliated cells, whereas only the Yc protein was intensely reactive over nonciliated cells. In the epididymis, immunoreactivity varied among the principal and basal cells of a given epididymal region for each GST antibody. In the case of principal cells, several of the GSTs showed a similar immunostaining pattern along the tubule. Although not identical in intensity of reaction, the Yc, Yb1, Ya and Yo proteins showed an increase in staining intensity from the proximal to distal segments of the epididymis. In contrast, the Yb2 protein was intensely expressed only in the distal caput with weak levels throughout the rest of the epididymis. The Yf reactivity was strongest from the distal initial segment to the distal caput and unreactive in the corpus and proximal cauda epididymidis.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- S Papp
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
37
|
Papp S, Robaire B, Hermo L. Developmental expression of the glutathione S-transferase Yo subunit in the rat testis and epididymis using light microscope immunocytochemistry. Anat Rec (Hoboken) 1994; 240:345-57. [PMID: 7825731 DOI: 10.1002/ar.1092400307] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Glutathione S-transferases (GSTs) are a family of isozymes that catalyze the conjugation of glutathione with various toxic electrophilic compounds. GSTs are composed of several classes based on the degree of sequence homology of their subunits. The Yo subunit, a member of the mu class, is expressed at high levels in the testis and epididymis. The purpose of this study was to immunolocalize the GST-Yo in these tissues during development. METHODS The testes and epididymides of rats aged 7, 15, 21, 28, 39, 42, 45, 49, and 56 days were fixed in Bouin's fixative, and immunostained for light microscopic analysis. RESULTS In the testis the cytoplasm of all germ cells was unreactive until day 39. At that time, step 18 spermatids appeared moderately reactive, while the few observed step 19 spermatids were intensely reactive as were their residual bodies. The presence of residual bodies indicates that spermiation takes place as early as day 39; however, the number of step 19 spermatids is low at this age. A progressive increase in the size of the tubule and number of elongating spermatids was seen between days 42 and 49. In addition, by day 49, a weak staining was observed in steps 12-15, moderate in steps 16-17, and intense in steps 18-19 spermatids. In terms of the intensity of staining, cell types stained, size of the tubules, and number of elongating spermatids, no difference was noted between day 49, 56, and adult animals. Thus Yo protein expression in germ cells reached maturity by day 49. The epithelial cells of the rete testis were intensely reactive at day 7 and remained so throughout development. In contrast, while the epithelial cells of the efferent ducts at day 7 were intensely reactive, they were weakly reactive by day 39 and remained so at later ages. Along the entire epididymis, the columnar epithelial cells showed a moderate apical/supranuclear reaction from day 7 to 28. By day 39 principal cells of the initial segment became weakly reactive, while those in the caput and corpus were moderately stained, a situation seen at later ages including adults. Only by day 49 did principal cells of the proximal cauda become moderately stained as seen in adult animals. Thus the expression of the Yo protein in the principal cells of the proximal cauda may be regulated by different factors than those of the caput and corpus epididymidis. Alternatively, the expression of the Yo subunit in principal cells of the proximal cauda may develop later since this region would be the last to receive luminally derived testicular products. In the initial segment, the decrease in staining of principal cells at day 39 may be due to an inhibiting factor emanating from the testis. Spermatozoa appeared in the lumen of each epididymal region well after the expression of Yo had reached its adult staining pattern indicating that they are not a factor. CONCLUSIONS Overall these results suggest that the expression of GST-Yo in the various cells of the testis and epididymis are controlled by different factors during postnatal development.
Collapse
Affiliation(s)
- S Papp
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
38
|
Maguire SM, Millar MR, Sharpe RM, Saunders PT. Stage-dependent expression of mRNA for cyclic protein 2 during spermatogenesis is modulated by elongate spermatids. Mol Cell Endocrinol 1993; 94:79-88. [PMID: 8375577 DOI: 10.1016/0303-7207(93)90054-n] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cyclic protein 2 (CP-2) is a product of the Sertoli cell which is secreted in a cyclical manner according to the stage of the spermatogenic cycle. This study has assessed the influence of the germ cell complement on expression of CP-2 mRNA. Adult rats were treated with 650 mg/kg methoxyacetic acid (MAA) to induce the specific depletion of > 80% of pachytene and later spermatocytes from most tubules, and expression of CP-2 mRNA was then assessed at various times after treatment when particular germ cell types were depleted selectively. CP-2 mRNA was specifically localised to the Sertoli cells of the seminiferous tubules by non-radioactive in situ hybridisation using a digoxigenin-labelled riboprobe. A stage specific variation in CP-2 mRNA levels was observed, with the mRNA being most abundant at stages IV-VII of the spermatogenic cycle. Northern analysis revealed that treatment with MAA led to an apparent increase in the amount of the major 1.7 kb CP-2 transcript when either pachytene spermatocytes or round spermatids were depleted. In contrast, the level of CP-2 mRNA was decreased by more than half at 21 days after MAA treatment. This decrease was confirmed by in situ hybridisation at 21 days after MAA treatment, when CP-2 mRNA expression was found to be decreased or absent from tubules at stages at which CP-2 mRNA is normally expressed (stages IV-VII) when elongate spermatids were depleted selectively from these tubules. These observations lead us to hypothesise that elongate spermatids positively modulate CP-2 mRNA expression in the Sertoli cell.
Collapse
Affiliation(s)
- S M Maguire
- MRC Reproductive Biology Unit, Edinburgh, UK
| | | | | | | |
Collapse
|
39
|
Abstract
High-dose boric acid (BA) exposure produces testicular lesions in adult rats characterized by inhibited spermiation that may progress to nonrecoverable atrophy. The mechanism for the testicular toxicity of BA is unknown. To examine possible direct effects, the present study evaluated selected aspects of various testicular cell culture systems after in vitro BA exposure. Specifically, the hallmarks of the BA testicular toxicity were addressed: the mild hormone effect, the initial inhibition of spermiation, and atrophy. No effect of BA on the steroidogenic function of isolated Leydig cells was observed, supporting the contention of a CNS-mediated rather than a direct hormone effect. Since increased testicular cyclic AMP (cAMP) produces inhibited spermiation, and a role for the serine proteases plasminogen activators (PAs) in spermiation has been proposed, we evaluated both Sertoli cell cAMP accumulation in Sertoli-germ cell cocultures and the stage-specific secretion of PA activity in cultured seminiferous tubules after in vitro BA exposure, respectively. The results showed that the inhibited spermiation is not due to BA effects on either process. To address the atrophy, we evaluated BA effects in Sertoli-germ cell cocultures on 1) morphology/germ cell attachment, which might identify a target cell; 2) Sertoli cell energy metabolism, because lactate, secreted by Sertoli cells, is a preferred energy source for germ cells; and 3) DNA/RNA synthesis, because germ cells synthesize DNA/RNA and BA impairs nucleic acid synthesis in liver and may do so in testis. Despite the absence of overt morphologic changes and germ cell loss, the most sensitive in vitro endpoint was DNA synthesis of mitotic/meiotic germ cells, with energy metabolism in Sertoli or germ cells affected to a lesser extent. A re-evaluation of testis sections from rats exposed to BA revealed a decrease in the early germ cell/Sertoli cell ratio prior to atrophy. Thus, although the mechanism for the inhibited spermiation is still undefined and is the subject of future work, these combined studies revealed some changes offering a plausible explanation for the atrophy aspect of the BA testicular lesion.
Collapse
Affiliation(s)
- W W Ku
- Developmental and Reproductive Toxicology Group, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | | | | |
Collapse
|
40
|
Rossi P, Dolci S, Albanesi C, Grimaldi P, Geremia R. Direct evidence that the mouse sex-determining gene Sry is expressed in the somatic cells of male fetal gonads and in the germ cell line in the adult testis. Mol Reprod Dev 1993; 34:369-73. [PMID: 7682420 DOI: 10.1002/mrd.1080340404] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Reverse transcriptase polymerase chain reaction (RT-PCR) analysis shows that Sry mRNA is expressed in male fetal urogenital ridges from 12.5 day p.c. embryos, but not in enriched populations of primordial germ cells from the same embryos, indicating that Sry is expressed in the somatic cells of the embryonal gonad at the time of testis determination. We also show that, in the adult male mouse testis, Sry mRNA is expressed at high levels in meiotic and postmeiotic germ cells and, at much lower levels, also in Sertoli cells. Treatment with cyclic adenosine monophosphate (cAMP) analogs of cultured Sertoli cells from postnatal testis completely abolishes Sry mRNA expression.
Collapse
Affiliation(s)
- P Rossi
- Dipartimento di Sanità Pubblica e Biologia Cellulare, Università degli Studi di Roma, Tor Vergata, Italy
| | | | | | | | | |
Collapse
|
41
|
Abstract
The complete cDNA coding for mouse P68 RNA helicase was cloned and its nucleotide sequence was determined. The sequence is about 95% identical to the human equivalent. Whereas the 5'-untranslated region is less conserved (71%), the 3'-ends of mouse and human mRNAs are nearly identical. Between stop codon and poly(A)-tail both sequences are 97% conserved. At the level of amino acid sequence, the similarity of both, mouse and human, DEAD box family proteins is as high as 98%. In situ hybridizations using cDNA subfragments as probes revealed a testis-selective expression of P68 RNA helicase mRNA. The signal was restricted to late pachytene spermatocytes and haploid spermatids. Northern blot analyses corroborated these results but suggested that expression of related mRNA species occurs in a variety of other tissues.
Collapse
Affiliation(s)
- L Lemaire
- Institut für Genetik, Heinrich-Heine-Universität, Düsseldorf, Germany
| | | |
Collapse
|
42
|
Smith A, Benavente R. Meiosis-specific protein selectively associated with sex chromosomes of rat pachytene spermatocytes. Proc Natl Acad Sci U S A 1992; 89:6938-42. [PMID: 1495983 PMCID: PMC49620 DOI: 10.1073/pnas.89.15.6938] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
During the first meiotic prophase of mammalian spermatogenesis, the sex chromosomes X and Y show a characteristic allocyclic behavior with respect to the autosomes. This is particularly evident during pachytene stage when sex chromosomes form the so-called sex vesicle. This structure is characterized by the condensed state of chromatin, transcriptional inactivity, and the limited extension of chromosome pairing, which is usually restricted to a short segment of sex chromosome axial elements. The molecular basis and functional significance of sex vesicle formation during mammalian spermatogenesis remain obscure. Here we report on the identification of a meiosis-specific sex vesicle protein we called XY40. Immunocytochemical localization on rat testis cryosections with a XY40-specific monoclonal antibody revealed that the labeling is confined to the axial elements of sex chromosomes. Biochemical characterization showed that protein XY40 (40 kDa; pI 5.7-5.8) can be extracted from rat pachytene spermatocytes and recovered in particles of 9.5 S with a native molecular mass of approximately 152 kDa. We speculate that protein XY40 may be involved in the allocyclic behavior of sex chromosomes during male meiotic prophase.
Collapse
Affiliation(s)
- A Smith
- Department of Zoology I, University of Würzburg, Federal Republic of Germany
| | | |
Collapse
|
43
|
Abstract
Our results on the formation of the ODF and perforatorium are diagrammatically summarized in Figures 30 and 31. The developmental expression of proteins making up these two cytoskeletal elements differs in timing, duration and intracellular localization. The ODF proteins are synthesized exclusively during the latter part of spermiogenesis, well after transcriptional activity in the haploid germ cell nucleus has ended. This implies that these major integral proteins of the tail are translationally regulated and that mechanisms must exist for the storage and eventual release of the mRNAs encoding these proteins. The perforatorial proteins, on the other hand, begin to be synthesized during the meiotic prophase reaching a peak of production in early spermiogenesis just before the initiation of the condensation of the spermatid's nucleus, at which time RNA synthesis stops. Another major difference between ODF and perforatorial protein production is that there seems to be a coordinated activity between the synthesis and the assembly of the ODF proteins, whereas there appears to be an almost 25 day delay between the initial meiotic synthesis and final condensation of perforatorial proteins in the subacrosomal space at the end of spermiogenesis. As for the intracellular localization of ODF and perforatorial proteins both have unprecedented distributions. The ODF proteins appear to be concentrated in a particular type of granular body which is especially abundant in the elongated spermatid at the time of peak ODF assembly. The perforatorial proteins, on the other hand, appear to be concentrated in the nuclei of pachytene spermatocytes and round spermatids until their displacement into the cytoplasm during nuclear condensation. Both forms of localization suggest a storage role for these proteins uniquely adapted by the spermatid to regulate the assemblies of the respective cytoskeletal elements.
Collapse
Affiliation(s)
- R J Oko
- Department of Anatomy, McGill University, Montréal, Québec, Canada
| | | |
Collapse
|
44
|
Raman R, Das P. Mammalian sex chromosomes. III. Activity of pseudoautosomal steroid sulfatase enzyme during spermatogenesis in Mus musculus. SOMATIC CELL AND MOLECULAR GENETICS 1991; 17:429-33. [PMID: 1763383 DOI: 10.1007/bf01233166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Parallel to the inactivation of the X chromosome in somatic cells of female, the male X in mammals is rendered inactive during spermatogenesis. Pseudoautosomal genes, those present on the X-Y meiotically pairable region of male, escape inactivation in female soma. It is suggested, but not demonstrated, that they may also be refractory to the inactivation signal in male germ cells. We have assayed activity of the enzyme steroid sulfatase, product of a pseudoautosomal gene, in testicular cells of the mouse and shown its presence in premeiotic, meiotic (pachytene), and postmeiotic (spermatid) cell types. It appears that, as in females, pseudoautosomal genes may escape inactivation in male germ cells also.
Collapse
Affiliation(s)
- R Raman
- Department of Zoology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
45
|
Ha H, Howard CA, Yeom YI, Abe K, Uehara H, Artzt K, Bennett D. Several testis-expressed genes in the mouse t-complex have expression differences between wild-type and t-mutant mice. DEVELOPMENTAL GENETICS 1991; 12:318-32. [PMID: 1718647 DOI: 10.1002/dvg.1020120409] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The t-complex of the mouse occupies the proximal half of chromosome 17 and contains genes which have profound effects on spermatogenesis. Mutations of several loci in the t-complex appear to interact to cause male sterility or transmission ratio distortion (TRD). By cDNA screening or chromosomal walking we have identified seven genes, which are expressed in the germ cells of testis and map to various regions of the t-complex. These genes were named t-complex testis-expressed (Tctex) genes. An analysis of their expression patterns in testes from +/+, +/t, and t/t mice was done by in situ hybridization and by northern blotting. Six genes begin to be expressed at the pachytene stage: Three of them are more abundant at pachytene stage, while three others are more abundant at postmeiotic stages. One gene is expressed at all the stages of spermatogenesis. Interestingly, four Tctex genes show differences in the amount of transcript between wild-type and t-mutant testes. The chromosomal location and expression pattern imply that Tctex genes might be candidate genes for sterility or TRD.
Collapse
Affiliation(s)
- H Ha
- Department of Zoology, University of Texas, Austin 78712-1064
| | | | | | | | | | | | | |
Collapse
|
46
|
Singer-Sam J, Robinson MO, Bellvé AR, Simon MI, Riggs AD. Measurement by quantitative PCR of changes in HPRT, PGK-1, PGK-2, APRT, MTase, and Zfy gene transcripts during mouse spermatogenesis. Nucleic Acids Res 1990; 18:1255-9. [PMID: 1690874 PMCID: PMC330442 DOI: 10.1093/nar/18.5.1255] [Citation(s) in RCA: 185] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A reverse transcriptase-polymerase chain reaction assay (RT-PCR) was used quantitatively to measure accumulated levels of RNA transcripts in total mouse RNAs derived from male germ cells at various spermatogenic stages. RNA levels for two X-linked enzymes, phosphoglycerate kinase (PGK-1) and hypoxanthine phosphoribosyl transferase (HPRT), both decrease during spermatogenesis, although the transcript levels decrease much more rapidly for PGK-1. RNA for the Y-linked ZFY (zinc finger protein) is elevated in all spermatogenic cell fractions tested, being particularly high in leptotene/zygotene spermatocytes and round spermatids. RNA for adenine phosphoribosyltransferase (APRT) increases 5-fold to a peak during late pachynema. RNA for PGK-2, undetectable in spermatogonial cells, increases at least 50-fold by the round spermatid stage. DNA (cytosine-5-)-methyltransferase (MTase) transcript levels are over an order of magnitude higher throughout spermatogenesis than in non-dividing liver cells.
Collapse
Affiliation(s)
- J Singer-Sam
- Division of Biology, Beckman Research Institute, City of Hope, Duarte, CA 91010
| | | | | | | | | |
Collapse
|
47
|
Oko R, Clermont Y. Light microscopic immunocytochemical study of fibrous sheath and outer dense fiber formation in the rat spermatid. Anat Rec (Hoboken) 1989; 225:46-55. [PMID: 2476045 DOI: 10.1002/ar.1092250108] [Citation(s) in RCA: 67] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The formation of the fibrous sheath (FS) and outer dense fibers (ODF), two major cytoskeletal components of the tail of spermatozoa, was analyzed in the seminiferous epithelium by immunoperoxidase techniques applied to paraffin-embedded testicular sections. Antibodies were prepared from purified FS and ODF fractions and from major 75 and 14.4 kDa FS polypeptides and major 32-26 14.4 kDa ODF polypeptides. The immunostaining results showed that the production of FS and ODF proteins appeared to be exclusive to step 9-19 spermatids and lasted over the duration of a full cycle of the seminiferous epithelium, or 12.8 days. During this period there was seemingly an initial lag of short duration between the synthesis and assembly of FS and ODF proteins followed by a long process of coordinated activity. Peak cytoplasmic immunoreactivity was reached in step 15 for FS proteins and midstep 16 for ODF proteins and remained elevated thereafter for approximately 80 hr for both FS and ODF proteins. The immunoreactivity was more uniform and diffused for FS proteins and granulated or clumpy for ODF proteins. Assembly of FS proteins along the axoneme proceeded in a distal to proximal direction while for ODF proteins assembly proceeded in a proximal to distal direction. The main route of elimination of residual cytoplasmic FS and ODF proteins appeared to take place through the cytoplasmic droplets and residual bodies, respectively. There appeared to be no variation in step reactivity between the major ODF polypeptides tested and only minor variation in step reactivity between the major FS polypeptides tested. However, although the 14.4 kDa polypeptides of FS and ODF share antigenic determinants, they do not appear to be identical, because they presented different immunolocalizations during spermiogenesis and different directions of assembly along the axoneme.
Collapse
Affiliation(s)
- R Oko
- Department of Anatomy, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
48
|
Bourgeois CA, Hubert J. Spatial relationship between the nucleolus and the nuclear envelope: structural aspects and functional significance. INTERNATIONAL REVIEW OF CYTOLOGY 1988; 111:1-52. [PMID: 3074957 DOI: 10.1016/s0074-7696(08)61730-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
49
|
Fujimoto H, Erickson RP, Toné S. Changes in polyadenylation of lactate dehydrogenase-X mRNA during spermatogenesis in mice. Mol Reprod Dev 1988; 1:27-34. [PMID: 2908441 DOI: 10.1002/mrd.1080010106] [Citation(s) in RCA: 33] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The expression of the mRNA for mouse testicular lactate dehydrogenase (LDH-X) was examined by RNA:cDNA hybridization in situ in the testis and by Northern analyses of meiotic and postmeiotic spermatogenic cell populations. Silver grains accumulated in cells inside the second layer from the periphery of the seminiferous tubule, confirming previous findings that LDH-X mRNA first appears in the spermatocyte and continues to accumulate until the late spermatid stage. Northern analyses showed that meiotic and postmeiotic cells contained 1.2 and 1.3 kb classes of hybridizing mRNA, respectively. RNase H digestion of oligo (dT)-hybridized RNA and poly(U)-Sepharose column chromatography with differential elution by formamide revealed that the difference in size of the two classes of mRNAs was due to the poly(A) tail length of the LDH-X mRNA. When the distribution of the LDH-X mRNA was examined across polysome gradients, both mRNAs were partially associated with polysomes. These results suggest that the changes in the polyadenylation of LDH-X mRNA were associated with the meiotic division during spermatogenesis in the mouse. They raise the possibility that the stable accumulation of the LDH-X mRNAs in the postmeiotic cells is enhanced by poly(A) tails of increased length.
Collapse
Affiliation(s)
- H Fujimoto
- Laboratory of Cell Biology, Mitsubishi-Kasei Institute of Life Science, Tokyo, Japan
| | | | | |
Collapse
|
50
|
Roberts TM, Pavalko FM, Ward S. Membrane and cytoplasmic proteins are transported in the same organelle complex during nematode spermatogenesis. J Cell Biol 1986; 102:1787-96. [PMID: 3517007 PMCID: PMC2114225 DOI: 10.1083/jcb.102.5.1787] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
During the development of pseudopodial spermatozoa of the nematode, Caenorhabditis elegans, protein synthesis stops before differentiation is completed. Colloidal gold conjugates of monoclonal antibody SP56, which binds to the surface of spermatozoa, and TR20, which recognizes the major sperm cytoplasmic protein (MSP), were used to label thin sections of testes embedded in Lowicryl K4M in order to follow polypeptides from their synthesis early in spermatogenesis to their segregation to specific compartments of the mature cell. Both antigens are synthesized in primary spermatocytes and are assembled into a unique double organelle, the fibrous body-membranous organelle (FB-MO) complex. However, the antigens are localized in different regions of this FB-MO complex. As described in detail, the assembly of proteins into the FB-MO complex allows both membrane and cytoplasmic components to be concentrated in the spermatids after meiosis. Then, the stepwise disassembly of this transient structure ensures delivery of each component to its final destination in the mature spermatozoan: MSP filaments in the fibrous body depolymerize, releasing MSP into the cytoplasm and the membranous organelles fuse with the plasma membrane, delivering SP56 antigen to the surface.
Collapse
|