1
|
Snider PL, Sierra Potchanant EA, Matias C, Edwards DM, Brault JJ, Conway SJ. The Loss of Tafazzin Transacetylase Activity Is Sufficient to Drive Testicular Infertility. J Dev Biol 2024; 12:32. [PMID: 39728085 DOI: 10.3390/jdb12040032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Barth syndrome (BTHS) is a rare, infantile-onset, X-linked mitochondriopathy exhibiting a variable presentation of failure to thrive, growth insufficiency, skeletal myopathy, neutropenia, and heart anomalies due to mitochondrial dysfunction secondary to inherited TAFAZZIN transacetylase mutations. Although not reported in BTHS patients, male infertility is observed in several Tafazzin (Taz) mouse alleles and in a Drosophila mutant. Herein, we examined the male infertility phenotype in a BTHS-patient-derived D75H point-mutant knockin mouse (TazPM) allele that expresses a mutant protein lacking transacetylase activity. Neonatal and adult TazPM testes were hypoplastic, and their epididymis lacked sperm. Histology and biomarker analysis revealed TazPM spermatogenesis is arrested prior to sexual maturation due to an inability to undergo meiosis and the generation of haploid spermatids. Moreover, TazPM testicular mitochondria were found to be structurally abnormal, and there was an elevation of p53-dependent apoptosis within TazPM seminiferous tubules. Immunoblot analysis revealed that TazPM gamete genome integrity was compromised, and both histone γ-H2Ax and Nucleoside diphosphate kinase-5 protein expression were absent in juvenile TazPM testes when compared to controls. We demonstrate that Taz-mediated transacetylase activity is required within mitochondria for normal spermatogenesis, and its absence results in meiotic arrest. We hypothesize that elevated TazPM spermatogonial apoptosis causes azoospermia and complete infertility.
Collapse
Affiliation(s)
- Paige L Snider
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Catalina Matias
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Donna M Edwards
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jeffrey J Brault
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
2
|
Ding D, Pang MH, Deng M, Nguyen T, Liu Y, Sun X, Xu Z, Zhang Y, Zhai Y, Yan Y, Ishibashi T. Testis-specific H2B.W1 disrupts nucleosome integrity by reducing DNA-histone interactions. Nucleic Acids Res 2024; 52:11612-11625. [PMID: 39329259 PMCID: PMC11514470 DOI: 10.1093/nar/gkae825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Multiple testis-specific histone variants are involved in the dynamic chromatin transitions during spermatogenesis. H2B.W1 (previously called H2BFWT) is an H2B variant specific to primate testis with hitherto unclear functions, although its single-nucleotide polymorphisms (SNPs) are closely associated with male non-obstructive infertility. Here, we found that H2B.W1 is only expressed in the mid-late spermatogonia stages, and H2B.W1 nucleosomes are defined by a more flexible structure originating from weakened interactions between histones and DNA. Furthermore, one of its SNPs, H2B.W1-H100R, which is associated with infertility, further destabilizes the nucleosomes and increases the nucleosome unwrapping rate by interfering with the R100 and H4 K91/R92 interaction. Our results suggest that destabilizing H2B.W1 containing nucleosomes might change the chromatin structure of spermatogonia, and that H2B.W1-H100R enhances the nucleosome-destabilizing effects, leading to infertility.
Collapse
Affiliation(s)
- Dongbo Ding
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
| | - Matthew Y H Pang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
| | - Mingxi Deng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
| | - Thi Thuy Nguyen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
| | - Yue Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
| | - Xulun Sun
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
| | - Zhichun Xu
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, HKSAR, China
| | - Yingyi Zhang
- Biological Cryo-EM Center, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
| | - Yuanliang Zhai
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, HKSAR, China
| | - Yan Yan
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
- Shenzhen PKU-HKUST Medical Center, Shenzhen, China
| | - Toyotaka Ishibashi
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, NT, HKSAR, China
- The Hong Kong University of Science and Technology Fok Ying Tung Research Institute, Nansha, Guangzhou, China
| |
Collapse
|
3
|
Chi RPA, Xu X, Li JL, Xu X, Hu G, Brown P, Willson C, Kirsanov O, Geyer C, Huang CL, Morgan M, DeMayo F. WNK1 is required during male pachynema to sustain fertility. iScience 2023; 26:107616. [PMID: 37694147 PMCID: PMC10485039 DOI: 10.1016/j.isci.2023.107616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/04/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
WNK1 is an important regulator in many physiological functions, yet its role in male reproduction is unexplored. In the male germline, WNK1 is upregulated in preleptotene spermatocytes indicating possible function(s) in spermatogenic meiosis. Indeed, deletion of Wnk1 in mid-pachytene spermatocytes using the Wnt7a-Cre mouse led to male sterility which resembled non-obstructive azoospermia in humans, where germ cells failed to complete spermatogenesis and produced no sperm. Mechanistically, we found elevated MTOR expression and signaling in the Wnk1-depleted spermatocytes. As MTOR is a central mediator of translation, we speculated that translation may be accelerated in these spermatocytes. Supporting this, we found the acrosome protein, ACRBP to be prematurely expressed in the spermatocytes with Wnk1 deletion. Our study uncovered an MTOR-regulating factor in the male germline with potential implications in translation, and future studies will aim to understand how WNK1 regulates MTOR activity and impact translation on a broader spectrum.
Collapse
Affiliation(s)
- Ru-pin Alicia Chi
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Xiaojiang Xu
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Xin Xu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Paula Brown
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Cynthia Willson
- Integrated Laboratory Systems LLC, Research Triangle Park, NC 27709, USA
| | - Oleksandr Kirsanov
- Department of Anatomy & Cell Biology at the Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Christopher Geyer
- Department of Anatomy & Cell Biology at the Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Chou-Long Huang
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa, IA 52242, USA
| | - Marcos Morgan
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Francesco DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| |
Collapse
|
4
|
Matsumura T, Katagiri K, Yao T, Ishikawa-Yamauchi Y, Nagata S, Hashimoto K, Sato T, Kimura H, Shinohara T, Sanbo M, Hirabayashi M, Ogawa T. Generation of rat offspring using spermatids produced through in vitro spermatogenesis. Sci Rep 2023; 13:12105. [PMID: 37495678 PMCID: PMC10372019 DOI: 10.1038/s41598-023-39304-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023] Open
Abstract
An in vitro spermatogenesis method using mouse testicular tissue to produce fertile sperm was established more than a decade ago. Although this culture method has generally not been effective in other animal species, we recently succeeded in improving the culture condition to induce spermatogenesis of rats up to the round spermatid stage. In the present study, we introduced acrosin-EGFP transgenic rats in order to clearly monitor the production of haploid cells during spermatogenesis in vitro. In addition, a metabolomic analysis of the culture media during cultivation revealed the metabolic dynamics of the testis tissue. By modifying the culture media based on these results, we were able to induce rat spermatogenesis repeatedly up to haploid cell production, including the formation of elongating spermatids, which was confirmed histologically and immunohistochemically. Finally, we performed a microinsemination experiment with in vitro produced spermatids, which resulted in the production of healthy and fertile offspring. This is the first demonstration of the in vitro production of functional haploid cells that yielded offspring in animals other than mice. These results are expected to provide a basis for the development of an in vitro spermatogenesis system applicable to many other mammals.
Collapse
Affiliation(s)
- Takafumi Matsumura
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
| | - Kumiko Katagiri
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
| | - Tatsuma Yao
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
- Research and Development Center, Fuso Pharmaceutical Industries, Ltd., 2-3-30 Morinomiya, Joto-ku, Osaka, 536-8523, Japan
| | - Yu Ishikawa-Yamauchi
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
| | - Shino Nagata
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama, Kanagawa, 230-0045, Japan
| | - Kiyoshi Hashimoto
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
- Department of Urology, Yokohama City University School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
| | - Takuya Sato
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan
| | - Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Makoto Sanbo
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Masumi Hirabayashi
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan.
| | - Takehiko Ogawa
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, 236-0004, Japan.
- Department of Urology, Yokohama City University School of Medicine, Yokohama, Kanagawa, 236-0004, Japan.
| |
Collapse
|
5
|
Ran L, Gao Z, Chen Q, Ran Y, Duan N, Hu G, Lu X, Xia R, Li Q, Cui F, Liu X, Xue B. Improving effects of telmisartan on spermatogenic disorder induced by fractionated low-dose irradiation in mice. Int Urol Nephrol 2023; 55:1427-1439. [PMID: 37093439 DOI: 10.1007/s11255-023-03601-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/12/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Male infertility is a hot problem worldwide, but there are few treatments, especially male infertility caused by irradiation is difficult to treat. The aim of this study was to investigate and evaluate novel drugs for the treatment of male infertility caused by irradiation. METHODS we randomly divided 18 male BALB/c mice into 3 groups: control, irradiated, and telmisartan. Both irradiated and telmisartan group completed whole-body 0.5 Gy five times irradiation, and the telmisartan group received intraperitoneal injection of telmisartan (1.2 mg/kg) daily on the next day after irradiation, and all groups were sampled on day 25 after irradiation. RESULTS Sperm motility results show that total sperm motility of irradiated group was significantly lower compared with control group, and testicular HE results showed that testis in irradiated group were severely damaged. Compared with irradiated group, the total sperm motility, sperm concentration, testicular index, Johnsen score, and the seminiferous tubule layer numbers were higher in telmisartan group (P < 0.05). The immunohistochemical staining showed γ-H2AX expression is higher in telmisartan group compared with irradiated group. And the relative mRNA expression of PLZF, GFRA1, STRA8, DMRT1, SPO11, SYCP2, OVOL2, CCNA1, TJP3, RUNX2, TXNDC2 TNP1, and PRM3 in telmisartan group was all significantly higher than irradiated group (P < 0.05). CONCLUSION In conclusion, in vivo experiments confirmed that telmisartan ameliorated the spermatogenic disorder in mice caused by fractionated low-dose irradiation via promoting spermatogenesis.
Collapse
Affiliation(s)
- Lingxiang Ran
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Zhixiang Gao
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Qiu Chen
- School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yuanshuai Ran
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Nengliang Duan
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Guangmo Hu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xingyu Lu
- School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Renlan Xia
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Qiaoqiao Li
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Fengmei Cui
- School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xiaolong Liu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| | - Boxin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
6
|
Philibert P, Déjardin S, Girard M, Durix Q, Gonzalez AA, Mialhe X, Tardat M, Poulat F, Boizet-Bonhoure B. Cocktails of NSAIDs and 17α Ethinylestradiol at Environmentally Relevant Doses in Drinking Water Alter Puberty Onset in Mice Intergenerationally. Int J Mol Sci 2023; 24:ijms24065890. [PMID: 36982971 PMCID: PMC10099742 DOI: 10.3390/ijms24065890] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) and 17α-ethinyl-estradiol (EE2) are among the most relevant endocrine-disrupting pharmaceuticals found in the environment, particularly in surface and drinking water due to their incomplete removal via wastewater treatment plants. Exposure of pregnant mice to NSAID therapeutic doses during the sex determination period has a negative impact on gonadal development and fertility in adults; however, the effects of their chronic exposure at lower doses are unknown. In this study, we investigated the impact of chronic exposure to a mixture containing ibuprofen, 2hydroxy-ibuprofen, diclofenac, and EE2 at two environmentally relevant doses (added to the drinking water from fetal life until puberty) on the reproductive tract in F1 exposed mice and their F2 offspring. In F1 animals, exposure delayed male puberty and accelerated female puberty. In post-pubertal F1 testes and ovaries, differentiation/maturation of the different gonad cell types was altered, and some of these modifications were observed also in the non-exposed F2 generation. Transcriptomic analysis of post-pubertal testes and ovaries of F1 (exposed) and F2 animals revealed significant changes in gene expression profiles and enriched pathways, particularly the inflammasome, metabolism and extracellular matrix pathways, compared with controls (non-exposed). This suggested that exposure to these drug cocktails has an intergenerational impact. The identified Adverse Outcome Pathway (AOP) networks for NSAIDs and EE2, at doses that are relevant to everyday human exposure, will improve the AOP network of the human reproductive system development concerning endocrine disruptor chemicals. It may serve to identify other putative endocrine disruptors for mammalian species based on the expression of biomarkers.
Collapse
Affiliation(s)
- Pascal Philibert
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, 34090 Montpellier, France
- Laboratoire de Biochimie et Biologie Moléculaire, Hôpital Carèmeau, CHU de Nîmes, 30900 Nîmes, France
| | - Stéphanie Déjardin
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, 34090 Montpellier, France
| | - Mélissa Girard
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, 34090 Montpellier, France
| | - Quentin Durix
- IExplore-RAM, Institut de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Université de Montpellier and Institut National de la Santé Et de la Recherche Médicale (INSERM), 34090 Montpellier, France
| | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, UMS Biocampus, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France
| | - Xavier Mialhe
- MGX-Montpellier GenomiX, UMS Biocampus, Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France
| | - Mathieu Tardat
- Biologie des Séquences Répétées, Institut de Génétique Humaine, Centre National de la Recherche Scientifique, Université de Montpellier, 34090 Montpellier, France
| | - Francis Poulat
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, 34090 Montpellier, France
| | - Brigitte Boizet-Bonhoure
- Développement et Pathologie de la Gonade, Institut de Génétique Humaine, Centre National de la Recherche Scientifique (CNRS), Université de Montpellier, 34090 Montpellier, France
| |
Collapse
|
7
|
Li Y, Meng R, Li S, Gu B, Xu X, Zhang H, Tan X, Shao T, Wang J, Xu D, Wang F. The ZFP541-KCTD19 complex is essential for pachytene progression by activating meiotic genes during mouse spermatogenesis. J Genet Genomics 2022; 49:1029-1041. [PMID: 35341968 DOI: 10.1016/j.jgg.2022.03.005] [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: 12/13/2021] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 12/29/2022]
Abstract
Meiosis is essential for fertility in sexually reproducing species and this sophisticated process has been extensively studied. Notwithstanding these efforts, key factors involved in meiosis have not been fully characterized. In this study, we investigate the regulatory roles of zinc finger protein 541 (ZFP541) and its interacting protein potassium channel tetramerization domain containing 19 (KCTD19) in spermatogenesis. ZFP541 is expressed from leptotene to the round spermatid stage, while the expression of KCTD19 is initiated in pachytene. Depletion of Zfp541 or Kctd19 leads to infertility in male mice and delays progression from early to mid/late pachynema. In addition, Zfp541-/- spermatocytes show abnormal programmed DNA double-strand break repair, impaired crossover formation and resolution, and asynapsis of the XY chromosomes. ZFP541 interacts with KCTD19, histone deacetylase 1/2 (HDAC1/2), and deoxynucleotidyl transferase terminal-interacting protein 1 (DNTTIP1). Moreover, ZFP541 binds to and activates the expression of genes involved in meiosis and post-meiosis including Kctd19; in turn, KCTD19 promotes the transcriptional activation activity of ZFP541. Taken together, our studies reveal that the ZFP541/KCTD19 signaling complex, acting as a key transcription regulator, plays an indispensable role in male fertility by regulating pachytene progression.
Collapse
Affiliation(s)
- Yushan Li
- The School of Public Health, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Ranran Meng
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Shanze Li
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Bowen Gu
- National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Xiaotong Xu
- National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Haihang Zhang
- National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Xinshui Tan
- National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Tianyu Shao
- National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Jiawen Wang
- National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Dan Xu
- National Institute of Biological Sciences Beijing, Beijing 102206, China
| | - Fengchao Wang
- National Institute of Biological Sciences Beijing, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
8
|
Understanding the Underlying Molecular Mechanisms of Meiotic Arrest during In Vitro Spermatogenesis in Rat Prepubertal Testicular Tissue. Int J Mol Sci 2022; 23:ijms23115893. [PMID: 35682573 PMCID: PMC9180380 DOI: 10.3390/ijms23115893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 12/10/2022] Open
Abstract
In vitro spermatogenesis appears to be a promising approach to restore the fertility of childhood cancer survivors. The rat model has proven to be challenging, since germ cell maturation is arrested in organotypic cultures. Here, we report that, despite a meiotic entry, abnormal synaptonemal complexes were found in spermatocytes, and in vitro matured rat prepubertal testicular tissues displayed an immature phenotype. RNA-sequencing analyses highlighted up to 600 differentially expressed genes between in vitro and in vivo conditions, including genes involved in blood-testis barrier (BTB) formation and steroidogenesis. BTB integrity, the expression of two steroidogenic enzymes, and androgen receptors were indeed altered in vitro. Moreover, most of the top 10 predicted upstream regulators of deregulated genes were involved in inflammatory processes or immune cell recruitment. However, none of the three anti-inflammatory molecules tested in this study promoted meiotic progression. By analysing for the first time in vitro matured rat prepubertal testicular tissues at the molecular level, we uncovered the deregulation of several genes and revealed that defective BTB function, altered steroidogenic pathway, and probably inflammation, could be at the origin of meiotic arrest.
Collapse
|
9
|
Delessard M, Stalin L, Rives-Feraille A, Moutard L, Saulnier J, Dumont L, Rives N, Rondanino C. Achievement of complete in vitro spermatogenesis in testicular tissues from prepubertal mice exposed to mono- or polychemotherapy. Sci Rep 2022; 12:7407. [PMID: 35523907 PMCID: PMC9076692 DOI: 10.1038/s41598-022-11286-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/18/2022] [Indexed: 02/08/2023] Open
Abstract
The assessment of the impact of chemotherapies on in vitro spermatogenesis in experimental models is required before considering the application of this fertility restoration strategy to prepubertal boys who received these treatments before testicular tissue cryopreservation. The present work investigated the effects of exposure of prepubertal mice to mono- (vincristine or cyclophosphamide) and polychemotherapy (a combination of vincristine and cyclophosphamide) on the first wave of in vitro spermatogenesis. When testicular tissue exposed to monochemotherapy was preserved, polychemotherapy led to severe alterations of the seminiferous epithelium and increased apoptosis in prepubertal testes prior in vitro maturation, suggesting a potential additive gonadotoxic effect. These alterations were also found in the testicular tissues of polychemotherapy-treated mice after 30 days of organotypic culture and were associated with a reduction in the germ cell/Sertoli cell ratio. The different treatments neither altered the ability of spermatogonia to differentiate in vitro into spermatozoa nor the yield of in vitro spermatogenesis. However, more spermatozoa with morphological abnormalities and fragmented DNA were produced after administration of polychemotherapy. This work therefore shows for the first time the possibility to achieve a complete in vitro spermatogenesis after an in vivo exposure of mice to a mono- or polychemotherapy before meiotic entry.
Collapse
Affiliation(s)
- Marion Delessard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Stalin
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Aurélie Rives-Feraille
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Moutard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Justine Saulnier
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Ludovic Dumont
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Nathalie Rives
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Christine Rondanino
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France.
| |
Collapse
|
10
|
Liu N, Qadri F, Busch H, Huegel S, Sihn G, Chuykin I, Hartmann E, Bader M, Rother F. Kpna6 deficiency causes infertility in male mice by disrupting spermatogenesis. Development 2021; 148:272018. [PMID: 34473250 PMCID: PMC8513612 DOI: 10.1242/dev.198374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 08/18/2021] [Indexed: 11/20/2022]
Abstract
Spermatogenesis is driven by an ordered series of events, which rely on trafficking of specific proteins between nucleus and cytoplasm. The karyopherin α family of proteins mediates movement of specific cargo proteins when bound to karyopherin β. Karyopherin α genes have distinct expression patterns in mouse testis, implying they may have unique roles during mammalian spermatogenesis. Here, we use a loss-of-function approach to determine specifically the role of Kpna6 in spermatogenesis and male fertility. We show that ablation of Kpna6 in male mice leads to infertility and has multiple cumulative effects on both germ cells and Sertoli cells. Kpna6-deficient mice exhibit impaired Sertoli cell function, including loss of Sertoli cells and a compromised nuclear localization of the androgen receptor. Furthermore, our data demonstrate devastating defects on spermiogenesis, including incomplete sperm maturation and a massive reduction in sperm number, accompanied by disturbed histone-protamine exchange, differential localization of the transcriptional regulator BRWD1 and altered expression of RFX2 target genes. Our work uncovers an essential role of Kpna6 in spermatogenesis and, hence, in male fertility. Summary: Two different mouse models delineate the morphological and functional impact of Kpna6 on spermatogenesis and Sertoli cell function and show that this protein is crucial for fertility in male mice.
Collapse
Affiliation(s)
- Na Liu
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | | | - Hauke Busch
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, Lübeck 23562, Germany
| | - Stefanie Huegel
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| | - Gabin Sihn
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Ilya Chuykin
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Department of Cell Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA
| | - Enno Hartmann
- Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| | - Franziska Rother
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany.,Institute for Biology, Center for Structural and Cellular Biology in Medicine, University of Lübeck, Lübeck 23562, Germany
| |
Collapse
|
11
|
Zhuang XJ, Feng X, Tang WH, Zhu JL, Li M, Li JS, Zheng XY, Li R, Liu P, Qiao J. FAM9B serves as a novel meiosis-related protein localized in meiotic chromosome cores and is associated with human gametogenesis. PLoS One 2021; 16:e0257248. [PMID: 34507348 PMCID: PMC8432983 DOI: 10.1371/journal.pone.0257248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023] Open
Abstract
Meiosis is a complex process involving the expression and interaction of numerous genes in a series of highly orchestrated molecular events. Fam9b localized in Xp22.3 has been found to be expressed in testes. However, FAM9B expression, localization, and its role in meiosis have not been previously reported. In this study, FAM9B expression was evaluated in the human testes and ovaries by RT-PCR, qPCR, and western blotting. FAM9B was found in the nuclei of primary spermatocytes in testes and specifically localized in the synaptonemal complex (SC) region of spermatocytes. FAM9B was also evident in the follicle cell nuclei and diffusely dispersed in the granular cell cytoplasm. FAM9B was partly co-localized with SYCP3, which is essential for both formation and maintenance of lateral SC elements. In addition, FAM9B had a similar distribution pattern and co-localization as γH2AX, which is a novel biomarker for DNA double-strand breaks during meiosis. All results indicate that FAM9B is a novel meiosis-associated protein that is co-localized with SYCP3 and γH2AX and may play an important role in SC formation and DNA recombination during meiosis. These findings offer a new perspective for understanding the molecular mechanisms involved in meiosis of human gametogenesis.
Collapse
Affiliation(s)
- Xin-jie Zhuang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- * E-mail: (PL); (XJZ)
| | - Xue Feng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
| | - Wen-hao Tang
- Department of Urology, The Third Hospital of Peking University, Beijing, China
| | - Jin-liang Zhu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
| | - Ming Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
| | - Jun-sheng Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
| | - Xiao-ying Zheng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
| | - Ping Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- * E-mail: (PL); (XJZ)
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Haidian District, Beijing, PR China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Haidian District, Beijing, PR China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Haidian District, Beijing, PR China
| |
Collapse
|
12
|
Nguyen-Powanda P, Robaire B. Aging and oxidative stress alter DNA repair mechanisms in male germ cells of superoxide dismutase-1 null mice. Biol Reprod 2021; 105:944-957. [PMID: 34098580 DOI: 10.1093/biolre/ioab114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/17/2021] [Accepted: 05/29/2021] [Indexed: 11/13/2022] Open
Abstract
The efficiency of antioxidant defense system decreases with aging, thus resulting in high levels of reactive oxygen species (ROS) and DNA damage in spermatozoa. This damage can lead to genetic disorders in the offspring. There are limited studies investigating the effects of the total loss of antioxidants, such as superoxide dismutase-1 (SOD1), in male germ cells as they progress through spermatogenesis. In this study, we evaluated the effects of aging and removing SOD1 (in male germ cells of SOD1-null (Sod1-/-) mice) in order to determine the potential mechanism(s) of DNA damage in these cells. Immunohistochemical analysis showed an increase in lipid peroxidation and DNA damage in the germ cells of aged wild-type (WT) and Sod1-/- mice of all age. Immunostaining of OGG1, a marker of base excision repair (BER), increased in aged WT and young Sod1-/- mice. In contrast, immunostaining intensity of LIGIV and RAD51, markers of non-homologous end-joining (NHEJ) and homologous recombination (HR), respectively, decreased in aged and Sod1-/- mice. Gene expression analysis showed similar results with altered mRNA expression of these key DNA repair transcripts in pachytene spermatocytes and round spermatids of aged and Sod1-/- mice. Our study indicates that DNA repair pathway markers of BER, NHEJ, and HR are differentially regulated as a function of aging and oxidative stress in spermatocytes and spermatids, and aging enhances the repair response to increased oxidative DNA damage, whereas impairments in other DNA repair mechanisms may contribute to the increase in DNA damage caused by aging and the loss of SOD1.
Collapse
Affiliation(s)
| | - Bernard Robaire
- Department of Obstetrics & Gynecology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
13
|
Matsumura T, Sato T, Abe T, Sanjo H, Katagiri K, Kimura H, Fujii T, Tanaka H, Hirabayashi M, Ogawa T. Rat in vitro spermatogenesis promoted by chemical supplementations and oxygen-tension control. Sci Rep 2021; 11:3458. [PMID: 33568686 PMCID: PMC7875995 DOI: 10.1038/s41598-021-82792-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
In vitro spermatogenesis (IVS) using air-liquid interphase organ culture method is possible with mouse testis tissues. The same method, however, has been hardly applicable to animals other than mice, only producing no or limited progression of spermatogenesis. In the present study, we challenged IVS of rats with modifications of culture medium, by supplementing chemical substances, including hormones, antioxidants, and lysophospholipids. In addition, reducing oxygen tension by placing tissues in an incubator of lower oxygen concentration and/or applying silicone cover ceiling on top of the tissue were effective for improving the spermatogenic efficiency. Through these modifications of the culture condition, rat spermatogenesis up to round spermatids was maintained over 70 days in the cultured tissue. Present results demonstrated a significant progress in rat IVS, revealing conditions commonly favorable for mice and rats as well as finding rat-specific optimizations. This is an important step towards successful IVS in many animal species, including humans.
Collapse
Affiliation(s)
- Takafumi Matsumura
- grid.268441.d0000 0001 1033 6139Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama, Kanagawa Japan
| | - Takuya Sato
- grid.268441.d0000 0001 1033 6139Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama, Kanagawa Japan
| | - Takeru Abe
- grid.268441.d0000 0001 1033 6139Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama, Kanagawa Japan
| | - Hiroyuki Sanjo
- grid.268441.d0000 0001 1033 6139Department of Urology, Yokohama City University School of Medicine, Yokohama, Kanagawa Japan
| | - Kumiko Katagiri
- grid.268441.d0000 0001 1033 6139Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama, Kanagawa Japan
| | - Hiroshi Kimura
- grid.265061.60000 0001 1516 6626Department of Mechanical Engineering, Tokai University, Hiratsuka, Kanagawa Japan
| | - Teruo Fujii
- grid.26999.3d0000 0001 2151 536XInstitute of Industrial Science, University of Tokyo, Bunkyo, Tokyo Japan
| | - Hiromitsu Tanaka
- grid.411871.a0000 0004 0647 5488Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki Japan
| | - Masumi Hirabayashi
- grid.467811.d0000 0001 2272 1771Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi Japan
| | - Takehiko Ogawa
- grid.268441.d0000 0001 1033 6139Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama, Kanagawa Japan ,grid.268441.d0000 0001 1033 6139Department of Urology, Yokohama City University School of Medicine, Yokohama, Kanagawa Japan
| |
Collapse
|
14
|
Zhang ZH, Jiang TX, Chen LB, Zhou W, Liu Y, Gao F, Qiu XB. Proteasome subunit α4s is essential for formation of spermatoproteasomes and histone degradation during meiotic DNA repair in spermatocytes. J Biol Chem 2021; 296:100130. [PMID: 33262216 PMCID: PMC7949063 DOI: 10.1074/jbc.ra120.016485] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/18/2020] [Accepted: 12/01/2020] [Indexed: 11/24/2022] Open
Abstract
Meiosis, which produces haploid progeny, is critical to ensuring both faithful genome transmission and genetic diversity. Proteasomes play critical roles at various stages of spermatogenesis, including meiosis, but the underlying mechanisms remain unclear. The atypical proteasomes, which contain the activator PA200, catalyze the acetylation-dependent degradation of the core histones in elongated spermatids and DNA repair in somatic cells. We show here that the testis-specific proteasome subunit α4s/PSMA8 is essential for male fertility by promoting proper formation of spermatoproteasomes, which harbor both PA200 and constitutive catalytic subunits. Immunostaining of a spermatocyte marker, SYCP3, indicated that meiosis was halted at the stage of spermatocytes in the α4s-deficient testes. α4s stimulated the in vitro degradation of the acetylated core histones, instead of nonacetylated histones, by the PA200-proteasome. Deletion of α4s blocked degradation of the core histones at DNA damage loci in spermatocytes, leading to meiotic arrest at metaphase I. Thus, α4s is required for histone degradation at meiotic DNA damage loci, proper progression of meiosis, and fertility in males by promoting proper formation of spermatoproteasomes. These results are important for understanding male infertility and might provide potential targets for male contraception or treatment of male infertility.
Collapse
Affiliation(s)
- Zi-Hui Zhang
- Key Laboratory of Cell Proliferation & Regulation Biology, Ministry of Education and College of Life Sciences, Beijing Normal University, Beijing, China
| | - Tian-Xia Jiang
- Key Laboratory of Cell Proliferation & Regulation Biology, Ministry of Education and College of Life Sciences, Beijing Normal University, Beijing, China.
| | - Lian-Bin Chen
- Key Laboratory of Cell Proliferation & Regulation Biology, Ministry of Education and College of Life Sciences, Beijing Normal University, Beijing, China
| | - Wenhui Zhou
- Medical Center for Human Reproduction, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yixun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiao-Bo Qiu
- Key Laboratory of Cell Proliferation & Regulation Biology, Ministry of Education and College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
15
|
Wen Z, Zhu H, Zhang A, Lin J, Zhang G, Liu D, Xiao Y, Ye C, Sun D, Wu B, Zhang J, Gao J. Cdc14a has a role in spermatogenesis, sperm maturation and male fertility. Exp Cell Res 2020; 395:112178. [PMID: 32679235 DOI: 10.1016/j.yexcr.2020.112178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 11/29/2022]
Abstract
Cdc14a is an evolutionarily conserved dual-specific protein phosphatase, and it plays different roles in different organisms. Cdc14a mutations in human have been reported to cause male infertility, while the specific role of Cdc14a in regulation of the male reproductive system remains elusive. In the present study, we established a knockout mouse model to study the function of Cdc14a in male reproductive system. Cdc14a-/- male mice were subfertile and they could only produce very few offspring. The number of sperm was decreased, the sperm motility was impaired, and the proportion of sperm with abnormal morphology was elevated in Cdc14a-/- mice. When we mated Cdc14a-/- male mice with wild-type (WT) female mice, fertilized eggs could be found in female fallopian tubes, however, the majority of these embryos died during development. Some empty spaces were observed in seminiferous tubule of Cdc14a-/- testes. Compared with WT male mice, the proportions of pachytene spermatocytes were increased and germ cells stained with γH2ax were decreased in Cdc14a-/- male mice, indicating that knockout of Cdc14a inhibited meiotic initiation. Subsequently, we analyzed the expression levels of some substrate proteins of Cdc14a, including Cdc25a, Wee1, and PR-Set7, and compared those with WT testes, in which the expression levels of these proteins were significantly increased in Cdc14a-/- testes. Our results revealed that Cdc14a-/- male mice are highly subfertile, and Cdc14a is essential for normal spermatogenesis and sperm function.
Collapse
Affiliation(s)
- Zongzhuang Wen
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Haixia Zhu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Aizhen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Jing Lin
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Guangkai Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Dongyue Liu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Yu Xiao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Chao Ye
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China
| | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, 300041, PR China.
| | - Bin Wu
- Department of Reproductive Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250100, PR China.
| | - Jian Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China.
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, 250100, PR China.
| |
Collapse
|
16
|
Tian Q, Guo SM, Xie SM, Yin Y, Zhou LQ. Rybp orchestrates spermatogenesis via regulating meiosis and sperm motility in mice. Cell Cycle 2020; 19:1492-1501. [PMID: 32324084 DOI: 10.1080/15384101.2020.1754585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Ring1 and Yin Yang 1-Binding Protein (RYBP) is a member of non-canonical polycomb repressive complex 1 to mediate monoubiquitination of histone H2A at lysine 119. It plays an important role in development, but its role in reproduction remains illusive. In this study, we used Rybp conditional knockout mouse model to genetically ablate Rybp in male germ cells. We found that Rybp deficiency during spermatogenesis led to smaller testes, loss of germline cells, disturbed meiosis, increased apoptosis of spermatocytes, decreased sperm motility, and reduced global H3K9me3, without impacting retrotransposon expression. Meanwhile, we depleted Rybp during oogenesis, but oocyte maturation and preimplantation development were normal. Our findings demonstrate that RYBP plays important roles in spermatogenesis through regulating meiosis and sperm motility.
Collapse
Affiliation(s)
- Qing Tian
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei, China
| | - Shi-Meng Guo
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei, China
| | - Shi-Ming Xie
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei, China
| | - Ying Yin
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei, China
| | - Li-Quan Zhou
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology , Wuhan, Hubei, China
| |
Collapse
|
17
|
Stenvall A, Larsson E, Holmqvist B, Strand SE, Jönsson BA. Quantitative γ-H2AX immunofluorescence method for DNA double-strand break analysis in testis and liver after intravenous administration of 111InCl 3. EJNMMI Res 2020; 10:22. [PMID: 32189079 PMCID: PMC7080928 DOI: 10.1186/s13550-020-0604-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/31/2020] [Indexed: 12/25/2022] Open
Abstract
Background It is well known that a severe cell injury after exposure to ionizing radiation is the induction of DNA double-strand breaks (DSBs). After exposure, an early response to DSBs is the phosphorylation of the histone H2AX molecule regions adjacent to the DSBs, referred to as γ-H2AX foci. The γ-H2AX assay after external exposure is a good tool for investigating the link between the absorbed dose and biological effect. However, less is known about DNA DSBs and γ-H2AX foci within the tissue microarchitecture after internal irradiation from radiopharmaceuticals. Therefore, in this study, we aimed to develop and validate a quantitative ex vivo model using γ-H2AX immunofluorescence staining and confocal laser scanning microscopy (CLSM) to investigate its applicability in nuclear medicine dosimetry research. Liver and testis were selected as the organs to study after intravenous administration of 111InCl3. Results In this study, we developed and validated a method that combines ex vivo γ-H2AX foci labeling of tissue sections with in vivo systemically irradiated mouse testis and liver tissues. The method includes CLSM imaging for intracellular cell-specific γ-H2AX foci detection and quantification and absorbed dose calculations. After exposure to ionizing radiation from 111InCl3, both hepatocytes and non-hepatocytes within the liver showed an absorbed dose-dependent elevation of γ-H2AX foci, whereas no such correlation was seen for the testis tissue. Conclusion It is possible to detect and quantify the radiation-induced γ-H2AX foci within the tissues of organs at risk after internal irradiation. We conclude that our method developed is an appropriate tool to study dose–response relationships in animal organs and human tissue biopsies after internal exposure to radiation.
Collapse
Affiliation(s)
- Anna Stenvall
- Department of Medical Radiation Physics, Clinical Sciences, Lund University, Lund, Sweden.
| | - Erik Larsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | | - Sven-Erik Strand
- Department of Medical Radiation Physics, Clinical Sciences, Lund University, Lund, Sweden
| | - Bo-Anders Jönsson
- Department of Medical Radiation Physics, Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
18
|
Effect of recombinant human vascular endothelial growth factor on testis tissue xenotransplants from prepubertal boys: a three-case study. Reprod Biomed Online 2019; 39:119-133. [DOI: 10.1016/j.rbmo.2019.02.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 11/23/2022]
|
19
|
Men Y, Zhao Y, Zhang P, Zhang H, Gao Y, Liu J, Feng Y, Li L, Shen W, Sun Z, Min L. Gestational exposure to low-dose zearalenone disrupting offspring spermatogenesis might be through epigenetic modifications. Basic Clin Pharmacol Toxicol 2019; 125:382-393. [PMID: 31058416 DOI: 10.1111/bcpt.13243] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022]
Abstract
Zearalenone (ZEA), a F-2 mycotoxin produced by Fusarium, has been found to be an endocrine disruptor through oestrogen receptor signalling pathway to impair spermatogenesis. The disruption on reproductive systems by ZEA exposure might be transgenerational. In our previous report, we have found that low dose (lower than no-observed effect level, NOEL) of ZEA impaired mouse spermatogenesis and decreased mouse semen quality. The purpose of the current investigation was to explore the impacts of low-dose ZEA on spermatogenesis in the offspring after prenatal exposure and the underlying mechanisms. And it demonstrated that prenatal low-dose ZEA exposure disrupted the meiosis process to inhibit the spermatogenesis in offspring and even to diminish the semen quality by the decrease in spermatozoa motility and concentration. The DNA methylation marker 5hmC was decreased, the histone methylation markers H3K9 and H3K27 were elevated, and oestrogen receptor alpha was reduced in the offspring testis after prenatal low-dose ZEA exposure. The data suggest that the disruption in spermatogenesis by prenatal low-dose ZEA exposure may be through the modifications on epigenetic pathways (DNA methylation and histone methylation) and the interactions with oestrogen receptor signalling pathway. Moreover, in the current study, the male offspring were indirectly exposed to low-dose ZEA through placenta and the spermatogenesis in offspring was disrupted which suggested that the toxicity of ZEA on reproductive systems was very severe. Therefore, we strongly recommend that greater attention should be paid to this mycotoxin to minimize its adverse impact on human spermatogenesis.
Collapse
Affiliation(s)
- Yuhao Men
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| | - Yong Zhao
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Pengfei Zhang
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yishan Gao
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| | - Jing Liu
- University Research Core, Qingdao Agricultural University, Qingdao, China
| | - Yanni Feng
- College of Veterinary Sciences, Qingdao Agricultural University, Qingdao, China
| | - Lan Li
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Zhongyi Sun
- Center for Reproductive Medicine, Shenzhen Hospital, Peking University, Shenzhen, China
| | - Lingjiang Min
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
20
|
Morgan M, Kabayama Y, Much C, Ivanova I, Di Giacomo M, Auchynnikava T, Monahan JM, Vitsios DM, Vasiliauskaitė L, Comazzetto S, Rappsilber J, Allshire RC, Porse BT, Enright AJ, O’Carroll D. A programmed wave of uridylation-primed mRNA degradation is essential for meiotic progression and mammalian spermatogenesis. Cell Res 2019; 29:221-232. [PMID: 30617251 PMCID: PMC6420129 DOI: 10.1038/s41422-018-0128-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/20/2018] [Indexed: 02/08/2023] Open
Abstract
Several developmental stages of spermatogenesis are transcriptionally quiescent which presents major challenges associated with the regulation of gene expression. Here we identify that the zygotene to pachytene transition is not only associated with the resumption of transcription but also a wave of programmed mRNA degradation that is essential for meiotic progression. We explored whether terminal uridydyl transferase 4- (TUT4-) or TUT7-mediated 3' mRNA uridylation contributes to this wave of mRNA degradation during pachynema. Indeed, both TUT4 and TUT7 are expressed throughout most of spermatogenesis, however, loss of either TUT4 or TUT7 does not have any major impact upon spermatogenesis. Combined TUT4 and TUT7 (TUT4/7) deficiency results in embryonic growth defects, while conditional gene targeting revealed an essential role for TUT4/7 in pachytene progression. Loss of TUT4/7 results in the reduction of miRNA, piRNA and mRNA 3' uridylation. Although this reduction does not greatly alter miRNA or piRNA expression, TUT4/7-mediated uridylation is required for the clearance of many zygotene-expressed transcripts in pachytene cells. We find that TUT4/7-regulated transcripts in pachytene spermatocytes are characterized by having long 3' UTRs with length-adjusted enrichment for AU-rich elements. We also observed these features in TUT4/7-regulated maternal transcripts whose dosage was recently shown to be essential for sculpting a functional maternal transcriptome and meiosis. Therefore, mRNA 3' uridylation is a critical determinant of both male and female germline transcriptomes. In conclusion, we have identified a novel requirement for 3' uridylation-programmed zygotene mRNA clearance in pachytene spermatocytes that is essential for male meiotic progression.
Collapse
Affiliation(s)
- Marcos Morgan
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 0627 3632grid.418924.2European Molecular Biology Laboratory (EMBL), Via Ramarini 32, 00015 Monterotondo, Italy ,0000 0004 1936 7988grid.4305.2Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF UK
| | - Yuka Kabayama
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 1936 7988grid.4305.2Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF UK
| | - Christian Much
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 0627 3632grid.418924.2European Molecular Biology Laboratory (EMBL), Via Ramarini 32, 00015 Monterotondo, Italy
| | - Ivayla Ivanova
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK
| | - Monica Di Giacomo
- 0000 0004 0627 3632grid.418924.2European Molecular Biology Laboratory (EMBL), Via Ramarini 32, 00015 Monterotondo, Italy
| | - Tatsiana Auchynnikava
- 0000 0004 1936 7988grid.4305.2Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF UK
| | - Jack Michael Monahan
- 0000 0000 9709 7726grid.225360.0European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD UK
| | | | - Lina Vasiliauskaitė
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 0627 3632grid.418924.2European Molecular Biology Laboratory (EMBL), Via Ramarini 32, 00015 Monterotondo, Italy
| | - Stefano Comazzetto
- 0000 0004 0627 3632grid.418924.2European Molecular Biology Laboratory (EMBL), Via Ramarini 32, 00015 Monterotondo, Italy
| | - Juri Rappsilber
- 0000 0004 1936 7988grid.4305.2Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF UK ,0000 0001 2292 8254grid.6734.6Institute of Biotechnology, Technische Universität Berlin, Berlin, 13355 Germany
| | - Robin Campbell Allshire
- 0000 0004 1936 7988grid.4305.2Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF UK
| | - Bo Torben Porse
- 0000 0001 0674 042Xgrid.5254.6Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, 2200 Denmark ,0000 0001 0674 042Xgrid.5254.6The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, 2200 Denmark ,0000 0001 0674 042Xgrid.5254.6Danish Stem Cell Centre (DanStem), Faculty of Health Sciences, University of Copenhagen, Copenhagen, 2200 Denmark
| | - Anton James Enright
- 0000 0000 9709 7726grid.225360.0European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD UK
| | - Dónal O’Carroll
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 1936 7988grid.4305.2Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF UK
| |
Collapse
|
21
|
Zheng Y, Lei Q, Jongejan A, Mulder CL, van Daalen SKM, Mastenbroek S, Hwang G, Jordan PW, Repping S, Hamer G. The influence of retinoic acid-induced differentiation on the radiation response of male germline stem cells. DNA Repair (Amst) 2018; 70:55-66. [PMID: 30179733 PMCID: PMC6237089 DOI: 10.1016/j.dnarep.2018.08.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/23/2018] [Accepted: 08/27/2018] [Indexed: 12/19/2022]
Abstract
Lifelong mammalian male fertility is maintained through an intricate balance between spermatogonial proliferation and differentiation. DNA damage in spermatogonia, for instance caused by chemo- or radiotherapy, can induce cell cycle arrest or germ cell apoptosis, possibly resulting in male infertility. Spermatogonia are generally more radiosensitive and prone to undergo apoptosis than somatic cells. Among spermatogonial subtypes the response to DNA damage is differentially modulated; undifferentiated spermatogonia, including the spermatogonial stem cells (SSCs), are relatively radio-resistant, whereas differentiating spermatogonia are very radiosensitive. To investigate the molecular mechanisms underlying this difference, we used an in vitro system consisting of mouse male germline stem (GS) cells that can be induced to differentiate. Using RNA-sequencing analysis, we analyzed the response of undifferentiated and differentiating GS cells to ionizing radiation (IR). At the RNA expression level, both undifferentiated and differentiating GS cells showed a very similar response to IR. Protein localization of several genes found to be involved in either spermatogonial differentiation or radiation response was investigated using mouse testis sections. For instance, we found that the transcription factor PDX1 was specifically expressed in undifferentiated spermatogonia and thus may be a novel marker for these cells. Interestingly, also at the protein level, undifferentiated GS cells showed a more pronounced upregulation of p53 in response to IR than differentiating GS cells. The higher p53 protein level in undifferentiated spermatogonia may preferentially induce cell cycle arrest, thereby giving these cells more time to repair inflicted DNA damage and increase their radio-resistance.
Collapse
Affiliation(s)
- Yi Zheng
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, China; Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Qijing Lei
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health Research Institute, Academic Medical Center Amsterdam, The Netherlands
| | - Callista L Mulder
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Saskia K M van Daalen
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Sebastiaan Mastenbroek
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Grace Hwang
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Philip W Jordan
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sjoerd Repping
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Geert Hamer
- Center for Reproductive Medicine, Amsterdam Research Institute Reproduction and Development, Academic Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Champroux A, Cocquet J, Henry-Berger J, Drevet JR, Kocer A. A Decade of Exploring the Mammalian Sperm Epigenome: Paternal Epigenetic and Transgenerational Inheritance. Front Cell Dev Biol 2018; 6:50. [PMID: 29868581 PMCID: PMC5962689 DOI: 10.3389/fcell.2018.00050] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
Abstract
The past decade has seen a tremendous increase in interest and progress in the field of sperm epigenetics. Studies have shown that chromatin regulation during male germline development is multiple and complex, and that the spermatozoon possesses a unique epigenome. Its DNA methylation profile, DNA-associated proteins, nucleo-protamine distribution pattern and non-coding RNA set up a unique epigenetic landscape which is delivered, along with its haploid genome, to the oocyte upon fertilization, and therefore can contribute to embryogenesis and to the offspring health. An emerging body of compelling data demonstrates that environmental exposures and paternal lifestyle can change the sperm epigenome and, consequently, may affect both the embryonic developmental program and the health of future generations. This short review will attempt to provide an overview of what is currently known about sperm epigenome and the existence of transgenerational epigenetic inheritance of paternally acquired traits that may contribute to the offspring phenotype.
Collapse
Affiliation(s)
- Alexandre Champroux
- GReD, Laboratoire “Génétique, Reproduction and Développement,” UMR Centre National de la Recherche Scientifique 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Julie Cocquet
- INSERM U1016, Institut Cochin, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Joëlle Henry-Berger
- GReD, Laboratoire “Génétique, Reproduction and Développement,” UMR Centre National de la Recherche Scientifique 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Joël R. Drevet
- GReD, Laboratoire “Génétique, Reproduction and Développement,” UMR Centre National de la Recherche Scientifique 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Ayhan Kocer
- GReD, Laboratoire “Génétique, Reproduction and Développement,” UMR Centre National de la Recherche Scientifique 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
23
|
Smart E, Lopes F, Rice S, Nagy B, Anderson RA, Mitchell RT, Spears N. Chemotherapy drugs cyclophosphamide, cisplatin and doxorubicin induce germ cell loss in an in vitro model of the prepubertal testis. Sci Rep 2018; 8:1773. [PMID: 29379115 PMCID: PMC5788858 DOI: 10.1038/s41598-018-19761-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/22/2017] [Indexed: 11/09/2022] Open
Abstract
Long term survival rates for childhood cancers is steadily increasing, however cancer survivors can experience fertility problems as a consequence of chemotherapy treatment. This is particularly problematic for young boys, for whom no fertility preservation treatment is yet established. Here, we have determined the effects on prepubertal mouse testis of three commonly used chemotherapy drugs; cyclophosphamide (using its active metabolite phosphoramide mustard), cisplatin and doxorubicin, exposing testicular fragments to a clinically relevant range of concentrations in vitro. All three drugs induced a specific and highly significant loss of germ cells, including spermatogonial stem cells. In contrast, there was no significant effect on somatic cells, for either Sertoli or interstitial cells. Time course analysis of cleaved Caspase-3 expression showed a significant increase in apoptosis eight hours prior to a detectable decrease in germ cell numbers following exposure to phosphoramide mustard or cisplatin, although this pattern was not seen following doxorubicin-exposure. Moreover, analysis of DNA damage at 16 h showed increased γH2AX expression in response to all three drugs. Overall, results show that cisplatin, doxorubicin and cyclophosphamide all specifically induce loss of germ cells, including of spermatogonial stem cells, in the prepubertal mouse testis at concentrations relevant to human therapeutic exposures.
Collapse
Affiliation(s)
- Ellie Smart
- Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom.,Center for Dermatology Research, University of Manchester, Manchester, United Kingdom
| | - Federica Lopes
- Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom
| | - Siobhan Rice
- Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom.,Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS,, United Kingdom
| | - Boglarka Nagy
- Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Norah Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom.
| |
Collapse
|
24
|
Agarwal AK, Tunison K, Dalal JS, Nagamma SS, Hamra FK, Sankella S, Shao X, Auchus RJ, Garg A. Metabolic, Reproductive, and Neurologic Abnormalities in Agpat1-Null Mice. Endocrinology 2017; 158:3954-3973. [PMID: 28973305 PMCID: PMC5695831 DOI: 10.1210/en.2017-00511] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/20/2017] [Indexed: 12/11/2022]
Abstract
Defects in the biosynthesis of phospholipids and neutral lipids are associated with cell membrane dysfunction, disrupted energy metabolism, and diseases including lipodystrophy. In these pathways, the 1-acylglycerol-3-phosphate O-acyltransferase (AGPAT) enzymes transfer a fatty acid to the sn-2 carbon of sn-1-acylglycerol-3-phosphate (lysophosphatidic acid) to form sn-1, 2-acylglycerol-3-phosphate [phosphatidic acid (PA)]. PA is a precursor for key phospholipids and diacylglycerol. AGPAT1 and AGPAT2 are highly homologous isoenzymes that are both expressed in adipocytes. Genetic defects in AGPAT2 cause congenital generalized lipodystrophy, indicating that AGPAT1 cannot compensate for loss of AGPAT2 in adipocytes. To further explore the physiology of AGPAT1, we characterized a loss-of-function mouse model (Agpat1-/-). The majority of Agpat1-/- mice died before weaning and had low body weight and low plasma glucose levels, independent of plasma insulin and glucagon levels, with reduced percentage of body fat but not generalized lipodystrophy. These mice also had decreased hepatic messenger RNA expression of Igf-1 and Foxo1, suggesting a decrease in gluconeogenesis. In male mice, sperm development was impaired, with a late meiotic arrest near the onset of round spermatid production, and gonadotropins were elevated. Female mice showed oligoanovulation yet retained responsiveness to gonadotropins. Agpat1-/- mice also demonstrated abnormal hippocampal neuron development and developed audiogenic seizures. In summary, Agpat1-/- mice developed widespread disturbances of metabolism, sperm development, and neurologic function resulting from disrupted phospholipid homeostasis. AGPAT1 appears to serve important functions in the physiology of multiple organ systems. The Agpat1-deficient mouse provides an important model in which to study the contribution of phospholipid and triacylglycerol synthesis to physiology and diseases.
Collapse
Affiliation(s)
- Anil K. Agarwal
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Katie Tunison
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Jasbir S. Dalal
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Sneha S. Nagamma
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - F. Kent Hamra
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Shireesha Sankella
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Xinli Shao
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Richard J. Auchus
- Department of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Abhimanyu Garg
- Division of Nutrition and Metabolic Diseases, Center for Human Nutrition, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
25
|
Pang J, Zhou Q, Sun X, Li L, Zhou B, Zeng F, Zhao Y, Shen W, Sun Z. Effect of low-dose zearalenone exposure on reproductive capacity of male mice. Toxicol Appl Pharmacol 2017; 333:60-67. [DOI: 10.1016/j.taap.2017.08.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/14/2017] [Accepted: 08/18/2017] [Indexed: 11/26/2022]
|
26
|
Dai MS, Hall SJ, Vantangoli Policelli MM, Boekelheide K, Spade DJ. Spontaneous testicular atrophy occurs despite normal spermatogonial proliferation in a Tp53 knockout rat. Andrology 2017; 5:1141-1152. [PMID: 28834365 DOI: 10.1111/andr.12409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 06/12/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
The tumor suppressor protein p53 (TP53) has many functions in cell cycle regulation, apoptosis, and DNA damage repair and is also involved in spermatogenesis in the mouse. To evaluate the role of p53 in spermatogenesis in the rat, we characterized testis biology in adult males of a novel p53 knockout rat (SD-Tp53tm1sage ). p53 knockout rats exhibited variable levels of testicular atrophy, including significantly decreased testis weights, atrophic seminiferous tubules, decreased seminiferous tubule diameter, and elevated spermatocyte TUNEL labeling rates, indicating a dysfunction in spermatogenesis. Phosphorylated histone H2AX protein levels and distribution were similar in the non-atrophic seminiferous tubules of both genotypes, showing evidence of pre-synaptic DNA double-strand breaks in leptotene and zygotene spermatocytes, preceding cell death in p53 knockout rat testes. Quantification of the spermatogonial stem cell (SSC) proliferation rate with bromodeoxyuridine (BrdU) labeling, in addition to staining with the undifferentiated type A spermatogonial marker GDNF family receptor alpha-1 (GFRA1), indicated that the undifferentiated spermatogonial population was normal in p53 knockout rats. Following exposure to 0.5 or 5 Gy X-ray, p53 knockout rats exhibited no germ cell apoptotic response beyond their unirradiated phenotype, while germ cell death in wild-type rat testes was elevated to a level similar to the unexposed p53 knockout rats. This study indicates that seminiferous tubule atrophy occurs following spontaneous, elevated levels of spermatocyte death in the p53 knockout rat. This phenomenon is variable across individual rats. These results indicate a critical role for p53 in rat germ cell survival and spermatogenesis.
Collapse
Affiliation(s)
- Matthew S Dai
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | | | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
27
|
Zhou L, Canagarajah B, Zhao Y, Baibakov B, Tokuhiro K, Maric D, Dean J. BTBD18 Regulates a Subset of piRNA-Generating Loci through Transcription Elongation in Mice. Dev Cell 2017; 40:453-466.e5. [PMID: 28292424 DOI: 10.1016/j.devcel.2017.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 11/18/2016] [Accepted: 02/09/2017] [Indexed: 11/27/2022]
Abstract
PIWI-interacting RNAs (piRNAs) are small non-coding RNAs essential for animal germ cell development. Despite intense investigation of post-transcriptional processing, chromatin regulators for piRNA biogenesis in mammals remain largely unexplored. Here we document that BTBD18 is a pachytene nuclear protein in mouse testes that occupies a subset of pachytene piRNA-producing loci. Ablation of Btbd18 in mice disrupts piRNA biogenesis, prevents spermiogenesis, and results in male sterility. Transcriptome profiling, chromatin accessibility, and RNA polymerase II occupancy demonstrate that BTBD18 facilitates expression of pachytene piRNA precursors by promoting transcription elongation. Thus, our study identifies BTBD18 as a specific controller for transcription activation through RNA polymerase II elongation at a subset of genomic piRNA loci.
Collapse
Affiliation(s)
- Liquan Zhou
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Bertram Canagarajah
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yangu Zhao
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Boris Baibakov
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Keizo Tokuhiro
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dragan Maric
- NINDS Flow Cytometry Core Facility, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Liu KH, Sun XF, Feng YZ, Cheng SF, Li B, Li YP, Shen W, Li L. The impact of Zearalenone on the meiotic progression and primordial follicle assembly during early oogenesis. Toxicol Appl Pharmacol 2017; 329:9-17. [PMID: 28552778 DOI: 10.1016/j.taap.2017.05.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 04/28/2017] [Accepted: 05/22/2017] [Indexed: 01/24/2023]
Abstract
Zearalenone (ZEA) is a mycotoxin produced by fusarium graminearum. It can cause abnormal reproductive function by acting as an environmental estrogen. Research has traditionally focused on acute and chronic injury on mammalian reproductive capacity after ZEA treatment. Little research has been done studying the effects of ZEA exposure on early oogenesis. In this study, we investigate the effects of ZEA exposure on meiotic entry, DNA double-strand breaks (DSBs), and primordial follicle assembly during murine early oogenesis. The results show that ZEA exposure significantly decreased the percentage of diplotene stage germ cells, and made more germ cells remain at zygotene or pachytene stages. Moreover, the mRNA expression level of meiosis-related genes was significantly reduced after ZEA treatment. ZEA exposure significantly increased DNA-DSBs at the diplotene stage. Meanwhile, DNA damage repair genes such as RAD51 and BRCA1 were activated. Furthermore, maternal exposure to ZEA significantly decreased the number of primordial follicles in newborn mouse ovaries. In conclusion, ZEA exposure impairs mouse female germ cell meiotic progression, DNA-DSBs, and primordial follicle assembly.
Collapse
Affiliation(s)
- Ke-Han Liu
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiao-Feng Sun
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Yan-Zhong Feng
- Institute of Animal Sciences, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Shun-Feng Cheng
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Bo Li
- Chengguo Station of Animal Husbandry and Veterinary, Laizhou 261437, China
| | - Ya-Peng Li
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Wei Shen
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Lan Li
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China.
| |
Collapse
|
29
|
Knock-Out Serum Replacement and Melatonin Effects on Germ Cell Differentiation in Murine Testicular Explant Cultures. Ann Biomed Eng 2017; 45:1783-1794. [PMID: 28488216 PMCID: PMC5489632 DOI: 10.1007/s10439-017-1847-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 04/27/2017] [Indexed: 12/11/2022]
Abstract
Finding robust culture conditions for in vitro maturation (IVM) of male germ cells is still a challenge. Recently, a testis organ culture method, using Knockout Serum Replacement (KSR), was suggested as a promising approach. However, the efficiency of that model is still not optimal. Hence, we have tried to establish the culture conditions in two laboratories, and to improve the reliability of the culture system to generate mature germ cells. Male mice at three days of age were sacrificed. Testes were cut into small pieces which were cultured atop agarose stands, using Minimum Essential Medium alpha supplemented with different supplements; melatonin, Glutamax, and different concentrations of KSR. The results showed that the duration of culture beyond 18 days had an impact on the number of differentiated germ cells. Supplementation with melatonin and Glutamax revealed a positive influence on the efficiency of male germ cell differentiation in vitro. Furthermore, the results confirmed that KSR had a positive effect on germ cell maturation and testosterone production, with a concentration of at least 10%. In conclusion, this study emphasizes the beneficial role of at least 10% KSR in the IVM of germ cells.
Collapse
|
30
|
Jha KN, Tripurani SK, Johnson GR. TSSK6 is required for γH2AX formation and the histone-to-protamine transition during spermiogenesis. J Cell Sci 2017; 130:1835-1844. [PMID: 28389581 DOI: 10.1242/jcs.202721] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/04/2017] [Indexed: 12/25/2022] Open
Abstract
Spermiogenesis includes transcriptional silencing, chromatin condensation and extensive morphological changes as spermatids transform into sperm. Chromatin condensation involves histone hyperacetylation, transitory DNA breaks, histone H2AX (also known as H2AFX) phosphorylation at Ser139 (γH2AX), and replacement of histones by protamines. Previously, we have reported that the spermatid protein kinase TSSK6 is essential for fertility in mice, but its specific role in spermiogenesis is unknown. Here, we show that TSSK6 expression is spatiotemporally coincident with γH2AX formation in the nuclei of developing mouse spermatids. RNA-sequencing analysis demonstrates that genetic ablation of Tssk6 does not impact gene expression or silencing in spermatids. However, loss of TSSK6 blocks γH2AX formation, even though the timing and level of the transient DNA breaks is unaltered. Further, Tssk6-knockout sperm contained increased levels of histones H3 and H4, and protamine 2 precursor and intermediate(s) indicative of a defective histone-to-protamine transition. These results demonstrate that TSSK6 is required for γH2AX formation during spermiogenesis, and also link γH2AX to the histone-to-protamine transition and male fertility.
Collapse
Affiliation(s)
- Kula N Jha
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Swamy K Tripurani
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Gibbes R Johnson
- Division of Biotechnology Review and Research IV, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
31
|
Kotzur T, Benavides-Garcia R, Mecklenburg J, Sanchez JR, Reilly M, Hermann BP. Granulocyte colony-stimulating factor (G-CSF) promotes spermatogenic regeneration from surviving spermatogonia after high-dose alkylating chemotherapy. Reprod Biol Endocrinol 2017; 15:7. [PMID: 28077131 PMCID: PMC5225630 DOI: 10.1186/s12958-016-0226-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/28/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The lifesaving chemotherapy and radiation treatments that allow patients to survive cancer can also result in a lifetime of side-effects, including male infertility. Infertility in male cancer survivors is thought to primarily result from killing of the spermatogonial stem cells (SSCs) responsible for producing spermatozoa since SSCs turn over slowly and are thereby sensitive to antineoplastic therapies. We previously demonstrated that the cytokine granulocyte colony-stimulating factor (G-CSF) can preserve spermatogenesis after alkylating chemotherapy (busulfan). METHODS Male mice were treated with G-CSF or controls before and/or after sterilizing busulfan treatment and evaluated immediately or 10-19 weeks later for effects on spermatogenesis. RESULTS We demonstrated that the protective effect of G-CSF on spermatogenesis was stable for at least 19 weeks after chemotherapy, nearly twice as long as previously shown. Further, G-CSF treatment enhanced spermatogenic measures 10 weeks after treatment in the absence of a cytotoxic insult, suggesting G-CSF acts as a mitogen in steady-state spermatogenesis. In agreement with this conclusion, G-CSF treatment for 3 days before busulfan treatment exacerbated the loss of spermatogenesis observed with G-CSF alone. Reciprocally, spermatogenic recovery was modestly enhanced in mice treated with G-CSF for 4 days after busulfan. These results suggested that G-CSF promoted spermatogonial proliferation, leading to enhanced spermatogenic regeneration from surviving SSCs. Similarly, there was a significant increase in proportion of PLZF+ undifferentiated spermatogonia that were Ki67+ (proliferating) 1 day after G-CSF treatment. CONCLUSIONS Together, these results clarify that G-CSF protects spermatogenesis after alkylating chemotherapy by stimulating proliferation of surviving spermatogonia, and indicate it may be useful as a retrospective fertility-restoring treatment.
Collapse
Affiliation(s)
- Travis Kotzur
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249 USA
| | - Roberto Benavides-Garcia
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249 USA
| | - Jennifer Mecklenburg
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249 USA
| | - Jamila R. Sanchez
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249 USA
| | - Matthew Reilly
- Departments of Biomedical Engineering and Ophthalmology, The Ohio State University, 1080 Carmack Road, Columbus, OH 43210 USA
| | - Brian P. Hermann
- Department of Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249 USA
| |
Collapse
|
32
|
Fujitani K, Otomo A, Wada M, Takamatsu N, Ito M. Sexually dimorphic expression of Dmrt1 and γH2AX in germ stem cells during gonadal development in Xenopus laevis. FEBS Open Bio 2016; 6:276-84. [PMID: 27239441 PMCID: PMC4821358 DOI: 10.1002/2211-5463.12035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/30/2015] [Accepted: 01/14/2016] [Indexed: 12/20/2022] Open
Abstract
In many animals, primordial germ cells (PGCs) migrate into developing gonads. There, they proliferate and differentiate into female and male germ stem cells (GSCs), oogonia and spermatogonia, respectively. Few studies have focused on the molecular mechanisms underlying the development of GSC sex determination. Here, we investigated the expression of the transcription factor Dmrt1 and a phosphorylated form of the histone variant H2AX (γH2AX) during gonadal development in Xenopus laevis. During early sexual differentiation, Dmrt1 was expressed in the GSCs of the ZW (female) and ZZ (male) gonads as well as somatic cells of the ZZ gonads. Notably, the PGCs and primary GSCs contained large, unstructured nuclei, whereas condensed, rounder nuclei appeared only in primary oogonia during tadpole development. After metamorphosis, Dmrt1 showed its expression in secondary spermatogonia, but not in secondary oogonia. Like Dmrt1, γH2AX was expressed in the nuclei of primary GSCs in early developing gonads. However, after metamorphosis, γH2AX expression continued in primary and secondary spermatogonia, but was barely detected in the condensed nuclei of primary oogonia. Taken together, these observations indicate that spermatogonia tend to retain PGC characteristics, compared to oogonia, which undergo substantial changes during gonadal differentiation in X. laevis. Our findings suggest that Dmrt1 and γH2AX may contribute to the maintenance of stem cell identity by controlling gene expression and epigenetic changes, respectively.
Collapse
Affiliation(s)
- Kazuko Fujitani
- Department of Bioscience School of Science Kitasato University Sagamihara Japan
| | - Asako Otomo
- Department of Molecular Life Sciences Tokai University School of Medicine Isehara Japan
| | - Mikako Wada
- Department of Bioscience School of Science Kitasato University Sagamihara Japan
| | - Nobuhiko Takamatsu
- Department of Bioscience School of Science Kitasato University Sagamihara Japan
| | - Michihiko Ito
- Department of Bioscience School of Science Kitasato University Sagamihara Japan
| |
Collapse
|
33
|
Clement TM, Inselman AL, Goulding EH, Willis WD, Eddy EM. Disrupting Cyclin Dependent Kinase 1 in Spermatocytes Causes Late Meiotic Arrest and Infertility in Mice. Biol Reprod 2015; 93:137. [PMID: 26490841 DOI: 10.1095/biolreprod.115.134940] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/15/2015] [Indexed: 01/22/2023] Open
Abstract
While cyclin dependent kinase 1 (CDK1) has a critical role in controlling resumption of meiosis in oocytes, its role has not been investigated directly in spermatocytes. Unique aspects of male meiosis led us to hypothesize that its role is different in male meiosis than in female meiosis. We generated a conditional knockout (cKO) of the Cdk1 gene in mouse spermatocytes to test this hypothesis. We found that CDK1-null spermatocytes undergo synapsis, chiasmata formation, and desynapsis as is seen in oocytes. Additionally, CDK1-null spermatocytes relocalize SYCP3 to centromeric foci, express H3pSer10, and initiate chromosome condensation. However, CDK1-null spermatocytes fail to form condensed bivalent chromosomes in prophase of meiosis I and instead are arrested at prometaphase. Thus, CDK1 has an essential role in male meiosis that is consistent with what is known about the role of CDK1 in female meiosis, where it is required for formation of condensed bivalent metaphase chromosomes and progression to the first meiotic division. We found that cKO spermatocytes formed fully condensed bivalent chromosomes in the presence of okadaic acid, suggesting that cKO chromosomes are competent to condense, although they do not do so in vivo. Additionally, arrested cKO spermatocytes exhibited irregular cell shape, irregular large nuclei, and large distinctive nucleoli. These cells persist in the seminiferous epithelium through the next seminiferous epithelial cycle with a lack of stage XII checkpoint-associated cell death. This indicates that CDK1 is required upstream of a checkpoint-associated cell death as well as meiotic metaphase progression in mouse spermatocytes.
Collapse
Affiliation(s)
- Tracy M Clement
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Amy L Inselman
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Eugenia H Goulding
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - William D Willis
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Edward M Eddy
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
34
|
Siklenka K, Erkek S, Godmann M, Lambrot R, McGraw S, Lafleur C, Cohen T, Xia J, Suderman M, Hallett M, Trasler J, Peters AHFM, Kimmins S. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science 2015; 350:aab2006. [PMID: 26449473 DOI: 10.1126/science.aab2006] [Citation(s) in RCA: 333] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 09/18/2015] [Indexed: 12/28/2022]
Abstract
A father's lifetime experiences can be transmitted to his offspring to affect health and development. However, the mechanisms underlying paternal epigenetic transmission are unclear. Unlike in somatic cells, there are few nucleosomes in sperm, and their function in epigenetic inheritance is unknown. We generated transgenic mice in which overexpression of the histone H3 lysine 4 (H3K4) demethylase KDM1A (also known as LSD1) during spermatogenesis reduced H3K4 dimethylation in sperm. KDM1A overexpression in one generation severely impaired development and survivability of offspring. These defects persisted transgenerationally in the absence of KDM1A germline expression and were associated with altered RNA profiles in sperm and offspring. We show that epigenetic inheritance of aberrant development can be initiated by histone demethylase activity in developing sperm, without changes to DNA methylation at CpG-rich regions.
Collapse
Affiliation(s)
- Keith Siklenka
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Serap Erkek
- Friedrich Miescher Institute for Biomedical Research (FMI), CH-4058 Basel, Switzerland. Faculty of Sciences, University of Basel, Basel, Switzerland
| | - Maren Godmann
- Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Romain Lambrot
- Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Serge McGraw
- Department of Pediatrics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Christine Lafleur
- Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Tamara Cohen
- Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Jianguo Xia
- Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada. Institute of Parasitology, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada
| | - Matthew Suderman
- MRC Integrative Epidemiology Unity, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Michael Hallett
- McGill Centre for Bioinformatics, School of Computer Science, Faculty of Science, McGill University, Montreal, Quebec, Canada
| | - Jacquetta Trasler
- Department of Pediatrics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada. Department of Human Genetics and Department of Pharmacology and Therapeutics, Research Institute of the McGill University Health Centre at the Montreal Children's Hospital, Montreal, Quebec, Canada
| | - Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research (FMI), CH-4058 Basel, Switzerland. Faculty of Sciences, University of Basel, Basel, Switzerland.
| | - Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada. Department of Animal Science, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
35
|
Pubertal exposure to di-(2-ethylhexyl)-phthalate inhibits G9a-mediated histone methylation during spermatogenesis in mice. Arch Toxicol 2015; 90:955-69. [DOI: 10.1007/s00204-015-1529-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 05/05/2015] [Indexed: 01/30/2023]
|
36
|
Zhao K, Chen Y, Yang R, Bai Y, Li C, Li H, Xiong C. miR-424/322 is downregulated in the semen of patients with severe DNA damage and may regulate sperm DNA damage. Reprod Fertil Dev 2015; 28:RD15052. [PMID: 25872494 DOI: 10.1071/rd15052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/06/2015] [Indexed: 01/09/2023] Open
Abstract
Sperm DNA integrity is an essential factor for accurate transmission of genetic information. Human sperm DNA damage is a common cause of male infertility but the exact mechanism remains poorly understood. Considering the vital role of microRNA (miRNA) in multiple pathophysiological processes, we hypothesised that testicular miRNA is involved in sperm DNA damage during spermatogenesis. Infertile patients with high sperm DNA fragment index (DFI; n=94) were selected from 1090 infertile men and a total of 18 testis-specific seminal miRNAs previously identified from human seminal plasma were chosen and tested. miR-29c and miR-424 were downregulated in men with high DFI. The inhibition of these two miRNAs in mice confirmed the role of miR-424 (murine homologue miR-322) in sperm DNA damage during spermatogenesis; by contrast, miR-29c exhibited a negative result. Thus, miR-424/322 is involved in sperm DNA damage. Furthermore, the dysregulation of this miRNA can induce DNA double-strand breaks during spermatogenesis.
Collapse
|
37
|
Bao J, Tang C, Li J, Zhang Y, Bhetwal BP, Zheng H, Yan W. RAN-binding protein 9 is involved in alternative splicing and is critical for male germ cell development and male fertility. PLoS Genet 2014; 10:e1004825. [PMID: 25474150 PMCID: PMC4256260 DOI: 10.1371/journal.pgen.1004825] [Citation(s) in RCA: 40] [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: 06/19/2014] [Accepted: 10/14/2014] [Indexed: 01/09/2023] Open
Abstract
As a member of the large Ran-binding protein family, Ran-binding protein 9 (RANBP9) has been suggested to play a critical role in diverse cellular functions in somatic cell lineages in vitro, and this is further supported by the neonatal lethality phenotype in Ranbp9 global knockout mice. However, the exact molecular actions of RANBP9 remain largely unknown. By inactivation of Ranbp9 specifically in testicular somatic and spermatogenic cells, we discovered that Ranbp9 was dispensable for Sertoli cell development and functions, but critical for male germ cell development and male fertility. RIP-Seq and proteomic analyses revealed that RANBP9 was associated with multiple key splicing factors and directly targeted >2,300 mRNAs in spermatocytes and round spermatids. Many of the RANBP9 target and non-target mRNAs either displayed aberrant splicing patterns or were dysregulated in the absence of Ranbp9. Our data uncovered a novel role of Ranbp9 in regulating alternative splicing in spermatogenic cells, which is critical for normal spermatogenesis and male fertility. Male fertility depends on successful production of functional sperm. Sperm are produced through spermatogenesis, a process of male germ cell proliferation and differentiation in the testis. Most of the genes involved in spermatogenesis are transcribed and processed into multiple isoforms, which are mainly achieved through alternative splicing. The testis-specific transcriptome, characterized by male germ cell-specific alternative splicing patterns, has been shown to be essential for successful spermatogenesis. However, how these male germ cells-specific alternative splicing events are regulated remains largely unknown. Here, we report that RANBP9 is involved in alternative splicing events that are critical for male germ cell development, and dysfunction of RANBP9 leads to disrupted spermatogenesis and compromised male fertility.
Collapse
Affiliation(s)
- Jianqiang Bao
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada, United States of America
| | - Chong Tang
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada, United States of America
| | - Jiachen Li
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada, United States of America
| | - Ying Zhang
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada, United States of America
| | - Bhupal P. Bhetwal
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada, United States of America
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada, United States of America
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada, United States of America
- * E-mail:
| |
Collapse
|
38
|
Singh P, Raman MJ. Dynamics of radiation induced γH2AX foci in chromatin subcompartments of mouse pachytene spermatocytes and round spermatids. Mol Reprod Dev 2014; 81:484-96. [DOI: 10.1002/mrd.22314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 02/18/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Priti Singh
- Cytogenetics Laboratory; Department of Zoology; Centre of Advanced Study; Banaras Hindu University; Varanasi India
| | - Mercy J. Raman
- Cytogenetics Laboratory; Department of Zoology; Centre of Advanced Study; Banaras Hindu University; Varanasi India
| |
Collapse
|
39
|
Abstract
The phosphorylation of histone H2AX at serine 139 (γ-H2AX) is one of the first steps of DNA damage response and its detection is widely used as a sensitive marker for DNA double-strand breaks induced by ionizing radiation or other genotoxic agents. Immuno-stained phosphorylated histone can be measured in single cells by flow cytometry or single γ-H2AX foci can be visualized and counted microscopically in histological or cytological preparations. Animal studies are well recognized as important tools to study mechanisms of in vivo response to genotoxic stress. Tissues are composed by many cell types differing for function, differentiation, and proliferative capacity. In particular, due to the complexity of spermatogenesis and the heterogeneity of testicular cell subpopulations, an accurate characterization of damage in this tissue is difficult and requires an approach which allows the identification of damage in the different cellular compartments. This chapter presents techniques for γ-H2AX detection in mouse bone marrow and testicular cells. Furthermore, advantages and weaknesses of flow cytometric and microscopic methods are described.
Collapse
|
40
|
Conditional inactivation of Miwi2 reveals that MIWI2 is only essential for prospermatogonial development in mice. Cell Death Differ 2014; 21:783-96. [PMID: 24464225 DOI: 10.1038/cdd.2014.5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/21/2013] [Accepted: 12/30/2013] [Indexed: 11/08/2022] Open
Abstract
The PIWI-piRNA pathway serves as a critical defense mechanism through which the genome of the male germline is protected from invasion by transposable elements (TEs). MIWI2/PIWIL4, a member of the murine PIWI subclade of the Argonaute family, has been shown to be expressed in primordial germ cells (PGCs) and prospermatogonia in fetal and prepubertal testes. Global inactivation of Miwi2 leads to male sterility due to an early meiotic arrest, which correlates with retrotransposon desuppression. However, it remains unclear whether MIWI2 functions beyond the PGC stage and whether MIWI2 has a role beyond TE suppression during male germ line development. Through conditional inactivation of Miwi2, we demonstrate herein that MIWI2 function is restricted to a narrow time window during male PGC reprograming and that Miwi2 is dispensable for postnatal male germline development and testicular function in mice. Moreover, persistent activation of LINE1 and IAP retrotransposons caused by Miwi2 inactivation is compatible with mitotic cell cycle progression of spermatogonia during the first wave of spermatogenesis, but can cause zygotene to pachytene arrest in early meiosis due to multiple defects including enhanced DNA double-strand breaks, aberrant histone modifications and altered mRNA transcriptome. Our data not only validate those from global Miwi2 KO studies, but also suggest that MIWI2 and MIWI2-associated piRNAs have functions beyond TE suppression.
Collapse
|
41
|
Rosner A, Moiseeva E, Rabinowitz C, Rinkevich B. Germ lineage properties in the urochordate Botryllus schlosseri - from markers to temporal niches. Dev Biol 2013; 384:356-74. [PMID: 24120376 DOI: 10.1016/j.ydbio.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 08/25/2013] [Accepted: 10/03/2013] [Indexed: 01/28/2023]
Abstract
The primordial germ cells (PGCs) in the colonial urochordate Botryllus schlosseri are sequestered in late embryonic stage. PGC-like populations, located at any blastogenic stage in specific niches, inside modules with curtailed lifespan, survive throughout the life of the colony by repeated weekly migration to newly formed buds. This cyclical migration and the lack of specific markers for PGC-like populations are obstacles to the study on PGCs. For that purpose, we isolated the Botryllus DDX1 (BS-DDX1) and characterized it by normal expression patterns and by specific siRNA knockdown experiments. Expression of BS-DDX1 concurrent with BS-Vasa, γ-H2AX, BS-cadherin and phospho-Smad1/5/8, demarcate PGC cells from soma cells and from more differentiated germ cells lineages, which enabled the detection of additional putative transient niches in zooids. Employing BS-cadherin siRNA knockdown, retinoic acid (RA) administration or β-estradiol administration affirmed the BS-Vasa(+)BS-DDX1(+)BS-cadherin(+)γ-H2AX(+)phospho-Smad1/5/8(+) population as the B. schlosseri PGC-like cells. By striving to understand the PGC-like cells trafficking between transient niches along blastogenic cycles, CM-DiI-stained PGC-like enriched populations from late blastogenic stage D zooids were injected into genetically matched colonial ramets at blastogenic stages A or C and their fates were observed for 9 days. Based on the accumulated data, we conceived a novel network of several transient and short lived 'germ line niches' that preserve PGCs homeostasis, protecting these cells from the weekly astogenic senescence processes, thus enabling the survival of the PGCs throughout the organism's life.
Collapse
Affiliation(s)
- Amalia Rosner
- National Institute of Oceanography, Israel Oceanography & Limnological Research, Tel Shikmona, P.O. Box 8030, Haifa 31080, Israel.
| | | | | | | |
Collapse
|
42
|
Liu C, Duan W, Zhang L, Xu S, Li R, Chen C, He M, Lu Y, Wu H, Yu Z, Zhou Z. Bisphenol A exposure at an environmentally relevant dose induces meiotic abnormalities in adult male rats. Cell Tissue Res 2013; 355:223-32. [DOI: 10.1007/s00441-013-1723-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 08/19/2013] [Indexed: 12/13/2022]
|
43
|
Smith TB, De Iuliis GN, Lord T, Aitken RJ. The senescence-accelerated mouse prone 8 as a model for oxidative stress and impaired DNA repair in the male germ line. Reproduction 2013; 146:253-62. [DOI: 10.1530/rep-13-0186] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The discovery of a truncated base excision repair pathway in human spermatozoa mediated by OGG1 has raised questions regarding the effect of mutations in critical DNA repair genes on the integrity of the paternal genome. The senescence-accelerated mouse prone 8 (SAMP8) is a mouse model containing a suite of naturally occurring mutations resulting in an accelerated senescence phenotype largely mediated by oxidative stress, which is further enhanced by a mutation in theOgg1gene, greatly reducing the ability of the enzyme to excise 8-hydroxy,2′-deoxyguanosine (8OHdG) adducts. An analysis of the reproductive phenotype of the SAMP8 males revealed a high level of DNA damage in caudal epididymal spermatozoa as measured by the alkaline Comet assay. Furthermore, these lesions were confirmed to be oxidative in nature, as demonstrated by significant increases in 8OHdG adduct formation in the SAMP8 testicular tissue (P<0.05) as well as in mature spermatozoa (P<0.001) relative to a control strain (SAMR1). Despite this high level of oxidative DNA damage in spermatozoa, reactive oxygen species generation was not elevated and motility of spermatozoa was found to be similar to that for the control strain with the exception of progressive motility, which exhibited a slight but significant decline with advancing age (P<0.05). When challenged with Fenton reagents (H2O2and Fe2+), the SAMP8 spermatozoa demonstrated a highly increased susceptibility to formation of 8OHdG adducts compared with the controls (P<0.001). These data highlight the role of oxidative stress and OGG1-dependent base excision repair mechanisms in defining the genetic integrity of mammalian spermatozoa.
Collapse
|
44
|
Di Giacomo M, Comazzetto S, Saini H, De Fazio S, Carrieri C, Morgan M, Vasiliauskaite L, Benes V, Enright AJ, O'Carroll D. Multiple epigenetic mechanisms and the piRNA pathway enforce LINE1 silencing during adult spermatogenesis. Mol Cell 2013; 50:601-8. [PMID: 23706823 DOI: 10.1016/j.molcel.2013.04.026] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/12/2013] [Accepted: 04/08/2013] [Indexed: 11/25/2022]
Abstract
Transposons present an acute challenge to the germline, and mechanisms that repress their activity are essential for transgenerational genomic integrity. LINE1 (L1) is the most successful retrotransposon and is epigenetically repressed by CpG DNA methylation. Here, we identify two additional important mechanisms by which L1 is repressed during spermatogenesis. We demonstrate that the Piwi protein Mili and the piRNA pathway are required to posttranscriptionally silence L1 in meiotic pachytene cells even in the presence of normal L1 DNA methylation. Strikingly, in the absence of both a functional piRNA pathway and DNA methylation, L1 elements are normally repressed in mitotic stages of spermatogenesis. Accordingly, we find that the euchromatic repressive histone H3 dimethylated lysine 9 modification cosuppresses L1 expression therein. We demonstrate the existence of multiple epigenetic mechanisms that in conjunction with the piRNA pathway sequentially enforce L1 silencing and genomic stability during mitotic and meiotic stages of adult spermatogenesis.
Collapse
Affiliation(s)
- Monica Di Giacomo
- Mouse Biology Unit, European Molecular Biology Laboratory, Via Ramarini 32, 00015 Monterotondo Scalo, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Doyle TJ, Bowman JL, Windell VL, McLean DJ, Kim KH. Transgenerational effects of di-(2-ethylhexyl) phthalate on testicular germ cell associations and spermatogonial stem cells in mice. Biol Reprod 2013; 88:112. [PMID: 23536373 DOI: 10.1095/biolreprod.112.106104] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent evidence has linked human phthalate exposure to abnormal reproductive and hormonal effects. Phthalates are plasticizers that confer flexibility and transparency to plastics, but they readily contaminate the body and the environment. In this study, timed pregnant CD1 outbred mice were treated with di-(2-ethylhexyl) phthalate (DEHP) from Embryonic Day 7 (E7) to E14. The subsequent generation (F1) offspring were then bred to produce the F2, F3, and F4 offspring, without any further DEHP treatment. This exposure scheme disrupted testicular germ cell association and decreased sperm count and motility in F1 to F4 offspring. By spermatogonial transplantation techniques, the exposure scheme also disrupted spermatogonial stem cell (SSC) function of F3 offspring. The W/W(V) recipient testes transplanted with F3 offspring germ cells from the DEHP-treated group had a dramatically lower percentage of donor germ cell-derived spermatogenic recovery in seminiferous tubules when compared to the recipient testes transplanted with CD1 control germ cells. Further characterization showed that the major block of donor germ cell-derived spermatogenesis was before the appearance of undifferentiated spermatogonia. Interestingly, the testes transplanted with the F3 offspring germ cells from the DEHP-treated group, when regenerated, replicated testis morphology similar to that observed in the testes from the F1 to F3 offspring of the DEHP-treated group, suggesting that the germ cell disorganization phenotype originates from the stem cells of F3 offspring. In conclusion, embryonic exposure to DEHP was found to disrupt testicular germ cell organization and SSC function in a transgenerational manner.
Collapse
Affiliation(s)
- Timothy J Doyle
- School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | | | | | | | | |
Collapse
|
46
|
Blanco-Rodríguez J. Programmed phosphorylation of histone H2AX precedes a phase of DNA double-strand break-independent synapsis in mouse meiosis. Reproduction 2012; 144:699-712. [PMID: 23035256 DOI: 10.1530/rep-12-0326] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Accurate homologue synapsis during meiosis is essential for faithful chromosome segregation and formation of viable gametes. The finding of Spo11-dependent gamma-H2AX (γH2AX) formation during leptotene and data on mutant mice have led to the notion that synapsis in mammals depends on meiotic DNA double-stranded break (DSB) repair. A second wave of ataxia telangiectasia mutated (ATM) and Rad3-related (ATR)-dependent γH2AX formation has been observed in Atm-null mice during zygotene, suggesting that this wave of phosphorylation also occurs in normal mice. Here I aimed to confirm and to analyse in deep this wave of phosphorylation. Immunostaining of spread spermatocytes shows that γH2AX accumulates on the short last axis stretches to pair. This accumulation appears within all the nuclei undergoing a specific step of late zygotene and disappears from every spermatocyte immediately after pairing completion. This γH2AX signal co-localises with ATR, is Spo11-independent and does not co-localise with free DNA 3'-end labelling. I conclude that ATR/γH2AX asynapsis signalling at the end of zygotene belongs to a physiologically programmed pathway operating at a specific meiotic step, and I propose that this pathway is involved in the triggering of a phase of DSB-independent chromosome pairing that leads to synapsis completion in normal mouse meiosis.
Collapse
Affiliation(s)
- Josefa Blanco-Rodríguez
- Departamento de Biología Celular, Facultad de Medicina, Universidad de Valladolid, Ramón y Cajal 7, 47005 Valladolid, Spain.
| |
Collapse
|
47
|
Cordelli E, Eleuteri P, Grollino MG, Benassi B, Blandino G, Bartoleschi C, Pardini MC, Di Caprio EV, Spanò M, Pacchierotti F, Villani P. Direct and delayed X-ray-induced DNA damage in male mouse germ cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2012; 53:429-439. [PMID: 22730201 DOI: 10.1002/em.21703] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 04/18/2012] [Accepted: 04/19/2012] [Indexed: 06/01/2023]
Abstract
Sperm DNA integrity is essential for the accurate transmission of paternal genetic information. Various stages of spermatogenesis are characterized by large differences in radiosensitivity. Differentiating spermatogonia are susceptible to radiation-induced cell killing, but some of them can repair DNA damage and progress through differentiation. In this study, we applied the neutral comet assay, immunodetection of phosphorylated H2AX (γ-H2AX) and the Sperm Chromatin Structure Assay (SCSA) to detect DNA strand breaks in testicular cells and spermatozoa at different times following in vivo X-ray irradiation. Radiation produced DNA strand breaks in testicular cells that were repaired within the first few hours after exposure. Spermatozoa were resistant to the induction of DNA damage, but non-targeted DNA lesions were detected in spermatozoa derived from surviving irradiated spermatogonia. These lesions formed while round spermatids started to elongate within the testicular seminiferous tubules. The transcription of pro-apoptotic genes at this time was also enhanced, suggesting that an apoptotic-like process was involved in DNA break production. Our results suggest that proliferating spermatogonia retain a memory of the radiation insult that is recognized at a later developmental stage and activates a process leading to DNA fragmentation.
Collapse
|
48
|
Pinpointing the expression of piRNAs and function of the PIWI protein subfamily during spermatogenesis in the mouse. Dev Biol 2011; 355:215-26. [PMID: 21539824 DOI: 10.1016/j.ydbio.2011.04.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 04/17/2011] [Indexed: 01/20/2023]
Abstract
PIWI proteins and piRNAs have been linked to transposon silencing in the primordial mouse testis, but their function in the adult testis remains elusive. Here we report the cytological characterization of piRNAs in the adult mouse testis and the phenotypic analysis of Miwi(-/-); Mili(-/-) mice. We show that piRNAs are specifically present in germ cells, especially abundant in spermatocytes and early round spermatids, regardless of the type of the genomic sequences to which they correspond. piRNAs and PIWI proteins are present in both the cytoplasm and nucleus. In the cytoplasm, they are enriched in the chromatoid body; whereas in the nucleus they are enriched in the dense body, a male-specific organelle associated with synapsis and the formation of the XY body during meiosis. Moreover, by generating Miwi(-/-); Mili(-/-) mice, which lack all PIWI proteins in the adult, we show that PIWI proteins and presumably piRNAs in the adult are required only for spermatogenesis. Spermatocytes without PIWI proteins are arrested at the pachytene stage, when the sex chromosomes undergo transcriptional silencing to form the XY body. These results pinpoint a function of the PIWI protein subfamily to meiosis during spermatogenesis.
Collapse
|
49
|
Paris L, Cordelli E, Eleuteri P, Grollino MG, Pasquali E, Ranaldi R, Meschini R, Pacchierotti F. Kinetics of γ-H2AX induction and removal in bone marrow and testicular cells of mice after X-ray irradiation. Mutagenesis 2011; 26:563-72. [DOI: 10.1093/mutage/ger017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
50
|
Lee J, Hirano T. RAD21L, a novel cohesin subunit implicated in linking homologous chromosomes in mammalian meiosis. J Cell Biol 2011; 192:263-76. [PMID: 21242291 PMCID: PMC3172173 DOI: 10.1083/jcb.201008005] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 12/22/2010] [Indexed: 11/26/2022] Open
Abstract
Cohesins are multi-subunit protein complexes that regulate sister chromatid cohesion during mitosis and meiosis. Here we identified a novel kleisin subunit of cohesins, RAD21L, which is conserved among vertebrates. In mice, RAD21L is expressed exclusively in early meiosis: it apparently replaces RAD21 in premeiotic S phase, becomes detectable on the axial elements in leptotene, and stays on the axial/lateral elements until mid pachytene. RAD21L then disappears, and is replaced with RAD21. This behavior of RAD21L is unique and distinct from that of REC8, another meiosis-specific kleisin subunit. Remarkably, the disappearance of RAD21L at mid pachytene correlates with the completion of DNA double-strand break repair and the formation of crossovers as judged by colabeling with molecular markers, γ-H2AX, MSH4, and MLH1. RAD21L associates with SMC3, STAG3, and either SMC1α or SMC1β. Our results suggest that cohesin complexes containing RAD21L may be involved in synapsis initiation and crossover recombination between homologous chromosomes.
Collapse
Affiliation(s)
- Jibak Lee
- Chromosome Dynamics Laboratory, RIKEN Advanced Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | |
Collapse
|