1
|
Xu H, Chi Y, Yin C, Li C, Chen Y, Liu Z, Liu X, Xie H, Chen ZJ, Zhao H, Wu K, Zhao S, Xing D. Three-dimensional genome structures of single mammalian sperm. Nat Commun 2025; 16:3805. [PMID: 40268951 PMCID: PMC12019598 DOI: 10.1038/s41467-025-59055-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
The three-dimensional (3D) organization of chromosomes is crucial for packaging a large mammalian genome into a confined nucleus and ensuring proper nuclear functions in somatic cells. However, the packaging of the much more condensed sperm genome is challenging to study with traditional imaging or sequencing approaches. In this study, we develop an enhanced chromosome conformation capture assay, and resolve the 3D whole-genome structures of single mammalian sperm. The reconstructed genome structures accurately delineate the species-specific nuclear morphologies for both human and mouse sperm. We discover that sperm genomes are divided into chromosomal territories and A/B compartments, similarly to somatic cells. However, neither human nor mouse sperm chromosomes contain topologically associating domains or chromatin loops. These results suggest that the fine-scale chromosomal organization of mammalian sperm fundamentally differs from that of somatic cells. The discoveries and methods established in this work will be valuable for future studies of sperm related infertility.
Collapse
Affiliation(s)
- Heming Xu
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Yi Chi
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Changjian Yin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Cheng Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Yujie Chen
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China
| | - Zhiyuan Liu
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China
| | - Xiaowen Liu
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Peking University, Beijing, China
| | - Hao Xie
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Keliang Wu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Shigang Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China.
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China.
| |
Collapse
|
2
|
Hao J, Xu H, Chang B, Ren J, Wang H, Ji L. Acupuncture mediates the "gut-testis axis" to improve asthenozoospermia. Front Endocrinol (Lausanne) 2025; 16:1514010. [PMID: 39959619 PMCID: PMC11827431 DOI: 10.3389/fendo.2025.1514010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/07/2025] [Indexed: 02/18/2025] Open
Abstract
Background Asthenozoospermia is a common cause of male infertility. Studies have shown that sperm quality and motility are affected by the gut-testis axis that can regulate testicular metabolism and function through the gut microbiota and its metabolites. Acupuncture is an important modality of complementary and alternative medicine. It can improve sperm motility, but it remains unclear whether acupuncture can enhance sperm vitality by influencing the gut-testis axis. Methods In this study, sperm quality, testicular pathology, and serum hormone levels were assessed using a cyclophosphamide-induced mouse model. Real-time PCR, a western blot analysis, and immunofluorescence techniques were used to assess the effects of acupuncture on the gut barrier and blood-testis barrier functions. In addition, gut microbiome and metabolomics were used to study the impact of acupuncture on the gut microbiota structure, serum, and testicular metabolites in asthenozoospermic mice. Further validation was obtained by performing a fecal microbiota transplantation (FMT). Results Acupuncture improved the sperm quality; ameliorated testicular pathology; increased serum testosterone (T), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) levels; and repaired gut and blood-testis barrier damage in asthenozoospermic mice. The abundances of Bacteroidota, Firmicutes, Faecalibaculum, and Dubosiella were associated with sperm motility, as shown by a gut microbiome analysis. Serum metabolomics revealed that differentially expressed metabolites (DEMs), such as cytosine and N-oleyl-leucine, were closely related to sperm motility. Testicular metabolomics analysis revealed DEMs, such as 5-fluorouridine and 1-acetylimidazole, were also associated with sperm motility. Furthermore, reproductive function improvements in asthenozoospermic mice through acupuncture were achieved via an FMT. Conclusion Acupuncture may alleviate asthenozoospermia symptoms by modulating the gut-testis axis and repairing the gut-testis barrier.
Collapse
Affiliation(s)
- Jianheng Hao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Huichao Xu
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Boya Chang
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Jia Ren
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Haijun Wang
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Laixi Ji
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
3
|
Trigg N, Schjenken JE, Martin JH, Skerrett-Byrne DA, Smyth SP, Bernstein IR, Anderson AL, Stanger SJ, Simpson ENA, Tomar A, Teperino R, Conine CC, De Iuliis GN, Roman SD, Bromfield EG, Dun MD, Eamens AL, Nixon B. Subchronic elevation in ambient temperature drives alterations to the sperm epigenome and accelerates early embryonic development in mice. Proc Natl Acad Sci U S A 2024; 121:e2409790121. [PMID: 39527742 PMCID: PMC11588121 DOI: 10.1073/pnas.2409790121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Forecasted increases in the prevalence and severity of extreme weather events accompanying changes in climatic behavior pose potential risk to the reproductive capacity of humans and animals of ecological and agricultural significance. While several studies have revealed that heat stress induced by challenges such as testicular insulation can elicit a marked negative effect on the male reproductive system, and particularly the production of spermatozoa, less is known about the immediate impact on male reproductive function following subchronic whole-body exposure to elevated ambient temperature. To address this knowledge gap, we exposed unrestrained male mice to heat stress conditions that emulate a heat wave (daily cycle of 8 h at 35 °C followed by 16 h at 25 °C) for a period of 7 d. Neither the testes or epididymides of heat-exposed male mice exhibited evidence of gross histological change, and similarly, spermatozoa of exposed males retained their functionality and ability to support embryonic development. However, the embryos generated from heat-exposed spermatozoa experienced pronounced changes in gene expression linked to acceleration of early embryo development, aberrant blastocyst hatching, and increased fetal:placental weight ratio. Such changes were causally associated with an altered sperm small noncoding RNA (sncRNA) profile, such that these developmental phenotypes were recapitulated by microinjection of wild-type embryos sired by control spermatozoa with RNAs extracted from heat-exposed spermatozoa. Such data highlight that even relatively modest excursions in ambient temperature can affect male reproductive function and identify the sperm sncRNA profile as a particular point of vulnerability to this imposed environmental stress.
Collapse
Affiliation(s)
- Natalie Trigg
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- Department of Genetics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Department of Pediatrics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Center for Reproductive and Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - John E. Schjenken
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Jacinta H. Martin
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - David A. Skerrett-Byrne
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Shannon P. Smyth
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- School of BioSciences Bio21 Molecular Sciences and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, VIC3010, Australia
| | - Ilana R. Bernstein
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Amanda L. Anderson
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Simone J. Stanger
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Ewan N. A. Simpson
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Archana Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg85764, Germany
- German Center for Diabetes Research, Deutsche Zentrum für Diabetesforschung, Neuherberg85764, Germany
| | - Colin C. Conine
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Department of Pediatrics Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Center for Reproductive and Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Geoffry N. De Iuliis
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Shaun D. Roman
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- NSW Health Pathology, Newcastle, NSW2300, Australia
| | - Elizabeth G. Bromfield
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
- School of BioSciences Bio21 Molecular Sciences and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, VIC3010, Australia
| | - Matthew D. Dun
- Cancer Signaling Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| | - Andrew L. Eamens
- School of Health, University of the Sunshine Coast, Maroochydore, QLD4558, Australia
| | - Brett Nixon
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW2308, Australia
- Infertility and Reproduction Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW2305, Australia
| |
Collapse
|
4
|
Mielnicka M, Tabaro F, Sureka R, Acurzio B, Paoletti R, Scavizzi F, Raspa M, Crevenna AH, Lapouge K, Remans K, Boulard M. Trim66's paternal deficiency causes intrauterine overgrowth. Life Sci Alliance 2024; 7:e202302512. [PMID: 38719749 PMCID: PMC11077763 DOI: 10.26508/lsa.202302512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
The tripartite motif-containing protein 66 (TRIM66, also known as TIF1-delta) is a PHD-Bromo-containing protein primarily expressed in post-meiotic male germ cells known as spermatids. Biophysical assays showed that the TRIM66 PHD-Bromodomain binds to H3 N-terminus only when lysine 4 is unmethylated. We addressed TRIM66's role in reproduction by loss-of-function genetics in the mouse. Males homozygous for Trim66-null mutations produced functional spermatozoa. Round spermatids lacking TRIM66 up-regulated a network of genes involved in histone acetylation and H3K4 methylation. Profiling of H3K4me3 patterns in the sperm produced by the Trim66-null mutant showed minor alterations below statistical significance. Unexpectedly, Trim66-null males, but not females, sired pups overweight at birth, hence revealing that Trim66 mutations cause a paternal effect phenotype.
Collapse
Affiliation(s)
- Monika Mielnicka
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Italy
| | - Francesco Tabaro
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Italy
| | - Rahul Sureka
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Italy
| | - Basilia Acurzio
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Italy
| | | | - Ferdinando Scavizzi
- National Research Council (IBBC), CNR-Campus International Development (EMMA-INFRAFRONTIER-IMPC), Monterotondo, Italy
| | - Marcello Raspa
- National Research Council (IBBC), CNR-Campus International Development (EMMA-INFRAFRONTIER-IMPC), Monterotondo, Italy
| | - Alvaro H Crevenna
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Italy
| | - Karine Lapouge
- European Molecular Biology Laboratory, Protein Expression and Purification Core Facility, Heidelberg, Germany
| | - Kim Remans
- European Molecular Biology Laboratory, Protein Expression and Purification Core Facility, Heidelberg, Germany
| | - Matthieu Boulard
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Italy
| |
Collapse
|
5
|
Caroppo E, Skinner MK. Could the sperm epigenome become a diagnostic tool for evaluation of the infertile man? Hum Reprod 2024; 39:478-485. [PMID: 38148019 DOI: 10.1093/humrep/dead266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/10/2023] [Indexed: 12/28/2023] Open
Abstract
Although male infertility is currently diagnosed when abnormal sperm parameters are found, the poor predictive ability of sperm parameters on natural fecundity and medically assisted reproduction outcome poses the need for improved diagnostic techniques for male infertility. The accumulating evidence about the role played by the sperm epigenome in modulation of the early phases of embryonic development has led researchers to focus on the epigenetic mechanisms within the sperm epigenome to find new molecular markers of male infertility. Indeed, sperm epigenome abnormalities could explain some cases of unexplained male infertility in men showing normal sperm parameters and were found to be associated with poor embryo development in IVF cycles. The present mini-review summarizes the current knowledge about this interesting topic, starting from a description of the epigenetic mechanisms of gene expression regulation (i.e. DNA methylation, histone modifications, and non-coding RNAs' activity). We also discuss possible mechanisms by which environmental factors might cause epigenetic changes in the human germline and affect embryonic development, as well as subsequent generations' phenotypes. Studies demonstrating sperm epigenome abnormalities in men with male infertility are reviewed, with particular emphasis on those with the more severe form of spermatogenic dysfunction. Observations demonstrate that the diagnostic and prognostic efficacy of sperm epigenome evaluation will help facilitate the management of men with male factor infertility.
Collapse
Affiliation(s)
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
6
|
Relovska S, Wang H, Zhang X, Fernández-Tussy P, Jeong KJ, Choi J, Suárez Y, McDonald JG, Fernández-Hernando C, Chung JJ. DHCR24-mediated sterol homeostasis during spermatogenesis is required for sperm mitochondrial sheath formation and impacts male fertility over time. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572851. [PMID: 38187697 PMCID: PMC10769317 DOI: 10.1101/2023.12.21.572851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Desmosterol and cholesterol are essential lipid components of the sperm plasma membrane. Cholesterol efflux is required for capacitation, a process through which sperm acquire fertilizing ability. In this study, using a transgenic mouse model overexpressing 24-dehydrocholesterol reductase (DHCR24), an enzyme in the sterol biosynthesis pathway responsible for the conversion of desmosterol to cholesterol, we show that disruption of sterol homeostasis during spermatogenesis led to defective sperm morphology characterized by incomplete mitochondrial packing in the midpiece, reduced sperm count and motility, and a decline in male fertility with increasing paternal age, without changes in body fat composition. Sperm depleted of desmosterol exhibit inefficiency in the acrosome reaction, metabolic dysfunction, and an inability to fertilize the egg. These findings provide molecular insights into sterol homeostasis for sperm capacitation and its impact on male fertility.
Collapse
Affiliation(s)
- Sona Relovska
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Huafeng Wang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Xinbo Zhang
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pablo Fernández-Tussy
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kyung Jo Jeong
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jeffrey G. McDonald
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jean-Ju Chung
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Gynecology and Obstetrics, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
7
|
Yin Q, Yang CH, Strelkova OS, Wu J, Sun Y, Gopalan S, Yang L, Dekker J, Fazzio TG, Li XZ, Gibcus J, Rando OJ. Revisiting chromatin packaging in mouse sperm. Genome Res 2023; 33:2079-2093. [PMID: 38129076 PMCID: PMC10760523 DOI: 10.1101/gr.277845.123] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023]
Abstract
Mammalian sperm show an unusual and heavily compacted genomic packaging state. In addition to its role in organizing the compact and hydrodynamic sperm head, it has been proposed that sperm chromatin architecture helps to program gene expression in the early embryo. Scores of genome-wide surveys in sperm have reported patterns of chromatin accessibility, nucleosome localization, histone modification, and chromosome folding. Here, we revisit these studies in light of recent reports that sperm obtained from the mouse epididymis are contaminated with low levels of cell-free chromatin. In the absence of proper sperm lysis, we readily recapitulate multiple prominent genome-wide surveys of sperm chromatin, suggesting that these profiles primarily reflect contaminating cell-free chromatin. Removal of cell-free DNA, and appropriate lysis conditions, are together required to reveal a sperm chromatin state distinct from most previous reports. Using ATAC-seq to explore relatively accessible genomic loci, we identify a landscape of open loci associated with early development and transcriptional control. Histone modification and chromosome folding profiles also strongly support the hypothesis that prior studies suffer from contamination, but technical challenges associated with reliably preserving the architecture of the compacted sperm head prevent us from confidently assaying true localization patterns for these epigenetic marks. Together, our studies show that our knowledge of chromosome packaging in mammalian sperm remains largely incomplete, and motivate future efforts to more accurately characterize genome organization in mature sperm.
Collapse
Affiliation(s)
- Qiangzong Yin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Chih-Hsiang Yang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Olga S Strelkova
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Jingyi Wu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
- Departments of Cell Biology and Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yu Sun
- Center for RNA Biology: From Genome to Therapeutics, Department of Biochemistry and Biophysics, Department of Urology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Sneha Gopalan
- Department of Molecular Cellular and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Liyan Yang
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Job Dekker
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
- Howard Hughes Medical Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Thomas G Fazzio
- Department of Molecular Cellular and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Xin Zhiguo Li
- Center for RNA Biology: From Genome to Therapeutics, Department of Biochemistry and Biophysics, Department of Urology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Johan Gibcus
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, USA;
| |
Collapse
|
8
|
Kotková L, Drábek J. Age-related changes in sperm DNA methylation and their forensic and clinical implications. Epigenomics 2023; 15:1157-1173. [PMID: 38031735 DOI: 10.2217/epi-2023-0307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
As a link between a stable genome and a dynamic environment, epigenetics is a promising tool for mapping age-related changes in human DNA. Methylated cytosine changes at specific loci are generally less studied in sperm DNA than in somatic cell DNA. Age-related methylation changes can be connected to various reproductive health problems and multiple disorders in offspring. In addition, they can be helpful in forensic fields, where testing of specific loci in semen samples found at sexual assault crime scenes can predict a perpetrator's age and narrow down the police investigation. This review focuses on age-related methylation changes in sperm. It covers the biological role of methylation, methylation testing techniques and the implications of methylation changes in forensics and clinical practice.
Collapse
Affiliation(s)
- Lucie Kotková
- Institute of Molecular & Translational Medicine, Faculty of Medicine & Dentistry, Palacky University Olomouc and University Hospital Olomouc, 77900, Czech Republic
| | - Jiří Drábek
- Institute of Molecular & Translational Medicine, Faculty of Medicine & Dentistry, Palacky University Olomouc and University Hospital Olomouc, 77900, Czech Republic
| |
Collapse
|
9
|
Hodge MJ, de Las Heras-Saldana S, Rindfleish SJ, Stephen CP, Pant SD. QTLs and Candidate Genes Associated with Semen Traits in Merino Sheep. Animals (Basel) 2023; 13:2286. [PMID: 37508063 PMCID: PMC10376747 DOI: 10.3390/ani13142286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Ram semen traits play a significant role in conception outcomes, which in turn may influence reproductive efficiency and the overall productivity and profitability of sheep enterprises. Since hundreds of ewes may be inseminated from a single ejaculate, it is important to evaluate semen quality prior to use in sheep breeding programs. Given that semen traits have been found to be heritable, genetic variation likely contributes to the variability observed in these traits. Identifying such genetic variants could provide novel insights into the molecular mechanisms underlying variability in semen traits. Therefore, this study aimed to identify quantitative trait loci (QTLs) associated with semen traits in Merino sheep. A genome-wide association study (GWAS) was undertaken using 4506 semen collection records from 246 Merino rams collected between January 2002 and May 2021. The R package RepeatABEL was used to perform a GWAS for semen volume, gross motility, concentration, and percent post-thaw motility. A total of 35 QTLs, located on 16 Ovis aries autosomes (OARs), were significantly associated with either of the four semen traits in this study. A total of 89, 95, 33, and 73 candidate genes were identified, via modified Bonferroni, within the QTLs significantly associated with volume, gross motility, concentration, and percent post-thaw motility, respectively. Among the candidate genes identified, SORD, SH2B1, and NT5E have been previously described to significantly influence spermatogenesis, spermatozoal motility, and high percent post-thaw motility, respectively. Several candidate genes identified could potentially influence ram semen traits based on existing evidence in the literature. As such, validation of these putative candidates may offer the potential to develop future strategies to improve sheep reproductive efficiency. Furthermore, Merino ram semen traits are lowly heritable (0.071-0.139), and thus may be improved by selective breeding.
Collapse
Affiliation(s)
- Marnie J Hodge
- School of Agricultural, Environmental and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
- Apiam Animal Health, Apiam Genetic Services, Dubbo, NSW 2830, Australia
| | - Sara de Las Heras-Saldana
- Animal Genetics and Breeding Unit, a Joint Venture of NSW Department of Primary Industries and University of New England, Armidale, NSW 2351, Australia
| | | | - Cyril P Stephen
- School of Agricultural, Environmental and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
- Gulbali Institute, Charles Sturt University, Boorooma Street, Wagga Wagga, NSW 2678, Australia
| | - Sameer D Pant
- School of Agricultural, Environmental and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia
- Gulbali Institute, Charles Sturt University, Boorooma Street, Wagga Wagga, NSW 2678, Australia
| |
Collapse
|
10
|
Capra E, Turri F, Lazzari B, Biffani S, Lange Consiglio A, Ajmone Marsan P, Stella A, Pizzi F. CpG DNA methylation changes during epididymal sperm maturation in bulls. Epigenetics Chromatin 2023; 16:20. [PMID: 37254160 DOI: 10.1186/s13072-023-00495-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/19/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND During epididymal transit spermatozoa acquire specific morphological features which enhance their ability to swim in a progressive manner and interact with the oocytes. At the same time, sperm cells undergo specific molecular rearrangements essential for the fertilizing sperm to drive a correct embryo development. To assess epigenetic sperm changes during epididymal maturation, the caput, corpus and cauda epididymis sperm tracts were isolated from eight bulls and characterized for different sperm quality parameters and for CpG DNA methylation using Reduced Representation Bisulfite Sequencing (RRBS) able to identify differentially methylated regions (DMRs) in higher CpG density regions. RESULTS Caput sperm showed significant variation in motility and sperm kinetics variables, whereas spermatozoa collected from the corpus presented morphology variation and significant alterations in variables related to acrosome integrity. A total of 57,583 methylated regions were identified across the eight bulls, showing a significantly diverse distribution for sperm collected in the three epididymal regions. Differential methylation was observed between caput vs corpus (n = 11,434), corpus vs cauda (n = 12,372) and caput vs cauda (n = 2790). During epididymal transit a high proportion of the epigenome was remodeled, showing several regions in which methylation decreases from caput to corpus and increases from corpus to cauda. CONCLUSIONS Specific CpG DNA methylation changes in sperm isolated from the caput, corpus, and cauda epididymis tracts are likely to refine the sperm epigenome during sperm maturation, potentially impacting sperm fertilization ability and spatial organization of the genome during early embryo development.
Collapse
Affiliation(s)
- Emanuele Capra
- Institute of Agricultural Biology and Biotechnology, National Research Council IBBA CNR, 26900, Lodi, Italy.
| | - F Turri
- Institute of Agricultural Biology and Biotechnology, National Research Council IBBA CNR, 26900, Lodi, Italy
| | - B Lazzari
- Institute of Agricultural Biology and Biotechnology, National Research Council IBBA CNR, 26900, Lodi, Italy
- Institute of Agricultural Biology and Biotechnology, National Research Council IBBA CNR, 20133, Milano, Italy
| | - S Biffani
- Institute of Agricultural Biology and Biotechnology, National Research Council IBBA CNR, 20133, Milano, Italy
| | - A Lange Consiglio
- Department of Veterinary Medicine and Animal Sciences (DIVAS), Università degli Studi di Milano, 26900, Lodi, Italy
| | - P Ajmone Marsan
- Department of Animal Science, Food and Technology-DIANA, and Romeo and Enrica Invernizzi Research Center on Sustainable Dairy Production-CREI, Università Cattolica del Sacro Cuore, 29122, Piacenza, Italy
| | - A Stella
- Institute of Agricultural Biology and Biotechnology, National Research Council IBBA CNR, 20133, Milano, Italy
| | - F Pizzi
- Institute of Agricultural Biology and Biotechnology, National Research Council IBBA CNR, 26900, Lodi, Italy
| |
Collapse
|
11
|
Ewe CK, Rechavi O. The third barrier to transgenerational inheritance in animals: somatic epigenetic resetting. EMBO Rep 2023; 24:e56615. [PMID: 36862326 PMCID: PMC10074133 DOI: 10.15252/embr.202256615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/02/2023] [Accepted: 02/15/2023] [Indexed: 03/03/2023] Open
Abstract
After early controversy, it is now increasingly clear that acquired responses to environmental factors may perpetuate across multiple generations-a phenomenon termed transgenerational epigenetic inheritance (TEI). Experiments with Caenorhabditis elegans, which exhibits robust heritable epigenetic effects, demonstrated small RNAs as key factors of TEI. Here, we discuss three major barriers to TEI in animals, two of which, the "Weismann barrier" and germline epigenetic reprogramming, have been known for decades. These are thought to effectively prevent TEI in mammals but not to the same extent in C. elegans. We argue that a third barrier-that we termed "somatic epigenetic resetting"-may further inhibit TEI and, unlike the other two, restricts TEI in C. elegans as well. While epigenetic information can overcome the Weismann barrier and transmit from the soma to the germline, it usually cannot "travel back" directly from the germline to the soma in subsequent generations. Nevertheless, heritable germline memory may still influence the animal's physiology by indirectly modifying gene expression in somatic tissues.
Collapse
Affiliation(s)
- Chee Kiang Ewe
- Department of Neurobiology, Wise Faculty of Life Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Oded Rechavi
- Department of Neurobiology, Wise Faculty of Life Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
12
|
Chan SY, Wan CWT, Law TYS, Chan DYL, Fok EKL. The Sperm Small RNA Transcriptome: Implications beyond Reproductive Disorder. Int J Mol Sci 2022; 23:15716. [PMID: 36555356 PMCID: PMC9779749 DOI: 10.3390/ijms232415716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Apart from the paternal half of the genetic material, the male gamete carries assorted epigenetic marks for optimal fertilization and the developmental trajectory for the early embryo. Recent works showed dynamic changes in small noncoding RNA (sncRNA) in spermatozoa as they transit through the testicular environment to the epididymal segments. Studies demonstrated the changes to be mediated by epididymosomes during the transit through the adluminal duct in the epididymis, and the changes in sperm sncRNA content stemmed from environmental insults significantly altering the early embryo development and predisposing the offspring to metabolic disorders. Here, we review the current knowledge on the establishment of the sperm sncRNA transcriptome and their role in male-factor infertility, evidence of altered offspring health in response to the paternal life experiences through sperm sncRNA species and, finally, their implications in assisted reproductive technology in terms of epigenetic inheritance.
Collapse
Affiliation(s)
- Sze Yan Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Crystal Wing Tung Wan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tin Yu Samuel Law
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - David Yiu Leung Chan
- Department of Obstetrics and Gynecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ellis Kin Lam Fok
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu 610017, China
| |
Collapse
|
13
|
Drevet JR, Hallak J, Nasr-Esfahani MH, Aitken RJ. Reactive Oxygen Species and Their Consequences on the Structure and Function of Mammalian Spermatozoa. Antioxid Redox Signal 2022; 37:481-500. [PMID: 34913729 DOI: 10.1089/ars.2021.0235] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Significance: Among the 200 or so cell types that comprise mammals, spermatozoa have an ambiguous relationship with the reactive oxygen species (ROS) inherent in the consumption of oxygen that supports aerobic metabolism. Recent Advances: In this review, we shall see that spermatozoa need the action of ROS to reach their structural and functional maturity, but that due to intrinsic unique characteristics, they are, perhaps more than any other cell type, susceptible to oxidative damage. Recent studies have improved our knowledge of how oxidative damage affects sperm structures and functions. The focus of this review will be on how genetic and epigenetic oxidative alterations to spermatozoa can have dramatic unintended consequences in terms of both the support and the suppression of sperm function. Critical Issues: Oxidative stress can have dramatic consequences not only for the spermatozoon itself, but also, and above all, on its primary objective, which is to carry out fertilization and to ensure, in part, that the embryonic development program should lead to a healthy progeny. Future Directions: Sperm oxidative DNA damage largely affects the integrity of the paternal genetic material to such an extent that the oocyte may have difficulties in correcting it. Diagnostic and therapeutic actions should be considered more systematically, especially in men with difficulties to conceive. Research is underway to determine whether the epigenetic information carried by spermatozoa is also subject to changes mediated by pro-oxidative situations. Antioxid. Redox Signal. 37, 481-500.
Collapse
Affiliation(s)
- Joël R Drevet
- Faculty of Medicine, GReD Institute, INSERM U1103-CNRS UMR6293-Université Clermont Auvergne, Clermont-Ferrand, France
| | - Jorge Hallak
- Androscience, Science and Innovation Center in Andrology and High-Complex Clinical and Research Andrology Laboratory, São Paulo, Brazil.,Division of Urology, University of São Paulo, São Paulo, Brazil.,Men's Health Study Group, Institute for Advanced Studies, University of São Paulo, São Paulo, Brazil.,Reproductive Toxicology Unit, Department of Pathology, University of São Paulo, São Paulo, Brazil
| | - Mohammad-Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Isfahan Fertility and Infertility Center, Isfahan, Iran
| | - Robert J Aitken
- Faculty of Science and Priority Research Center for Reproductive Sciences, The University of Newcastle, Callaghan, Australia.,Faculty of Health and Medicine, Priority Research Center for Reproductive Sciences, The University of Newcastle, Callaghan, Australia.,Hunter Medical Research Institute, New Lambton Heights, Australia
| |
Collapse
|
14
|
Chen H, Scott-Boyer MP, Droit A, Robert C, Belleannée C. Sperm Heterogeneity Accounts for Sperm DNA Methylation Variations Observed in the Caput Epididymis, Independently From DNMT/TET Activities. Front Cell Dev Biol 2022; 10:834519. [PMID: 35392175 PMCID: PMC8981467 DOI: 10.3389/fcell.2022.834519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/04/2022] [Indexed: 11/15/2022] Open
Abstract
Following their production in the testis, spermatozoa enter the epididymis where they gain their motility and fertilizing abilities. This post-testicular maturation coincides with sperm epigenetic profile changes that influence progeny outcome. While recent studies highlighted the dynamics of small non-coding RNAs in maturing spermatozoa, little is known regarding sperm methylation changes and their impact at the post-fertilization level. Fluorescence-activated cell sorting (FACS) was used to purify spermatozoa from the testis and different epididymal segments (i.e., caput, corpus and cauda) of CAG/su9-DsRed2; Acr3-EGFP transgenic mice in order to map out sperm methylome dynamics. Reduced representation bisulfite sequencing (RRBS-Seq) performed on DNA from these respective sperm populations indicated that high methylation changes were observed between spermatozoa from the caput vs. testis with 5,546 entries meeting our threshold values (q value <0.01, methylation difference above 25%). Most of these changes were transitory during epididymal sperm maturation according to the low number of entries identified between spermatozoa from cauda vs. testis. According to enzymatic and sperm/epididymal fluid co-incubation assays, (de)methylases were not found responsible for these sperm methylation changes. Instead, we identified that a subpopulation of caput spermatozoa displayed distinct methylation marks that were susceptible to sperm DNAse treatment and accounted for the DNA methylation profile changes observed in the proximal epididymis. Our results support the paradigm that a fraction of caput spermatozoa has a higher propensity to bind extracellular DNA, a phenomenon responsible for the sperm methylome variations observed at the post-testicular level. Further investigating the degree of conservation of this sperm heterogeneity in human will eventually provide new considerations regarding sperm selection procedures used in fertility clinics.
Collapse
Affiliation(s)
- Hong Chen
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
- Center for Research in Reproduction, Development and Intergenerational Health, Quebec, QC, Canada
| | | | - Arnaud Droit
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Claude Robert
- Center for Research in Reproduction, Development and Intergenerational Health, Quebec, QC, Canada
- Faculty of Animal Sciences, Université Laval, Quebec, QC, Canada
| | - Clémence Belleannée
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
- Center for Research in Reproduction, Development and Intergenerational Health, Quebec, QC, Canada
- *Correspondence: Clémence Belleannée,
| |
Collapse
|
15
|
Barney R, Stalker K, Lutes A, Bayles A, Aston K, Jenkins T. Assessment of seminal cell-free DNA as a potential contaminate in studies of human sperm DNA methylation. Andrology 2022; 10:702-709. [PMID: 35179315 DOI: 10.1111/andr.13163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/10/2022] [Accepted: 01/30/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Human seminal cell-free DNA (cfDNA) methylation patterns have not yet been thoroughly explored, however, recent work in mouse has suggested that some cfDNA encountered in the epididymis may contaminate DNA methylation studies assessing the mature sperm. Such contamination could clearly prove to be a significant confounder, for many reasons, in epigenetic studies of male factor infertility. OBJECTIVES To explore the nature of seminal cfDNA methylation and the likelihood that it would be retained following standard semen sample processing for epigenetic analysis. MATERIALS AND METHODS We assessed 12 semen samples collected at Utah Fertility Center. For each sample, seminal cfDNA was isolated from the sperm pellet. The sperm was split into three aliquots including one exposed to DNase to remove any additional cfDNA (termed "pure sperm"), one not exposed to DNase, and one exposed to DNase but reintroduced to seminal cfDNA. We additionally assessed blood DNA as our benchmark for somatic cell DNA methylation patterns. DNA methylation was measured via Illumina's 850k array and assessed for differential regional methylation. RESULTS 46,352 differentially methylated regions (FDR > 40) were identified between pure sperm and seminal cfDNA. We found at these sites that the average DNA methylation in cfDNA always fell somewhere between the average methylation in sperm and in blood. We also assessed each sperm treatment groups at all 46,352 regions of interest and found no significant differences at any of these sites. DISCUSSION AND CONCLUSION Our data suggest that seminal cfDNA is a clear mixture of both somatic and germline DNA and that cfDNA is not a contaminating feature in sperm DNA methylation studies following standard protocols in human sperm DNA extraction. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ryan Barney
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Kelaney Stalker
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Andrew Lutes
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA.,Utah Fertility Center, Pleasant Grove, UT, USA
| | | | - Kenneth Aston
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Tim Jenkins
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA.,Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
16
|
Chromatin alterations during the epididymal maturation of mouse sperm refine the paternally inherited epigenome. Epigenetics Chromatin 2022; 15:2. [PMID: 34991687 PMCID: PMC8734183 DOI: 10.1186/s13072-021-00433-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Paternal lifestyle choices and male exposure history have a critical influence on the health and fitness of the next generation. Accordingly, defining the processes of germline programming is essential to resolving how the epigenetic memory of paternal experiences transmits to their offspring. Established dogma holds that all facets of chromatin organization and histone posttranslational modification are complete before sperm exits the testes. However, recent clinical and animal studies suggest that patterns of DNA methylation change during epididymal maturation. In this study, we used complementary proteomic and deep-sequencing approaches to test the hypothesis that sperm posttranslational histone modifications change during epididymal transit. RESULTS Using proteomic analysis to contrast immature spermatozoa and mature sperm isolated from the mouse epididymis, we find progressive changes in multiple histone posttranslational modifications, including H3K4me1, H3K27ac, H3K79me2, H3K64ac, H3K122ac, H4K16ac, H3K9me2, and H4K20me3. Interestingly, some of these changes only occurred on histone variant H3.3, and most involve chromatin modifications associated with gene enhancer activity. In contrast, the bivalent chromatin modifications, H3K4me3, and H3K27me3 remained constant. Using chromatin immunoprecipitation coupled with deep sequencing, we find that changes in histone h3, lysine 27 acetylation (H3K27ac) involve sharpening broad diffuse regions into narrow peaks centered on the promoter regions of genes driving embryonic development. Significantly, many of these regions overlap with broad domains of H3K4me3 in oocytes and ATAC-seq signatures of open chromatin identified in MII oocytes and sperm. In contrast, histone h3, lysine 9 dimethylation (H3K9me2) becomes enriched within the promoters of genes driving meiosis and in the distal enhancer regions of tissue-specific genes sequestered at the nuclear lamina. Maturing sperm contain the histone deacetylase enzymes HDAC1 and HDAC3, suggesting the NuRD complex may drive some of these changes. Finally, using Western blotting, we detected changes in chromatin modifications between caput and caudal sperm isolated from rams (Ovis aries), inferring changes in histone modifications are a shared feature of mammalian epididymal maturation. CONCLUSIONS These data extend our understanding of germline programming and reveal that, in addition to trafficking noncoding RNAs, changes in histone posttranslational modifications are a core feature of epididymal maturation.
Collapse
|