101
|
Borowiec AS, Sion B, Chalmel F, D Rolland A, Lemonnier L, De Clerck T, Bokhobza A, Derouiche S, Dewailly E, Slomianny C, Mauduit C, Benahmed M, Roudbaraki M, Jégou B, Prevarskaya N, Bidaux G. Cold/menthol TRPM8 receptors initiate the cold-shock response and protect germ cells from cold-shock-induced oxidation. FASEB J 2016; 30:3155-70. [PMID: 27317670 PMCID: PMC5001517 DOI: 10.1096/fj.201600257r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/23/2016] [Indexed: 12/21/2022]
Abstract
Testes of most male mammals present the particularity of being externalized from the body and are consequently slightly cooler than core body temperature (4-8°C below). Although, hypothermia of the testis is known to increase germ cells apoptosis, little is known about the underlying molecular mechanisms, including cold sensors, transduction pathways, and apoptosis triggers. In this study, using a functional knockout mouse model of the cold and menthol receptors, dubbed transient receptor potential melastatine 8 (TRPM8) channels, we found that TRPM8 initiated the cold-shock response by differentially modulating cold- and heat-shock proteins. Besides, apoptosis of germ cells increased in proportion to the cooling level in control mice but was independent of temperature in knockout mice. We also observed that the rate of germ cell death correlated positively with the reactive oxygen species level and negatively with the expression of the detoxifying enzymes. This result suggests that the TRPM8 sensor is a key determinant of germ cell fate under hypothermic stimulation.-Borowiec, A.-S., Sion, B., Chalmel, F., Rolland, A. D., Lemonnier, L., De Clerck, T., Bokhobza, A., Derouiche, S., Dewailly, E., Slomianny, C., Mauduit, C., Benahmed, M., Roudbaraki, M., Jégou, B., Prevarskaya, N., Bidaux, G. Cold/menthol TRPM8 receptors initiate the cold-shock response and protect germ cells from cold-shock-induced oxidation.
Collapse
Affiliation(s)
| | - Benoit Sion
- Pharmacologie Fondamentale et Clinique de la Douleur, INSERM, U1107, Neuro-Dol, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
| | | | | | - Loïc Lemonnier
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Tatiana De Clerck
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Alexandre Bokhobza
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Sandra Derouiche
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Etienne Dewailly
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Christian Slomianny
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Claire Mauduit
- Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, INSERM, U1065, Nice, France; and
| | - Mohamed Benahmed
- Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, INSERM, U1065, Nice, France; and
| | - Morad Roudbaraki
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Bernard Jégou
- INSERM, U1085-Irset, Campus de Beaulieu, Rennes, France
| | - Natalia Prevarskaya
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France
| | - Gabriel Bidaux
- Physiologie Cellulaire (PHYCEL), INSERM, U1003, Université Lille, Lille, France; Laboratoire de Physique des Lasers, Atomes et Molécules (PhLAM), UMR8523, Biophotonic Team, Villeneuve d'Ascq, France
| |
Collapse
|
102
|
Lima AC, Jung M, Rusch J, Usmani A, Lopes AM, Conrad DF. Multispecies Purification of Testicular Germ Cells. Biol Reprod 2016; 95:85. [PMID: 27557646 PMCID: PMC5176363 DOI: 10.1095/biolreprod.116.140566] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
Advanced methods of cellular purification are required to apply genome technology to the study of spermatogenesis. One approach, based on flow cytometry of murine testicular cells stained with Hoechst-33342 (Ho-FACS), has been extensively optimized and currently allows the isolation of nine germ cell types. This staining technique is straightforward to implement, is highly effective at purifying specific germ cell types, and yields sufficient cell numbers for high-throughput studies. Ho-FACS is a technique that does not require species-specific markers, but whose applicability to other species is largely unexplored. We hypothesized that, because of the similar cell physiology of spermatogenesis across mammals, Ho-FACS could be used to produce highly purified subpopulations of germ cells in mammals other than mouse. To test this hypothesis, we applied Ho-FACS to four mammalian species that are widely used in testis research: Rattus norvegicus, Cavia porcellus, Canis familiaris, and Sus scrofadomesticus. We successfully isolated four germ cell populations from these species with average purity of 79% for spermatocytes, 90% for spermatids, and 66% for spermatogonia. Additionally, we compare the performance of mechanical and chemical dissociation for each species, and propose an optimized gating strategy to better discriminate round and elongating spermatids in the mouse, which can potentially be applied to other species. Our work indicates that spermatogenesis may be uniquely accessible among mammalian developmental systems, as a single set of reagents may be sufficient to isolate germ cell populations from many different mammalian species, opening new avenues in the fields of development and male reproductive biology.
Collapse
Affiliation(s)
- Ana C. Lima
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
- Graduate Program in Areas of Basic and Applied Biology (GABBA), Abel Salazar Institute of Biomedical Sciences, University of Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal–IPATIMUP, Porto, Portugal
| | - Min Jung
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Jannette Rusch
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Abul Usmani
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Alexandra M. Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal–IPATIMUP, Porto, Portugal
| | - Donald F. Conrad
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
- Correspondence: Donald F. Conrad, Department of Genetics, Washington University School of Medicine, Campus Box 8232, St. Louis, MO 63110. E-mail:
| |
Collapse
|
103
|
Regulatory complexity revealed by integrated cytological and RNA-seq analyses of meiotic substages in mouse spermatocytes. BMC Genomics 2016; 17:628. [PMID: 27519264 PMCID: PMC4983049 DOI: 10.1186/s12864-016-2865-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 06/28/2016] [Indexed: 01/24/2023] Open
Abstract
Background The continuous and non-synchronous nature of postnatal male germ-cell development has impeded stage-specific resolution of molecular events of mammalian meiotic prophase in the testis. Here the juvenile onset of spermatogenesis in mice is analyzed by combining cytological and transcriptomic data in a novel computational analysis that allows decomposition of the transcriptional programs of spermatogonia and meiotic prophase substages. Results Germ cells from testes of individual mice were obtained at two-day intervals from 8 to 18 days post-partum (dpp), prepared as surface-spread chromatin and immunolabeled for meiotic stage-specific protein markers (STRA8, SYCP3, phosphorylated H2AFX, and HISTH1T). Eight stages were discriminated cytologically by combinatorial antibody labeling, and RNA-seq was performed on the same samples. Independent principal component analyses of cytological and transcriptomic data yielded similar patterns for both data types, providing strong evidence for substage-specific gene expression signatures. A novel permutation-based maximum covariance analysis (PMCA) was developed to map co-expressed transcripts to one or more of the eight meiotic prophase substages, thereby linking distinct molecular programs to cytologically defined cell states. Expression of meiosis-specific genes is not substage-limited, suggesting regulation of substage transitions at other levels. Conclusions This integrated analysis provides a general method for resolving complex cell populations. Here it revealed not only features of meiotic substage-specific gene expression, but also a network of substage-specific transcription factors and relationships to potential target genes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2865-1) contains supplementary material, which is available to authorized users.
Collapse
|
104
|
Mulugeta E, Wassenaar E, Sleddens-Linkels E, van IJcken WFJ, Heard E, Grootegoed JA, Just W, Gribnau J, Baarends WM. Genomes of Ellobius species provide insight into the evolutionary dynamics of mammalian sex chromosomes. Genome Res 2016; 26:1202-10. [PMID: 27510564 PMCID: PMC5052041 DOI: 10.1101/gr.201665.115] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 07/11/2016] [Indexed: 11/24/2022]
Abstract
The X and Y sex chromosomes of placental mammals show hallmarks of a tumultuous evolutionary past. The X Chromosome has a rich and conserved gene content, while the Y Chromosome has lost most of its genes. In the Transcaucasian mole vole Ellobius lutescens, the Y Chromosome including Sry has been lost, and both females and males have a 17,X diploid karyotype. Similarly, the closely related Ellobius talpinus, has a 54,XX karyotype in both females and males. Here, we report the sequencing and assembly of the E. lutescens and E. talpinus genomes. The results indicate that the loss of the Y Chromosome in E. lutescens and E. talpinus occurred in two independent events. Four functional homologs of mouse Y-Chromosomal genes were detected in both female and male E. lutescens, of which three were also detected in the E. talpinus genome. One of these is Eif2s3y, known as the only Y-derived gene that is crucial for successful male meiosis. Female and male E. lutescens can carry one and the same X Chromosome with a largely conserved gene content, including all genes known to function in X Chromosome inactivation. The availability of the genomes of these mole vole species provides unique models to study the dynamics of sex chromosome evolution.
Collapse
Affiliation(s)
- Eskeatnaf Mulugeta
- Department of Developmental Biology, Erasmus MC, 3015CN, Rotterdam, The Netherlands; Institut Curie, Genetics and Developmental Biology Unit, 75248, Paris, France
| | - Evelyne Wassenaar
- Department of Developmental Biology, Erasmus MC, 3015CN, Rotterdam, The Netherlands
| | | | | | - Edith Heard
- Institut Curie, Genetics and Developmental Biology Unit, 75248, Paris, France
| | - J Anton Grootegoed
- Department of Developmental Biology, Erasmus MC, 3015CN, Rotterdam, The Netherlands
| | - Walter Just
- Institute of Human Genetics, University of Ulm, 89081, Ulm, Germany
| | - Joost Gribnau
- Department of Developmental Biology, Erasmus MC, 3015CN, Rotterdam, The Netherlands
| | - Willy M Baarends
- Department of Developmental Biology, Erasmus MC, 3015CN, Rotterdam, The Netherlands
| |
Collapse
|
105
|
Landeen EL, Muirhead CA, Wright L, Meiklejohn CD, Presgraves DC. Sex Chromosome-wide Transcriptional Suppression and Compensatory Cis-Regulatory Evolution Mediate Gene Expression in the Drosophila Male Germline. PLoS Biol 2016; 14:e1002499. [PMID: 27404402 PMCID: PMC4942098 DOI: 10.1371/journal.pbio.1002499] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/06/2016] [Indexed: 12/22/2022] Open
Abstract
The evolution of heteromorphic sex chromosomes has repeatedly resulted in the evolution of sex chromosome-specific forms of regulation, including sex chromosome dosage compensation in the soma and meiotic sex chromosome inactivation in the germline. In the male germline of Drosophila melanogaster, a novel but poorly understood form of sex chromosome-specific transcriptional regulation occurs that is distinct from canonical sex chromosome dosage compensation or meiotic inactivation. Previous work shows that expression of reporter genes driven by testis-specific promoters is considerably lower—approximately 3-fold or more—for transgenes inserted into X chromosome versus autosome locations. Here we characterize this transcriptional suppression of X-linked genes in the male germline and its evolutionary consequences. Using transgenes and transpositions, we show that most endogenous X-linked genes, not just testis-specific ones, are transcriptionally suppressed several-fold specifically in the Drosophila male germline. In wild-type testes, this sex chromosome-wide transcriptional suppression is generally undetectable, being effectively compensated by the gene-by-gene evolutionary recruitment of strong promoters on the X chromosome. We identify and experimentally validate a promoter element sequence motif that is enriched upstream of the transcription start sites of hundreds of testis-expressed genes; evolutionarily conserved across species; associated with strong gene expression levels in testes; and overrepresented on the X chromosome. These findings show that the expression of X-linked genes in the Drosophila testes reflects a balance between chromosome-wide epigenetic transcriptional suppression and long-term compensatory adaptation by sex-linked genes. Our results have broad implications for the evolution of gene expression in the Drosophila male germline and for genome evolution. Expression of sex-linked genes in the Drosophila male germline reflects a balance between an X chromosome-wide transcriptional suppression and long-term, gene-wise evolutionary recruitment of strong, compensatory promoter elements. The evolution of different sex chromosomes (e.g., X and Y) has occurred many times in animals and plants. One consequence of having different chromosome copy numbers between the sexes (XY males and XX females) is the evolution of sex chromosome-specific regulation, both in the soma (i.e., X chromosome dosage compensation) and in the male germline (i.e., meiotic sex chromosome inactivation). Understanding how the X is regulated in the male germline has implications for gene expression, the evolution of sex chromosome-specific gene content, and speciation. Surprisingly, how the X chromosome is regulated in the Drosophila melanogaster male germline remains unclear. We have characterized X suppression, a novel form of X chromosome transcriptional regulation specific to the Drosophila male germline. Our results reveal that transcription of the X is suppressed 2- to 4-fold for endogenous genes. We show that the X chromosome has evolved strong testis-specific promoters via the gene-by-gene recruitment of sequence elements that counteract transcriptional suppression of the X chromosome. These findings reveal a novel form of X chromosome regulation and lead to a new model for the control of gene expression in the Drosophila male germline.
Collapse
Affiliation(s)
- Emily L. Landeen
- Department of Biology, University of Rochester, Rochester, New York, United States of America
- * E-mail: (ELL); (DCP)
| | - Christina A. Muirhead
- Department of Biology, University of Rochester, Rochester, New York, United States of America
- The Ronin Institute, Montclair, New Jersey, United States of America
| | - Lori Wright
- Department of Biology, University of Rochester, Rochester, New York, United States of America
| | - Colin D. Meiklejohn
- School of Biological Sciences, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Daven C. Presgraves
- Department of Biology, University of Rochester, Rochester, New York, United States of America
- * E-mail: (ELL); (DCP)
| |
Collapse
|
106
|
Chalmel F, Rolland AD. Linking transcriptomics and proteomics in spermatogenesis. Reproduction 2016; 150:R149-57. [PMID: 26416010 DOI: 10.1530/rep-15-0073] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Spermatogenesis is a complex and tightly regulated process leading to the continuous production of male gametes, the spermatozoa. This developmental process requires the sequential and coordinated expression of thousands of genes, including many that are testis-specific. The molecular networks underlying normal and pathological spermatogenesis have been widely investigated in recent decades, and many high-throughput expression studies have studied genes and proteins involved in male fertility. In this review, we focus on studies that have attempted to correlate transcription and translation during spermatogenesis by comparing the testicular transcriptome and proteome. We also discuss the recent development and use of new transcriptomic approaches that provide a better proxy for the proteome, from both qualitative and quantitative perspectives. Finally, we provide illustrations of how testis-derived transcriptomic and proteomic data can be integrated to address new questions and how the 'proteomics informed by transcriptomics' technique, by combining RNA-seq and MS-based proteomics, can contribute significantly to the discovery of new protein-coding genes or new protein isoforms expressed during spermatogenesis.
Collapse
Affiliation(s)
- Frédéric Chalmel
- Inserm U1085-IrsetUniversité de Rennes 1, F-35042 Rennes, France
| | | |
Collapse
|
107
|
Vanderperre B, Cermakova K, Escoffier J, Kaba M, Bender T, Nef S, Martinou JC. MPC1-like Is a Placental Mammal-specific Mitochondrial Pyruvate Carrier Subunit Expressed in Postmeiotic Male Germ Cells. J Biol Chem 2016; 291:16448-61. [PMID: 27317664 DOI: 10.1074/jbc.m116.733840] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 12/13/2022] Open
Abstract
Selective transport of pyruvate across the inner mitochondrial membrane by the mitochondrial pyruvate carrier (MPC) is a fundamental step that couples cytosolic and mitochondrial metabolism. The recent molecular identification of the MPC complex has revealed two interacting subunits, MPC1 and MPC2. Although in yeast, an additional subunit, MPC3, can functionally replace MPC2, no alternative MPC subunits have been described in higher eukaryotes. Here, we report for the first time the existence of a novel MPC subunit termed MPC1-like (MPC1L), which is present uniquely in placental mammals. MPC1L shares high sequence, structural, and topological homology with MPC1. In addition, we provide several lines of evidence to show that MPC1L is functionally equivalent to MPC1: 1) when co-expressed with MPC2, it rescues pyruvate import in a MPC-deleted yeast strain; 2) in mammalian cells, it can associate with MPC2 to form a functional carrier as assessed by bioluminescence resonance energy transfer; 3) in MPC1 depleted mouse embryonic fibroblasts, MPC1L rescues the loss of pyruvate-driven respiration and stabilizes MPC2 expression; and 4) MPC1- and MPC1L-mediated pyruvate imports show similar efficiency. However, we show that MPC1L has a highly specific expression pattern and is localized almost exclusively in testis and more specifically in postmeiotic spermatids and sperm cells. This is in marked contrast to MPC1/MPC2, which are ubiquitously expressed throughout the organism. To date, the biological importance of this alternative MPC complex during spermatogenesis in placental mammals remains unknown. Nevertheless, these findings open up new avenues for investigating the structure-function relationship within the MPC complex.
Collapse
Affiliation(s)
| | | | - Jessica Escoffier
- the Swiss Centre for Applied Human Toxicology, and the Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva 4, Switzerland
| | - Mayis Kaba
- the Swiss Centre for Applied Human Toxicology, and the Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva 4, Switzerland
| | | | - Serge Nef
- the Swiss Centre for Applied Human Toxicology, and the Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva 4, Switzerland
| | | |
Collapse
|
108
|
Contrasting Levels of Molecular Evolution on the Mouse X Chromosome. Genetics 2016; 203:1841-57. [PMID: 27317678 DOI: 10.1534/genetics.116.186825] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/08/2016] [Indexed: 11/18/2022] Open
Abstract
The mammalian X chromosome has unusual evolutionary dynamics compared to autosomes. Faster-X evolution of spermatogenic protein-coding genes is known to be most pronounced for genes expressed late in spermatogenesis, but it is unclear if these patterns extend to other forms of molecular divergence. We tested for faster-X evolution in mice spanning three different forms of molecular evolution-divergence in protein sequence, gene expression, and DNA methylation-across different developmental stages of spermatogenesis. We used FACS to isolate individual cell populations and then generated cell-specific transcriptome profiles across different stages of spermatogenesis in two subspecies of house mice (Mus musculus), thereby overcoming a fundamental limitation of previous studies on whole tissues. We found faster-X protein evolution at all stages of spermatogenesis and faster-late protein evolution for both X-linked and autosomal genes. In contrast, there was less expression divergence late in spermatogenesis (slower late) on the X chromosome and for autosomal genes expressed primarily in testis (testis-biased). We argue that slower-late expression divergence reflects strong regulatory constraints imposed during this critical stage of sperm development and that these constraints are particularly acute on the tightly regulated sex chromosomes. We also found slower-X DNA methylation divergence based on genome-wide bisulfite sequencing of sperm from two species of mice (M. musculus and M. spretus), although it is unclear whether slower-X DNA methylation reflects development constraints in sperm or other X-linked phenomena. Our study clarifies key differences in patterns of regulatory and protein evolution across spermatogenesis that are likely to have important consequences for mammalian sex chromosome evolution, male fertility, and speciation.
Collapse
|
109
|
Yang F, Silber S, Leu NA, Oates RD, Marszalek JD, Skaletsky H, Brown LG, Rozen S, Page DC, Wang PJ. TEX11 is mutated in infertile men with azoospermia and regulates genome-wide recombination rates in mouse. EMBO Mol Med 2016; 7:1198-210. [PMID: 26136358 PMCID: PMC4568952 DOI: 10.15252/emmm.201404967] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Genome-wide recombination is essential for genome stability, evolution, and speciation. Mouse Tex11, an X-linked meiosis-specific gene, promotes meiotic recombination and chromosomal synapsis. Here, we report that TEX11 is mutated in infertile men with non-obstructive azoospermia and that an analogous mutation in the mouse impairs meiosis. Genetic screening of a large cohort of idiopathic infertile men reveals that TEX11 mutations, including frameshift and splicing acceptor site mutations, cause infertility in 1% of azoospermic men. Functional evaluation of three analogous human TEX11 missense mutations in transgenic mouse models identified one mutation (V748A) as a potential infertility allele and found two mutations non-causative. In the mouse model, an intronless autosomal Tex11 transgene functionally substitutes for the X-linked Tex11 gene, providing genetic evidence for the X-to-autosomal retrotransposition evolution phenomenon. Furthermore, we find that TEX11 protein levels modulate genome-wide recombination rates in both sexes. These studies indicate that TEX11 alleles affecting expression level or substituting single amino acids may contribute to variations in recombination rates between sexes and among individuals in humans.
Collapse
Affiliation(s)
- Fang Yang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sherman Silber
- Infertility Center of St. Louis, St. Luke's Hospital, St. Louis, MO, USA
| | - N Adrian Leu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert D Oates
- Department of Urology, Boston University Medical Center, Boston, MA, USA
| | - Janet D Marszalek
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA, USA
| | - Helen Skaletsky
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA, USA
| | - Laura G Brown
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA, USA
| | - Steve Rozen
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA, USA Duke-Nus Graduate Medical School Singapore, Singapore City, Singapore
| | - David C Page
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA, USA Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - P Jeremy Wang
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
110
|
Meiotic behaviour of evolutionary sex-autosome translocations in Bovidae. Chromosome Res 2016; 24:325-38. [PMID: 27136937 DOI: 10.1007/s10577-016-9524-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/12/2016] [Accepted: 04/17/2016] [Indexed: 10/21/2022]
Abstract
The recurrent occurrence of sex-autosome translocations during mammalian evolution suggests common mechanisms enabling a precise control of meiotic synapsis, recombination and inactivation of sex chromosomes. We used immunofluorescence and FISH to study the meiotic behaviour of sex chromosomes in six species of Bovidae with evolutionary sex-autosome translocations (Tragelaphus strepsiceros, Taurotragus oryx, Tragelaphus imberbis, Tragelaphus spekii, Gazella leptoceros and Nanger dama ruficollis). The autosomal regions of fused sex chromosomes showed normal synapsis with their homologous counterparts. Synapsis in the pseudoautosomal region (PAR) leads to the formation of characteristic bivalent (in T. imberbis and T. spekii with X;BTA13/Y;BTA13), trivalent (in T. strepsiceros and T. oryx with X/Y;BTA13 and G. leptoceros with X;BTA5/Y) and quadrivalent (in N. dama ruficollis with X;BTA5/Y;BTA16) structures at pachynema. However, when compared with other mammals, the number of pachynema lacking MLH1 foci in the PAR was relatively high, especially in T. imberbis and T. spekii, species with both sex chromosomes involved in sex autosome translocations. Meiotic transcriptional inactivation of the sex-autosome translocations assessed by γH2AX staining was restricted to their gonosomal regions. Despite intraspecies differences, the evolutionary fixation of sex-autosome translocations among bovids appears to involve general mechanisms ensuring sex chromosome pairing, synapsis, recombination and inactivation.
Collapse
|
111
|
Payer B. Developmental regulation of X-chromosome inactivation. Semin Cell Dev Biol 2016; 56:88-99. [PMID: 27112543 DOI: 10.1016/j.semcdb.2016.04.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/13/2016] [Accepted: 04/21/2016] [Indexed: 12/01/2022]
Abstract
With the emergence of sex-determination by sex chromosomes, which differ in composition and number between males and females, appeared the need to equalize X-chromosomal gene dosage between the sexes. Mammals have devised the strategy of X-chromosome inactivation (XCI), in which one of the two X-chromosomes is rendered transcriptionally silent in females. In the mouse, the best-studied model organism with respect to XCI, this inactivation process occurs in different forms, imprinted and random, interspersed by periods of X-chromosome reactivation (XCR), which is needed to switch between the different modes of XCI. In this review, I describe the recent advances with respect to the developmental control of XCI and XCR and in particular their link to differentiation and pluripotency. Furthermore, I review the mechanisms, which influence the timing and choice, with which one of the two X-chromosomes is chosen for inactivation during random XCI. This has an impact on how females are mosaics with regard to which X-chromosome is active in different cells, which has implications on the severity of diseases caused by X-linked mutations.
Collapse
Affiliation(s)
- Bernhard Payer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology and Universitat Pompeu Fabra (UPF), Dr. Aiguader, 88, Barcelona 08003, Spain.
| |
Collapse
|
112
|
Shum EY, Jones SH, Shao A, Chousal JN, Krause MD, Chan WK, Lou CH, Espinoza JL, Song HW, Phan MH, Ramaiah M, Huang L, McCarrey JR, Peterson KJ, De Rooij DG, Cook-Andersen H, Wilkinson MF. The Antagonistic Gene Paralogs Upf3a and Upf3b Govern Nonsense-Mediated RNA Decay. Cell 2016; 165:382-95. [PMID: 27040500 PMCID: PMC4826573 DOI: 10.1016/j.cell.2016.02.046] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/02/2016] [Accepted: 02/20/2016] [Indexed: 01/11/2023]
Abstract
Gene duplication is a major evolutionary force driving adaptation and speciation, as it allows for the acquisition of new functions and can augment or diversify existing functions. Here, we report a gene duplication event that yielded another outcome--the generation of antagonistic functions. One product of this duplication event--UPF3B--is critical for the nonsense-mediated RNA decay (NMD) pathway, while its autosomal counterpart--UPF3A--encodes an enigmatic protein previously shown to have trace NMD activity. Using loss-of-function approaches in vitro and in vivo, we discovered that UPF3A acts primarily as a potent NMD inhibitor that stabilizes hundreds of transcripts. Evidence suggests that UPF3A acquired repressor activity through simple impairment of a critical domain, a rapid mechanism that may have been widely used in evolution. Mice conditionally lacking UPF3A exhibit "hyper" NMD and display defects in embryogenesis and gametogenesis. Our results support a model in which UPF3A serves as a molecular rheostat that directs developmental events.
Collapse
Affiliation(s)
- Eleen Y Shum
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Samantha H Jones
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Ada Shao
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Jennifer N Chousal
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Matthew D Krause
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Wai-Kin Chan
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chih-Hong Lou
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Josh L Espinoza
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Hye-Won Song
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Mimi H Phan
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Madhuvanthi Ramaiah
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Lulu Huang
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78219, USA
| | - Kevin J Peterson
- Department of Biology, Dartmouth College, Hanover, NH 03755, USA
| | - Dirk G De Rooij
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 Utrecht, the Netherlands
| | - Heidi Cook-Andersen
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Miles F Wilkinson
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92103, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92103, USA.
| |
Collapse
|
113
|
Paracrine Wnt/β-catenin signaling mediates proliferation of undifferentiated spermatogonia in the adult mouse testis. Proc Natl Acad Sci U S A 2016; 113:E1489-97. [PMID: 26929341 DOI: 10.1073/pnas.1601461113] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Spermatogonial stem cells (SSCs) fuel the production of male germ cells but the mechanisms behind SSC self-renewal, proliferation, and differentiation are still poorly understood. Using the Wnt target gene Axin2 and genetic lineage-tracing experiments, we found that undifferentiated spermatogonia, comprising SSCs and transit amplifying progenitor cells, respond to Wnt/β-catenin signals. Genetic elimination of β-catenin indicates that Wnt/β-catenin signaling promotes the proliferation of these cells. Signaling is likely initiated by Wnt6, which is uniquely expressed by neighboring Sertoli cells, the only somatic cells in the seminiferous tubule that support germ cells and act as a niche for SSCs. Therefore, unlike other stem cell systems where Wnt/β-catenin signaling is implicated in self-renewal, the Wnt pathway in the testis specifically contributes to the proliferation of SSCs and progenitor cells.
Collapse
|
114
|
Hu YC, Namekawa SH. Functional significance of the sex chromosomes during spermatogenesis. Reproduction 2016; 149:R265-77. [PMID: 25948089 DOI: 10.1530/rep-14-0613] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Mammalian sex chromosomes arose from an ordinary pair of autosomes. Over hundreds of millions of years, they have evolved into highly divergent X and Y chromosomes and have become increasingly specialized for male reproduction. Both sex chromosomes have acquired and amplified testis-specific genes, suggestive of roles in spermatogenesis. To understand how the sex chromosome genes participate in the regulation of spermatogenesis, we review genes, including single-copy, multi-copy, and ampliconic genes, whose spermatogenic functions have been demonstrated in mouse genetic studies. Sex chromosomes are subject to chromosome-wide transcriptional silencing in meiotic and postmeiotic stages of spermatogenesis. We also discuss particular sex-linked genes that escape postmeiotic silencing and their evolutionary implications. The unique gene contents and genomic structures of the sex chromosomes reflect their strategies to express genes at various stages of spermatogenesis and reveal the driving forces that shape their evolution.Free Chinese abstract: A Chinese translation of this abstract is freely available at http://www.reproduction-online.org/content/149/6/R265/suppl/DC1.Free Japanese abstract: A Japanese translation of this abstract is freely available at http://www.reproduction-online.org/content/149/6/R265/suppl/DC2.
Collapse
Affiliation(s)
- Yueh-Chiang Hu
- Division of Developmental BiologyDivision of Reproductive SciencesCincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Satoshi H Namekawa
- Division of Developmental BiologyDivision of Reproductive SciencesCincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA Division of Developmental BiologyDivision of Reproductive SciencesCincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| |
Collapse
|
115
|
Protein markers of synaptic behavior and chromatin remodeling of the neo-XY body in phyllostomid bats. Chromosoma 2015; 125:701-8. [PMID: 26661581 DOI: 10.1007/s00412-015-0566-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/22/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
Abstract
The XX/XY system is the rule among mammals. However, many exceptions from this general pattern have been discovered since the last decades. One of these non-conventional sex chromosome mechanisms is the multiple sex chromosome system, which is evolutionary fixed among many bat species of the family Phyllostomidae, and has arisen by a translocation between one original gonosome (X or Y chromosome), and an autosome, giving rise to a "neo-XY body." The aim of this work is to study the synaptic behavior and the chromatin remodeling of multiple sex chromosomes in different species of phyllostomid bats using electron microscopy and molecular markers. Testicular tissues from adult males of the species Artibeus lituratus, Artibeus planirostris, Uroderma bilobatum, and Vampyrodes caraccioli from the eastern Amazonia were analyzed by optical/electron microscopy and immunofluorescence of meiotic proteins involved in synapsis (SYCP3 and SYCE3), sister-chromatid cohesion (SMC3), and chromatin silencing (BRCA1, γ-H2AX, and RNApol 2). The presence of asynaptic axes-labeled by BRCA1 and γ-H2AX-at meiotic prophase in testes that have a normal development of spermatogenesis, suggests that the basic mechanism that arrests spreading of transcriptional silencing (meiotic sex chromosome inactivation (MSCI)) to the autosomal segments may be per se the formation of a functional synaptonemal complex between homologous or non-homologous regions, and thus, this SC barrier might be probably related to the preservation of fertility in these systems.
Collapse
|
116
|
Royo H, Seitz H, ElInati E, Peters AHFM, Stadler MB, Turner JMA. Silencing of X-Linked MicroRNAs by Meiotic Sex Chromosome Inactivation. PLoS Genet 2015; 11:e1005461. [PMID: 26509798 PMCID: PMC4624941 DOI: 10.1371/journal.pgen.1005461] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/23/2015] [Indexed: 11/18/2022] Open
Abstract
During the pachytene stage of meiosis in male mammals, the X and Y chromosomes are transcriptionally silenced by Meiotic Sex Chromosome Inactivation (MSCI). MSCI is conserved in therian mammals and is essential for normal male fertility. Transcriptomics approaches have demonstrated that in mice, most or all protein-coding genes on the X chromosome are subject to MSCI. However, it is unclear whether X-linked non-coding RNAs behave in a similar manner. The X chromosome is enriched in microRNA (miRNA) genes, with many exhibiting testis-biased expression. Importantly, high expression levels of X-linked miRNAs (X-miRNAs) have been reported in pachytene spermatocytes, indicating that these genes may escape MSCI, and perhaps play a role in the XY-silencing process. Here we use RNA FISH to examine X-miRNA expression in the male germ line. We find that, like protein-coding X-genes, X-miRNAs are expressed prior to prophase I and are thereafter silenced during pachynema. X-miRNA silencing does not occur in mouse models with defective MSCI. Furthermore, X-miRNAs are expressed at pachynema when present as autosomally integrated transgenes. Thus, we conclude that silencing of X-miRNAs during pachynema in wild type males is MSCI-dependent. Importantly, misexpression of X-miRNAs during pachynema causes spermatogenic defects. We propose that MSCI represents a chromosomal mechanism by which X-miRNAs, and other potential X-encoded repressors, can be silenced, thereby regulating genes with critical late spermatogenic functions. During male germ cell formation, the X and the Y chromosomes are inactivated. This process is conserved and it is essential for germ cell generation. It is believed that X/Y silencing affects all protein-coding genes, but the status of miRNAs and other non-coding genes needs further investigation. MicroRNAs from the X-chromosome (X-miRNAs) have been reported as potential silencing escapers, and they have been proposed to play a role in the inactivation mechanism itself. By looking at the individual cell level, we show unambiguously that X-miRNAs are subject to X/Y silencing, a finding that contradicts the current literature. Moreover, we generated mouse mutants in which we forced expression of X-miRNAs during X/Y silencing, and this lead to germ cell death. We propose that X/Y silencing can influence transcription of essential germ cell genes by regulating X-repressors.
Collapse
Affiliation(s)
- Hélène Royo
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
- The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
| | - Hervé Seitz
- Institute of Human Genetics, UPR 1142, CNRS, Montpellier, France
| | - Elias ElInati
- The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
| | | | - Michael B. Stadler
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - James M. A. Turner
- The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
- * E-mail:
| |
Collapse
|
117
|
Young JC, Wakitani S, Loveland KL. TGF-β superfamily signaling in testis formation and early male germline development. Semin Cell Dev Biol 2015; 45:94-103. [PMID: 26500180 DOI: 10.1016/j.semcdb.2015.10.029] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 12/11/2022]
Abstract
The TGF-β ligand superfamily contains at least 40 members, many of which are produced and act within the mammalian testis to facilitate formation of sperm. Their progressive expression at key stages and in specific cell types determines the fertility of adult males, influencing testis development and controlling germline differentiation. BMPs are essential for the interactive instructions between multiple cell types in the early embryo that drive initial specification of gamete precursors. In the nascent foetal testis, several ligands including Nodal, TGF-βs, Activins and BMPs, serve as key masculinizing switches by regulating male germline pluripotency, somatic and germline proliferation, and testicular vascularization and architecture. In postnatal life, local production of these factors determine adult testis size by regulating Sertoli cell multiplication and differentiation, in addition to specifying germline differentiation and multiplication. Because TGF-β superfamily signaling is integral to testis formation, it affects processes that underlie testicular pathologies, including testicular cancer, and its potential to contribute to subfertility is beginning to be understood.
Collapse
Affiliation(s)
- Julia C Young
- Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Shoichi Wakitani
- Hudson Institute of Medical Research, Clayton, Victoria, Australia; Laboratory of Veterinary Biochemistry and Molecular Biology, University of Miyazaki, Japan
| | - Kate L Loveland
- Hudson Institute of Medical Research, Clayton, Victoria, Australia; School of Clinical Sciences, Monash University, Clayton, Victoria, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
118
|
Xist imprinting is promoted by the hemizygous (unpaired) state in the male germ line. Proc Natl Acad Sci U S A 2015; 112:14415-22. [PMID: 26489649 DOI: 10.1073/pnas.1519528112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The long noncoding X-inactivation-specific transcript (Xist gene) is responsible for mammalian X-chromosome dosage compensation between the sexes, the process by which one of the two X chromosomes is inactivated in the female soma. Xist is essential for both the random and imprinted forms of X-chromosome inactivation. In the imprinted form, Xist is paternally marked to be expressed in female embryos. To investigate the mechanism of Xist imprinting, we introduce Xist transgenes (Tg) into the male germ line. Although ectopic high-level Xist expression on autosomes can be compatible with viability, transgenic animals demonstrate reduced fitness, subfertility, defective meiotic pairing, and other germ-cell abnormalities. In the progeny, paternal-specific expression is recapitulated by the 200-kb Xist Tg. However, Xist imprinting occurs efficiently only when it is in an unpaired or unpartnered state during male meiosis. When transmitted from a hemizygous father (+/Tg), the Xist Tg demonstrates paternal-specific expression in the early embryo. When transmitted by a homozygous father (Tg/Tg), the Tg fails to show imprinted expression. Thus, Xist imprinting is directed by sequences within a 200-kb X-linked region, and the hemizygous (unpaired) state of the Xist region promotes its imprinting in the male germ line.
Collapse
|
119
|
Sosa E, Flores L, Yan W, McCarrey JR. Escape of X-linked miRNA genes from meiotic sex chromosome inactivation. Development 2015; 142:3791-800. [PMID: 26395485 DOI: 10.1242/dev.127191] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/08/2015] [Indexed: 01/25/2023]
Abstract
Past studies have indicated that transcription of all X-linked genes is repressed by meiotic sex chromosome inactivation (MSCI) during the meiotic phase of spermatogenesis in mammals. However, more recent studies have shown an increase in steady-state levels of certain X-linked miRNAs in pachytene spermatocytes, suggesting that either synthesis of these miRNAs increases or that degradation of these miRNAs decreases dramatically in these cells. To distinguish between these possibilities, we performed RNA-FISH to detect nascent transcripts from multiple miRNA genes in various spermatogenic cell types. Our results show definitively that Type I X-linked miRNA genes are subject to MSCI, as are all or most X-linked mRNA genes, whereas Type II and III X-linked miRNA genes escape MSCI by continuing ongoing, active transcription in primary spermatocytes. We corroborated these results by co-localization of RNA-FISH signals with both a corresponding DNA-FISH signal and an immunofluorescence signal for RNA polymerase II. We also found that X-linked miRNA genes that escape MSCI locate non-randomly to the periphery of the XY body, whereas genes that are subject to MSCI remain located within the XY body in pachytene spermatocytes, suggesting that the mechanism of escape of X-linked miRNA genes from MSCI involves their relocation to a position outside of the repressive chromatin domain associated with the XY body. The fact that Type II and III X-linked miRNA genes escape MSCI suggests an immediacy of function of the encoded miRNAs specifically required during the meiotic stages of spermatogenesis.
Collapse
Affiliation(s)
- Enrique Sosa
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Luis Flores
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Wei Yan
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
120
|
Regulation of germ cell function by SUMOylation. Cell Tissue Res 2015; 363:47-55. [PMID: 26374733 DOI: 10.1007/s00441-015-2286-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/11/2015] [Indexed: 01/30/2023]
Abstract
Oogenesis and spermatogenesis are tightly regulated complex processes that are critical for fertility. Germ cells undergo meiosis to generate haploid cells necessary for reproduction. Errors in meiosis, including the generation of chromosomal abnormalities, can result in reproductive defects and infertility. Meiotic proteins are regulated by post-translational modifications including SUMOylation, the covalent attachment of small ubiquitin-like modifier (SUMO) proteins. Here, we review the role of SUMO proteins in controlling germ cell development and maturation based on recent findings from mouse models. Several studies have characterized the localization of SUMO proteins in male and female germ cells. However, a deeper understanding of how SUMOylation regulates proteins with essential roles in oogenesis and spermatogenesis will provide useful insight into the underlying mechanisms of germ cell development and fertility.
Collapse
|
121
|
Güneş S, Kulaç T. The role of epigenetics in spermatogenesis. Turk J Urol 2015; 39:181-7. [PMID: 26328105 DOI: 10.5152/tud.2013.037] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/30/2013] [Indexed: 11/22/2022]
Abstract
Male germ cells have a unique morphology and function to facilitate fertilization. Sperm deoxyribonucleic acid (DNA) is highly condensed to protect the paternal genome during transfer from male to oocyte. Sperm cells undergo extensive epigenetic modifications during differentiation to become a mature spermatozoon. Epigenetic modifications, including DNA methylation, histone modifications, and chromatin remodeling are substantial regulators of spermatogenesis. DNA hypermethylation is associated with gene silencing. Meanwhile, hypomethylation is associated with gene expression. In sperm cells, promoters of developmental genes are highly hypomethylated. Proper DNA methylation is essential for embryo development. Histone modifications are chemical modifications that change the DNA-binding capacity of histones and the accessibility of regulatory factors to the DNA, thereby altering gene expression. Phosphorylation, methylation, acetylation, and ubiquitination are primary modifications of lysine and serine residues on histone tails. In addition to somatic histones, testis-specific histone variants are expressed, including histone H2B in mature sperm. The replacement of histones with protamines is a crucial step in spermatogenesis. Histone hyper-acetylation induces a loose chromatin structure and facilitates topoisomerase-induced DNA strand breaks. As a result, histones are replaced with transition proteins. Next, the transition proteins are replaced with protamines that induce compaction of sperm DNA. This review provides an overview of epigenetic changes during spermatogenesis.
Collapse
Affiliation(s)
- Sezgin Güneş
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Tuba Kulaç
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
122
|
Lambrot R, Lafleur C, Kimmins S. The histone demethylase KDM1A is essential for the maintenance and differentiation of spermatogonial stem cells and progenitors. FASEB J 2015; 29:4402-16. [PMID: 26243864 DOI: 10.1096/fj.14-267328] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 06/22/2015] [Indexed: 12/22/2022]
Abstract
Little is known of the fundamental processes governed by epigenetic mechanisms in the supplier cells of spermatogenesis, the spermatogonial stem cells (SSCs). The histone H3 lysine demethylase KDM1A is expressed in spermatogonia. We hypothesized that KDM1A serves in transcriptional regulation of SSCs and fertility. Using a conditional deletion of Kdm1a [conditional knockout (cKO)] in mouse spermatogonia, we determined that Kdm1a is essential for spermatogenesis as adult cKO males completely lack germ cells. Analysis of postnatal testis development revealed that undifferentiated and differentiating spermatogonial populations form in Kdm1a-cKO animals, yet the majority fail to enter meiosis. Loss of germ cells in the cKO was rapid with none remaining by postnatal day (PND) 21. To gain insight into the mechanistic implications of Kdm1a ablation, we isolated PND 6 spermatogonia enriched for SSCs and analyzed their transcriptome by RNA sequencing. Loss of Kdm1a was associated with altered transcription of 1206 genes. Importantly, differentially expressed genes between control and Kdm1a-cKO animals included those that are essential for SSC and progenitor maintenance and spermatogonial differentiation. The complete loss of fertility and failure to establish spermatogenesis indicate that Kdm1a is a master controller of gene transcription in spermatogonia and is required for SSC and progenitor maintenance and differentiation.
Collapse
Affiliation(s)
- Romain Lambrot
- *Department of Animal Science, McGill University, Sainte Anne de Bellevue, Quebec, Canada; and Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Christine Lafleur
- *Department of Animal Science, McGill University, Sainte Anne de Bellevue, Quebec, Canada; and Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Sarah Kimmins
- *Department of Animal Science, McGill University, Sainte Anne de Bellevue, Quebec, Canada; and Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
123
|
Sayadi A, Jeyakani J, Seet SH, Wei CL, Bourque G, Bard FA, Jenkins NA, Copeland NG, Bard-Chapeau EA. Functional features of EVI1 and EVI1Δ324 isoforms of MECOM gene in genome-wide transcription regulation and oncogenicity. Oncogene 2015; 35:2311-21. [DOI: 10.1038/onc.2015.286] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 06/09/2015] [Accepted: 06/13/2015] [Indexed: 11/09/2022]
|
124
|
Sin HS, Kartashov AV, Hasegawa K, Barski A, Namekawa SH. Poised chromatin and bivalent domains facilitate the mitosis-to-meiosis transition in the male germline. BMC Biol 2015. [PMID: 26198001 PMCID: PMC4508805 DOI: 10.1186/s12915-015-0159-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background The male germline transcriptome changes dramatically during the mitosis-to-meiosis transition to activate late spermatogenesis genes and to transiently suppress genes commonly expressed in somatic lineages and spermatogenesis progenitor cells, termed somatic/progenitor genes. Results These changes reflect epigenetic regulation. Induction of late spermatogenesis genes during spermatogenesis is facilitated by poised chromatin established in the stem cell phases of spermatogonia, whereas silencing of somatic/progenitor genes during meiosis and postmeiosis is associated with formation of bivalent domains which also allows the recovery of the somatic/progenitor program after fertilization. Importantly, during spermatogenesis mechanisms of epigenetic regulation on sex chromosomes are different from autosomes: X-linked somatic/progenitor genes are suppressed by meiotic sex chromosome inactivation without deposition of H3K27me3. Conclusions Our results suggest that bivalent H3K27me3 and H3K4me2/3 domains are not limited to developmental promoters (which maintain bivalent domains that are silent throughout the reproductive cycle), but also underlie reversible silencing of somatic/progenitor genes during the mitosis-to-meiosis transition in late spermatogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0159-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ho-Su Sin
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 49229, USA.,Present address: Department of Developmental Biology, Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Andrey V Kartashov
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 49229, USA
| | - Kazuteru Hasegawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 49229, USA.,Present address: Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Artem Barski
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 49229, USA.
| | - Satoshi H Namekawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 49229, USA.
| |
Collapse
|
125
|
Seipin deficiency increases chromocenter fragmentation and disrupts acrosome formation leading to male infertility. Cell Death Dis 2015; 6:e1817. [PMID: 26181198 PMCID: PMC4650735 DOI: 10.1038/cddis.2015.188] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 11/16/2022]
Abstract
The Berardinelli–Seip congenital lipodystrophy type 2 (Bscl2, seipin) gene is involved in adipogenesis. Bscl2−/− males were infertile but had normal mating behavior. Both Bscl2−/− cauda epididymis sperm count and sperm motility were ~20 × less than control. Bscl2−/− seminiferous tubules had relatively normal presence of spermatogonia and spermatocytes but had reduced spermatids and sperm. Spatiotemporal expression analyses in Bscl2+/+ testes demonstrated prominent Bscl2 transcriptional activity in spermatocytes with a plateau reached around postnatal day 28. Seipin protein localization was most abundant in postmeiotic spermatids, suggesting translational repression of Bscl2 mRNA in spermatocytes. In situ end-labeling plus detected increased spermatid apoptosis in Bscl2−/− testis and annexin V detected increased percentage of positive Bscl2−/− round spermatids compared with control. Immunofluorescence of marker proteins synaptonemal complex proteins 3 and 1 (SYCP3 and SYCP1), and H3K9me3 (histone H3 trimethylated at lysine 9) in germ cell spreads detected normal meiotic chromosome pairing and homologous chromosome synapsis in Bscl2−/− spermatocytes, but significantly increased percentages of round spermatids with chromocenter fragmentation and late spermatids and sperm with chromatin vacuoles, indicating defective chromatin condensation in Bscl2−/− spermatids. Bscl2−/− late spermatids were disorganized within the seminiferous epithelium, despite normal appearance of Sertoli cells detected by vimentin immunofluorescence. Peanut agglutinin staining revealed various abnormalities of acrosomes in Bscl2−/− late spermatids, including the absence, irregular-shaped, and fragmented acrosomes, indicating defective acrosome formation in Bscl2−/− late spermatids, which may affect late spermatid orientation in the seminiferous epithelium. Mitotracker strongly stained the midpiece of control sperm but only very weakly labeled the midpiece of Bscl2−/− sperm, indicating defective mitochondrial activity that most likely contributed to reduced Bscl2−/− sperm motility. These data demonstrate novel roles of seipin in spermatid chromatin integrity, acrosome formation, and mitochondrial activity. Increased spermatid apoptosis, increased chromocenter fragmentation, defective chromatin condensation, abnormal acrosome formation, and defective mitochondrial activity contributed to decreased sperm production and defective sperm that resulted in Bscl2−/− male infertility.
Collapse
|
126
|
Kato Y, Alavattam KG, Sin HS, Meetei AR, Pang Q, Andreassen PR, Namekawa SH. FANCB is essential in the male germline and regulates H3K9 methylation on the sex chromosomes during meiosis. Hum Mol Genet 2015; 24:5234-49. [PMID: 26123487 DOI: 10.1093/hmg/ddv244] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/22/2015] [Indexed: 11/13/2022] Open
Abstract
Fanconi anemia (FA) is a recessive X-linked and autosomal genetic disease associated with bone marrow failure and increased cancer, as well as severe germline defects such as hypogonadism and germ cell depletion. Although deficiencies in FA factors are commonly associated with germ cell defects, it remains unknown whether the FA pathway is involved in unique epigenetic events in germ cells. In this study, we generated Fancb mutant mice, the first mouse model of X-linked FA, and identified a novel function of the FA pathway in epigenetic regulation during mammalian gametogenesis. Fancb mutant mice were infertile and exhibited primordial germ cell (PGC) defects during embryogenesis. Further, Fancb mutation resulted in the reduction of undifferentiated spermatogonia in spermatogenesis, suggesting that FANCB regulates the maintenance of undifferentiated spermatogonia. Additionally, based on functional studies, we dissected the pathway in which FANCB functions during meiosis. The localization of FANCB on sex chromosomes is dependent on MDC1, a binding partner of H2AX phosphorylated at serine 139 (γH2AX), which initiates chromosome-wide silencing. Also, FANCB is required for FANCD2 localization during meiosis, suggesting that the role of FANCB in the activation of the FA pathway is common to both meiosis and somatic DNA damage responses. H3K9me2, a silent epigenetic mark, was decreased on sex chromosomes, whereas H3K9me3 was increased on sex chromosomes in Fancb mutant spermatocytes. Taken together, these results indicate that FANCB functions at critical stages of germ cell development and reveal a novel function of the FA pathway in the regulation of H3K9 methylation in the germline.
Collapse
Affiliation(s)
- Yasuko Kato
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 4929, USA
| | - Kris G Alavattam
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 4929, USA
| | - Ho-Su Sin
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 4929, USA
| | - Amom Ruhikanta Meetei
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 4929, USA
| | - Qishen Pang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 4929, USA
| | - Paul R Andreassen
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 4929, USA
| | - Satoshi H Namekawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 4929, USA
| |
Collapse
|
127
|
Otaka K, Hiradate Y, Kobayashi N, Shirakata Y, Tanemura K. Distribution of the sex chromosome during mouse spermatogenesis in testis tissue sections. J Reprod Dev 2015; 61:375-81. [PMID: 26073979 PMCID: PMC4623142 DOI: 10.1262/jrd.2015-013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During mammalian spermatogenesis, spermatogenic cells undergo mitotic division and are subsequently divided into haploid spermatids by meiotic division, but the dynamics of sex chromosomes during spermatogenesis are unclear in vivo. To gain insight into the distribution of sex chromosomes in the testis, we examined the localization of sex chromosomes before and after meiosis in mouse testis sections. Here, we developed a method of fluorescence in situ hybridization (FISH) using specific probes for the X and Y chromosomes to obtain their positional information in histological testis sections. FISH analysis revealed the sex chromosomal position during spermatogenesis in each stage of seminiferous epithelia and in each spermatogenic cell. In the spermatogonia and leptotene spermatocytes, sex chromosomes were distantly positioned in the cell. In the zygotene and pachytene spermatocytes at prophase I, X and Y chromosomes had a random
distribution. After meiosis, the X and Y spermatids were random in every seminiferous epithelium. We also detected aneuploidy of sex chromosomes in spermatogenic cells using our developed FISH analysis. Our results provide further insight into the distribution of sex chromosomes during spermatogenesis, which could help to elucidate a specific difference between X and Y spermatids and sex chromosome-specific behavior.
Collapse
Affiliation(s)
- Kosuke Otaka
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Miyagi 981-8555, Japan
| | | | | | | | | |
Collapse
|
128
|
Broering TJ, Wang YL, Pandey RN, Hegde RS, Wang SC, Namekawa SH. BAZ1B is dispensable for H2AX phosphorylation on Tyrosine 142 during spermatogenesis. Biol Open 2015; 4:873-84. [PMID: 25979708 PMCID: PMC4571090 DOI: 10.1242/bio.011734] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Meiosis is precisely regulated by the factors involved in DNA damage response in somatic cells. Among them, phosphorylation of H2AX on Serine 139 (γH2AX) is an essential signal for the silencing of unsynapsed sex chromosomes during male meiosis. However, it remains unknown how adjacent H2AX phosphorylation on Tyrosine 142 (pTyr142) is regulated in meiosis. Here we investigate the meiotic functions of BAZ1B (WSTF), the only known Tyr142 kinase in somatic cells, using mice possessing a conditional deletion of BAZ1B. Although BAZ1B deletion causes ectopic γH2AX signals on synapsed autosomes during the early pachytene stage, BAZ1B is dispensable for fertility and critical events during spermatogenesis. BAZ1B deletion does not alter events on unsynapsed axes and pericentric heterochromatin formation. Furthermore, BAZ1B is dispensable for localization of the ATP-dependent chromatin remodeling protein SMARCA5 (SNF2h) during spermatogenesis despite the complex formation between BAZ1B and SMARCA5, known as the WICH complex, in somatic cells. Notably, pTyr142 is regulated independently of BAZ1B and is dephosphorylated on the sex chromosomes during meiosis in contrast with the presence of adjacent γH2AX. Dephosphorylation of pTyr142 is regulated by MDC1, a binding partner of γH2AX. These results reveal the distinct regulation of two adjacent phosphorylation sites of H2AX during meiosis, and suggest that another kinase mediates Tyr142 phosphorylation.
Collapse
Affiliation(s)
- Tyler J Broering
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yuan-Liang Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Ram Naresh Pandey
- Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rashmi S Hegde
- Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Shao-Chun Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Satoshi H Namekawa
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
129
|
Periodic retinoic acid-STRA8 signaling intersects with periodic germ-cell competencies to regulate spermatogenesis. Proc Natl Acad Sci U S A 2015; 112:E2347-56. [PMID: 25902548 DOI: 10.1073/pnas.1505683112] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Mammalian spermatogenesis--the transformation of stem cells into millions of haploid spermatozoa--is elaborately organized in time and space. We explored the underlying regulatory mechanisms by genetically and chemically perturbing spermatogenesis in vivo, focusing on spermatogonial differentiation, which begins a series of amplifying divisions, and meiotic initiation, which ends these divisions. We first found that, in mice lacking the retinoic acid (RA) target gene Stimulated by retinoic acid gene 8 (Stra8), undifferentiated spermatogonia accumulated in unusually high numbers as early as 10 d after birth, whereas differentiating spermatogonia were depleted. We thus conclude that Stra8, previously shown to be required for meiotic initiation, also promotes (but is not strictly required for) spermatogonial differentiation. Second, we found that injection of RA into wild-type adult males induced, independently, precocious spermatogonial differentiation and precocious meiotic initiation; thus, RA acts instructively on germ cells at both transitions. Third, the competencies of germ cells to undergo spermatogonial differentiation or meiotic initiation in response to RA were found to be distinct, periodic, and limited to particular seminiferous stages. Competencies for both transitions begin while RA levels are low, so that the germ cells respond as soon as RA levels rise. Together with other findings, our results demonstrate that periodic RA-STRA8 signaling intersects with periodic germ-cell competencies to regulate two distinct, cell-type-specific responses: spermatogonial differentiation and meiotic initiation. This simple mechanism, with one signal both starting and ending the amplifying divisions, contributes to the prodigious output of spermatozoa and to the elaborate organization of spermatogenesis.
Collapse
|
130
|
Federici F, Mulugeta E, Schoenmakers S, Wassenaar E, Hoogerbrugge JW, van der Heijden GW, van Cappellen WA, Slotman JA, van IJcken WFJ, Laven JSE, Grootegoed JA, Baarends WM. Incomplete meiotic sex chromosome inactivation in the domestic dog. BMC Genomics 2015; 16:291. [PMID: 25884295 PMCID: PMC4399420 DOI: 10.1186/s12864-015-1501-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/30/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In mammalian meiotic prophase, homologous chromosome recognition is aided by formation and repair of programmed DNA double-strand breaks (DSBs). Subsequently, stable associations form through homologous chromosome synapsis. In male mouse meiosis, the largely heterologous X and Y chromosomes synapse only in their short pseudoautosomal regions (PARs), and DSBs persist along the unsynapsed non-homologous arms of these sex chromosomes. Asynapsis of these arms and the persistent DSBs then trigger transcriptional silencing through meiotic sex chromosome inactivation (MSCI), resulting in formation of the XY body. This inactive state is partially maintained in post-meiotic haploid spermatids (postmeiotic sex chromatin repression, PSCR). For the human, establishment of MSCI and PSCR have also been reported, but X-linked gene silencing appears to be more variable compared to mouse. To gain more insight into the regulation and significance of MSCI and PSCR among different eutherian species, we have performed a global analysis of XY pairing dynamics, DSB repair, MSCI and PSCR in the domestic dog (Canis lupus familiaris), for which the complete genome sequence has recently become available, allowing a thorough comparative analyses. RESULTS In addition to PAR synapsis between X and Y, we observed extensive self-synapsis of part of the dog X chromosome, and rapid loss of known markers of DSB repair from that part of the X. Sequencing of RNA from purified spermatocytes and spermatids revealed establishment of MSCI. However, the self-synapsing region of the X displayed higher X-linked gene expression compared to the unsynapsed area in spermatocytes, and was post-meiotically reactivated in spermatids. In contrast, genes in the PAR, which are expected to escape MSCI, were expressed at very low levels in both spermatocytes and spermatids. Our comparative analysis was then used to identify two X-linked genes that may escape MSCI in spermatocytes, and 21 that are specifically re-activated in spermatids of human, mouse and dog. CONCLUSIONS Our data indicate that MSCI is incomplete in the dog. This may be partially explained by extensive, but transient, self-synapsis of the X chromosome, in association with rapid completion of meiotic DSB repair. In addition, our comparative analysis identifies novel candidate male fertility genes.
Collapse
Affiliation(s)
- Federica Federici
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Eskeatnaf Mulugeta
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands. .,Present address: Institut Curie, Genetics and Developmental Biology, Unit 11 et 13 rue Pierre et Marie Curie, 75248, Paris, Cedex 05, France.
| | - Sam Schoenmakers
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands. .,Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Evelyne Wassenaar
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Jos W Hoogerbrugge
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Godfried W van der Heijden
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands. .,Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Wiggert A van Cappellen
- Department of Pathology, Erasmus Optical Imaging Centre, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Johan A Slotman
- Department of Pathology, Erasmus Optical Imaging Centre, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Wilfred F J van IJcken
- Erasmus Center for Biomics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| | - Joop S E Laven
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - J Anton Grootegoed
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Willy M Baarends
- Department of Developmental Biology, Erasmus MC, University Medical Center, PO BOX 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
131
|
Song HW, Bettegowda A, Oliver D, Yan W, Phan MH, de Rooij DG, Corbett MA, Wilkinson MF. shRNA off-target effects in vivo: impaired endogenous siRNA expression and spermatogenic defects. PLoS One 2015; 10:e0118549. [PMID: 25790000 PMCID: PMC4366048 DOI: 10.1371/journal.pone.0118549] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 01/20/2015] [Indexed: 12/23/2022] Open
Abstract
RNA interference (RNAi) is widely used to determine the function of genes. We chose this approach to assess the collective function of the highly related reproductive homeobox 3 (Rhox3) gene paralogs. Using a Rhox3 short hairpin (sh) RNA with 100% complementarity to all 8 Rhox3 paralogs, expressed from a CRE-regulated transgene, we successfully knocked down Rhox3 expression in male germ cells in vivo. These Rhox3-shRNA transgenic mice had dramatic defects in spermatogenesis, primarily in spermatocytes and round spermatids. To determine whether this phenotype was caused by reduced Rhox3 expression, we generated mice expressing the Rhox3-shRNA but lacking the intended target of the shRNA—Rhox3. These double-mutant mice had a phenotype indistinguishable from Rhox3-shRNA-expressing mice that was different from mice lacking the Rhox3 paralogs, indicating that the Rhox3 shRNA disrupts spermatogenesis independently of Rhox3. Rhox3-shRNA transgenic mice displayed few alterations in the expression of protein-coding genes, but instead exhibited reduced levels of all endogenous siRNAs we tested. This supported a model in which the Rhox3 shRNA causes spermatogenic defects by sequestering one or more components of the endogenous small RNA biogenesis machinery. Our study serves as a warning for those using shRNA approaches to investigate gene functions in vivo.
Collapse
Affiliation(s)
- Hye-Won Song
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Anilkumar Bettegowda
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Daniel Oliver
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Mimi H. Phan
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Dirk G. de Rooij
- Center for Reproductive Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark A. Corbett
- School of Pediatrics and Reproductive Health, The University of Adelaide, Adelaide, Australia
| | - Miles F. Wilkinson
- Department of Reproductive Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
132
|
Hasegawa K, Sin HS, Maezawa S, Broering TJ, Kartashov AV, Alavattam KG, Ichijima Y, Zhang F, Bacon WC, Greis KD, Andreassen PR, Barski A, Namekawa SH. SCML2 establishes the male germline epigenome through regulation of histone H2A ubiquitination. Dev Cell 2015; 32:574-88. [PMID: 25703348 PMCID: PMC4391279 DOI: 10.1016/j.devcel.2015.01.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 12/23/2014] [Accepted: 01/16/2015] [Indexed: 01/03/2023]
Abstract
Gametogenesis is dependent on the expression of germline-specific genes. However, it remains unknown how the germline epigenome is distinctly established from that of somatic lineages. Here we show that genes commonly expressed in somatic lineages and spermatogenesis-progenitor cells undergo repression in a genome-wide manner in late stages of the male germline and identify underlying mechanisms. SCML2, a germline-specific subunit of a Polycomb repressive complex 1 (PRC1), establishes the unique epigenome of the male germline through two distinct antithetical mechanisms. SCML2 works with PRC1 and promotes RNF2-dependent ubiquitination of H2A, thereby marking somatic/progenitor genes on autosomes for repression. Paradoxically, SCML2 also prevents RNF2-dependent ubiquitination of H2A on sex chromosomes during meiosis, thereby enabling unique epigenetic programming of sex chromosomes for male reproduction. Our results reveal divergent mechanisms involving a shared regulator by which the male germline epigenome is distinguished from that of the soma and progenitor cells.
Collapse
Affiliation(s)
- Kazuteru Hasegawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Ho-Su Sin
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - So Maezawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Tyler J Broering
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Andrey V Kartashov
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Kris G Alavattam
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Yosuke Ichijima
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Fan Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - W Clark Bacon
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Kenneth D Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Paul R Andreassen
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Artem Barski
- Division of Allergy and Immunology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA
| | - Satoshi H Namekawa
- Division of Reproductive Sciences, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 49267, USA.
| |
Collapse
|
133
|
Abstract
During spermiogenesis, the postmeiotic phase of mammalian spermatogenesis, transcription is progressively repressed as nuclei of haploid spermatids are compacted through a dramatic chromatin reorganization involving hyperacetylation and replacement of most histones with protamines. Although BRDT functions in transcription and histone removal in spermatids, it is unknown whether other BET family proteins play a role. Immunofluorescence of spermatogenic cells revealed BRD4 in a ring around the nuclei of spermatids containing hyperacetylated histones. The ring lies directly adjacent to the acroplaxome, the cytoskeletal base of the acrosome, previously linked to chromatin reorganization. The BRD4 ring does not form in acrosomal mutant mice. Chromatin immunoprecipitation followed by sequencing in spermatids revealed enrichment of BRD4 and acetylated histones at the promoters of active genes. BRD4 and BRDT show distinct and synergistic binding patterns, with a pronounced enrichment of BRD4 at spermatogenesis-specific genes. Direct association of BRD4 with acetylated H4 decreases in late spermatids as acetylated histones are removed from the condensing nucleus in a wave following the progressing acrosome. These data provide evidence of a prominent transcriptional role for BRD4 and suggest a possible removal mechanism for chromatin components from the genome via the progressing acrosome as transcription is repressed and chromatin is compacted during spermiogenesis.
Collapse
|
134
|
Polycomb protein SCML2 associates with USP7 and counteracts histone H2A ubiquitination in the XY chromatin during male meiosis. PLoS Genet 2015; 11:e1004954. [PMID: 25634095 PMCID: PMC4310598 DOI: 10.1371/journal.pgen.1004954] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 12/12/2014] [Indexed: 11/19/2022] Open
Abstract
Polycomb group proteins mediate transcriptional silencing in diverse developmental processes. Sex chromosomes undergo chromosome-wide transcription silencing during male meiosis. Here we report that mouse SCML2 (Sex comb on midleg-like 2), an X chromosome-encoded polycomb protein, is specifically expressed in germ cells, including spermatogonia, spermatocytes, and round spermatids. SCML2 associates with phosphorylated H2AX and localizes to the XY body in spermatocytes. Loss of SCML2 in mice causes defective spermatogenesis, resulting in sharply reduced sperm production. SCML2 interacts with and recruits a deubiquitinase, USP7, to the XY body in spermatocytes. In the absence of SCML2, USP7 fails to accumulate on the XY body, whereas H2A monoubiquitination is dramatically augmented in the XY chromatin. Our results demonstrate that the SCML2/USP7 complex constitutes a novel molecular pathway in modulating the epigenetic state of sex chromosomes during male meiosis.
Collapse
|
135
|
Wallingford MC, Filkins R, Adams D, Walentuk M, Salicioni AM, Visconti PE, Mager J. Identification of a novel isoform of the leukemia-associated MLLT1 (ENL/LTG19) protein. Gene Expr Patterns 2014; 17:11-5. [PMID: 25481096 DOI: 10.1016/j.gep.2014.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/17/2014] [Accepted: 11/22/2014] [Indexed: 11/17/2022]
Abstract
Genome wide transcriptional profiles offer abundant information regarding mRNA levels in specific tissues, organs or developmental stages. Although these data sets do not offer spatial or cell type-specific information, they can be extremely useful for gene discovery when analyzed by the appropriate techniques. Previously, we proposed and validated the use of combinatorial dataset analysis techniques to identify novel genes required during pre-implantation development. Now we build upon this work to identify genes that have dynamic expression during gametogenesis. Here we present detailed analysis of the expression pattern of leukemia-associated, myeloid/lymphoid or mixed-lineage leukemia; translocated to 1 (Mllt1) gene. We document a novel splice isoform of Mllt1 and confirm that both Mllt1 mRNA isoforms are translated. We provide data supporting that MLLT1 protein isoforms display distinct stage-specific expression during spermiogenesis and adult tissues. Finally, we evaluated genes neighboring the Mllt1 locus, and show dynamic stage specific expression patterns of other genes Catsperd, Prr22, Rfx2 and Slc25a41. We document testes expressed alternative isoforms of Prr22 and Rfx2. These results indicate that transcriptome data mining, combined with specific expression analysis provides a wealth of novel gene expression information.
Collapse
Affiliation(s)
- Mary C Wallingford
- Department of Bioengineering, University of Washington, Seattle, WA 01003, United States
| | - Rachel Filkins
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA 01003, United States
| | - Danielle Adams
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA 01003, United States
| | - Melanie Walentuk
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA 01003, United States
| | - Ana Maria Salicioni
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA 01003, United States
| | - Pablo E Visconti
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA 01003, United States
| | - Jesse Mager
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
136
|
Zhang T, Murphy MW, Gearhart MD, Bardwell VJ, Zarkower D. The mammalian Doublesex homolog DMRT6 coordinates the transition between mitotic and meiotic developmental programs during spermatogenesis. Development 2014; 141:3662-71. [PMID: 25249458 DOI: 10.1242/dev.113936] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In mammals, a key transition in spermatogenesis is the exit from spermatogonial differentiation and mitotic proliferation and the entry into spermatocyte differentiation and meiosis. Although several genes that regulate this transition have been identified, how it is controlled and coordinated remains poorly understood. Here, we examine the role in male gametogenesis of the Doublesex-related gene Dmrt6 (Dmrtb1) in mice and find that Dmrt6 plays a crucial role in directing germ cells through the mitotic-to-meiotic germ cell transition. DMRT6 protein is expressed in late mitotic spermatogonia. In mice of the C57BL/6J strain, a null mutation in Dmrt6 disrupts spermatogonial differentiation, causing inappropriate expression of spermatogonial differentiation factors, including SOHLH1, SOHLH2 and DMRT1 as well as the meiotic initiation factor STRA8, and causing most late spermatogonia to undergo apoptosis. In mice of the 129Sv background, most Dmrt6 mutant germ cells can complete spermatogonial differentiation and enter meiosis, but they show defects in meiotic chromosome pairing, establishment of the XY body and processing of recombination foci, and they mainly arrest in mid-pachynema. mRNA profiling of Dmrt6 mutant testes together with DMRT6 chromatin immunoprecipitation sequencing suggest that DMRT6 represses genes involved in spermatogonial differentiation and activates genes required for meiotic prophase. Our results indicate that Dmrt6 plays a key role in coordinating the transition in gametogenic programs from spermatogonial differentiation and mitosis to spermatocyte development and meiosis.
Collapse
Affiliation(s)
- Teng Zhang
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA Molecular, Cellular, Developmental Biology and Genetics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark W Murphy
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Micah D Gearhart
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vivian J Bardwell
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - David Zarkower
- Developmental Biology Center, Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| |
Collapse
|
137
|
Zhang Y, Song B, Du WD, He XJ, Ruan J, Zhou FS, Zuo XB, Ye L, Xie XS, Cao YX. Genetic association study ofRNF8andBRDTvariants with non-obstructive azoospermia in the Chinese Han population. Syst Biol Reprod Med 2014; 61:26-31. [DOI: 10.3109/19396368.2014.979513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
138
|
Aloisio GM, Nakada Y, Saatcioglu HD, Peña CG, Baker MD, Tarnawa ED, Mukherjee J, Manjunath H, Bugde A, Sengupta AL, Amatruda JF, Cuevas I, Hamra FK, Castrillon DH. PAX7 expression defines germline stem cells in the adult testis. J Clin Invest 2014; 124:3929-44. [PMID: 25133429 PMCID: PMC4153705 DOI: 10.1172/jci75943] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/01/2014] [Indexed: 12/22/2022] Open
Abstract
Spermatogenesis is a complex, multistep process that maintains male fertility and is sustained by rare germline stem cells. Spermatogenic progression begins with spermatogonia, populations of which express distinct markers. The identity of the spermatogonial stem cell population in the undisturbed testis is controversial due to a lack of reliable and specific markers. Here we identified the transcription factor PAX7 as a specific marker of a rare subpopulation of A(single) spermatogonia in mice. PAX7+ cells were present in the testis at birth. Compared with the adult testis, PAX7+ cells constituted a much higher percentage of neonatal germ cells. Lineage tracing in healthy adult mice revealed that PAX7+ spermatogonia self-maintained and produced expanding clones that gave rise to mature spermatozoa. Interestingly, in mice subjected to chemotherapy and radiotherapy, both of which damage the vast majority of germ cells and can result in sterility, PAX7+ spermatogonia selectively survived, and their subsequent expansion contributed to the recovery of spermatogenesis. Finally, PAX7+ spermatogonia were present in the testes of a diverse set of mammals. Our data indicate that the PAX7+ subset of A(single) spermatogonia functions as robust testis stem cells that maintain fertility in normal spermatogenesis in healthy mice and mediate recovery after severe germline injury, such as occurs after cancer therapy.
Collapse
Affiliation(s)
- Gina M. Aloisio
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Yuji Nakada
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Hatice D. Saatcioglu
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher G. Peña
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Michael D. Baker
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Edward D. Tarnawa
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jishnu Mukherjee
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Hema Manjunath
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Abhijit Bugde
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Anita L. Sengupta
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - James F. Amatruda
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ileana Cuevas
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - F. Kent Hamra
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| | - Diego H. Castrillon
- Department of Pathology, Department of
Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility,
Department of Cell Biology, Departments of Internal
Medicine, Molecular Biology, and Pediatrics, and Department of Pharmacology,
UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
139
|
Identification and characterization of germ cell genes expressed in the F9 testicular teratoma stem cell line. PLoS One 2014; 9:e103837. [PMID: 25153150 PMCID: PMC4143169 DOI: 10.1371/journal.pone.0103837] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/02/2014] [Indexed: 02/06/2023] Open
Abstract
The F9 cell line, which was derived from a mouse testicular teratoma that originated from pluripotent germ cells, has been used as a model for differentiation. However, it is largely unknown whether F9 cells possess the characteristics of male germ cells. In the present study, we investigated spermatogenic stage- and cell type-specific gene expression in F9 cells. Analysis of previous microarray data showed that a large number of stage-regulated germ cell genes are expressed in F9 cells. Specifically, genes that are prominently expressed in spermatogonia and have transcriptional regulatory functions appear to be enriched in F9 cells. Our in silico and in vitro analyses identified several germ cell-specific or -predominant genes that are expressed in F9 cells. Among them, strong promoter activities were observed in the regions upstream of the spermatogonial genes, Dmrt1 (doublesex and mab-3 related transcription factor 1), Stra8 (stimulated by retinoic acid gene 8) and Tex13 (testis expressed gene 13), in F9 cells. A detailed analysis of the Tex13 promoter allowed us to identify an enhancer and a region that is implicated in germ cell-specificity. We also found that Tex13 expression is regulated by DNA methylation. Finally, analysis of GFP (green fluorescent protein) TEX13 localization revealed that the protein distributes heterogeneously in the cytoplasm and nucleus, suggesting that TEX13 shuttles between these two compartments. Taken together, our results demonstrate that F9 cells express numerous spermatogonial genes and could be used for transcriptional studies focusing on such genes. As an example of this, we use F9 cells to provide comprehensive expressional information about Tex13, and report that this gene appears to encode a germ cell-specific protein that functions in the nucleus during early spermatogenesis.
Collapse
|
140
|
Broering TJ, Alavattam KG, Sadreyev RI, Ichijima Y, Kato Y, Hasegawa K, Camerini-Otero RD, Lee JT, Andreassen PR, Namekawa SH. BRCA1 establishes DNA damage signaling and pericentric heterochromatin of the X chromosome in male meiosis. ACTA ACUST UNITED AC 2014; 205:663-75. [PMID: 24914237 PMCID: PMC4050732 DOI: 10.1083/jcb.201311050] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The major role of BRCA1 in meiosis is not in meiotic recombination but instead in promotion of the dramatic chromatin changes required for formation and function of the XY body. During meiosis, DNA damage response (DDR) proteins induce transcriptional silencing of unsynapsed chromatin, including the constitutively unsynapsed XY chromosomes in males. DDR proteins are also implicated in double strand break repair during meiotic recombination. Here, we address the function of the breast cancer susceptibility gene Brca1 in meiotic silencing and recombination in mice. Unlike in somatic cells, in which homologous recombination defects of Brca1 mutants are rescued by 53bp1 deletion, the absence of 53BP1 did not rescue the meiotic failure seen in Brca1 mutant males. Further, BRCA1 promotes amplification and spreading of DDR components, including ATR and TOPBP1, along XY chromosome axes and promotes establishment of pericentric heterochromatin on the X chromosome. We propose that BRCA1-dependent establishment of X-pericentric heterochromatin is critical for XY body morphogenesis and subsequent meiotic progression. In contrast, BRCA1 plays a relatively minor role in meiotic recombination, and female Brca1 mutants are fertile. We infer that the major meiotic role of BRCA1 is to promote the dramatic chromatin changes required for formation and function of the XY body.
Collapse
Affiliation(s)
- Tyler J Broering
- Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Kris G Alavattam
- Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Ruslan I Sadreyev
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114 Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114 Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114 Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114
| | - Yosuke Ichijima
- Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Yasuko Kato
- Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Kazuteru Hasegawa
- Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - R Daniel Camerini-Otero
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jeannie T Lee
- Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114 Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114 Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114 Howard Hughes Medical Institute, Department of Molecular Biology, Massachusetts General Hospital, Department of Pathology, and Department of Genetics, Harvard Medical School, Boston, MA 02114
| | - Paul R Andreassen
- Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Satoshi H Namekawa
- Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229Division of Reproductive Sciences and Division of Developmental Biology, Perinatal Institute, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229 Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
141
|
Maclary E, Hinten M, Harris C, Kalantry S. Long nonoding RNAs in the X-inactivation center. Chromosome Res 2014; 21:601-614. [PMID: 24297756 DOI: 10.1007/s10577-013-9396-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The X-inactivation center is a hotbed of functional long noncoding RNAs in eutherian mammals. These RNAs are thought to help orchestrate the epigenetic transcriptional states of the two X-chromosomes in females as well as of the single X-chromosome in males. To balance X-linked gene expression between the sexes, females undergo transcriptional silencing of most genes on one of the two X-chromosomes in a process termed X-chromosome inactivation. While one X-chromosome is inactivated, the other X-chromosome remains active. Moreover, with a few notable exceptions, the originally established epigenetic transcriptional profiles of the two X-chromosomes is maintained as such through many rounds of cell division, essentially for the life of the organism. The stable and divergent transcriptional fates of the two X-chromosomes, despite residing in a shared nucleoplasm, make X-inactivation a paradigm of epigenetic transcriptional regulation. Originally proposed in 1961 by Mary Lyon, the X-inactivation hypothesis has been validated through much experimentation. In the last 25 years, the discovery and functional characterization has firmly established X-linked long noncoding RNAs as key players in choreographing X-chromosome inactivation.
Collapse
Affiliation(s)
- Emily Maclary
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Michael Hinten
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Clair Harris
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Sundeep Kalantry
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105
| |
Collapse
|
142
|
High-resolution mapping of chromatin packaging in mouse embryonic stem cells and sperm. Dev Cell 2014; 30:11-22. [PMID: 24998598 DOI: 10.1016/j.devcel.2014.05.024] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/28/2014] [Accepted: 05/30/2014] [Indexed: 11/24/2022]
Abstract
Mammalian embryonic stem cells (ESCs) and sperm exhibit unusual chromatin packaging that plays important roles in cellular function. Here, we extend a recently developed technique, based on deep paired-end sequencing of lightly digested chromatin, to assess footprints of nucleosomes and other DNA-binding proteins genome-wide in murine ESCs and sperm. In ESCs, we recover well-characterized features of chromatin such as promoter nucleosome depletion and further identify widespread footprints of sequence-specific DNA-binding proteins such as CTCF, which we validate in knockdown studies. We document global differences in nuclease accessibility between ESCs and sperm, finding that the majority of histone retention in sperm preferentially occurs in large gene-poor genomic regions, with only a small subset of nucleosomes being retained over promoters of developmental regulators. Finally, we describe evidence that CTCF remains associated with the genome in mature sperm, where it could play a role in organizing the sperm genome.
Collapse
|
143
|
Vernet N, Mahadevaiah SK, Yamauchi Y, Decarpentrie F, Mitchell MJ, Ward MA, Burgoyne PS. Mouse Y-linked Zfy1 and Zfy2 are expressed during the male-specific interphase between meiosis I and meiosis II and promote the 2nd meiotic division. PLoS Genet 2014; 10:e1004444. [PMID: 24967676 PMCID: PMC4072562 DOI: 10.1371/journal.pgen.1004444] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 05/02/2014] [Indexed: 11/19/2022] Open
Abstract
Mouse Zfy1 and Zfy2 encode zinc finger transcription factors that map to the short arm of the Y chromosome (Yp). They have previously been shown to promote meiotic quality control during pachytene (Zfy1 and Zfy2) and at the first meiotic metaphase (Zfy2). However, from these previous studies additional roles for genes encoded on Yp during meiotic progression were inferred. In order to identify these genes and investigate their function in later stages of meiosis, we created three models with diminishing Yp and Zfy gene complements (but lacking the Y-long-arm). Since the Y-long-arm mediates pairing and exchange with the X via their pseudoautosomal regions (PARs) we added a minute PAR-bearing X chromosome derivative to enable formation of a sex bivalent, thus avoiding Zfy2-mediated meiotic metaphase I (MI) checkpoint responses to the unpaired (univalent) X chromosome. Using these models we obtained definitive evidence that genetic information on Yp promotes meiosis II, and by transgene addition identified Zfy1 and Zfy2 as the genes responsible. Zfy2 was substantially more effective and proved to have a much more potent transactivation domain than Zfy1. We previously established that only Zfy2 is required for the robust apoptotic elimination of MI spermatocytes in response to a univalent X; the finding that both genes potentiate meiosis II led us to ask whether there was de novo Zfy1 and Zfy2 transcription in the interphase between meiosis I and meiosis II, and this proved to be the case. X-encoded Zfx was also expressed at this stage and Zfx over-expression also potentiated meiosis II. An interphase between the meiotic divisions is male-specific and we previously hypothesised that this allows meiosis II critical X and Y gene reactivation following sex chromosome silencing in meiotic prophase. The interphase transcription and meiosis II function of Zfx, Zfy1 and Zfy2 validate this hypothesis. The mouse Y chromosome genes Zfy1 and Zfy2 were first identified in the late 1980s during the search for the gene on the Y that triggers male development; they encode proteins that regulate the expression of other genes to which they bind via a ‘zinc finger’ domain. We have now discovered that these genes play important roles during spermatogenesis. Zfy2 proved to be essential for the efficient operation of a ‘checkpoint’ during the first meiotic division that identifies and kills cells that would otherwise produce sperm with an unbalanced chromosome set. Female meiosis, which does not have an equivalent checkpoint, generates a significant proportion of eggs with an unbalanced chromosome set. In the present study we show that Zfy2 also has a major role in ensuring that the second meiotic division occurs, with Zfy1 and a related gene, Zfx, on the X chromosome providing some support. In order to fulfil this function all three genes are expressed in the ‘interphase’ stage between the two divisions. In female meiosis there is no interphase stage between the two meiotic divisions but in this case essential functions during the divisions are supported by stored RNAs, so an interphase is not needed.
Collapse
Affiliation(s)
- Nadège Vernet
- MRC National Institute for Medical Research, London, United Kingdom
- Department of functional genomics and cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- * E-mail: ,
| | | | - Yasuhiro Yamauchi
- Institute for Biogenesis Research, University of Hawaii Medical School, Honolulu, Hawaii, United States of America
| | | | - Michael J. Mitchell
- Aix Marseille Université, GMGF, Marseille, France
- Inserm UMR_S 910, Marseille, France
| | - Monika A. Ward
- Institute for Biogenesis Research, University of Hawaii Medical School, Honolulu, Hawaii, United States of America
| | - Paul S. Burgoyne
- MRC National Institute for Medical Research, London, United Kingdom
| |
Collapse
|
144
|
Abstract
In several different taxa, there is indubitable evidence of transcriptional silencing of the X and Y chromosomes in male meiotic cells of spermatogenesis. However, the so called meiotic sex chromosome inactivation (MSCI) has been recently a hot bed for debate in Drosophila melanogaster. This review covers cytological and genetic observations, data from transgenic constructs with testis-specific promoters, global expression profiles obtained from mutant, wild-type, larvae and adult testes as well as from cells of different stages of spermatogenesis. There is no dispute on that D. melanogaster spermatogenesis presents a down-regulation of X chromosome that does not result from the lack of dosage compensation. However, the issue is currently focused on the level of reduction of X-linked expression, the precise time it occurs and how many genes are affected. The deep examination of data and experiments in this review exposes the limitations intrinsic to the methods of studying MSCI in D. melanogaster. The current methods do not allow us to affirm anything else than the X chromosome down-regulation in meiosis (MSCI). Therefore, conclusion about level, degree or precise timing is inadequate until new approaches are implemented to know the details of MSCI or other processes involved for D. melanogaster model.
Collapse
Affiliation(s)
- Maria D Vibranovski
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil 05508
| |
Collapse
|
145
|
Bergs JW, Neuendorff N, van der Heijden G, Wassenaar E, Rexin P, Elsässer HP, Moll R, Baarends WM, Brehm A. Differential expression and sex chromosome association of CHD3/4 and CHD5 during spermatogenesis. PLoS One 2014; 9:e98203. [PMID: 24849318 PMCID: PMC4029951 DOI: 10.1371/journal.pone.0098203] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 04/30/2014] [Indexed: 01/20/2023] Open
Abstract
ATP-dependent nucleosome remodelers of the CHD family play important roles in chromatin regulation during development and differentiation. The ubiquitously expressed CHD3 and CHD4 proteins are essential for stem cell function and serve to orchestrate gene expression in different developmental settings. By contrast, the closely related CHD5 is predominantly expressed in neural tissue and its role is believed to be restricted to neural differentiation. Indeed, loss of CHD5 contributes to neuroblastoma. In this study, we first demonstrate that CHD5 is a nucleosome-stimulated ATPase. We then compare CHD3/4 and CHD5 expression in mouse brain and show that CHD5 expression is restricted to a subset of cortical and hippocampal neurons whereas CHD3/4 expression is more widespread. We also uncover high levels of CHD5 expression in testis. CHD5 is transiently expressed in differentiating germ cells. Expression is first detected in nuclei of post-meiotic round spermatids, reaches a maximum in stage VIII spermatids and then falls to undetectable levels in stage IX spermatids. Surprisingly, CHD3/4 and CHD5 show complementary expression patterns during spermatogenesis with CHD3/4 levels progressively decreasing as CHD5 expression increases. In spermatocytes, CHD3/4 localizes to the pseudoautosomal region, the X centromeric region and then spreads into the XY body chromatin. In postmeiotic cells, CHD5 colocalises with macroH2A1.2 in association with centromeres and part of the Y chromosome. The subnuclear localisations of CHD4 and CHD5 suggest specific roles in regulation of sex chromosome chromatin and pericentromeric chromatin structure prior to the histone-protamine switch.
Collapse
Affiliation(s)
- Judith W. Bergs
- Institute for Molecular Biology and Tumor Research (IMT), Philipps Universität Marburg, Marburg, Germany
| | - Nina Neuendorff
- Institute for Molecular Biology and Tumor Research (IMT), Philipps Universität Marburg, Marburg, Germany
| | - Godfried van der Heijden
- Department of Reproduction and Development, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Evelyne Wassenaar
- Department of Reproduction and Development, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Peter Rexin
- Institut für Pathologie, Philipps Universität Marburg, Marburg, Germany
| | - Hans-Peter Elsässer
- Institut für Zytobiologie und Zytopathologie, Philipps Universität Marburg, Marburg, Germany
| | - Roland Moll
- Institut für Pathologie, Philipps Universität Marburg, Marburg, Germany
| | - Willy M. Baarends
- Department of Reproduction and Development, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Alexander Brehm
- Institute for Molecular Biology and Tumor Research (IMT), Philipps Universität Marburg, Marburg, Germany
| |
Collapse
|
146
|
Bonache S, Algaba F, Franco E, Bassas L, Larriba S. Altered gene expression signature of early stages of the germ line supports the pre-meiotic origin of human spermatogenic failure. Andrology 2014; 2:596-606. [PMID: 24803180 DOI: 10.1111/j.2047-2927.2014.00217.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/18/2014] [Accepted: 03/25/2014] [Indexed: 11/29/2022]
Abstract
The molecular basis of spermatogenic failure (SpF) is still largely unknown. Accumulating evidence suggests that a series of specific events such as meiosis, are determined at the early stage of spermatogenesis. This study aims to assess the expression profile of pre-meiotic genes of infertile testicular biopsies that might help to define the molecular phenotype associated with human deficiency of sperm production. An accurate quantification of testicular mRNA levels of genes expressed in spermatogonia was carried out by RT-qPCR in individuals showing SpF owing to germ cell maturation defects, Sertoli cell-only syndrome or conserved spermatogenesis. In addition, the gene expression profile of SpF was compared with that of testicular tumour, which is considered to be a severe developmental disease of germ cell differentiation. Protein expression from selected genes was evaluated by immunohistochemistry. Our results indicate that SpF is accompanied by differences in expression of certain genes associated with spermatogonia in the absence of any apparent morphological and/or numerical change in this specific cell type. In SpF testicular samples, we observed down-regulation of genes involved in cell cycle (CCNE1 and POLD1), transcription and post-transcription regulation (DAZL, RBM15 and DICER1), protein degradation (FBXO32 and TM9SF2) and homologous recombination in meiosis (MRE11A and RAD50) which suggests that the expression of these genes is critical for a proper germ cell development. Interestingly, a decrease in the CCNE1, DAZL, RBM15 and STRA8 cellular transcript levels was also observed, suggesting that the gene expression capacity of spermatogonia is altered in SpF contributing to an unsuccessful sperm production. Altogether, these data point to the spermatogenic derangement being already determined at, or arising in, the initial stages of the germ line.
Collapse
Affiliation(s)
- S Bonache
- Human Molecular Genetics Group, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | |
Collapse
|
147
|
Evans E, Hogarth C, Mitchell D, Griswold M. Riding the spermatogenic wave: profiling gene expression within neonatal germ and sertoli cells during a synchronized initial wave of spermatogenesis in mice. Biol Reprod 2014; 90:108. [PMID: 24719255 DOI: 10.1095/biolreprod.114.118034] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Continual sperm production relies on germ cells undergoing spermatogenesis asynchronously. As a result, the testis always contains a mixed population of germ cells at different stages of their differentiation process. The heterogeneous nature of the testis makes profiling gene expression within Sertoli cells or specific populations of germ cells impossible when a wild-type testis is assessed. We recently reported a unique method for synchronizing spermatogenesis without affecting fertility by manipulating RA levels within the neonatal testis. Using this protocol, combined with the RiboTag transgenic mouse line, we have mapped the Sertoli and germ cell translatome during the initial synchronized wave of spermatogenesis. Using microarray analysis, we identified 392 and 194 germ cell and Sertoli cells transcripts, respectively, that dynamically change during spermatogonial differentiation, division, and the onset of meiosis. Functional annotation clustering revealed that transcripts enriched in germ cells were mostly associated with meiosis (21 transcripts), chromatin organization (12 transcripts), and cell cycle (3 transcripts). In addition, glycoproteins (65 transcripts), cell adhesion (15 transcripts), and cell junction (13 transcripts) transcripts were overrepresented in the Sertoli cell-enriched list. These datasets represent the first transcriptional analysis of spermatogonial differentiation, division, and meiotic onset. These data suggest that several of the genes encoding meiotic proteins are expressed and are actively being translated well before germ cells enter meiosis. In addition, this study provides novel candidate genes, Asf1b and Esyt3, that may be involved in the regulation of spermatogonial chromatin reorganization, germ-Sertoli cell interactions, and/or blood-testis barrier formation.
Collapse
Affiliation(s)
- Elizabeth Evans
- School of Molecular Biosciences and The Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Cathryn Hogarth
- School of Molecular Biosciences and The Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Debra Mitchell
- School of Molecular Biosciences and The Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Michael Griswold
- School of Molecular Biosciences and The Center for Reproductive Biology, Washington State University, Pullman, Washington
| |
Collapse
|
148
|
Campbell P, Nachman MW. X-y interactions underlie sperm head abnormality in hybrid male house mice. Genetics 2014; 196:1231-40. [PMID: 24504187 PMCID: PMC3982709 DOI: 10.1534/genetics.114.161703] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 01/30/2014] [Indexed: 12/23/2022] Open
Abstract
The genetic basis of hybrid male sterility in house mice is complex, highly polygenic, and strongly X linked. Previous work suggested that there might be interactions between the Mus musculus musculus X and the M. m. domesticus Y with a large negative effect on sperm head morphology in hybrid males with an F1 autosomal background. To test this, we introgressed the M. m. domesticus Y onto a M. m. musculus background and measured the change in sperm morphology, testis weight, and sperm count across early backcross generations and in 11th generation backcross males in which the opportunity for X-autosome incompatibilities is effectively eliminated. We found that abnormality in sperm morphology persists in M. m. domesticus Y introgression males, and that this phenotype is rescued by M. m. domesticus introgressions on the X chromosome. In contrast, the severe reductions in testis weight and sperm count that characterize F1 males were eliminated after one generation of backcrossing. These results indicate that X-Y incompatibilities contribute specifically to sperm morphology. In contrast, X-autosome incompatibilities contribute to low testis weight, low sperm count, and sperm morphology. Restoration of normal testis weight and sperm count in first generation backcross males suggests that a small number of complex incompatibilities between loci on the M. m. musculus X and the M. m. domesticus autosomes underlie F1 male sterility. Together, these results provide insight into the genetic architecture of F1 male sterility and help to explain genome-wide patterns of introgression across the house mouse hybrid zone.
Collapse
Affiliation(s)
- Polly Campbell
- Corresponding author: Department of Zoology, 508 Life Sciences West, Oklahoma State University, Stillwater, OK 74078. E-mail:
| | | |
Collapse
|
149
|
Dai L, Peng C, Montellier E, Lu Z, Chen Y, Ishii H, Debernardi A, Buchou T, Rousseaux S, Jin F, Sabari BR, Deng Z, Allis CD, Ren B, Khochbin S, Zhao Y. Lysine 2-hydroxyisobutyrylation is a widely distributed active histone mark. Nat Chem Biol 2014; 10:365-70. [PMID: 24681537 DOI: 10.1038/nchembio.1497] [Citation(s) in RCA: 336] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 03/05/2014] [Indexed: 12/15/2022]
Abstract
We report the identification of a new type of histone mark, lysine 2-hydroxyisobutyrylation (Khib), and identify the mark at 63 human and mouse histone Khib sites, including 27 unique lysine sites that are not known to be modified by lysine acetylation (Kac) and lysine crotonylation (Kcr). This histone mark was initially identified by MS and then validated by chemical and biochemical methods. Histone Khib shows distinct genomic distributions from histone Kac or histone Kcr during male germ cell differentiation. Using chromatin immunoprecipitation sequencing, gene expression analysis and immunodetection, we show that in male germ cells, H4K8hib is associated with active gene transcription in meiotic and post-meiotic cells. In addition, H4K8ac-associated genes are included in and constitute only a subfraction of H4K8hib-labeled genes. The histone Khib mark is conserved and widely distributed, has high stoichiometry and induces a large structural change. These findings suggest its critical role on the regulation of chromatin functions.
Collapse
Affiliation(s)
- Lunzhi Dai
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Chao Peng
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Emilie Montellier
- INSERM, U823, Université Joseph Fourier-Grenoble 1, Institut Albert Bonniot, Faculté de Médecine, La Tronche, France
| | - Zhike Lu
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Yue Chen
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - Haruhiko Ishii
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California-San Diego School of Medicine, La Jolla, California, USA
| | - Alexandra Debernardi
- INSERM, U823, Université Joseph Fourier-Grenoble 1, Institut Albert Bonniot, Faculté de Médecine, La Tronche, France
| | - Thierry Buchou
- INSERM, U823, Université Joseph Fourier-Grenoble 1, Institut Albert Bonniot, Faculté de Médecine, La Tronche, France
| | - Sophie Rousseaux
- INSERM, U823, Université Joseph Fourier-Grenoble 1, Institut Albert Bonniot, Faculté de Médecine, La Tronche, France
| | - Fulai Jin
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California-San Diego School of Medicine, La Jolla, California, USA
| | - Benjamin R Sabari
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, New York, USA
| | - Zhiyou Deng
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| | - C David Allis
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, New York, USA
| | - Bing Ren
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California-San Diego School of Medicine, La Jolla, California, USA
| | - Saadi Khochbin
- INSERM, U823, Université Joseph Fourier-Grenoble 1, Institut Albert Bonniot, Faculté de Médecine, La Tronche, France
| | - Yingming Zhao
- Ben May Department of Cancer Research, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
150
|
Boussouar F, Goudarzi A, Buchou T, Shiota H, Barral S, Debernardi A, Guardiola P, Brindle P, Martinez G, Arnoult C, Khochbin S, Rousseaux S. A specific CBP/p300-dependent gene expression programme drives the metabolic remodelling in late stages of spermatogenesis. Andrology 2014; 2:351-9. [PMID: 24522976 DOI: 10.1111/j.2047-2927.2014.00184.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/27/2013] [Accepted: 12/30/2013] [Indexed: 11/28/2022]
Abstract
Histone hyperacetylation is thought to drive the replacement of histones by transition proteins that occur in elongating spermatids (ElS) after a general shut down of transcription. The molecular machineries underlying this histone hyperacetylation remain still undefined. Here, we focused our attention on the role of Cbp and p300 in histone hyperacetylation and in the preceding late-gene transcriptional activity in ElS. A strategy was designed to partially deplete Cbp and p300 in ElS. These cells progressed normally through spermiogenesis and showed normal histone hyperacetylation and removal. However, a genome-wide transcriptomic analysis, performed in the round spermatids (RS) and ElS, revealed the existence of a gene regulatory circuit encompassing genes presenting high expression levels in pre-meiotic cells, undergoing a repressed state in spermatocytes and early post-meiotic cells, but becoming reactivated in ElS, just prior to the global shutdown of transcription. Interestingly, this group of genes was over-represented within the genes affected by Cbp/p300 knock down and were all involved in metabolic remodelling. This study revealed the occurrence of a tightly regulated Cbp/p300-dependent gene expression programme that drives a specific metabolic state both in progenitor spermatogenic cells and in late transcriptionally active spermatids and confirmed a special link between Cpb/p300 and cell metabolism programming previously shown in somatic cells.
Collapse
Affiliation(s)
- F Boussouar
- INSERM, U823, Institut Albert Bonniot, Université Joseph Fourier - Grenoble 1, Grenoble, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|