1
|
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
|
2
|
Georgakopoulos I, Kouloulias V, Ntoumas GN, Desse D, Koukourakis I, Kougioumtzopoulou A, Kanakis G, Zygogianni A. Radiotherapy and Testicular Function: A Comprehensive Review of the Radiation-Induced Effects with an Emphasis on Spermatogenesis. Biomedicines 2024; 12:1492. [PMID: 39062064 PMCID: PMC11274587 DOI: 10.3390/biomedicines12071492] [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: 05/14/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
This comprehensive review explores the existing literature on the effects of radiotherapy on testicular function, focusing mainly on spermatogenic effects, but also with a brief report on endocrine abnormalities. Data from animal experiments as well as results on humans either from clinical studies or from accidental radiation exposure are included to demonstrate a complete perspective on the level of vulnerability of the testes and their various cellular components to irradiation. Even relatively low doses of radiation, produced either from direct testicular irradiation or more commonly from scattered doses, may often lead to detrimental effects on sperm count and quality. Leydig cells are more radioresistant; however, they can still be influenced by the doses used in clinical practice. The potential resultant fertility complications of cancer radiotherapy should be always discussed with the patient before treatment initiation, and all available and appropriate fertility preservation measures should be taken to ensure the future reproductive potential of the patient. The topic of potential hereditary effects of germ cell irradiation remains a controversial field with ethical implications, requiring future research.
Collapse
Affiliation(s)
- Ioannis Georgakopoulos
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Vas. Sofias 76, 115 28 Athens, Greece; (G.-N.N.); (I.K.); (A.Z.)
| | - Vassilios Kouloulias
- Radiotherapy Unit, 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, Rimini 1, 124 62 Athens, Greece; (V.K.); (A.K.)
| | - Georgios-Nikiforos Ntoumas
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Vas. Sofias 76, 115 28 Athens, Greece; (G.-N.N.); (I.K.); (A.Z.)
| | - Dimitra Desse
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Vas. Sofias 76, 115 28 Athens, Greece; (G.-N.N.); (I.K.); (A.Z.)
| | - Ioannis Koukourakis
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Vas. Sofias 76, 115 28 Athens, Greece; (G.-N.N.); (I.K.); (A.Z.)
| | - Andromachi Kougioumtzopoulou
- Radiotherapy Unit, 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, Rimini 1, 124 62 Athens, Greece; (V.K.); (A.K.)
| | - George Kanakis
- Department of Endocrinology, Athens Naval & VA Hospital, 115 21 Athens, Greece;
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynaecology, Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Anna Zygogianni
- Radiation Oncology Unit, 1st Department of Radiology, Medical School, Aretaieion Hospital, National and Kapodistrian University of Athens, Vas. Sofias 76, 115 28 Athens, Greece; (G.-N.N.); (I.K.); (A.Z.)
| |
Collapse
|
3
|
Pasquini M, Chiani F, Gambadoro A, Di Pietro C, Paoletti R, Orsini T, Putti S, Scavizzi F, La Sala G, Ermakova O. The Odad3 Gene Is Necessary for Spermatozoa Development and Male Fertility in Mice. Cells 2024; 13:1053. [PMID: 38920681 PMCID: PMC11201558 DOI: 10.3390/cells13121053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Odad3 gene loss-of-function mutation leads to Primary Ciliary Dyskinesia (PCD), a disease caused by motile cilia dysfunction. Previously, we demonstrated that knockout of the Odad3 gene in mice replicates several features of PCD, such as hydrocephalus, defects in left-right body symmetry, and male infertility, with a complete absence of sperm in the reproductive tract. The majority of Odad3 knockout animals die before sexual maturation due to severe hydrocephalus and failure to thrive, which precludes fertility studies. Here, we performed the expression analysis of the Odad3 gene during gonad development and in adult testes. We showed that Odad3 starts its expression during the first wave of spermatogenesis, specifically at the meiotic stage, and that its expression is restricted to the germ cells in the adult testes, suggesting that Odad3 plays a role in spermatozoa formation. Subsequently, we conditionally deleted the Odad3 gene in adult males and demonstrated that even partial ablation of the Odad3 gene leads to asthenoteratozoospermia with multiple morphological abnormalities of sperm flagella (MMAF) in mice. The analysis of the seminiferous tubules in Odad3-deficient mice revealed defects in spermatogenesis with accumulation of seminiferous tubules at the spermiogenesis and spermiation phases. Furthermore, analysis of fertility in heterozygous Odad3+/- knockout mice revealed a reduction in sperm count and motility as well as abnormal sperm morphology. Additionally, Odad3+/- males exhibited a shorter fertile lifespan. Overall, these results suggest the important role of Odad3 and Odad3 gene dosage in male fertility. These findings may have an impact on the genetic and fertility counseling practice of PCD patients carrying Odad3 loss-of-function mutations.
Collapse
Affiliation(s)
- Miriam Pasquini
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Francesco Chiani
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Alessia Gambadoro
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Chiara Di Pietro
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Renata Paoletti
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
| | - Tiziana Orsini
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Sabrina Putti
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Ferdinando Scavizzi
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Gina La Sala
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| | - Olga Ermakova
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy; (M.P.); (F.C.); (A.G.); (C.D.P.); (R.P.); (T.O.); (S.P.); (F.S.)
- European Mouse Mutant Archive (EMMA), INFRAFRONTIER, Monterotondo Mouse Clinic, National Research Council of Italy (CNR), Adriano Buzzati-Traverso Campus, Via Ramarini, 32, 00015 Monterotondo, Italy
| |
Collapse
|
4
|
Mitchell T, Lin J, Hicks S, James J, Rangan P, Forni P. Loss of function of male-specific lethal 3 (Msl3) does not affect spermatogenesis in rodents. Dev Dyn 2024; 253:453-466. [PMID: 37847071 PMCID: PMC11021377 DOI: 10.1002/dvdy.669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/12/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Male-specific lethal 3 (Msl3) is a member of the chromatin-associated male-specific lethal MSL complex, which is responsible for the transcriptional upregulation of genes on the X chromosome in males of Drosophila. Although the dosage complex operates differently in mammals, the Msl3 gene is conserved from flies to humans. Msl3 is required for meiotic entry during Drosophila oogenesis. Recent reports indicate that also in primates, Msl3 is expressed in undifferentiated germline cells before meiotic entry. However, if Msl3 plays a role in the meiotic entry of mammals has yet to be explored. RESULTS To understand, if Msl3a plays a role in the meiotic entry of mammals, we used mouse spermatogenesis as a study model. Analyses of single-cell RNA-seq data revealed that, in mice, Msl3 is mostly expressed in meiotic cells. To test the role of Msl3 in meiosis, we used a male germline-specific Stra8-iCre driver and a newly generated Msl3flox conditional knock-out mouse line. Msl3 conditional loss-of-function in spermatogonia did not cause spermatogenesis defects or changes in the expression of genes related to meiosis. CONCLUSIONS Our data suggest that, in mice, Msl3 exhibits delayed expression compared to Drosophila and primates, and loss-of-function mutations disrupting the chromodomain of Msl3 alone do not impede meiotic entry in rodents.
Collapse
Affiliation(s)
- T.A. Mitchell
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| | - J.M. Lin
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| | - S.M. Hicks
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - J.R. James
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - P. Rangan
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - P.E. Forni
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
5
|
Liu Y, Lin Z, Yan J, Zhang X, Tong MH. A Rad50-null mutation in mouse germ cells causes reduced DSB formation, abnormal DSB end resection and complete loss of germ cells. Development 2024; 151:dev202312. [PMID: 38512324 DOI: 10.1242/dev.202312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
The conserved MRE11-RAD50-NBS1/Xrs2 complex is crucial for DNA break metabolism and genome maintenance. Although hypomorphic Rad50 mutation mice showed normal meiosis, both null and hypomorphic rad50 mutation yeast displayed impaired meiosis recombination. However, the in vivo function of Rad50 in mammalian germ cells, particularly its in vivo role in the resection of meiotic double strand break (DSB) ends at the molecular level remains elusive. Here, we have established germ cell-specific Rad50 knockout mouse models to determine the role of Rad50 in mitosis and meiosis of mammalian germ cells. We find that Rad50-deficient spermatocytes exhibit defective meiotic recombination and abnormal synapsis. Mechanistically, using END-seq, we demonstrate reduced DSB formation and abnormal DSB end resection occurs in mutant spermatocytes. We further identify that deletion of Rad50 in gonocytes leads to complete loss of spermatogonial stem cells due to genotoxic stress. Taken together, our results reveal the essential role of Rad50 in mammalian germ cell meiosis and mitosis, and provide in vivo views of RAD50 function in meiotic DSB formation and end resection at the molecular level.
Collapse
Affiliation(s)
- Yuefang Liu
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhen Lin
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junyi Yan
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xi Zhang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming-Han Tong
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
6
|
Wilson CA, Batzel P, Postlethwait JH. Direct male development in chromosomally ZZ zebrafish. Front Cell Dev Biol 2024; 12:1362228. [PMID: 38529407 PMCID: PMC10961373 DOI: 10.3389/fcell.2024.1362228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/20/2024] [Indexed: 03/27/2024] Open
Abstract
The genetics of sex determination varies across taxa, sometimes even within a species. Major domesticated strains of zebrafish (Danio rerio), including AB and TU, lack a strong genetic sex determining locus, but strains more recently derived from nature, like Nadia (NA), possess a ZZ male/ZW female chromosomal sex-determination system. AB fish pass through a juvenile ovary stage, forming oocytes that survive in fish that become females but die in fish that become males. To understand mechanisms of gonad development in NA zebrafish, we studied histology and single cell transcriptomics in developing ZZ and ZW fish. ZW fish developed oocytes by 22 days post-fertilization (dpf) but ZZ fish directly formed testes, avoiding a juvenile ovary phase. Gonads of some ZW and WW fish, however, developed oocytes that died as the gonad became a testis, mimicking AB fish, suggesting that the gynogenetically derived AB strain is chromosomally WW. Single-cell RNA-seq of 19dpf gonads showed similar cell types in ZZ and ZW fish, including germ cells, precursors of gonadal support cells, steroidogenic cells, interstitial/stromal cells, and immune cells, consistent with a bipotential juvenile gonad. In contrast, scRNA-seq of 30dpf gonads revealed that cells in ZZ gonads had transcriptomes characteristic of testicular Sertoli, Leydig, and germ cells while ZW gonads had granulosa cells, theca cells, and developing oocytes. Hematopoietic and vascular cells were similar in both sex genotypes. These results show that juvenile NA zebrafish initially develop a bipotential gonad; that a factor on the NA W chromosome, or fewer than two Z chromosomes, is essential to initiate oocyte development; and without the W factor, or with two Z doses, NA gonads develop directly into testes without passing through the juvenile ovary stage. Sex determination in AB and TU strains mimics NA ZW and WW zebrafish, suggesting loss of the Z chromosome during domestication. Genetic analysis of the NA strain will facilitate our understanding of the evolution of sex determination mechanisms.
Collapse
|
7
|
Jensen CFS, Mamsen LS, Wang D, Fode M, Giwercman A, Jørgensen N, Ohl DA, Fedder J, Hoffmann ER, Yding Andersen C, Sønksen J. Results from the first autologous grafting of adult human testis tissue: a case report. Hum Reprod 2024; 39:303-309. [PMID: 38140699 DOI: 10.1093/humrep/dead243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/21/2023] [Indexed: 12/24/2023] Open
Abstract
Fertility restoration using autologous testicular tissue transplantation is relevant for infertile men surviving from childhood cancer and, possibly, in men with absent or incomplete spermatogenesis resulting in the lack of spermatozoa in the ejaculate (non-obstructive azoospermia, NOA). Currently, testicular tissue from pre-pubertal boys extracted before treatment with gonadotoxic cancer therapy can be cryopreserved with good survival of spermatogonial stem cells. However, strategies for fertility restoration, after successful cancer treatment, are still experimental and no clinical methods have yet been developed. Similarly, no clinically available treatments can help men with NOA to become biological fathers after failed attempts of testicular surgical sperm retrieval. We present a case of a 31-year-old man with NOA who had three pieces of testis tissue (each ∼2 × 4 × 2 mm3) extracted and cryopreserved in relation to performing microdissection testicular sperm extraction (mTESE). Approximately 2 years after mTESE, the thawed tissue pieces were engrafted in surgically created pockets bilaterally under the scrotal skin. Follow-up was performed after 2, 4, and 6 months with assessment of reproductive hormones and ultrasound of the scrotum. After 6 months, all engrafted tissue was extracted and microscopically analyzed for the presence of spermatozoa. Furthermore, parts of the extracted tissue were analyzed histologically and by immunohistochemical analysis. Active blood flow in the engrafted tissue was demonstrated by doppler ultrasound after 6 months. No spermatozoa were found in the extracted tissue. Histological and immunohistochemical analysis demonstrated graft survival with intact clear tubules and normal cell organization. Sertoli cells and spermatocytes with normal morphology were located near the basement membrane. MAGE-A and VASA positive spermatogonia/spermatocytes were detected together with SOX9 positive Sertoli cells. Spermatocytes and/or Sertoli cells positive for γH2AX was also detected. In summary, following autologous grafting of frozen-thawed testis tissue under the scrotal skin in a man with NOA, we demonstrated graft survival after 6 months. No mature spermatozoa were detected; however, this is likely due to the pre-existing spermatogenic failure.
Collapse
Affiliation(s)
- Christian Fuglesang S Jensen
- Department of Urology, Copenhagen University Hospital-Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Danyang Wang
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Laboratory of Reproductive Biology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Mikkel Fode
- Department of Urology, Copenhagen University Hospital-Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Aleksander Giwercman
- Department of Translational Medicine and Reproductive Medicine Centre, Lunds University and Skane University Hospital, Malmö, Sweden
| | - Niels Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Dana A Ohl
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Jens Fedder
- Centre of Andrology & Fertility Clinic, Department D, Odense University Hospital, Odense C, Denmark
| | - Eva R Hoffmann
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Claus Yding Andersen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Laboratory of Reproductive Biology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Jens Sønksen
- Department of Urology, Copenhagen University Hospital-Herlev and Gentofte Hospital, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Spandole-Dinu S, Catrina AM, Voinea OC, Andone A, Radu S, Haidoiu C, Călborean O, Hertzog RG, Popescu DM. Evaluating the radioprotective effect of green barley juice on male rats. Int J Radiat Biol 2024; 100:281-288. [PMID: 37769021 DOI: 10.1080/09553002.2023.2264923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 09/23/2023] [Indexed: 09/30/2023]
Abstract
PURPOSE DNA damage accounts for most biological effects of ionizing radiation. Antioxidants are known for their protective effect by preventing DNA damage. This pilot study aimed to evaluate the potential radioprotective effect of Natural SOD®, a green barley juice rich in antioxidants, on DNA damage in the testes and lymphocytes of Wistar rats exposed to ionizing radiation. MATERIALS AND METHODS Male Wistar rats (n = 15) were selected and equally divided into three groups. Rats in one of the groups were pretreated orally with Natural SOD® for 14 days, while rats in another group were sham-pretreated with saline solution. Rats in both these groups were afterwards subjected to a single dose of 6 Gy X-ray whole-body irradiation. The control group did not receive any treatment and was not irradiated. Shortly after X-ray exposure, all rats were sacrificed and testes and blood were collected. Gamma-H2AX and histopathological assessment in the testes, along with comet assay of lymphocytes were performed. RESULTS Histopathological examination of the testes showed no significant architectural alterations. Immunofluorescent staining of γ-H2AX revealed more DNA double-strand break sites in testicular cells from sham animals compared to Natural SOD® pretreated rats. Alkaline comet assay results showed increased DNA damage in lymphocytes of irradiated rats compared to the control group with little differences between the pretreated groups. Animals pretreated with Natural SOD showed slightly reduced DNA damage compared to sham-pretreated rats. These findings suggest a potential protective effect of Natural SOD® against radiation-induced DNA damage. CONCLUSIONS Natural SOD® exhibited a potential prophylactic radioprotective effect in rats, particularly in testes. Further investigations to determine medium and long-term effects of X-ray in animals administered Natural SOD® are needed to better estimate the radioprotective effect.
Collapse
Affiliation(s)
- Sonia Spandole-Dinu
- Experimental Radiobiology Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| | - Ana-Maria Catrina
- Neurobiology Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| | - Oana Cristina Voinea
- Experimental Pharmacotoxicology Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
- Pathology Department, Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| | - Alina Andone
- Experimental Radiobiology Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| | - Speranța Radu
- Experimental Radiobiology Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| | - Cerasela Haidoiu
- Neurobiology Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| | - Octavian Călborean
- Experimental Radiobiology Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| | - Radu Gabriel Hertzog
- National Center for Expertise and Intervention in Public Health for CBRN agents, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| | - Diana Mihaela Popescu
- Regenerative Medicine Laboratory, Cantacuzino National Medical Military Institute for Research and Development, Bucharest, Romania
| |
Collapse
|
9
|
Endo T, Kobayashi K, Matsumura T, Emori C, Ozawa M, Kawamoto S, Okuzaki D, Shimada K, Miyata H, Shimada K, Kodani M, Ishikawa-Yamauchi Y, Motooka D, Hara E, Ikawa M. Multiple ageing effects on testicular/epididymal germ cells lead to decreased male fertility in mice. Commun Biol 2024; 7:16. [PMID: 38177279 PMCID: PMC10766604 DOI: 10.1038/s42003-023-05685-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
In mammals, females undergo reproductive cessation with age, whereas male fertility gradually declines but persists almost throughout life. However, the detailed effects of ageing on germ cells during and after spermatogenesis, in the testis and epididymis, respectively, remain unclear. Here we comprehensively examined the in vivo male fertility and the overall organization of the testis and epididymis with age, focusing on spermatogenesis, and sperm function and fertility, in mice. We first found that in vivo male fertility decreased with age, which is independent of mating behaviors and testosterone levels. Second, overall sperm production in aged testes was decreased; about 20% of seminiferous tubules showed abnormalities such as germ cell depletion, sperm release failure, and perturbed germ cell associations, and the remaining 80% of tubules contained lower number of germ cells because of decreased proliferation of spermatogonia. Further, the spermatozoa in aged epididymides exhibited decreased total cell numbers, abnormal morphology/structure, decreased motility, and DNA damage, resulting in low fertilizing and developmental rates. We conclude that these multiple ageing effects on germ cells lead to decreased in vivo male fertility. Our present findings are useful to better understand the basic mechanism behind the ageing effect on male fertility in mammals including humans.
Collapse
Affiliation(s)
- Tsutomu Endo
- Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University, Tokyo, Japan.
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| | - Kiyonori Kobayashi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Chihiro Emori
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Manabu Ozawa
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shimpei Kawamoto
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Okuzaki
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kentaro Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Mayo Kodani
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yu Ishikawa-Yamauchi
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Daisuke Motooka
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Eiji Hara
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masahito Ikawa
- Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
10
|
López-Jiménez P, Berenguer I, Pérez-Moreno I, de Aledo JG, Parra MT, Page J, Gómez R. The Organotypic Culture of Mouse Seminiferous Tubules as a Reliable Methodology for the Study of Meiosis In Vitro. Methods Mol Biol 2024; 2818:147-160. [PMID: 39126472 DOI: 10.1007/978-1-0716-3906-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Male mouse meiosis has been traditionally studied using descriptive methods like histological sections and spreading or squashing techniques, which allow the observation of fixed meiocytes in either wildtype or genetically modified mice. For these studies, the sacrifice of the males and the extraction of the testicles are required to obtain the material of study. Other functional in vivo studies include the administration of intravenous or intraperitoneal drugs, or the exposure to mutagenic agents or generators of DNA damage, in order to study their impact on meiosis progression. However, in these studies, the exposure times or drug concentration are important limitations to consider when acknowledging animal welfare. Recently, several approaches have been proposed to offer alternative methodologies that allow the in vitro study of spermatocytes with a considerable reduction in the use of animals. Here we revisit and validate an optimal technique of organotypic culture of fragments of seminiferous tubules for meiotic studies. This technique is a trustable methodology to develop functional studies that preserve the histological configuration of the seminiferous tubule, aim homogeneity of the procedures (the use of the same animal for different study conditions), and allow procedures that would compromise the animal welfare. Therefore, this methodology is highly recommendable for the study of meiosis and spermatogenesis, while it supports the principle of 3R's for animal research.
Collapse
Affiliation(s)
- Pablo López-Jiménez
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Meiosis group, MRC Laboratory of Medical Sciences, London, UK
| | - Inés Berenguer
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa (CBMSO), Madrid, Spain
| | - Irene Pérez-Moreno
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | | | - María Teresa Parra
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Jesús Page
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| | - Rocío Gómez
- Departamento de Biología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| |
Collapse
|
11
|
Chen Y, Lin Z, Zhang X, Wu M, Tong MH. Isolation of Homogeneous Sub-populations of Spermatocytes from Mouse Testis. Methods Mol Biol 2024; 2818:115-132. [PMID: 39126470 DOI: 10.1007/978-1-0716-3906-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Mammalian meiosis is a highly specialized cell division process, resulting in the production of genetically unique haploid cells. However, the molecular mechanisms governing meiosis remain largely unknown, primarily due to the difficulty in isolating pure sub-populations of spermatocytes. Definitive molecular, biochemical, and functional investigations of the meiosis process require the isolation of these individual homogeneous sub-populations of spermatocytes. Here, we present an approach that enables the purification of homogeneous spermatocytes from mouse testis at desired sub-stages. This approach consists of two strategic steps. The first is to synchronize spermatogenesis, aiming to minimize the diversity and complexity of testicular germ cells. The second involves utilizing mouse models with germ cell-specific fluorescent markers to differentiate the desired subtype from other cells in the testis. By employing fluorescence-activated cell sorting (FACS), this approach yields highly pure populations of spermatocytes at each sub-stage. When combined with other massively parallel sequencing techniques and in vitro cell culture methods, this approach will enhance our comprehension of the molecular mechanisms underlying mammalian meiosis and promote in vitro gametogenesis.
Collapse
Affiliation(s)
- Yao Chen
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Zhen Lin
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xi Zhang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Meixia Wu
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ming-Han Tong
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
12
|
Wilson CA, Batzel P, Postlethwait JH. Direct Male Development in Chromosomally ZZ Zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573483. [PMID: 38234788 PMCID: PMC10793451 DOI: 10.1101/2023.12.27.573483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The genetics of sex determination varies across taxa, sometimes even within a species. Major domesticated strains of zebrafish ( Danio rerio ), including AB and TU, lack a strong genetic sex determining locus, but strains more recently derived from nature, like Nadia (NA), possess a ZZ male/ZW female chromosomal sex-determination system. AB strain fish pass through a juvenile ovary stage, forming oocytes that survive in fish that become females but die in fish that become males. To understand mechanisms of gonad development in NA zebrafish, we studied histology and single cell transcriptomics in developing ZZ and ZW fish. ZW fish developed oocytes by 22 days post-fertilization (dpf) but ZZ fish directly formed testes, avoiding a juvenile ovary phase. Gonads of some ZW and WW fish, however, developed oocytes that died as the gonad became a testis, mimicking AB fish, suggesting that the gynogenetically derived AB strain is chromosomally WW. Single-cell RNA-seq of 19dpf gonads showed similar cell types in ZZ and ZW fish, including germ cells, precursors of gonadal support cells, steroidogenic cells, interstitial/stromal cells, and immune cells, consistent with a bipotential juvenile gonad. In contrast, scRNA-seq of 30dpf gonads revealed that cells in ZZ gonads had transcriptomes characteristic of testicular Sertoli, Leydig, and germ cells while ZW gonads had granulosa cells, theca cells, and developing oocytes. Hematopoietic and vascular cells were similar in both sex genotypes. These results show that juvenile NA zebrafish initially develop a bipotential gonad; that a factor on the NA W chromosome or fewer than two Z chromosomes is essential to initiate oocyte development; and without the W factor or with two Z doses, NA gonads develop directly into testes without passing through the juvenile ovary stage. Sex determination in AB and TU strains mimics NA ZW and WW zebrafish, suggesting loss of the Z chromosome during domestication. Genetic analysis of the NA strain will facilitate our understanding of the evolution of sex determination mechanisms.
Collapse
|
13
|
Mitchell T, Lin JM, James JR, Hicks SM, Rangan P, Forni PE. Loss Of Chromodomain of Male-Specific Lethal 3 (MSL3) Does Not Affect Spermatogenesis In Rodents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532933. [PMID: 36993289 PMCID: PMC10055081 DOI: 10.1101/2023.03.16.532933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Msl3 is a member of the chromatin-associated male-specific lethal MSL complex which is responsible for the transcriptional upregulation of genes on the X chromosome in males Drosophila. Although the dosage complex operates differently in mammals, the Msl3 gene is conserved from flies to humans. Msl3 is required for meiotic entry during Drosophila oogenesis. Recent reports indicate that also in primates, Msl3 is expressed in undifferentiated germline cells before meiotic entry. However, if Msl3 plays a role in the meiotic entry of mammals has yet to be explored. To study this, we used mouse spermatogenesis as a study model. Analyses of single cells RNA-seq data revealed that, in mice, Msl3 is mostly expressed in meiotic cells. To test the role of Msl3 in meiosis, we used a male germline-specific Stra8-iCre driver and a newly generated Msl3flox conditional knock-out mouse line. Msl3 conditional loss-of-function in spermatogonia did not cause spermatogenesis defects or changes in the expression of genes related to meiosis. Our data suggest that, in mice, Msl3 exhibits delayed expression compared to Drosophila and primates, and loss-of-function mutations disrupting the chromodomain of Msl3 alone do not impede meiotic entry in rodents.
Collapse
|
14
|
Xu L, Pierce JL, Sanchez A, Chen KS, Shukla AA, Fustino NJ, Stuart SH, Bagrodia A, Xiao X, Guo L, Krailo MD, Shaikh F, Billmire DF, Pashankar F, Bestrashniy J, Oosterhuis JW, Gillis AJM, Xie Y, Teot L, Mora J, Poynter JN, Rakheja D, Looijenga LHJ, Draper BW, Frazier AL, Amatruda JF. Integrated genomic analysis reveals aberrations in WNT signaling in germ cell tumors of childhood and adolescence. Nat Commun 2023; 14:2636. [PMID: 37149691 PMCID: PMC10164134 DOI: 10.1038/s41467-023-38378-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 04/26/2023] [Indexed: 05/08/2023] Open
Abstract
Germ cell tumors (GCTs) are neoplasms of the testis, ovary and extragonadal sites that occur in infants, children, adolescents and adults. Post-pubertal (type II) malignant GCTs may present as seminoma, non-seminoma or mixed histologies. In contrast, pre-pubertal (type I) GCTs are limited to (benign) teratoma and (malignant) yolk sac tumor (YST). Epidemiologic and molecular data have shown that pre- and post-pubertal GCTs arise by distinct mechanisms. Dedicated studies of the genomic landscape of type I and II GCT in children and adolescents are lacking. Here we present an integrated genomic analysis of extracranial GCTs across the age spectrum from 0-24 years. Activation of the WNT pathway by somatic mutation, copy-number alteration, and differential promoter methylation is a prominent feature of GCTs in children, adolescents and young adults, and is associated with poor clinical outcomes. Significantly, we find that small molecule WNT inhibitors can suppress GCT cells both in vitro and in vivo. These results highlight the importance of WNT pathway signaling in GCTs across all ages and provide a foundation for future efforts to develop targeted therapies for these cancers.
Collapse
Affiliation(s)
- Lin Xu
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Joshua L Pierce
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Angelica Sanchez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kenneth S Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Abhay A Shukla
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicholas J Fustino
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Blank Children's Hospital, Des Moines, IA, USA
| | - Sarai H Stuart
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Xue Xiao
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mark D Krailo
- Department of Preventative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
- Children's Oncology Group, Monrovia, CA, USA
| | - Furqan Shaikh
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - Farzana Pashankar
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Ad J M Gillis
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Yang Xie
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population & Data Sciences, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lisa Teot
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Jaume Mora
- Sant Joan de Déu Barcelona Children's Hospital, Barcelona, Spain
| | - Jenny N Poynter
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Dinesh Rakheja
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Bruce W Draper
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA, USA
| | - A Lindsay Frazier
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - James F Amatruda
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
- Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Talibova G, Bilmez Y, Ozturk S. Increased double-strand breaks in aged mouse male germ cells may result from changed expression of the genes essential for homologous recombination or nonhomologous end joining repair. Histochem Cell Biol 2023; 159:127-147. [PMID: 36241856 DOI: 10.1007/s00418-022-02157-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2022] [Indexed: 11/26/2022]
Abstract
DNA double-strand breaks (DSBs) are commonly appearing deleterious DNA damages, which progressively increase in male germ cells during biological aging. There are two main pathways for repairing DSBs: homologous recombination (HR) and classical nonhomologous end joining (cNHEJ). Knockout and functional studies revealed that, while RAD51 and RPA70 proteins are indispensable for HR-based repair, KU80 and XRCC4 are the key proteins in cNHEJ repair. As is known, γH2AX contributes to these pathways through recruiting repair-related proteins to damaged site. The underlying reasons of increased DSBs in male germ cells during aging are not fully addressed yet. In this study, we aimed to analyze the spatiotemporal expression of the Rad51, Rpa70, Ku80, and Xrcc4 genes in the postnatal mouse testes, classified into young, prepubertal, pubertal, postpubertal, and aged groups according to their reproductive features and histological structures. We found that expression of these genes significantly decreased in the aged group compared with the other groups (P < 0.05). γH2AX staining showed that DSB levels in the germ cells from spermatogonia to elongated spermatids as well as in the Sertoli cells remarkably increased in the aged group (P < 0.05). The RAD51, RPA70, KU80, and XRCC4 protein levels exhibited predominant changes in the germ and Sertoli cells among groups (P < 0.05). These findings suggest that altered expression of the Rad51, Rpa70, Ku80, and Xrcc4 genes in the germ and Sertoli cells may be associated with increasing DSBs during biological aging, which might result in fertility loss.
Collapse
Affiliation(s)
- Gunel Talibova
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Yesim Bilmez
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
16
|
Gao Y, Zhang D, Wang P, Qu X, Xu J, Yu Y, Zhou X. Acrylamide-induced meiotic arrest of spermatocytes in adolescent mice by triggering excessive DNA strand breaks: Potential therapeutic effects of resveratrol. Hum Exp Toxicol 2023; 42:9603271231188293. [PMID: 37550604 DOI: 10.1177/09603271231188293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Background: Baked carbohydrate-rich foods are the main source of acrylamide (AA) in the general population and are widely consumed by teenagers. Considering the crucial development of the reproductive system during puberty, the health risks posed by AA in adolescent males have raised public concern.Methods: In this study, we exposed 3-week-old male pubertal mice to AA for 4 weeks to evaluate its effect on spermatogenesis using computer-assisted sperm analysis (CASA) and historical analysis. Flow cytometric analysis and meiocyte spreading assay were conducted to assess meiosis in mice. The expression of meiosis-related proteins and double-strand break (DSB) proteins were evaluated by immunoblot analyses. Additionally, isolated spermatocytes were used to explore the role of resveratrol in AA-induced damages of meiosis.Results: Our results showed that AA decreased the testicular and epididymal indexes, reduced sperm count and motility, and induced morphological disruption of the testes in pubertal mice. Subsequent meiotic analysis revealed that AA increased the proportion of 4C spermatocytes and decreased the proportion of 1C spermatids. The expression levels of meiosis-related proteins (SYCP3, Cyclin A1 and CDK2) were downregulated, and signaling proteins (γH2AX, p-CHK2 and p-ATM) expression levels were upregulated in AA-treated mice testes. Similar expression patterns were observed in primary spermatocytes treated with AA and these effects were reversed significantly by resveratrol.Conclusions: Our results indicate that AA induces meiotic arrest via persistent activation of DSBs, which may contribute to AA-compromised spermatogenesis. Resveratrol could serve as a potential therapeutic agent against AA-induced meiotic toxicity. These data highlight the importance of natural product supplementation for treating AA-related reproductive toxicity.
Collapse
Affiliation(s)
- Y Gao
- Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - D Zhang
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - P Wang
- Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - X Qu
- Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - J Xu
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Y Yu
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - X Zhou
- Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
17
|
Le W, Zhou F, Xiang J, Weng Y, Wu D, Xu J, Zhang J. Preliminary Study on 53BP1-Mediated DNA Double-Strand Break Response in Spermatogonial Stem Cells. Reprod Sci 2022; 30:1572-1584. [PMID: 36446980 DOI: 10.1007/s43032-022-01122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/30/2022] [Indexed: 11/30/2022]
Abstract
53BP1 mediates DNA repair process in somatic cells; however, the function of 53BP1 in germline stem cells still remains unclear. In the present study, animals and cells DNA damage repair (DDR) model was established by irradiation and HU treatment; immunofluorescence staining and laser confocal microscopy were used to detect the expression of 53BP1, p-CHK2, and p-P53 in the DDR process of mSSCs. 53BP1 knockdown expression mSSCs cell line conducted by Trp53bp1-shRNA was established and EdU staining was adopted to analyze cell cycle and cell proliferation. Moreover, NHEJ reporter vector was applied to detect the repair efficacy after Trp53bp1 knocked-down (KD) expression. Results showed that 53BP1 could form foci signals in mSSCs during DDR process both in vivo and in vitro, which was independent of γH2AX. 53BP1 downstream protein, p-P53, and p-CHK2 were involved and dynamically expressed in DDR response. Knocking down of Trp53bp1 expression in mSSCs could not dramatically inhibit cell proliferation, but may increase cell sensitivity to HU. The NHEJ repair efficacy was sharply decreased in Trp53bp-KD SSCs via flow cytometry analysis. We revealed the specific mechanism of 53BP1 in SSCs DDR process, which is expected to provide a new theoretical basis and insights for the diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Wei Le
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, China
| | - Fang Zhou
- Department of Surgery, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 Xianxia Road, Shanghai, 200050, China
| | - Jun Xiang
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, China
| | - Yiming Weng
- Department of Reproductive Medicine, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, China
| | - Denglong Wu
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, 200065, China
| | - Jun Xu
- Department of Regenerative Medicine, East Hospital, Tongji University School of Medicine, 1239 Siping Road, Shanghai, 200120, China
| | - Jinfu Zhang
- Department of Reproductive Medicine, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 540 Xinhua Road, Shanghai, 200052, China.
| |
Collapse
|
18
|
Talibova G, Bilmez Y, Ozturk S. DNA double-strand break repair in male germ cells during spermatogenesis and its association with male infertility development. DNA Repair (Amst) 2022; 118:103386. [DOI: 10.1016/j.dnarep.2022.103386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022]
|
19
|
Fukunaga H, Yokoya A, Prise KM. A Brief Overview of Radiation-Induced Effects on Spermatogenesis and Oncofertility. Cancers (Basel) 2022; 14:cancers14030805. [PMID: 35159072 PMCID: PMC8834293 DOI: 10.3390/cancers14030805] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Spermatogenesis is one of the most important processes for the propagation of life; however, the testes’ ability to form sperm via this differentiation process is highly radiosensitive and easily impacted by exposure to environmental, occupational, or therapeutic radiation. Furthermore, the possibility that radiation effects on the gonads can be passed on from generation to generation should not be overlooked. This review focuses on the radiation-induced effects on spermatogenesis and the transgenerational effects. We also explore the potential of novel radiobiological approaches to improve male fertility preservation during radiotherapy. Abstract The genotoxicity of radiation on germ cells may be passed on to the next generation, thus its elucidation is not only a scientific issue but also an ethical, legal, and social issue in modern society. In this article, we briefly overview the effects of radiation on spermatogenesis and its associated genotoxicity, including the latest findings in the field of radiobiology. The potential role of transgenerational effects is still poorly understood, and further research in this area is desirable. Furthermore, from the perspective of oncofertility, we discuss the historical background and clinical importance of preserving male fertility during radiation treatment and the potential of microbeam radiotherapy. We hope that this review will contribute to stimulating further discussions and investigations for therapies for pediatric and adolescent/young adult patients.
Collapse
Affiliation(s)
- Hisanori Fukunaga
- Center for Environmental and Health Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Correspondence:
| | - Akinari Yokoya
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, Ibaraki 319-1106, Japan;
- Graduate School of Science and Engineering, Ibaraki University, Ibaraki 310-8512, Japan
| | - Kevin M. Prise
- Patrick G Johnstone Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK;
| |
Collapse
|
20
|
Yuan H, Sun J, Wang S, Xiang Z, Yang F, Yan Y, Duan Y, Li L, Wu X, Si W. Primary culture of germ cells that portray stem cell characteristics and recipient preparation for autologous transplantation in the rhesus monkey. J Cell Mol Med 2022; 26:1567-1578. [PMID: 35104031 PMCID: PMC8899175 DOI: 10.1111/jcmm.17197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 12/20/2022] Open
Abstract
Fertility preservation for prepubertal cancer patients prior to oncologic treatment is an emerging issue, and non‐human primates are considered to constitute suitable models due to the limited availability of human testicular tissues. However, the feasibility of spermatogonial stem cell (SSC) propagation in vitro and autologous testicular germ cell transplantation in vivo requires further exploration in monkeys. Herein, we characterized germ cells in macaque testes at 6 months (M), 18 M and 60 M of age, and effectively isolated the spermatogenic cells (including the spermatogonia) from macaque testes with high purity (over 80%) using combined approaches of STA‐PUT separation, Percoll gradients and differential plating. We also generated recipient monkey testes with ablated endogenous spermatogenesis using the alkylating agent busulfan in six macaques, and successfully mimicked autologous cell transplantation in the testes under ultrasonographic guidance. The use of trypan blue led to successful intratubular injection in 4 of 4 testes. Although SSCs in culture showed no significant propagation, we were able to maintain monkey testicular germ cells with stem cell characteristics for up to 3 weeks. Collectively, these data provided meaningful information for future fertility preservation and SSC studies on both non‐human primates and humans.
Collapse
Affiliation(s)
- Huaqin Yuan
- Cancer Center of Nanjing GaoChun People's Hospital, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jiachen Sun
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Shengnan Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ziyi Xiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Fan Yang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Yaping Yan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yanchao Duan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Lufan Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xin Wu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Si
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
21
|
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: 1.0] [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
|
22
|
Islam KN, Modi MM, Siegfried KR. The Zebrafish Meiotic Cohesin Complex Protein Smc1b Is Required for Key Events in Meiotic Prophase I. Front Cell Dev Biol 2021; 9:714245. [PMID: 34434933 PMCID: PMC8381726 DOI: 10.3389/fcell.2021.714245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
The eukaryotic structural maintenance of chromosomes (SMC) proteins are involved in key processes of chromosome structure and dynamics. SMC1β was identified as a component of the meiotic cohesin complex in vertebrates, which aids in keeping sister chromatids together prior to segregation in meiosis II and is involved in association of homologous chromosomes in meiosis I. The role of SMC1β in meiosis has primarily been studied in mice, where mutant male and female mice are infertile due to germ cell arrest at pachytene and metaphase II stages, respectively. Here, we investigate the function of zebrafish Smc1b to understand the role of this protein more broadly in vertebrates. We found that zebrafish smc1b is necessary for fertility and has important roles in meiosis, yet has no other apparent roles in development. Therefore, smc1b functions primarily in meiosis in both fish and mammals. In zebrafish, we showed that smc1b mutant spermatocytes initiated telomere clustering in leptotene, but failed to complete this process and progress into zygotene. Furthermore, mutant spermatocytes displayed a complete failure of synapsis between homologous chromosomes and homolog pairing only occurred at chromosome ends. Interestingly, meiotic DNA double strand breaks occurred in the absence of Smc1b despite failed pairing and synapsis. Overall, our findings point to an essential role of Smc1b in the leptotene to zygotene transition during zebrafish spermatogenesis. In addition, ovarian follicles failed to form in smc1b mutants, suggesting an essential role in female meiosis as well. Our results indicate that there are some key differences in Smc1b requirement in meiosis among vertebrates: while Smc1b is not required for homolog pairing and synapsis in mice, it is essential for these processes in zebrafish.
Collapse
Affiliation(s)
- Kazi Nazrul Islam
- Biology Department, University of Massachusetts Boston, Boston, MA, United States
| | - Maitri Mitesh Modi
- Biology Department, University of Massachusetts Boston, Boston, MA, United States
| | | |
Collapse
|
23
|
Varuzhanyan G, Ladinsky MS, Yamashita SI, Abe M, Sakimura K, Kanki T, Chan DC. Fis1 ablation in the male germline disrupts mitochondrial morphology and mitophagy, and arrests spermatid maturation. Development 2021; 148:271183. [PMID: 34355730 PMCID: PMC8380467 DOI: 10.1242/dev.199686] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/13/2021] [Indexed: 11/20/2022]
Abstract
Male germline development involves choreographed changes to mitochondrial number, morphology and organization. Mitochondrial reorganization during spermatogenesis was recently shown to require mitochondrial fusion and fission. Mitophagy, the autophagic degradation of mitochondria, is another mechanism for controlling mitochondrial number and physiology, but its role during spermatogenesis is largely unknown. During post-meiotic spermatid development, restructuring of the mitochondrial network results in packing of mitochondria into a tight array in the sperm midpiece to fuel motility. Here, we show that disruption of mouse Fis1 in the male germline results in early spermatid arrest that is associated with increased mitochondrial content. Mutant spermatids coalesce into multinucleated giant cells that accumulate mitochondria of aberrant ultrastructure and numerous mitophagic and autophagic intermediates, suggesting a defect in mitophagy. We conclude that Fis1 regulates mitochondrial morphology and turnover to promote spermatid maturation.
Collapse
Affiliation(s)
- Grigor Varuzhanyan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena CA 91125, USA
| | - Mark S Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena CA 91125, USA
| | - Shun-Ichi Yamashita
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Tomotake Kanki
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena CA 91125, USA
| |
Collapse
|
24
|
Aitken RJ, Bakos HW. Should we be measuring DNA damage in human spermatozoa? New light on an old question. Hum Reprod 2021; 36:1175-1185. [PMID: 33532854 DOI: 10.1093/humrep/deab004] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Assessments of sperm DNA damage are controversial because of perceived uncertainties over the relationship with pregnancy and the limited range of therapies available should positive results be returned. In this article, we highlight recent data supporting a chain of associations between oxidative stress in the male germ line, DNA damage in spermatozoa, defective DNA repair in the oocyte, the mutational load carried by the resulting embryo and the long-term health trajectory of the offspring. Any condition capable of generating oxidative damage in spermatozoa (age, obesity, smoking, prolonged abstinence, varicocele, chemical exposures, radiation etc.) is capable of influencing offspring health in this manner, creating a range of pathologies in the progeny including neuropsychiatric disorders and cancer. If sperm DNA damage is detected, there are several therapeutic interventions that can be introduced to improve DNA quality prior to the use of these cells in ART. We therefore argue that infertility specialists should be engaged in the diagnosis and remediation of sperm DNA damage as a matter of best practice, in order to minimize the risk of adverse health outcomes in children conceived using ART.
Collapse
Affiliation(s)
- R John Aitken
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Hassan W Bakos
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW 2308, Australia
- Monash IVF Group Limited, Level 2, 1 Fennell Street, Parramatta, NSW 2151 Australia
| |
Collapse
|
25
|
Mohan UP, P B TP, Iqbal STA, Arunachalam S. Mechanisms of doxorubicin-mediated reproductive toxicity - A review. Reprod Toxicol 2021; 102:80-89. [PMID: 33878324 DOI: 10.1016/j.reprotox.2021.04.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
The anticancer drug doxorubicin has been associated with several adverse side-effects including reproductive toxicity in both genders. The current review has complied the mechanisms of doxorubicin induced reproductive toxicity. The articles cited in the review were searched using Google Scholar, PubMed, Scopus, Science Direct. Doxorubicin treatment has been found to cause a decrease in testicular mass along with histopathological deformities, oligospermia and abnormalities in sperm morphology. Apart from severely affecting the normal physiological role of both Leydig cells and Sertoli cells, doxorubicin also causes chromosome abnormalities and affects DNA methylase enzyme. Testicular lipid metabolism has been found to be negatively affected by doxorubicin treatment resulting in altered profile of sphingolipids glycerophospholipids and neutral lipids. Dysregulation of 3β-hydroxysteroid dehydrogenase (3β-HSD) and 17β- hydroxysteroid dehydrogenase (17β-HSD) are strongly linked to testicular exposure to doxorubicin. Further, oxidative stress along with endoplasmic reticulum stress are also found to aggravate the male reproductive functioning in doxorubicin treated conditions. Several antioxidant enzymes such as superoxide dismutase, catalase, glutathione peroxidase (GPx) are downregulated by doxorubicin. It also disturbs the hormones of the hypothalamic-pituitary-gonadal (HPG)-axis including testosterone, luteinizing hormone, follicle stimulating hormone etc. In females, the drug disturbs folliculogenesis and oogenesis leading to failure of ovulation and uterine cycle. In rodent model the drug shortens pro-estrous and estrous phases. It was also found that doxorubicin causes mitochondrial dysfunction in oocytes with impaired calcium signaling along with ER stress. The goal of the present review is to comprehends various pathways due to which doxorubicin treatment promotes toxicity in male and female reproductive system.
Collapse
Affiliation(s)
- Uma Priya Mohan
- Centre for Cardiovascular and Adverse Drug Reactions, Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu, PIN 626126, India
| | | | | | - Sankarganesh Arunachalam
- Centre for Cardiovascular and Adverse Drug Reactions, Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamil Nadu, PIN 626126, India.
| |
Collapse
|
26
|
Cruz A, Sullivan DB, Doty KF, Hess RA, Canisso IF, Reddi PP. Acrosomal marker SP-10 (gene name Acrv1) for staging of the cycle of seminiferous epithelium in the stallion. Theriogenology 2020; 156:214-221. [PMID: 32758798 DOI: 10.1016/j.theriogenology.2020.06.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/28/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022]
Abstract
The acrosome plays a critical role in sperm-oocyte interactions during fertilization. SP-10 is an acrosomal matrix protein, which is evolutionarily conserved among mammals. The SP-10 antibody has been shown to be useful for staging the seminiferous cycle in the mouse and human. A canonical acrosomal marker; however, has never been used for staging in the horse. The objectives of the present study were to investigate the presence of SP-10 within the horse acrosome using an anti-mouse SP-10 antibody, to classify spermatids based on the shape of the acrosome, and then to use that information to assign stages of the cycle of the seminiferous epithelium. Testes from mature stallions with history of normospermic ejaculates were used for immunohistochemistry. We found that the mouse SP-10 antibody stained the horse acrosome vividly in testis cross-sections, indicating evolutionary conservation. Previous methods based on morphology alone without the aid of an antibody marker showed 8 stages in the horse seminiferous epithelium. Morphological detail of the acrosome afforded by the SP-10 marker in this study identified 16 steps of spermatids. This, in turn, led to the identification of 12 distinct stages in the cycle of the seminiferous epithelium of the horse wherein stage I shows recently formed round spermatids and stage XII includes meiotic divisions; a classification that is consistent with other animal models. The SP-10 antibody marks the acrosome in a way that enables researchers in the field to identify stages of spermatogenesis in the horse easily. In conclusion, we demonstrated that immunolabeling for SP-10 can be an objective approach to stage the cycle of the seminiferous epithelium in normospermic stallions; future studies will determine if SP-10 could be used to assess testicular dysfunction.
Collapse
Affiliation(s)
- Anamaria Cruz
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana Champaign, USA
| | - Derek B Sullivan
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana Champaign, USA
| | - Karen F Doty
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana Champaign, USA
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana Champaign, USA
| | - Igor F Canisso
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana Champaign, USA; Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois, Urbana Champaign, USA
| | - Prabhakara P Reddi
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana Champaign, USA.
| |
Collapse
|
27
|
Ceyhan Y, Zhang M, Guo J, Sandoval CG, Vacher J, Kaftanovskaya EM, Agoulnik AI, Agoulnik IU. Deletion of inositol polyphosphate 4-phosphatase type-II B affects spermatogenesis in mice. PLoS One 2020; 15:e0233163. [PMID: 32413098 PMCID: PMC7228085 DOI: 10.1371/journal.pone.0233163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Inositol polyphosphate-4-phosphatase type II (INPP4B) is a dual-specificity phosphatase that acts as a tumor suppressor in multiple cancers. INPP4B dephosphorylates phospholipids at the 4th position of the inositol ring and inhibits AKT and PKC signaling by hydrolyzing of PI(3,4)P2 and PI(4,5)P2, respectively. INPP4B protein phosphatase targets include phospho-tyrosines on Akt and phospho-serine and phospho-threonine on PTEN. INPP4B is highly expressed in testes, suggesting its role in testes development and physiology. The objective of this study was to determine whether Inpp4b deletion impacts testicular function in mice. In testis, Inpp4b expression was the highest in postmeiotic germ cells in both mice and men. The testes of Inpp4b knockout male mice were significantly smaller compared to the testes of wild-type (WT) males. Inpp4b-/- males produced fewer mature sperm cells compared to WT, and this difference increased with age and high fat diet (HFD). Reduction in early steroidogenic enzymes and luteinizing hormone (LH) receptor gene expression was detected, although androgen receptor (AR) protein level was similar in WT and Inpp4b-/- testes. Germ cell apoptosis was significantly increased in the knockout mice, while expression of meiotic marker γH2A.X was decreased. Our data demonstrate that INPP4B plays a role in maintenance of male germ cell differentiation and protects testis functions against deleterious effects of aging and high fat diet.
Collapse
Affiliation(s)
- Yasemin Ceyhan
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
| | - Manqi Zhang
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, United States of America
| | - Jingtao Guo
- Department of Oncological Sciences and Huntsman Cancer Institute, Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Surgery (Andrology/Urology), Center for Reconstructive Urology and Men’s Health, University of Utah Health Sciences Center, Salt Lake City, UT, United States of America
| | - Carlos G. Sandoval
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
| | - Jean Vacher
- Department of Medicine, Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Elena M. Kaftanovskaya
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
| | - Alexander I. Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
- Biomolecular Science Institute, Florida International University, Miami, FL, United States of America
| | - Irina U. Agoulnik
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
- Biomolecular Science Institute, Florida International University, Miami, FL, United States of America
- * E-mail:
| |
Collapse
|
28
|
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: 2.5] [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
|
29
|
MEIOSIN Directs the Switch from Mitosis to Meiosis in Mammalian Germ Cells. Dev Cell 2020; 52:429-445.e10. [DOI: 10.1016/j.devcel.2020.01.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/10/2019] [Accepted: 01/09/2020] [Indexed: 01/12/2023]
|
30
|
Xu C, Shah MA, Mipam T, Wu S, Yi C, Luo H, Yuan M, Chai Z, Zhao W, Cai X. Bovid microRNAs involved in the process of spermatogonia differentiation into spermatocytes. Int J Biol Sci 2020; 16:239-250. [PMID: 31929752 PMCID: PMC6949159 DOI: 10.7150/ijbs.38232] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 09/28/2019] [Indexed: 12/17/2022] Open
Abstract
The male infertility of cattleyak resulted from spermatogenic arrest has greatly restricted the effective utilization of the heterosis from crossbreeding of cattle and yak. Based on our previous studies, the significant divergences of the transcriptomic and proteomic sequencing between yak and cattleyak prompt us to investigate the critical roles of microRNAs in post-transcriptional regulation of gene expression during spermatogenesis. TUNEL-POD analysis presented sharply decreased spermatogenic cell types and the increased apoptotic spermatogonia in cattleyak. The STA-PUT velocity sedimentation was employed to obtain spermatogonia and spermatocytes from cattle, yak and cattleyak and these spermatogenic cells were verified by the morphological and phenotypic identification. MicroRNA microarray showed that 27 differentially expressed miRNAs were simultaneously identified both in cattleyak vs cattle and in cattleyak vs yak comparisons. Further analysis revealed that the down-regulation of bta-let-7 families, bta-miR-125 and bta-miR-23a might impair the RA-induced differentiation of spermatogonia. Target gene analysis for differentially expressed miRNAs revealed that miRNAs targeted major players involved in vesicle-mediated transport, regulation of protein kinase activity and Pathways in cancer. In addition, spermatogonia transfection analysis revealed that the down-regulation of bta-miR-449a in the cattleyak might block the transition of male germ cells from the mitotic cycle to the meiotic program. The present study provided valuable information for future elucidating the regulatory roles of miRNAs involved in spermatogenic arrest of cattleyak.
Collapse
Affiliation(s)
- Chuanfei Xu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China.,School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Mujahid Ali Shah
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - TserangDonko Mipam
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China
| | - Shixin Wu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Chuanping Yi
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Hui Luo
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Meng Yuan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China.,School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| |
Collapse
|
31
|
Qi L, Li J, Le W, Zhang J. Low-dose ionizing irradiation triggers apoptosis of undifferentiated spermatogonia in vivo and in vitro. Transl Androl Urol 2019; 8:591-600. [PMID: 32038955 DOI: 10.21037/tau.2019.10.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background The present study aimed to investigate the mechanism of low-dose ionizing radiation (IR) induced apoptosis of undifferentiated spermatogonia in vivo and in vitro. Methods Following 50 mGy IR, testicular tissues were collected from the adult DBA/2 mice at 1, 2 and 24 h; mice in the control group received pseudo-irradiation. Immunofluorescence (IF) staining and TUNEL were performed to assess DNA damage and apoptosis, respectively, in the irradiated testicular tissues. Furthermore, the spermatogonia were also irradiated in vitro, and the expression of apoptosis-related proteins was detected by Western blotting. TUNEL and flow cytometry were applied to assess cell apoptosis. Results γH2AX (a marker of DNA damage) was up-regulated in the seminiferous tubules at 1 and 2 h after IR, but it was reduced following the DNA repair. This was consistent with the finding that apoptosis of germline cells was present in the seminiferous tubules after IR, especially at 1 h (IF and TUNEL). Apoptosis was also present in the PLZF(+) spermatogonia, particularly at 1 h after IR. Apoptotic cells decreased with the increase in DNA repair time after IR. Moreover, the caspase-3 protein was expressed in the undifferentiated spermatogonia following IR. The expression of caspase-3, P53, Ku70 and DNA-PKcs in the cultured spermatogonia was also up-regulated following IR in vitro, but their expression decreased gradually over time after IR, which was supported by the findings from flow cytometry, and the apoptosis of spermatogonia peaked at 24 h post IR. Conclusions IR may induce the apoptosis of spermatogonia at early stage in vivo, but the apoptosis of spermatogonia secondary to IR occurs at a relatively later time point (24 h) in vitro mainly. The apoptosis of spermatogonia is improved over time after IR.
Collapse
Affiliation(s)
- Lixin Qi
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jiaxuan Li
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Wei Le
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Jinfu Zhang
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200050, China
| |
Collapse
|
32
|
Varuzhanyan G, Rojansky R, Sweredoski MJ, Graham RL, Hess S, Ladinsky MS, Chan DC. Mitochondrial fusion is required for spermatogonial differentiation and meiosis. eLife 2019; 8:51601. [PMID: 31596236 PMCID: PMC6805159 DOI: 10.7554/elife.51601] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 09/27/2019] [Indexed: 01/22/2023] Open
Abstract
Differentiating cells tailor their metabolism to fulfill their specialized functions. We examined whether mitochondrial fusion is important for metabolic tailoring during spermatogenesis. Acutely after depletion of mitofusins Mfn1 and Mfn2, spermatogenesis arrests due to failure to accomplish a metabolic shift during meiosis. This metabolic shift includes increased mitochondrial content, mitochondrial elongation, and upregulation of oxidative phosphorylation (OXPHOS). With long-term mitofusin loss, all differentiating germ cell types are depleted, but proliferation of stem-like undifferentiated spermatogonia remains unaffected. Thus, compared with undifferentiated spermatogonia, differentiating spermatogonia and meiotic spermatocytes have cell physiologies that require high levels of mitochondrial fusion. Proteomics in fibroblasts reveals that mitofusin-null cells downregulate respiratory chain complexes and mitochondrial ribosomal subunits. Similarly, mitofusin depletion in immortalized spermatocytes or germ cells in vivo results in reduced OXPHOS subunits and activity. We reveal that by promoting OXPHOS, mitofusins enable spermatogonial differentiation and a metabolic shift during meiosis.
Collapse
Affiliation(s)
- Grigor Varuzhanyan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Rebecca Rojansky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Michael J Sweredoski
- Proteome Exploration Laboratory of the Beckman Institute, California Institute of Technology, Pasadena, United States
| | - Robert Lj Graham
- Proteome Exploration Laboratory of the Beckman Institute, California Institute of Technology, Pasadena, United States
| | - Sonja Hess
- Proteome Exploration Laboratory of the Beckman Institute, California Institute of Technology, Pasadena, United States
| | - Mark S Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
33
|
Zhao D, Shen C, Gao T, Li H, Guo Y, Li F, Liu C, Liu Y, Chen X, Zhang X, Wu Y, Yu Y, Lin M, Yuan Y, Chen X, Huang X, Yang S, Yu J, Zhang J, Zheng B. Myotubularin related protein 7 is essential for the spermatogonial stem cell homeostasis via PI3K/AKT signaling. Cell Cycle 2019; 18:2800-2813. [PMID: 31478454 DOI: 10.1080/15384101.2019.1661174] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Myotubularin related protein 7 (MTMR7), a key member of the MTMR family, depicts phosphatase activity and is involved in myogenesis and tumor growth. We have previously identified MTMR7 in the proteomic profile of mouse spermatogonial stem cell (SSC) maturation and differentiation, implying that MTMR7 is associated with neonatal testicular development. In this study, to further explore the distribution and function of MTMR7 in mouse testis, we studied the effect of Mtmr7 knockdown on neonatal testicular development by testicular and SSC culture methods. Our results revealed that MTMR7 is exclusively located in early germ cells. Deficiency of MTMR7 by morpholino in neonatal testis caused excessive SSC proliferation, which was attributable to the aberrant PI3K/AKT signaling activation. Altogether, our study demonstrates that MTMR7 maintains SSC homeostasis by inhibiting PI3K/AKT signaling activation.
Collapse
Affiliation(s)
- Dan Zhao
- Fourth Affiliated Hospital of Jiangsu University , Zhenjiang , China
| | - Cong Shen
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou , China.,State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Tingting Gao
- Center of Clinical Reproductive Medicine, the Affiliated Changzhou Matemity and Child Health Care Hospital of Nanjing Medical University , Changzhou , China
| | - Hong Li
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou , China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China.,The Affiliated Wuxi Matemity and Child Health Care Hospital of Nanjing Medical University , Wuxi , China
| | - Feng Li
- Fourth Affiliated Hospital of Jiangsu University , Zhenjiang , China.,Reproductive Medicine Center, Northern Jiangsu Province Hospital , Yangzhou , China
| | - Chenchen Liu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Yuanyuan Liu
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou , China.,State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Xia Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , China
| | - Xi Zhang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Yangyang Wu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Yi Yu
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou , China
| | - Meng Lin
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Yan Yuan
- Human Reproductive and Genetic center, Affiliated Hospital of Jiangnan University , Wuxi , China
| | - Xiaofang Chen
- Fourth Affiliated Hospital of Jiangsu University , Zhenjiang , China
| | - Xiaoyan Huang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Shenmin Yang
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou , China
| | - Jun Yu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangsu University, Jiangsu University , Zhenjiang , China
| | - Jun Zhang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| | - Bo Zheng
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, the Affiliated Suzhou Hospital of Nanjing Medical University , Suzhou , China.,State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University , Nanjing , China
| |
Collapse
|
34
|
Ketchum CC, Larsen CD, McNeil A, Meyer-Ficca ML, Meyer RG. Early histone H4 acetylation during chromatin remodeling in equine spermatogenesis. Biol Reprod 2019; 98:115-129. [PMID: 29186293 DOI: 10.1093/biolre/iox159] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 11/23/2017] [Indexed: 01/08/2023] Open
Abstract
Chromatin remodeling during spermatogenesis culminates in the exchange of nucleosomes for transition proteins and protamines as an important part of spermatid development to give rise to healthy sperm. Comparative immunofluorescence analyses of equine and murine testis histological sections were used to characterize nucleoprotein exchange in the stallion. Histone H4 hyperacetylation is considered a key event of histone removal during the nucleoprotein transition to a protamine-based sperm chromatin structure. In the stallion, but not the mouse, H4 was already highly acetylated in lysine residues K5, K8, and K12 in round spermatids almost immediately after meiotic division. Time courses of transition protein 1 (TP1), protamine 1, H2A histone family member Z (H2AFZ), and testis-specific histone H2B variant (TH2B) expression in stallion spermatogenesis were similar to the mouse where protamine 1 and TP1 were only expressed in elongating spermatids much later in spermatid development. The additional acetylation of H4 in K16 position (H4K16ac) was detected during a brief phase of spermatid elongation in both species, concomitant with the phosphorylation of the noncanonical histone variant H2AFX resulting from DNA strand break-mediated DNA relaxation. The results suggest that H4K16 acetylation, which is dependent on DNA damage signaling, may be more important for nucleosome replacement in spermiogenesis than indicated by data obtained in rodents and highlight the value of the stallion as an alternative animal model for investigating human spermatogenesis. A revised classification system of the equine spermatogenic cycle for simplified comparison with the mouse is proposed to this end.
Collapse
Affiliation(s)
- Chelsea C Ketchum
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah Experimental Station, Utah State University, Logan, Utah, USA.,Utah Experimental Station, Utah State University, Logan, Utah, USA
| | - Casey D Larsen
- School of Veterinary Medicine (Washington-Idaho-Montana-Utah Regional Veterinary Medical Program, WIMU), Utah State University, Logan, Utah, USA
| | - Alexis McNeil
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah Experimental Station, Utah State University, Logan, Utah, USA
| | - Mirella L Meyer-Ficca
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah Experimental Station, Utah State University, Logan, Utah, USA.,School of Veterinary Medicine (Washington-Idaho-Montana-Utah Regional Veterinary Medical Program, WIMU), Utah State University, Logan, Utah, USA
| | - Ralph G Meyer
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah Experimental Station, Utah State University, Logan, Utah, USA.,Utah Experimental Station, Utah State University, Logan, Utah, USA.,School of Veterinary Medicine (Washington-Idaho-Montana-Utah Regional Veterinary Medical Program, WIMU), Utah State University, Logan, Utah, USA
| |
Collapse
|
35
|
Kilarkaje N, Al-Hussaini H. Type 1 diabetes upregulates metastasis-associated protein 1- phosphorylated histone 2AX signaling in the testis. Eur J Pharmacol 2019; 846:30-37. [DOI: 10.1016/j.ejphar.2019.01.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/12/2018] [Accepted: 01/15/2019] [Indexed: 01/02/2023]
|
36
|
Transition from a meiotic to a somatic-like DNA damage response during the pachytene stage in mouse meiosis. PLoS Genet 2019; 15:e1007439. [PMID: 30668564 PMCID: PMC6358097 DOI: 10.1371/journal.pgen.1007439] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 02/01/2019] [Accepted: 11/28/2018] [Indexed: 11/24/2022] Open
Abstract
Homologous recombination (HR) is the principal mechanism of DNA repair acting during meiosis and is fundamental for the segregation of chromosomes and the increase of genetic diversity. Nevertheless, non-homologous end joining (NHEJ) mechanisms can also act during meiosis, mainly in response to exogenously-induced DNA damage in late stages of first meiotic prophase. In order to better understand the relationship between these two repair pathways, we studied the response to DNA damage during male mouse meiosis after gamma radiation. We clearly discerned two types of responses immediately after treatment. From leptotene to early pachytene, exogenous damage triggered the massive presence of γH2AX throughout the nucleus, which was associated with DNA repair mediated by HR components (DMC1 and RAD51). This early pathway finished with the sequential removal of DMC1 and RAD51 and was no longer inducible at mid pachytene. However, from mid-pachytene to diplotene, γH2AX appeared as large discrete foci. This late repair pattern was mediated initially by NHEJ, involving Ku70 and XRCC4, which were constitutively present, and 53BP1, which appeared at sites of damage soon after irradiation. Nevertheless, 24 hours after irradiation, a HR pathway involving RAD51 but not DMC1 mostly replaced NHEJ. Additionally, we observed the occurrence of synaptonemal complex bridges between bivalents, most likely representing chromosome translocation events that may involve DMC1, RAD51 or 53BP1. Our results reinforce the idea that the early “meiotic” repair pathway that acts by default at the beginning of meiosis is replaced from mid-pachytene onwards by a “somatic-like” repair pattern. This shift might be important to resolve DNA damage (either endogenous or exogenous) that could not be repaired by the early meiotic mechanisms, for instance those in the sex chromosomes, which lack a homologous chromosome to repair with. This transition represents another layer of functional changes that occur in meiotic cells during mid pachytene, in addition to epigenetic reprograming, reactivation of transcription, changes in the gene expression profile and acquisition of competence to proceed to metaphase. DNA repair is critical for both somatic and meiotic cells. During meiosis, hundreds of DNA double strand breaks (DSBs) are introduced endogenously. To repair this damage, meiotic cells use a specialized version of the homologous recombination (HR) pathway that uses specific meiotic recombinases, such as DMC1, to promote repair with the homologous chromosome instead of the sister chromatid. This process is important to ensure chromosome segregation during meiosis and, as a side consequence, increases the genetic diversity of offspring. Nevertheless, under specific circumstances, meiotic cells can use other DNA repair mechanisms such as non-homologous end joining (NHEJ), which is error-prone. We investigated the response of mouse spermatocytes to increased DNA damage caused by gamma radiation, which is commonly used in cancer therapy. We found that the excess of DSBs produced by irradiation is processed by the meiotic HR recombination pathway in spermatocytes at the early stages of first meiotic prophase. However, this response is not inducible from the mid-pachytene stage onwards. From this point on, spermatocytes rely on a response that shares many features with that of somatic cells. In this response, the NHEJ pathway is first used to repair DNA damage but is subsequently replaced by a HR mechanism that does not use DMC1. Instead, it relies only on RAD51, which is known to function in both somatic and meiosis cells and, contrary to DMC1, has a preference for the sister chromatid. This switch from a meiotic to a somatic-like response is accompanied by a conspicuous change in the epigenetic response to DNA damage, reinforcing the idea that a functional transition occurs in meiotic cells during the mid-pachytene stage.
Collapse
|
37
|
Singh P, Aggarwal LM, Parry SA, Raman MJ. Radiation dosimetry and repair kinetics of DNA damage foci in mouse pachytene spermatocyte and round spermatid stages. Mutagenesis 2019; 33:231-239. [PMID: 30239864 DOI: 10.1093/mutage/gey007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 04/19/2018] [Indexed: 12/23/2022] Open
Abstract
Accurate quantification of DNA double strand breaks (DSB) in testicular germ cells is difficult because of cellular heterogeneity and the presence of endogenous γH2AX. Here, we used confocal microscopy to quantify DNA damage and repair kinetics following γ-irradiation (0.5-4 Gy) in three major mouse male germ cell stages, early and late pachytene spermatocytes and round spermatids (RSs), following a defined post irradiation time course. Dose-response curves showing linear best fit validated γH2AX focus as a rapid biodosimetric tool in these substages in response to whole body in vivo exposure. Stage specific foci yield/dose and repair kinetics demonstrated differential radiosensitivity and repair efficiency: early pachytenes (EP) repaired most rapidly and completely followed by late pachytene (LP) and RSs. Repair kinetics for all three stages followed 'exponential decay' in response to each radiation dose. In pachytenes immediate colocalisation of γH2AX and 53BP1, which participates in non-homologous end-joining repair pathway, was followed by dissociation from the major focal area of γH2AX by 4 h demonstrating ongoing DSB repair. These results confirm the differential radiosensitivity and repair kinetics of DSBs in male germ cells at different stages. Taken together, our results provide a simple and accurate method for assessing DNA damage and repair kinetics during spermatogenesis.
Collapse
Affiliation(s)
- Priti Singh
- Cytogenetics Laboratory, Department of Zoology, Centre of Advanced Study, India
| | - Lalit Mohan Aggarwal
- Department of Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Stephen A Parry
- Cornell Statistical Consulting Unit, Cornell University, Ithaca, NY, USA
| | - Mercy J Raman
- Cytogenetics Laboratory, Department of Zoology, Centre of Advanced Study, India
| |
Collapse
|
38
|
Teletin M, Vernet N, Yu J, Klopfenstein M, Jones JW, Féret B, Kane MA, Ghyselinck NB, Mark M. Two functionally redundant sources of retinoic acid secure spermatogonia differentiation in the seminiferous epithelium. Development 2019; 146:dev.170225. [PMID: 30487180 PMCID: PMC6340151 DOI: 10.1242/dev.170225] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022]
Abstract
In mammals, all-trans retinoic acid (ATRA) is instrumental to spermatogenesis. It is synthesized by two retinaldehyde dehydrogenases (RALDH) present in both Sertoli cells (SCs) and germ cells (GCs). In order to determine the relative contributions of each source of ATRA, we have generated mice lacking all RALDH activities in the seminiferous epithelium (SE). We show that both the SC- and GC-derived sources of ATRA cooperate to initiate and propagate spermatogenetic waves at puberty. In adults, they exert redundant functions and, against all expectations, the GC-derived source does not perform any specific roles despite contributing to two-thirds of the total amount of ATRA present in the testis. The production from SCs is sufficient to maintain the periodic expression of genes in SCs, as well and the cycle and wave of the SE, which account for the steady production of spermatozoa. The production from SCs is also specifically required for spermiation. Importantly, our study shows that spermatogonia differentiation depends upon the ATRA synthesized by RALDH inside the SE, whereas initiation of meiosis and expression of STRA8 by spermatocytes can occur without ATRA. Summary: All-trans retinoic acid made by Sertoli cells is instrumental to spermatogenesis and is specifically required for spermatid release.
Collapse
Affiliation(s)
- Marius Teletin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, F-67404 Illkirch Cedex, France.,Service de Biologie de la Reproduction, Hôpitaux Universitaires de Strasbourg (HUS), France
| | - Nadège Vernet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Muriel Klopfenstein
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Betty Féret
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Norbert B Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Manuel Mark
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, F-67404 Illkirch Cedex, France .,Service de Biologie de la Reproduction, Hôpitaux Universitaires de Strasbourg (HUS), France
| |
Collapse
|
39
|
Matsuda S, Wanibuchi S, Kasahara T. Quantitative analysis of γH2AX reveals distinct responses in multiple mouse organs after administration of mitomycin C or ethyl methanesulfonate. Mutagenesis 2018; 33:371-378. [DOI: 10.1093/mutage/gey040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/25/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Shun Matsuda
- Safety Evaluation Center, Ecology and Quality Management Division, CSR Division, FUJIFILM Corporation, 210 Nakanuma, Minamiashigara, Kanagawa, Japan
| | - Sayaka Wanibuchi
- Safety Evaluation Center, Ecology and Quality Management Division, CSR Division, FUJIFILM Corporation, 210 Nakanuma, Minamiashigara, Kanagawa, Japan
| | - Toshihiko Kasahara
- Safety Evaluation Center, Ecology and Quality Management Division, CSR Division, FUJIFILM Corporation, 210 Nakanuma, Minamiashigara, Kanagawa, Japan
| |
Collapse
|
40
|
Hwang G, Verver DE, Handel MA, Hamer G, Jordan PW. Depletion of SMC5/6 sensitizes male germ cells to DNA damage. Mol Biol Cell 2018; 29:3003-3016. [PMID: 30281394 PMCID: PMC6333175 DOI: 10.1091/mbc.e18-07-0459] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The structural maintenance of chromosomes complex SMC5/6 is thought to be essential for DNA repair and chromosome segregation during mitosis and meiosis. To determine the requirements of the SMC5/6 complex during mouse spermatogenesis we combined a conditional knockout allele for Smc5, with four germ cell–specific Cre-recombinase transgenes, Ddx4-Cre, Stra8-Cre, Spo11-Cre, and Hspa2-Cre, to mutate Smc5 in spermatogonia, in spermatocytes before meiotic entry, during early meiotic stages, and during midmeiotic stages, respectively. Conditional mutation of Smc5 resulted in destabilization of the SMC5/6 complex. Despite this, we observed only mild defects in spermatogenesis. Mutation of Smc5 mediated by Ddx4-Cre and Stra8-Cre resulted in partial loss of preleptotene spermatocytes; however, spermatogenesis progresses and mice are fertile. Mutation of Smc5 via Spo11-Cre or Hspa2-Cre did not result in detectable defects of spermatogenesis. Upon exposure to gamma irradiation or etoposide treatment, each conditional Smc5 mutant demonstrated an increase in the number of enlarged round spermatids with multiple acrosomes and supernumerary chromosome content. We propose that the SMC5/6 complex is not acutely required for premeiotic DNA replication and meiotic progression during mouse spermatogenesis; however, when germ cells are challenged by exogenous DNA damage, the SMC5/6 complex ensures genome integrity, and thus, fertility.
Collapse
Affiliation(s)
- G Hwang
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| | - D E Verver
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - M A Handel
- The Jackson Laboratory, Bar Harbor, ME 04609
| | - G Hamer
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - P W Jordan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
41
|
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: 3.0] [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
|
42
|
Jangiam W, Udomtanakunchai C, Reungpatthanaphong P, Tungjai M, Honikel L, Gordon CR, Rithidech KN. Late Effects of Low-Dose Radiation on the Bone Marrow, Lung, and Testis Collected From the Same Exposed BALB/cJ Mice. Dose Response 2018; 16:1559325818815031. [PMID: 30622448 PMCID: PMC6302279 DOI: 10.1177/1559325818815031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 01/15/2023] Open
Abstract
We used 3 biological metrics highly relevant to health risks, that is, cell death, inflammation, and global DNA methylation, to determine the late effects of low doses (0.05 or 0.1 Gy) of 137Cs γ rays on the bone marrow, lung, and testis collected at 6 months post-irradiation from the same exposed BALB/cJ mouse. This integrative approach has not been used for such a purpose. Mice exposed to 0 or 1 Gy of radiation served as a sham or positive control group, respectively. The results could deliver information for better health risk assessment across tissues, including better scientific basis for radiation protection and clinical application. We found no changes in the levels of all studied biological metrics (except a significant increase in the levels of an anti-inflammatory cytokine, ie, interleukin 10) in tissues of 0.05-Gy exposed mice, when compared to those in sham controls. In contrast, significantly increased levels of cell death and inflammation, including a significant loss of global 5-hydroxymethylcytosine, were found in all tissues of the same mice exposed to 0.1 or 1.0 Gy. Our data demonstrated not only no harm but also hormesis in the 0.05-Gy exposed mice. However, the hormetic effect appears to be dependent on biological metrics and tissue.
Collapse
Affiliation(s)
- Witawat Jangiam
- Pathology Department, Stony Brook University, Stony Brook, NY, USA
- Department of Chemical Engineering, Burapha University, Chonburi, Thailand
| | - Chatchanok Udomtanakunchai
- Pathology Department, Stony Brook University, Stony Brook, NY, USA
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Paiboon Reungpatthanaphong
- Pathology Department, Stony Brook University, Stony Brook, NY, USA
- Department of Applied Radiation and Isotopes, Faculty of Sciences, Kasetsart University, Bangkok, Thailand
| | - Montree Tungjai
- Pathology Department, Stony Brook University, Stony Brook, NY, USA
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Louise Honikel
- Pathology Department, Stony Brook University, Stony Brook, NY, USA
| | - Chris R. Gordon
- Pathology Department, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
43
|
Reza AMMT, Choi YJ, Han SG, Song H, Park C, Hong K, Kim JH. Roles of microRNAs in mammalian reproduction: from the commitment of germ cells to peri-implantation embryos. Biol Rev Camb Philos Soc 2018; 94:415-438. [PMID: 30151880 PMCID: PMC7379200 DOI: 10.1111/brv.12459] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are active regulators of numerous biological and physiological processes including most of the events of mammalian reproduction. Understanding the biological functions of miRNAs in the context of mammalian reproduction will allow a better and comparative understanding of fertility and sterility in male and female mammals. Herein, we summarize recent progress in miRNA‐mediated regulation of mammalian reproduction and highlight the significance of miRNAs in different aspects of mammalian reproduction including the biogenesis of germ cells, the functionality of reproductive organs, and the development of early embryos. Furthermore, we focus on the gene expression regulatory feedback loops involving hormones and miRNA expression to increase our understanding of germ cell commitment and the functioning of reproductive organs. Finally, we discuss the influence of miRNAs on male and female reproductive failure, and provide perspectives for future studies on this topic.
Collapse
Affiliation(s)
- Abu Musa Md Talimur Reza
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Yun-Jung Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|
44
|
Zhao J, Zhao J, Xu G, Wang Z, Gao J, Cui S, Liu J. Deletion of Spata2 by CRISPR/Cas9n causes increased inhibin alpha expression and attenuated fertility in male mice. Biol Reprod 2018; 97:497-513. [PMID: 29025062 DOI: 10.1093/biolre/iox093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 08/25/2017] [Indexed: 12/22/2022] Open
Abstract
As somatic cells in the testis seminiferous tubule, Sertoli cells provide the medium for spermatogenesis. One of the important functions of Sertoli cells is synthesizing and secreting cell factors to affect the production of sperm; however, much of those molecular regulation mechanisms remain unknown. Here, we confirm the localization of protein SPATA2 (spermatogenesis-associated protein 2), which had previously been shown to be highly expressed in Sertoli cells of the adult mouse testis. To further conduct a functional study, we generated SPATA2 global knockout mice via use of the CRISPR/Cas9n gene editing technology. The 120-day-old knockout mice testes showed almost a 40% decrease in size and weight and variations in the histomorphology of the seminiferous epithelium, with a 40% decrease in sperm count. Further examination revealed that the proliferation of germ cells in the seminiferous tubules was attenuated by 28%. In addition, we found that SPATA2 deletion led to an approximately 70% increase in the inhibin alpha-subunit mRNA and protein level in the testes compared to that of wild-type mice. Our data revealed the impact of SPATA2 on male fertility and suggested that SPATA2 ensures the normal secretory function of Sertoli cells.
Collapse
Affiliation(s)
- Jie Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jianjun Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Guojin Xu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Zhijuan Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jie Gao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Sheng Cui
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jiali Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| |
Collapse
|
45
|
Motoyama S, Takeiri A, Tanaka K, Harada A, Matsuzaki K, Taketo J, Matsuo S, Fujii E, Mishima M. Advantages of evaluating γH2AX induction in non-clinical drug development. Genes Environ 2018; 40:10. [PMID: 29785231 PMCID: PMC5950202 DOI: 10.1186/s41021-018-0098-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/26/2018] [Indexed: 01/17/2023] Open
Abstract
γH2AX, the phosphorylated form of a histone variant H2AX at Ser 139, is already widely used as a biomarker to research the fundamental biology of DNA damage and repair and to assess the risk of environmental chemicals, pollutants, radiation, and so on. It is also beginning to be used in the early non-clinical stage of pharmaceutical drug development as an in vitro tool for screening and for mechanistic studies on genotoxicity. Here, we review the available information on γH2AX-based test systems that can be used to develop drugs and present our own experience of practically applying these systems during the non-clinical phase of drug development. Furthermore, the potential application of γH2AX as a tool for in vivo non-clinical safety studies is also discussed.
Collapse
Affiliation(s)
- Shigeki Motoyama
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Akira Takeiri
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Kenji Tanaka
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Asako Harada
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Kaori Matsuzaki
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Junko Taketo
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Saori Matsuo
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Etsuko Fujii
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Masayuki Mishima
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| |
Collapse
|
46
|
Le W, Qi L, Xu C, Xiang Z, Mao Z, Zhang J, Xu J, Wu D. Preliminary study of the homologous recombination repair pathway in mouse spermatogonial stem cells. Andrology 2018; 6:488-497. [PMID: 29577652 DOI: 10.1111/andr.12481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 12/19/2022]
Abstract
The present study was designed to detect DNA repair response through the homologous recombination pathway in mouse spermatogonial stem cells. Mouse spermatogonial stem cells (mSSCs) were obtained from the adult DBA/2 mouse testes by MACS sorting. mSSCs and mice animals were divided into four groups (30 min, 2, 24 h, control) and treated with ionizing irradiation while the control group received pseudo-irradiation. Proteins involved in the homologous recombination pathway (γH2AX, ATM, RAD51, CtIP, and RPA2) were assessed in mSSCs both in vitro and in vivo. Moreover, the non-homologous end-joining or homologous recombination (NHEJ/HR) reporter plasmids were transfected into mSSCs to assess NHEJ/HR pathway activity after DNA double-strand break (DSB). γH2AX, a classical DNA DSB marker, was absent in mSSCs both in vivo and in vitro after DSB repair, but was highly expressed in other tissue stem cells. In addition, ATM and phosphorylated ATM (p-ATM) were involved in DNA damage response (DDR) in mSSCs. p-ATM foci were overexpressed immediately after irradiation (30 min and 2 h), but gradually decreased over the repair time. The HR pathway-related proteins, CtIP and RPA2 were negatively regulated after treatment in Western blot (WB). NHEJ/HR reporter plasmid transfection indicated that the HR pathway played a minor role in mSSCs during DDR, consistent with the WB findings. This study demonstrates that mSSCs may have a unique response to DNA damage since crucial proteins involved in HR pathway were negatively regulated after DSB. In addition, the expression level of p-ATM, but not γH2AX, was increased after DSB, suggesting that DNA damage repair in mSSCs might be a γH2AX-independent response. Furthermore, the HR pathway may play a minor role during DDR in mSSCs.
Collapse
Affiliation(s)
- W Le
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - L Qi
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - C Xu
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Z Xiang
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Z Mao
- Tongji University School of Life Sciences and Technology, Shanghai, China
| | - J Zhang
- Department of Reproductive Medicine and Andrology, Tongren Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - J Xu
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - D Wu
- Department of Urology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Chianese R, Viggiano A, Urbanek K, Cappetta D, Troisi J, Scafuro M, Guida M, Esposito G, Ciuffreda LP, Rossi F, Berrino L, Fasano S, Pierantoni R, De Angelis A, Meccariello R. Chronic exposure to low dose of bisphenol A impacts on the first round of spermatogenesis via SIRT1 modulation. Sci Rep 2018; 8:2961. [PMID: 29440646 PMCID: PMC5811609 DOI: 10.1038/s41598-018-21076-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 01/18/2018] [Indexed: 12/22/2022] Open
Abstract
Spermatogenesis depends on endocrine, autocrine and paracrine communications along the hypothalamus-pituitary-gonad axis. Bisphenol A (BPA), an estrogen-mimic endocrine disrupting chemical, is an environmental contaminant used to manufacture polycarbonate plastics and epoxy resins with toxic effects for male reproduction. Here we investigated whether the chronic exposure to low BPA doses affects spermatogenesis through the modulation of SIRT1, a NAD+-dependent deacetylase involved in the progression of spermatogenesis, with outcomes on apoptosis, oxidative stress, metabolism and energy homeostasis. BPA exposure via placenta first, and lactation and drinking water later, affected the body weight gain in male offspring at 45 postnatal days and the first round of spermatogenesis, with impairment of blood testis barrier, reactive oxygen species production, DNA damage and decreased expression of SIRT1. The analysis of SIRT1 downstream molecular pathways revealed the increase of acetyl-p53Lys370, γH2AX foci, the decrease of oxidative stress defenses and the higher apoptotic rate in the testis of treated animals, with partial rescue at sex maturation. In conclusion, SIRT1 pathways disruption after BPA exposure can have serious consequences on the first round of spermatogenesis.
Collapse
Affiliation(s)
- Rosanna Chianese
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Andrea Viggiano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende 1, 84081, Baronissi, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Jacopo Troisi
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende 1, 84081, Baronissi, Italy
- Theoreo srl Spin-off Company of the University of Salerno, Salerno, Italy
| | - Marika Scafuro
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Maurizio Guida
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende 1, 84081, Baronissi, Italy
- Theoreo srl Spin-off Company of the University of Salerno, Salerno, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Silvia Fasano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Riccardo Pierantoni
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Rosaria Meccariello
- Department of Movement Sciences and Wellbeing, University of Naples "Parthenope", Via Medina 40, 80133, Naples, Italy.
| |
Collapse
|
48
|
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: 8.3] [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
|
49
|
Yu C, Li Y, Liu M, Gao M, Li C, Yan H, Li C, Sun L, Mo L, Wu C, Qi X, Ren J. Critical Role of Hepatic Cyp450s in the Testis-Specific Toxicity of (5R)-5-Hydroxytriptolide in C57BL/6 Mice. Front Pharmacol 2017; 8:832. [PMID: 29209210 PMCID: PMC5702336 DOI: 10.3389/fphar.2017.00832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/31/2017] [Indexed: 12/17/2022] Open
Abstract
Low solubility, tissue accumulation, and toxicity are chief obstacles to developing triptolide derivatives, so a better understanding of the pharmacokinetics and toxicity of triptolide derivatives will help with these limitations. To address this, we studied pharmacokinetics and toxicity of (5R)-5-hydroxytriptolide (LLDT-8), a novel triptolide derivative immunosuppressant in a conditional knockout (KO) mouse model with liver-specific deletion of CYP450 reductase. Compared to wild type (WT) mice, after LLDT-8 treatment, KO mice suffered severe testicular toxicity (decreased testicular weight, spermatocytes apoptosis) unlike WT mice. Moreover, KO mice had greater LLDT-8 exposure as confirmed with elevated AUC and Cmax, increased drug half-life, and greater tissue distribution. γ-H2AX, a marker of meiosis process, its localization and protein level in testis showed a distinct meiosis block induced by LLDT-8. RNA polymerase II (Pol II), an essential factor for RNA storage and synapsis in spermatogenesis, decreased in testes of KO mice after LLDT-8 treatment. Germ-cell line based assays confirmed that LLDT-8 selectively inhibited Pol II in spermatocyte-like cells. Importantly, the analysis of androgen receptor (AR) related genes showed that LLDT-8 did not change AR-related signaling in testes. Thus, hepatic CYP450s were responsible for in vivo metabolism and clearance of LLDT-8 and aggravated testicular injury may be due to increased LLDT-8 exposure in testis and subsequent Pol II reduction.
Collapse
Affiliation(s)
- Cunzhi Yu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yu Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mingxia Liu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Man Gao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chenggang Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hong Yan
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunzhu Li
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lihan Sun
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Liying Mo
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Meyer RG, Ketchum CC, Meyer-Ficca ML. Heritable sperm chromatin epigenetics: a break to remember†. Biol Reprod 2017; 97:784-797. [DOI: 10.1093/biolre/iox137] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023] Open
|