1
|
Zhang YW, Wu SX, Wang GW, Wan RD, Yang QE. Single-cell analysis identifies critical regulators of spermatogonial development and differentiation in cattle-yak bulls. J Dairy Sci 2024; 107:7317-7336. [PMID: 38642661 DOI: 10.3168/jds.2023-24442] [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: 11/22/2023] [Accepted: 03/11/2024] [Indexed: 04/22/2024]
Abstract
Spermatogenesis is a continuous process in which functional sperm are produced through a series of mitotic and meiotic divisions and morphological changes in germ cells. The aberrant development and fate transitions of spermatogenic cells cause hybrid sterility in mammals. Cattle-yak, a hybrid animal between taurine cattle (Bos taurus) and yak (Bos grunniens), exhibits male-specific sterility due to spermatogenic failure. In the present study, we performed single-cell RNA sequencing analysis to identify differences in testicular cell composition and the developmental trajectory of spermatogenic cells between yak and cattle-yak. The composition and molecular signatures of spermatogonial subtypes were dramatically different between these 2 animals, and the expression of genes associated with stem cell maintenance, cell differentiation and meiotic entry was altered in cattle-yak, indicating the impairment of undifferentiated spermatogonial fate decisions. Cell communication analysis revealed that signaling within different spermatogenic cell subpopulations was weakened, and progenitor spermatogonia were unable to or delayed receiving and sending signals for transformation to the next stage in cattle-yak. Simultaneously, the communication between niche cells and germ cells was also abnormal. Collectively, we obtained the expression profiles of transcriptome signatures of different germ cells and testicular somatic cell populations at the single-cell level and identified critical regulators of spermatogonial differentiation and meiosis in yak and sterile cattle-yak. The findings of this study shed light on the genetic mechanisms that lead to hybrid sterility and speciation in bovid species.
Collapse
Affiliation(s)
- Yi-Wen Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shi-Xin Wu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guo-Wen Wang
- Qinghai Academy of Animal Husbandry and Veterinary Sciences, Xining, Qinghai 810016, China
| | - Rui-Dong Wan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi-En Yang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810000, China; University of Chinese Academy of Sciences, Beijing 100049, China; Qinghai Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810001, China.
| |
Collapse
|
2
|
Yang YF, Ma HL, Wang X, Nie M, Mao JF, Wu XY. Clinical manifestations and spermatogenesis outcomes in Chinese patients with congenital hypogonadotropic hypogonadism caused by inherited or de novo FGFR1 mutations. Asian J Androl 2024; 26:426-432. [PMID: 38227553 PMCID: PMC11280213 DOI: 10.4103/aja202366] [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: 12/14/2022] [Accepted: 11/02/2023] [Indexed: 01/18/2024] Open
Abstract
Fibroblast growth factor receptor 1 ( FGFR1 ) mutations are associated with congenital hypogonadotropic hypogonadism (CHH) through inheritance or spontaneous occurrence. We detected FGFR1 mutations in a Chinese cohort of 210 CHH patients at Peking Union Medical College Hospital (Beijing, China) using next-generation and Sanger sequencing. We assessed missense variant pathogenicity using six bioinformatics tools and compared clinical features and treatment outcomes between inherited and de novo mutation groups. Among 19 patients with FGFR1 mutations, three were recurrent, and 16 were novel variants. Sixteen of the novel mutations were likely pathogenic according to the American College of Medical Genetics and Genomics (ACMG) guidelines, with the prevalent P366L variant. The majority of FGFR1 mutations was inherited (57.9%), with frameshift mutations exclusive to the de novo mutation group. The inherited mutation group had a lower incidence of cryptorchidism, short stature, and skeletal deformities. In the inherited mutation group, luteinizing hormone (LH) levels were 0.5 IU l -1 , follicle-stimulating hormone (FSH) levels were 1.0 IU l -1 , and testosterone levels were 1.3 nmol l -1 . In contrast, the de novo group had LH levels of 0.2 IU l -1 , FSH levels of 0.5 IU l -1 , and testosterone levels of 0.9 nmol l -1 , indicating milder hypothalamus-pituitary-gonadal axis (HPGA) functional deficiency in the inherited group. The inherited mutation group showed a tendency toward higher spermatogenesis rates. In conclusion, this study underscores the predominance of inherited FGFR1 mutations and their association with milder HPGA dysfunction compared to de novo mutations, contributing to our understanding of the genetic and clinical aspects of FGFR1 mutations.
Collapse
Affiliation(s)
- Yu-Fan Yang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Hai-Lu Ma
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Xi Wang
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Min Nie
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Jiang-Feng Mao
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Xue-Yan Wu
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
3
|
Bernstein IR, Nixon B, Lyons JM, Damyanova KB, De Oliveira CS, Mabotuwana NS, Stanger SJ, Kaiko GE, Ying TH, Oatley JM, Skillen NM, Lochrin AJ, Peters JL, Lord T. The hypoxia-inducible factor EPAS1 is required for spermatogonial stem cell function in regenerative conditions. iScience 2023; 26:108424. [PMID: 38077147 PMCID: PMC10700845 DOI: 10.1016/j.isci.2023.108424] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/21/2023] [Accepted: 11/07/2023] [Indexed: 05/10/2025] Open
Abstract
In this study we explored the role of hypoxia and the hypoxia-inducible transcription factor EPAS1 in regulating spermatogonial stem cell (SSC) function in the mouse testis. We have demonstrated that SSCs reside in hypoxic microenvironments in the testis through utilization of the oxygen-sensing probe pimonidazole, and by confirming the stable presence of EPAS1, which is degraded at >5% O2. Through the generation of a germline-specific Epas1 knockout mouse line, and through modulation of EPAS1 levels in primary cultures of spermatogonia with the small drug molecule Daprodustat, we have demonstrated that EPAS1 is required for robust SSC function in regenerative conditions (post-transplantation and post-chemotherapy), via the regulation of key cellular processes such as metabolism. These findings shed light on the relationship between hypoxia and male fertility and will potentially facilitate optimization of in vitro culture conditions for infertility treatment pipelines using SSCs, such as those directed at pediatric cancer survivors.
Collapse
Affiliation(s)
- Ilana R. Bernstein
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, Infertility and Reproduction Program, New Lambton Heights, NSW 2305, Australia
| | - Jess M. Lyons
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Katerina B. Damyanova
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Camila S. De Oliveira
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Nishani S. Mabotuwana
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Simone J. Stanger
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Gerard E. Kaiko
- Hunter Medical Research Institute, Immune Health Research Program, New Lambton Heights, NSW 2305, Australia
- School of Biomedical Sciences and Pharmacy, College of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Tan Hui Ying
- Hunter Medical Research Institute, Immune Health Research Program, New Lambton Heights, NSW 2305, Australia
- School of Biomedical Sciences and Pharmacy, College of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Jon M. Oatley
- School of Molecular Biosciences, Centre for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Nicole M. Skillen
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Alyssa J. Lochrin
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Jera L. Peters
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, Infertility and Reproduction Program, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
4
|
Dunleavy JEM, Graffeo M, Wozniak K, O'Connor AE, Merriner DJ, Nguyen J, Schittenhelm RB, Houston BJ, O'Bryan MK. The katanin A-subunits KATNA1 and KATNAL1 act co-operatively in mammalian meiosis and spermiogenesis to achieve male fertility. Development 2023; 150:dev201956. [PMID: 37882691 PMCID: PMC10690054 DOI: 10.1242/dev.201956] [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: 05/07/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
Katanins, a class of microtubule-severing enzymes, are potent M-phase regulators in oocytes and somatic cells. How the complex and evolutionarily crucial, male mammalian meiotic spindle is sculpted remains unknown. Here, using multiple single and double gene knockout mice, we reveal that the canonical katanin A-subunit KATNA1 and its close paralogue KATNAL1 together execute multiple aspects of meiosis. We show KATNA1 and KATNAL1 collectively regulate the male meiotic spindle, cytokinesis and midbody abscission, in addition to diverse spermatid remodelling events, including Golgi organisation, and acrosome and manchette formation. We also define KATNAL1-specific roles in sperm flagellum development, manchette regulation and sperm-epithelial disengagement. Finally, using proteomic approaches, we define the KATNA1, KATNAL1 and KATNB1 mammalian testis interactome, which includes a network of cytoskeletal and vesicle trafficking proteins. Collectively, we reveal that the presence of multiple katanin A-subunit paralogs in mammalian spermatogenesis allows for 'customised cutting' via neofunctionalisation and protective buffering via gene redundancy.
Collapse
Affiliation(s)
- Jessica E. M. Dunleavy
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Maddison Graffeo
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kathryn Wozniak
- Monash Biomedicine Discovery Institute and The Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Anne E. O'Connor
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - D. Jo Merriner
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Joseph Nguyen
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Ralf B. Schittenhelm
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Brendan J. Houston
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Moira K. O'Bryan
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
5
|
Walton KL, Goney MP, Peppas Z, Stringer JM, Winship A, Hutt K, Goodchild G, Maskey S, Chan KL, Brûlé E, Bernard DJ, Stocker WA, Harrison CA. Inhibin Inactivation in Female Mice Leads to Elevated FSH Levels, Ovarian Overstimulation, and Pregnancy Loss. Endocrinology 2022; 163:6543938. [PMID: 35255139 PMCID: PMC9272799 DOI: 10.1210/endocr/bqac025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 11/21/2022]
Abstract
Inhibins are members of the transforming growth factor-β family, composed of a common α-subunit disulfide-linked to 1 of 2 β-subunits (βA in inhibin A or βB in inhibin B). Gonadal-derived inhibin A and B act in an endocrine manner to suppress the synthesis of follicle-stimulating hormone (FSH) by pituitary gonadotrope cells. Roles for inhibins beyond the pituitary, however, have proven difficult to delineate because deletion of the inhibin α-subunit gene (Inha) results in unconstrained expression of activin A and activin B (homodimers of inhibin β-subunits), which contribute to gonadal tumorigenesis and lethal cachectic wasting. Here, we generated mice with a single point mutation (Arg233Ala) in Inha that prevents proteolytic processing and the formation of bioactive inhibin. In vitro, this mutation blocked inhibin maturation and bioactivity, without perturbing activin production. Serum FSH levels were elevated 2- to 3-fold in InhaR233A/R233A mice due to the loss of negative feedback from inhibins, but no pathological increase in circulating activins was observed. While inactivation of inhibin A and B had no discernible effect on male reproduction, female InhaR233A/R233A mice had increased FSH-dependent follicle development and enhanced natural ovulation rates. Nevertheless, inhibin inactivation resulted in significant embryo-fetal resorptions and severe subfertility and was associated with disrupted maternal ovarian function. Intriguingly, heterozygous Inha+/R233A females had significantly enhanced fecundity, relative to wild-type littermates. These studies have revealed novel effects of inhibins in the establishment and maintenance of pregnancy and demonstrated that partial inactivation of inhibin A/B is an attractive approach for enhancing female fertility.
Collapse
Affiliation(s)
- Kelly L Walton
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
- Correspondence: Kelly L Walton, PhD, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia 4072.
| | - Monica P Goney
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Zoe Peppas
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Jessica M Stringer
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Amy Winship
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Karla Hutt
- Department of Anatomy and Developmental Biology Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Georgia Goodchild
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Shreya Maskey
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Karen L Chan
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Emilie Brûlé
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Daniel J Bernard
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - William A Stocker
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Australia
| | - Craig A Harrison
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Correspondence: Craig A Harrison, PhD, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia 3168.
| |
Collapse
|
6
|
Houston BJ, O'Connor AE, Wang D, Goodchild G, Merriner DJ, Luan H, Conrad DF, Nagirnaja L, Aston KI, Kliesch S, Wyrwoll MJ, Friedrich C, Tüttelmann F, Harrison C, O'Bryan MK, Walton K. Human INHBB Gene Variant (c.1079T>C:p.Met360Thr) Alters Testis Germ Cell Content, but Does Not Impact Fertility in Mice. Endocrinology 2022; 163:6504015. [PMID: 35022746 DOI: 10.1210/endocr/bqab269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Indexed: 11/19/2022]
Abstract
Testicular-derived inhibin B (α/β B dimers) acts in an endocrine manner to suppress pituitary production of follicle-stimulating hormone (FSH), by blocking the actions of activins (β A/B/β A/B dimers). Previously, we identified a homozygous genetic variant (c.1079T>C:p.Met360Thr) arising from uniparental disomy of chromosome 2 in the INHBB gene (β B-subunit of inhibin B and activin B) in a man suffering from infertility (azoospermia). In this study, we aimed to test the causality of the p.Met360Thr variant in INHBB and testis function. Here, we used CRISPR/Cas9 technology to generate InhbbM364T/M364T mice, where mouse INHBB p.Met364 corresponds with human p.Met360. Surprisingly, we found that the testes of male InhbbM364T/M364T mutant mice were significantly larger compared with those of aged-matched wildtype littermates at 12 and 24 weeks of age. This was attributed to a significant increase in Sertoli cell and round spermatid number and, consequently, seminiferous tubule area in InhbbM364T/M364T males compared to wildtype males. Despite this testis phenotype, male InhbbM364T/M364T mutant mice retained normal fertility. Serum hormone analyses, however, indicated that the InhbbM364T variant resulted in reduced circulating levels of activin B but did not affect FSH production. We also examined the effect of this p.Met360Thr and an additional INHBB variant (c.314C>T: p.Thr105Met) found in another infertile man on inhibin B and activin B in vitro biosynthesis. We found that both INHBB variants resulted in a significant disruption to activin B in vitro biosynthesis. Together, this analysis supports that INHBB variants that limit activin B production have consequences for testis composition in males.
Collapse
Affiliation(s)
- Brendan J Houston
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Australia
| | - Anne E O'Connor
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Australia
| | - Degang Wang
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- The Affiliated Zhongshan Boai Hospital of Southern Medical University, Guangdong, China
| | - Georgia Goodchild
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - D Jo Merriner
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
| | - Haitong Luan
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Don F Conrad
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
- Genetics of Male Infertility Initiative, GEMINI, Portland, OR, USA
| | - Liina Nagirnaja
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
- Genetics of Male Infertility Initiative, GEMINI, Portland, OR, USA
| | - Kenneth I Aston
- Genetics of Male Infertility Initiative, GEMINI, Portland, OR, USA
- Department of Surgery (Urology Division) University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sabine Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Margot J Wyrwoll
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Corinna Friedrich
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Craig Harrison
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Moira K O'Bryan
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, Australia
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Australia
| | - Kelly Walton
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| |
Collapse
|
7
|
Dunleavy JEM, O'Connor AE, Okuda H, Merriner DJ, O'Bryan MK. KATNB1 is a master regulator of multiple katanin enzymes in male meiosis and haploid germ cell development. Development 2021; 148:273717. [PMID: 34822718 DOI: 10.1242/dev.199922] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Katanin microtubule-severing enzymes are crucial executers of microtubule regulation. Here, we have created an allelic loss-of-function series of the katanin regulatory B-subunit KATNB1 in mice. We reveal that KATNB1 is the master regulator of all katanin enzymatic A-subunits during mammalian spermatogenesis, wherein it is required to maintain katanin A-subunit abundance. Our data shows that complete loss of KATNB1 from germ cells is incompatible with sperm production, and we reveal multiple new spermatogenesis functions for KATNB1, including essential roles in male meiosis, acrosome formation, sperm tail assembly, regulation of both the Sertoli and germ cell cytoskeletons during sperm nuclear remodelling, and maintenance of seminiferous epithelium integrity. Collectively, our findings reveal that katanins are able to differentially regulate almost all key microtubule-based structures during mammalian male germ cell development, through the complexing of one master controller, KATNB1, with a 'toolbox' of neofunctionalised katanin A-subunits.
Collapse
Affiliation(s)
- Jessica E M Dunleavy
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Anne E O'Connor
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Hidenobu Okuda
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia
| | - D Jo Merriner
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Moira K O'Bryan
- School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
8
|
Nie M, Yu B, Chen R, Sun B, Mao J, Wang X, Zhang H, Wu X. Novel rare variants in FGFR1 and clinical characteristics analysis in a series of congenital hypogonadotropic hypogonadism patients. Clin Endocrinol (Oxf) 2021; 95:153-162. [PMID: 33548149 DOI: 10.1111/cen.14436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE We aimed to analyse FGFR1 rare variants in a series of Chinese congenital hypogonadotropic hypogonadism (CHH) patients. In addition, we intended to understand the clinical characteristics and the response to treatment of CHH patients with FGFR1 rare variants. PATIENTS AND METHODS A total of 357 CHH patients were recruited at Peking Union Medical College Hospital. We used Sanger sequencing to analyse FGFR1 gene. In silico analysis was carried out to study the pathogenicity of novel missense variants. The clinical, endocrinological and therapeutic effects from patients carrying FGFR1 rare variants were analysed retrospectively. RESULTS Thimissense mutations.rty patients in this series were found to harbour 29 FGFR1 rare variants, with 8 recurrent and 21 novel variants. After comprehensive analysis, 18 out of 21 novel variants were classified as likely pathogenic (LP) ones. These variants are widely spread throughout the FGFR1 gene and almost all FGFR1 functional domains, which exhibited no hot spot. Cryptorchidism, cleft palate and dental abnormality incidence in this CHH series that possessed FGFR1 LP variants were approximately 38.5%, 7.6% and 3.8%, respectively. Among patients who accepted the fertility-promoting treatment, 8 out of 10 patients succeeded in spermatogenesis. CONCLUSIONS Eighteen novel LP variants were found to expand the spectrum of FGFR1 rare variants. In CHH patients possessing FGFR1 variants, we found that the rate of spermatogenesis was high following fertility-promoting therapy and the existence of cryptorchidism may represent the underlying factors which affect spermatogenesis.
Collapse
Affiliation(s)
- Min Nie
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Bingqing Yu
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Rongrong Chen
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bang Sun
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiangfeng Mao
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xi Wang
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueyan Wu
- NHC Key Laboratory of Endocrinology (Peking Union Medical College Hospital), Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Azhar M, Altaf S, Uddin I, Cheng J, Wu L, Tong X, Qin W, Bao J. Towards Post-Meiotic Sperm Production: Genetic Insight into Human Infertility from Mouse Models. Int J Biol Sci 2021; 17:2487-2503. [PMID: 34326689 PMCID: PMC8315030 DOI: 10.7150/ijbs.60384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Declined quality and quantity of sperm is currently the major cause of patients suffering from infertility. Male germ cell development is spatiotemporally regulated throughout the whole developmental process. While it has been known that exogenous factors, such as environmental exposure, diet and lifestyle, et al, play causative roles in male infertility, recent progress has revealed abundant genetic mutations tightly associated with defective male germline development. In mammals, male germ cells undergo dramatic morphological change (i.e., nuclear condensation) and chromatin remodeling during post-meiotic haploid germline development, a process termed spermiogenesis; However, the molecular machinery players and functional mechanisms have yet to be identified. To date, accumulated evidence suggests that disruption in any step of haploid germline development is likely manifested as fertility issues with low sperm count, poor sperm motility, aberrant sperm morphology or combined. With the continually declined cost of next-generation sequencing and recent progress of CRISPR/Cas9 technology, growing studies have revealed a vast number of disease-causing genetic variants associated with spermiogenic defects in both mice and humans, along with mechanistic insights partially attained and validated through genetically engineered mouse models (GEMMs). In this review, we mainly summarize genes that are functional at post-meiotic stage. Identification and characterization of deleterious genetic variants should aid in our understanding of germline development, and thereby further improve the diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Muhammad Azhar
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Saba Altaf
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Islam Uddin
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Jinbao Cheng
- The 901th hospital of Joint logistics support Force of PLA, Anhui, China
| | - Limin Wu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Xianhong Tong
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, China
| | - Jianqiang Bao
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui, China
| |
Collapse
|
10
|
Comparison of Clinical Characteristics and Spermatogenesis in CHH Patients Caused by PROKR2 and FGFR1 Mutations. Reprod Sci 2021; 28:3219-3227. [PMID: 33983622 DOI: 10.1007/s43032-021-00609-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Abstract
A retrospective study was conducted to investigate the effect of gonadotropin or pulsatile gonadotropin-releasing hormone (GnRH) therapy on spermatogenesis in congenital hypogonadotropic hypogonadism (CHH) patients with PROKR2 (prokineticin receptor 2) or FGFR1 (fibroblast growth factor receptor 1) mutations. Clinical features, gonadotropin levels, testicular volume (TV), and sperm concentration in response to gonadotropin and pulsatile GnRH therapy were compared between groups with PROKR2 and FGFR1 mutations. Twelve patients with PROKR2 gene mutation and fourteen patients with FGFR1 gene mutation were included. The incidence of cryptorchidism in PROKR2 and FGFR1 groups was 16.7% and 50%, respectively (p = 0.110). The baseline TV in the PROKR2 group was larger than that in FGFR1 group (2.0 vs. 1.63, p = 0.047). The initial LH, FSH, and testosterone levels were similar between the two groups. Based on the analysis of achieving spermatogenesis using Kaplan-Meier and log-rank tests, the PROKR2 group demonstrated shorter period of seminal spermatozoa appearance than the FGFR1 group (χ2 = 8.297, p = 0.004); the median duration of achieving spermatogenesis in the PROKR2 and FGFR1 groups was 9 and 16 months, respectively. The PROKR2 mutation group exhibited shorter required time to achieve different sperm concentration thresholds (5, 10, and 15 million/mL) than the FGFR1 mutation group (p = 0.012, 0.024, and 0.040). In conclusion, the PROKR2 group achieved spermatogenesis easily than the FGFR1 group, possibly due to the lower prevalence of cryptorchidism and larger baseline testicular volume in the PROKR2 group.
Collapse
|
11
|
Houston BJ, Nagirnaja L, Merriner DJ, O'Connor AE, Okuda H, Omurtag K, Smith C, Aston KI, Conrad DF, O'Bryan MK. The Sertoli cell expressed gene secernin-1 (Scrn1) is dispensable for male fertility in the mouse. Dev Dyn 2021; 250:922-931. [PMID: 33442887 DOI: 10.1002/dvdy.299] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Male infertility is a prevalent clinical presentation for which there is likely a strong genetic component due to the thousands of genes required for spermatogenesis. Within this study we investigated the role of the gene Scrn1 in male fertility. Scrn1 is preferentially expressed in XY gonads during the period of sex determination and in adult Sertoli cells based on single cell RNA sequencing. We investigated the expression of Scrn1 in juvenile and adult tissues and generated a knockout mouse model to test its role in male fertility. RESULTS Scrn1 was expressed at all ages examined in the post-natal testis; however, its expression peaked at postnatal days 7-14 and SCRN1 protein was clearly localized to Sertoli cells. Scrn1 deletion was achieved via removal of exon 3, and its loss had no effect on male fertility or sex determination. Knockout mice were capable of siring litters of equal size to wild type counterparts and generated equal numbers of sperm with comparable motility and morphology characteristics. CONCLUSIONS Scrn1 was found to be dispensable for male fertility, but this study identifies SCRN1 as a novel marker of the Sertoli cell cytoplasm.
Collapse
Affiliation(s)
- Brendan J Houston
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia.,School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Liina Nagirnaja
- Division of Genetics, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - D Jo Merriner
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Anne E O'Connor
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Hidenobu Okuda
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Kenan Omurtag
- Division of Reproductive Endocrinology and Infertility, School of Medicine, Washington University, St Louis, Missouri, USA
| | - Craig Smith
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kenneth I Aston
- Department of Surgery (Urology), University of Utah, Salt Lake City, Utah, USA
| | - Donald F Conrad
- Division of Genetics, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Moira K O'Bryan
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia.,School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
12
|
Programmed Cell Death 2-Like ( Pdcd2l) Is Required for Mouse Embryonic Development. G3-GENES GENOMES GENETICS 2020; 10:4449-4457. [PMID: 33055224 PMCID: PMC7718740 DOI: 10.1534/g3.120.401714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Globozoospermia is a rare form of male infertility where men produce round-headed sperm that are incapable of fertilizing an oocyte naturally. In a previous study where we undertook a whole exome screen to define novel genetic causes of globozoospermia, we identified homozygous mutations in the gene PDCD2L. Two brothers carried a p.(Leu225Val) variant predicted to introduce a novel splice donor site, thus presenting PDCD2L as a potential regulator of male fertility. In this study, we generated a Pdcd2l knockout mouse to test its role in male fertility. Contrary to the phenotype predicted from its testis-enriched expression pattern, Pdcd2l null mice died during embryogenesis. Specifically, we identified that Pdcd2l is essential for post-implantation embryonic development. Pdcd2l−/− embryos were resorbed at embryonic days 12.5-17.5 and no knockout pups were born, while adult heterozygous Pdcd2l males had comparable fertility to wildtype males. To specifically investigate the role of PDCD2L in germ cells, we employed Drosophila melanogaster as a model system. Consistent with the mouse data, global knockdown of trus, the fly ortholog of PDCD2L, resulted in lethality in flies at the third instar larval stage. However, germ cell-specific knockdown with two germ cell drivers did not affect male fertility. Collectively, these data suggest that PDCD2L is not essential for male fertility. By contrast, our results demonstrate an evolutionarily conserved role of PDCD2L in development.
Collapse
|
13
|
Crespo M, Damont A, Blanco M, Lastrucci E, Kennani SE, Ialy-Radio C, Khattabi LE, Terrier S, Louwagie M, Kieffer-Jaquinod S, Hesse AM, Bruley C, Chantalat S, Govin J, Fenaille F, Battail C, Cocquet J, Pflieger D. Multi-omic analysis of gametogenesis reveals a novel signature at the promoters and distal enhancers of active genes. Nucleic Acids Res 2020; 48:4115-4138. [PMID: 32182340 PMCID: PMC7192594 DOI: 10.1093/nar/gkaa163] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/30/2020] [Accepted: 03/07/2020] [Indexed: 12/17/2022] Open
Abstract
Epigenetic regulation of gene expression is tightly controlled by the dynamic modification of histones by chemical groups, the diversity of which has largely expanded over the past decade with the discovery of lysine acylations, catalyzed from acyl-coenzymes A. We investigated the dynamics of lysine acetylation and crotonylation on histones H3 and H4 during mouse spermatogenesis. Lysine crotonylation appeared to be of significant abundance compared to acetylation, particularly on Lys27 of histone H3 (H3K27cr) that accumulates in sperm in a cleaved form of H3. We identified the genomic localization of H3K27cr and studied its effects on transcription compared to the classical active mark H3K27ac at promoters and distal enhancers. The presence of both marks was strongly associated with highest gene expression. Assessment of their co-localization with transcription regulators (SLY, SOX30) and chromatin-binding proteins (BRD4, BRDT, BORIS and CTCF) indicated systematic highest binding when both active marks were present and different selective binding when present alone at chromatin. H3K27cr and H3K27ac finally mark the building of some sperm super-enhancers. This integrated analysis of omics data provides an unprecedented level of understanding of gene expression regulation by H3K27cr in comparison to H3K27ac, and reveals both synergistic and specific actions of each histone modification.
Collapse
Affiliation(s)
- Marion Crespo
- Univ. Grenoble Alpes, CEA, Inserm, IRIG-BGE, 38000 Grenoble, France
| | - Annelaure Damont
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, MetaboHUB, 91191 Gif-sur-Yvette, France
| | - Melina Blanco
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, 75014 Paris, France
| | | | - Sara El Kennani
- Univ. Grenoble Alpes, CEA, Inserm, IRIG-BGE, 38000 Grenoble, France.,CNRS UMR 5309, Inserm U1209, Université Grenoble Alpes, Institute for Advanced Biosciences, 38000 Grenoble, France
| | - Côme Ialy-Radio
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, 75014 Paris, France
| | - Laila El Khattabi
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, 75014 Paris, France
| | - Samuel Terrier
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, MetaboHUB, 91191 Gif-sur-Yvette, France
| | | | | | - Anne-Marie Hesse
- Univ. Grenoble Alpes, CEA, Inserm, IRIG-BGE, 38000 Grenoble, France
| | | | - Sophie Chantalat
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, 2 rue Gaston Crémieux, CP 5706, 91057 Evry Cedex, France
| | - Jérôme Govin
- Univ. Grenoble Alpes, CEA, Inserm, IRIG-BGE, 38000 Grenoble, France.,CNRS UMR 5309, Inserm U1209, Université Grenoble Alpes, Institute for Advanced Biosciences, 38000 Grenoble, France
| | - François Fenaille
- Service de Pharmacologie et d'Immunoanalyse, Laboratoire d'Etude du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, MetaboHUB, 91191 Gif-sur-Yvette, France
| | - Christophe Battail
- Univ. Grenoble Alpes, CEA, INSERM, Biosciences and Biotechnology Institute of Grenoble, Biology of Cancer and Infection UMR_S 1036, 38000 Grenoble, France
| | - Julie Cocquet
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, 75014 Paris, France
| | - Delphine Pflieger
- Univ. Grenoble Alpes, CEA, Inserm, IRIG-BGE, 38000 Grenoble, France.,CNRS, IRIG-BGE, 38000 Grenoble, France
| |
Collapse
|
14
|
A framework for high-resolution phenotyping of candidate male infertility mutants: from human to mouse. Hum Genet 2020; 140:155-182. [PMID: 32248361 DOI: 10.1007/s00439-020-02159-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/27/2020] [Indexed: 12/18/2022]
Abstract
Male infertility is a heterogeneous condition of largely unknown etiology that affects at least 7% of men worldwide. Classical genetic approaches and emerging next-generation sequencing studies support genetic variants as a frequent cause of male infertility. Meanwhile, the barriers to transmission of this disease mean that most individual genetic cases will be rare, but because of the large percentage of the genome required for spermatogenesis, the number of distinct causal mutations is potentially large. Identifying bona fide causes of male infertility thus requires advanced filtering techniques to select for high-probability candidates, including the ability to test causality in animal models. The mouse remains the gold standard for defining the genotype-phenotype connection in male fertility. Here, we present a best practice guide consisting of (a) major points to consider when interpreting next-generation sequencing data performed on infertile men, and, (b) a systematic strategy to categorize infertility types and how they relate to human male infertility. Phenotyping infertility in mice can involve investigating the function of multiple cell types across the testis and epididymis, as well as sperm function. These findings will feed into the diagnosis and treatment of male infertility as well as male health broadly.
Collapse
|
15
|
Lee AS, Rusch J, Lima AC, Usmani A, Huang N, Lepamets M, Vigh-Conrad KA, Worthington RE, Mägi R, Wu X, Aston KI, Atkinson JP, Carrell DT, Hess RA, O'Bryan MK, Conrad DF. Rare mutations in the complement regulatory gene CSMD1 are associated with male and female infertility. Nat Commun 2019; 10:4626. [PMID: 31604923 PMCID: PMC6789153 DOI: 10.1038/s41467-019-12522-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/11/2019] [Indexed: 12/27/2022] Open
Abstract
Infertility in men and women is a complex genetic trait with shared biological bases between the sexes. Here, we perform a series of rare variant analyses across 73,185 women and men to identify genes that contribute to primary gonadal dysfunction. We report CSMD1, a complement regulatory protein on chromosome 8p23, as a strong candidate locus in both sexes. We show that CSMD1 is enriched at the germ-cell/somatic-cell interface in both male and female gonads. Csmd1-knockout males show increased rates of infertility with significantly increased complement C3 protein deposition in the testes, accompanied by severe histological degeneration. Knockout females show significant reduction in ovarian quality and breeding success, as well as mammary branching impairment. Double knockout of Csmd1 and C3 causes non-additive reduction in breeding success, suggesting that CSMD1 and the complement pathway play an important role in the normal postnatal development of the gonads in both sexes.
Collapse
Affiliation(s)
- Arthur S Lee
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jannette Rusch
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ana C Lima
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Abul Usmani
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ni Huang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Maarja Lepamets
- Estonian Genome Center, University of Tartu, 51010, Tartu, Estonia
| | - Katinka A Vigh-Conrad
- Oregon National Primate Center, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Ronald E Worthington
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL, 62025, USA
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, 51010, Tartu, Estonia
| | - Xiaobo Wu
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kenneth I Aston
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Douglas T Carrell
- Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Rex A Hess
- College of Veterinary Medicine, University of Illinois, Urbana-Champaign, IL, 61802, USA
| | - Moira K O'Bryan
- The School of Biological Sciences, Monash University, Clayton, Victoria, 3800, Australia
| | - Donald F Conrad
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Oregon National Primate Center, Oregon Health and Science University, Beaverton, OR, 97006, USA.
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, OR, 97239, USA.
| |
Collapse
|
16
|
Lim S, Kierzek M, O'Connor AE, Brenker C, Merriner DJ, Okuda H, Volpert M, Gaikwad A, Bianco D, Potter D, Prabhakar R, Strünker T, O'Bryan MK. CRISP2 Is a Regulator of Multiple Aspects of Sperm Function and Male Fertility. Endocrinology 2019; 160:915-924. [PMID: 30759213 DOI: 10.1210/en.2018-01076] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/08/2019] [Indexed: 11/19/2022]
Abstract
The cysteine-rich secretory proteins (CRISPs) are a group of proteins that show a pronounced expression biased to the male reproductive tract. Although sperm encounter CRISPs at virtually all phases of sperm development and maturation, CRISP2 is the sole CRISP produced during spermatogenesis, wherein it is incorporated into the developing sperm head and tail. In this study we tested the necessity for CRISP2 in male fertility using Crisp2 loss-of-function mouse models. In doing so, we revealed a role for CRISP2 in establishing the ability of sperm to undergo the acrosome reaction and in establishing a normal flagellum waveform. Crisp2-deficient sperm possess a stiff midpiece and are thus unable to manifest the rapid form of progressive motility seen in wild type sperm. As a consequence, Crisp2-deficient males are subfertile. Furthermore, a yeast two-hybrid screen and immunoprecipitation studies reveal that CRISP2 can bind to the CATSPER1 subunit of the Catsper ion channel, which is necessary for normal sperm motility. Collectively, these data define CRISP2 as a determinant of male fertility and explain previous clinical associations between human CRISP2 expression and fertility.
Collapse
Affiliation(s)
- Shuly Lim
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Michelina Kierzek
- Center of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Anne E O'Connor
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- The School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Christoph Brenker
- Center of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - D Jo Merriner
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- The School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Hidenobu Okuda
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- The School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Marianna Volpert
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Avinash Gaikwad
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- The School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Deborah Bianco
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - David Potter
- Monash Micro Imaging, Monash University, Clayton, Victoria, Australia
| | - Ranganathan Prabhakar
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia
| | - Timo Strünker
- Center of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Moira K O'Bryan
- The Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- The School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
17
|
Liu Z, Mao J, Xu H, Wang X, Huang B, Zheng J, Nie M, Zhang H, Wu X. Gonadotropin-Induced Spermatogenesis in CHH Patients with Cryptorchidism. Int J Endocrinol 2019; 2019:6743489. [PMID: 31929795 PMCID: PMC6935817 DOI: 10.1155/2019/6743489] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/22/2019] [Indexed: 11/18/2022] Open
Abstract
Congenital hypogonadotropic hypogonadism (CHH) patients with cryptorchidism history usually have poor spermatogenesis outcome, while researches focusing on this population are rare. This study retrospectively evaluated gonadotropin-induced spermatogenesis outcome in CHH patients with cryptorchidism (n = 40). One hundred and eighty-three CHH patients without cryptorchidism were served as control. All patients received combined gonadotropins therapy (HCG and HMG) and were followed up for at least 6 months. The median follow-up period was 24 (15, 33) months (totally 960 person-months). Sperm (>0/ml) initially appeared in semen at a median of estimated 24 months (95% confidence interval (CI) 17.8-30.2). Twenty (20/40, 50%) patients succeeded in producing sperms, and the average time to produce first sperm was 19 ± 8 months. Five pregnancies were achieved in 9 (5/9, 56%) couples who desired for children. Compared with CHH patients without cryptorchidism (n = 183), cryptorchid patients had longer median time for sperm appearance in semen (24 months vs. 15 months, P < 0.001), lower rate of spermatogenesis (50% vs. 67%, P=0.032), and lower mean sperm concentration (1.9 (0.5, 8.6) million/ml vs. 11.1(1.0, 25.0) million/ml, P=0.006) at the last visit. In conclusion, CHH patients with cryptorchidism require a longer period for gonadotropin-induced spermatogenesis. The successful rate and sperm concentration were lower than patients without cryptorchidism.
Collapse
Affiliation(s)
- Zhaoxiang Liu
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jiangfeng Mao
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
| | - Hongli Xu
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
| | - Xi Wang
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
| | - Bingkun Huang
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
| | - Junjie Zheng
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
| | - Min Nie
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
| | - Hongbing Zhang
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xueyan Wu
- Key Laboratory of Endocrinology, Department of Endocrinology, Peking Union Medical College Hospital, National Health Commission of People's Republic of China, Beijing, China
| |
Collapse
|
18
|
Saucedo L, Sobarzo C, Brukman NG, Guidobaldi HA, Lustig L, Giojalas LC, Buffone MG, Vazquez-Levin MH, Marín-Briggiler C. Involvement of fibroblast growth factor 2 (FGF2) and its receptors in the regulation of mouse sperm physiology. Reproduction 2018; 156:163-172. [DOI: 10.1530/rep-18-0133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/01/2018] [Indexed: 01/22/2023]
Abstract
Fibroblast growth factor 2 (FGF2) and its receptors (FGFRs) have been described in several tissues, where they regulate cellular proliferation, differentiation, motility and apoptosis. Although FGF2/FGFRs expression in the male reproductive tract has been reported, there is scarce evidence on their presence in the female reproductive tract and their involvement in the modulation of sperm function. Therefore, the objective of this study was to determine the expression of FGF2 in the female reproductive tract and to assess the role of the FGF2/FGFRs system in the regulation of sperm physiology using the murine model. FGF2 was detected in uterus and oviduct protein extracts, and it was immunolocalized in epithelial cells of the uterus,isthmusandampulla, as well as in thecumulus oophorus-oocyte complex. The receptors FGFR1, FGFR2, FGFR3 and FGFR4 were immunodetected in the flagellum and acrosomal region of sperm recovered from thecaudaepididymis. Analysis of testis sections showed the expression of FGFRs in germ cells at different stages of the spermatogenesis, suggesting the testicular origin of the sperm FGFRs. Sperm incubation with recombinant FGF2 (rFGF2) led to increased sperm motility and velocity and to enhanced intracellular Ca2+levels and acrosomal loss compared to the control. In conclusion, this study shows that FGF2 is expressed in tissues of the female reproductive tract. Also, the fact that functional FGFRs are present in mouse sperm and that rFGF2 affects sperm motility and acrosomal exocytosis, suggests the involvement of this system in thein vivoregulation of sperm function.
Collapse
|
19
|
Saucedo L, Rumpel R, Sobarzo C, Schreiner D, Brandes G, Lustig L, Vazquez-Levin MH, Grothe C, Marín-Briggiler C. Deficiency of fibroblast growth factor 2 (FGF-2) leads to abnormal spermatogenesis and altered sperm physiology. J Cell Physiol 2018; 233:9640-9651. [PMID: 30054911 DOI: 10.1002/jcp.26876] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/23/2018] [Indexed: 01/08/2023]
Abstract
In previous studies, we described the presence of fibroblast growth factor 2 (FGF-2) and its receptors (FGFRs) in human testis and sperm, which are involved in spermatogenesis and in motility regulation. The aim of the present study was to analyze the role of FGF-2 in the maintenance of sperm physiology using FGF-2 knockout (KO) mice. Our results showed that in wild-type (WT) animals, FGF-2 is expressed in germ cells of the seminiferous epithelium, in epithelial cells of the epididymis, and in the flagellum and acrosomal region of epididymal sperm. In the FGF-2 KO mice, we found alterations in spermatogenesis kinetics, higher numbers of spermatids per testis, and enhanced daily sperm production compared with the WT males. No difference in the percentage of sperm motility was detected, but a significant increase in sperm concentration and in sperm head abnormalities was observed in FGF-2 KO animals. Sperm from KO mice depicted reduced phosphorylation on tyrosine residues (a phenomenon that was associated with sperm capacitation) and increased acrosomal loss after incubation under capacitating conditions. However, the FGF-2 KO males displayed no apparent fertility defects, since their mating with WT females showed no differences in the time to delivery, litter size, and pup weight in comparison with WT males. Overall, our findings suggest that FGF-2 exerts a role in mammalian spermatogenesis and that the lack of FGF-2 leads to dysregulated sperm production and altered sperm morphology and function. FGF-2-deficient mice constitute a model for the study of the complex mechanisms underlying mammalian spermatogenesis.
Collapse
Affiliation(s)
- Lucía Saucedo
- Instituto de Biología y Medicina Experimental (IBYME), National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Regina Rumpel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Cristian Sobarzo
- Instituto de Investigaciones Biomédicas, National Research Council of Argentina (CONICET), University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Dietmar Schreiner
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Gudrun Brandes
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Livia Lustig
- Instituto de Investigaciones Biomédicas, National Research Council of Argentina (CONICET), University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Mónica Hebe Vazquez-Levin
- Instituto de Biología y Medicina Experimental (IBYME), National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Claudia Grothe
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Clara Marín-Briggiler
- Instituto de Biología y Medicina Experimental (IBYME), National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| |
Collapse
|
20
|
Dunleavy JEM, Okuda H, O’Connor AE, Merriner DJ, O’Donnell L, Jamsai D, Bergmann M, O’Bryan MK. Katanin-like 2 (KATNAL2) functions in multiple aspects of haploid male germ cell development in the mouse. PLoS Genet 2017; 13:e1007078. [PMID: 29136647 PMCID: PMC5705150 DOI: 10.1371/journal.pgen.1007078] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 11/28/2017] [Accepted: 10/16/2017] [Indexed: 11/19/2022] Open
Abstract
The katanin microtubule-severing proteins are essential regulators of microtubule dynamics in a diverse range of species. Here we have defined critical roles for the poorly characterised katanin protein KATNAL2 in multiple aspects of spermatogenesis: the initiation of sperm tail growth from the basal body, sperm head shaping via the manchette, acrosome attachment, and ultimately sperm release. We present data suggesting that depending on context, KATNAL2 can partner with the regulatory protein KATNB1 or act autonomously. Moreover, our data indicate KATNAL2 may regulate δ- and ε-tubulin rather than classical α-β-tubulin microtubule polymers, suggesting the katanin family has a greater diversity of function than previously realised. Together with our previous research, showing the essential requirement of katanin proteins KATNAL1 and KATNB1 during spermatogenesis, our data supports the concept that in higher order species the presence of multiple katanins has allowed for subspecialisation of function within complex cellular settings such as the seminiferous epithelium. Male infertility affects one in twenty men of reproductive age in western countries. Despite this, the biochemical basis of common defects, including reduced sperm count and abnormal sperm structure and function, remains poorly defined. Microtubules are cellular “scaffolds” that serve critical roles in all cells, including developing male germ cells wherein they facilitate mitosis and meiosis (cell division), sperm head remodelling and sperm tail formation. The precise regulation of microtubule number, length and movement is thus, essential for male fertility. Within this manuscript, we have used spermatogenesis to define the function of the putative microtubule-severing protein katanin-like 2 (KATNAL2). We show that mice with compromised KATNAL2 function are male sterile as a consequence of defects in the structural remodelling of germ cells. Notably, we show the function of microtubule-based structures involved in sperm head shaping and tail formation are disrupted. Further, we show for the first time, that KATNAL2 can function both independently or in concert with the katanin regulatory protein KATNB1 and that it can target the poorly characterized tubulin subunits delta and epsilon. Our research has immediate relevance to the origins of human male infertility and provides novel insights into aspects of microtubule regulation relevant to numerous tissues and species.
Collapse
Affiliation(s)
- Jessica E. M. Dunleavy
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and The Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria; Australia
| | - Hidenobu Okuda
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
| | - Anne E. O’Connor
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
| | - D. Jo Merriner
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
| | - Liza O’Donnell
- Hudson Institute of Medical Research and Department of Molecular and Translational Science, Monash University, Melbourne, Victoria; Australia
| | - Duangporn Jamsai
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and The Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria; Australia
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Hesse; Germany
| | - Moira K. O’Bryan
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
- * E-mail:
| |
Collapse
|
21
|
PLAG1 deficiency impairs spermatogenesis and sperm motility in mice. Sci Rep 2017; 7:5317. [PMID: 28706261 PMCID: PMC5509656 DOI: 10.1038/s41598-017-05676-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023] Open
Abstract
Deficiency in pleomorphic adenoma gene 1 (PLAG1) leads to reduced fertility in male mice, but the mechanism by which PLAG1 contributes to reproduction is unknown. To investigate the involvement of PLAG1 in testicular function, we determined (i) the spatial distribution of PLAG1 in the testis using X-gal staining; (ii) transcriptomic consequences of PLAG1 deficiency in knock-out and heterozygous mice compared to wild-type mice using RNA-seq; and (iii) morphological and functional consequences of PLAG1 deficiency by determining testicular histology, daily sperm production and sperm motility in knock-out and wild-type mice. PLAG1 was sparsely expressed in germ cells and in Sertoli cells. Genes known to be involved in spermatogenesis were downregulated in the testes of knock-out mice, as well as Hsd17b3, which encodes a key enzyme in androgen biosynthesis. In the absence of Plag1, a number of genes involved in immune processes and epididymis-specific genes were upregulated in the testes. Finally, loss of PLAG1 resulted in significantly lowered daily sperm production, in reduced sperm motility, and in several animals, in sloughing of the germinal epithelium. Our results demonstrate that the subfertility seen in male PLAG1-deficient mice is, at least in part, the result of significantly reduced sperm output and sperm motility.
Collapse
|
22
|
Elevated paternal glucocorticoid exposure alters the small noncoding RNA profile in sperm and modifies anxiety and depressive phenotypes in the offspring. Transl Psychiatry 2016; 6:e837. [PMID: 27300263 PMCID: PMC4931607 DOI: 10.1038/tp.2016.109] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/25/2016] [Indexed: 12/21/2022] Open
Abstract
Recent studies have suggested that physiological and behavioral traits may be transgenerationally inherited through the paternal lineage, possibly via non-genomic signals derived from the sperm. To investigate how paternal stress might influence offspring behavioral phenotypes, a model of hypothalamic-pituitary-adrenal (HPA) axis dysregulation was used. Male breeders were administered water supplemented with corticosterone (CORT) for 4 weeks before mating with untreated female mice. Female, but not male, F1 offspring of CORT-treated fathers displayed altered fear extinction at 2 weeks of age. Only male F1 offspring exhibited altered patterns of ultrasonic vocalization at postnatal day 3 and, as adults, showed decreased time in open on the elevated-plus maze and time in light on the light-dark apparatus, suggesting a hyperanxiety-like behavioral phenotype due to paternal CORT treatment. Interestingly, expression of the paternally imprinted gene Igf2 was increased in the hippocampus of F1 male offspring but downregulated in female offspring. Male and female F2 offspring displayed increased time spent in the open arm of the elevated-plus maze, suggesting lower levels of anxiety compared with control animals. Only male F2 offspring showed increased immobility time on the forced-swim test and increased latency to feed on the novelty-supressed feeding test, suggesting a depression-like phenotype in these animals. Collectively, these data provide evidence that paternal CORT treatment alters anxiety and depression-related behaviors across multiple generations. Analysis of the small RNA profile in sperm from CORT-treated males revealed marked effects on the expression of small noncoding RNAs. Sperm from CORT-treated males contained elevated levels of three microRNAs, miR-98, miR-144 and miR-190b, which are predicted to interact with multiple growth factors, including Igf2 and Bdnf. Sustained elevation of glucocorticoids is therefore involved in the transmission of paternal stress-induced traits across generations in a process involving small noncoding RNA signals transmitted by the male germline.
Collapse
|
23
|
Liu Z, Mao J, Wu X, Xu H, Wang X, Huang B, Zheng J, Nie M, Zhang H. Efficacy and Outcome Predictors of Gonadotropin Treatment for Male Congenital Hypogonadotropic Hypogonadism: A Retrospective Study of 223 Patients. Medicine (Baltimore) 2016; 95:e2867. [PMID: 26945370 PMCID: PMC4782854 DOI: 10.1097/md.0000000000002867] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Gonadotropin induces masculinization and spermatogenesis in men with congenital hypogonadotropic hypogonadism (CHH). However, large cohort studies for the efficacy and reliable predictors of this therapy need to be conducted. The aim of this study was to investigate the efficacy of gonadotropin treatment in a large cohort of male CHH patients and analyze putative predictors for successful spermatogenesis. This retrospective study included 223 CHH azoospermic patients without puberty development treated between 2005 and 2014. All patients received combined human chorionic gonadotropin (HCG) and human menopausal gonadotropin (HMG) and were followed up for >6 months (5109 person-months). Serum total testosterone level, testicular size, spermatogenesis, and pregnancy outcome were recorded at each visit. After gonadotropin therapy, testicular size was enlarged from 2.1 ± 1.6 to 8.1 ± 4.6 mL (P < 0.001) and serum total testosterone was elevated from 0.9 ± 0.5 to 15.1 ± 8.2 nmol/L (P < 0.001). Spermatogenesis (>0/mL) occurred at a median period of 15 months (95% confidence interval 13.5-16.5). Larger basal testicular volume (P = 0.012) and noncryptorchidism history (P = 0.028) are independent predictors for earlier sperm appearance. Sixty four percent (143/223) of patients succeeded in producing sperms and the average time for initial sperm detection was 14 ± 8 months. However, their sperm concentrations (11.7 [2.1, 24.4] million/mL) and sperm progressive motility (A + B 36.9% ± 20.2%) are significantly lower than World Health Organization standards. Of the 34 patients who desired for fathering children, 19 patients impregnanted their partners during the treatment. Gonadotropin therapy induces spermatogenesis in male CHH patients. A larger basal testicular size and noncryptorchidism history are favorable indicators for earlier spermatogenesis.
Collapse
Affiliation(s)
- Zhaoxiang Liu
- From the Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital (ZL, JM, XW, HX, XW, BH, JZ, MN), and Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Peking Union Medical College and Chinese Academy of Medical Sciences (HZ), Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lim SL, Qu ZP, Kortschak RD, Lawrence DM, Geoghegan J, Hempfling AL, Bergmann M, Goodnow CC, Ormandy CJ, Wong L, Mann J, Scott HS, Jamsai D, Adelson DL, O’Bryan MK. HENMT1 and piRNA Stability Are Required for Adult Male Germ Cell Transposon Repression and to Define the Spermatogenic Program in the Mouse. PLoS Genet 2015; 11:e1005620. [PMID: 26496356 PMCID: PMC4619860 DOI: 10.1371/journal.pgen.1005620] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/30/2015] [Indexed: 02/04/2023] Open
Abstract
piRNAs are critical for transposable element (TE) repression and germ cell survival during the early phases of spermatogenesis, however, their role in adult germ cells and the relative importance of piRNA methylation is poorly defined in mammals. Using a mouse model of HEN methyltransferase 1 (HENMT1) loss-of-function, RNA-Seq and a range of RNA assays we show that HENMT1 is required for the 2’ O-methylation of mammalian piRNAs. HENMT1 loss leads to piRNA instability, reduced piRNA bulk and length, and ultimately male sterility characterized by a germ cell arrest at the elongating germ cell phase of spermatogenesis. HENMT1 loss-of-function, and the concomitant loss of piRNAs, resulted in TE de-repression in adult meiotic and haploid germ cells, and the precocious, and selective, expression of many haploid-transcripts in meiotic cells. Precocious expression was associated with a more active chromatin state in meiotic cells, elevated levels of DNA damage and a catastrophic deregulation of the haploid germ cell gene expression. Collectively these results define a critical role for HENMT1 and piRNAs in the maintenance of TE repression in adult germ cells and setting the spermatogenic program. Piwi-interacting RNAs (piRNAs) are small non-coding RNAs found in great abundance within both embryonic and adult male germ cells. Within embryonic germ cells, piRNAs have a well-recognized role in transposable element (TE) silencing, however, their role in adult cells remains poorly defined. Here we demonstrate that HENMT1 dysfunction and the resultant piRNA instability dramatically impacts multiple aspects of adult germ cell biology. Specifically, pachytene piRNAs are required to maintain TE silencing in adult germ cells and to set the spermatogenic gene expression program. piRNA loss leads to a more active chromatin state in the regulatory regions of numerous normally haploid germ cell genes and their precocious expression during meiosis, followed by a catastrophic deregulation of gene expression in haploid cells and male sterility.
Collapse
Affiliation(s)
- Shu Ly Lim
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Zhi Peng Qu
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - R. Daniel Kortschak
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - David M. Lawrence
- Australian Cancer Research Foundation Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Joel Geoghegan
- Australian Cancer Research Foundation Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Anna-Lena Hempfling
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Christopher C. Goodnow
- Australian Phenomics Facility, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Christopher J. Ormandy
- The Garvan Institute of Medical Research, Sydney, Darlinghurst, New South Wales, Australia
| | - Lee Wong
- The Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Jeff Mann
- Murdoch Childrens Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Hamish S. Scott
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
- Australian Cancer Research Foundation Cancer Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
- Department of Molecular Pathology, Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | - Duangporn Jamsai
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - David L. Adelson
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Moira K. O’Bryan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
25
|
Mao J, Xu H, Wang X, Huang B, Liu Z, Zhen J, Nie M, Min L, Wu X. Congenital combined pituitary hormone deficiency patients have better responses to gonadotrophin-induced spermatogenesis than idiopathic hypogonadotropic hypogonadism patients. Hum Reprod 2015; 30:2031-7. [PMID: 26141714 DOI: 10.1093/humrep/dev158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/04/2015] [Indexed: 12/18/2022] Open
Abstract
STUDY QUESTION Do patients with congenital combined pituitary hormone deficiency (CCPHD) have different responses to gonadotrophin-induced spermatogenesis compared with those with idiopathic hypogonadotropic hypogonadism (IHH)? SUMMARY ANSWER CCPHD patients have a better response to gonadotrophin therapy than IHH patients. WHAT IS KNOWN ALREADY Gonadotrophins are effective in inducing spermatogenesis in patients with hypogonadotropic hypogonadism. DESIGN, SIZE AND DURATION This retrospective cohort study included 75 patients, 53 of whom had IHH and 22 CCPHD. They were diagnosed, treated and followed up between January 2008 and December 2013. PARTICIPANTS/MATERIALS, SETTING AND METHODS Combined gonadotrophin therapy, consisting of human chorionic gonadotrophin and human menopausal gonadotrophin, was administered for 24 months. The success rate of spermatogenesis (≥1 sperm in ejaculate), serum total testosterone level, testicle size and sperm concentration during the treatment, as well as the first time sperm were detected in the ejaculate, were compared between the two diagnostic groups. All patients were treated in Peking Union Medical College Hospital. MAIN RESULTS AND THE ROLE OF CHANCE Spermatogenesis was successfully induced in 85% of IHH patients and 100% of CCPHD patients after 24-month combined gonadotrophin treatment (P = 0.03). In comparison with IHH, CCPHD patients had larger mean testicle sizes during the gonadotrophin treatment at 6, 12, 18 and 24 months (all P < 0.05). The initial time for sperm appearance in IHH group (n = 45) and CCPHD group (n = 22) was 13.2 ± 5.9 versus 10.4 ± 3.8 months (P = 0.045). Generally, CCPHD patients had higher sperm counts [median (quartiles)] than IHH patients during the treatment, but the difference was only statistically significant at 12 months of treatment, 3.3 (1.8, 12.0) versus 1.0 (0.0, 4.6) million/ml, P = 0.001. There was a higher level of serum total testosterone [mean (SD)] in the CCPHD group than the IHH group (676 ± 245 versus 555 ± 209 ng/dl, P = 0.035). LIMITATIONS, REASONS FOR CAUTION First, the inherent nature of a retrospective designed study was a main shortcoming. Secondly, pathological gene mutations in IHH and CCPHD patients should be further investigated. Clarification of the underlying mechanisms between cryptorchidism and mutated genes may provide more information for the divergent therapeutic responses between two groups. Only a minority of patients were actively seeking to have children so information about fertility is limited. WIDER IMPLICATIONS OF THE FINDINGS CCPHD patients had a lower incidence of cryptorchidism and a better response to gonadotrophin therapy than IHH patients, reflecting multiple defects on the different levels of reproduction axis in IHH. Furthermore, growth hormone is not indispensable for spermatogenesis in CCPHD patients. STUDY FUNDING/COMPETING INTERESTS The study was supported by Natural Science Foundation of China (No: 81100416). None of the authors has any conflicts of interest to declare.
Collapse
Affiliation(s)
- Jiangfeng Mao
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Hongli Xu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Xi Wang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Bingkun Huang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Zhaoxiang Liu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Junjie Zhen
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Min Nie
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| | - Le Min
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xueyan Wu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Beijing 100730, China
| |
Collapse
|
26
|
Itman C, Bielanowicz A, Goh H, Lee Q, Fulcher AJ, Moody SC, Doery JCG, Martin J, Eyre S, Hedger MP, Loveland KL. Murine Inhibin α-Subunit Haploinsufficiency Causes Transient Abnormalities in Prepubertal Testis Development Followed by Adult Testicular Decline. Endocrinology 2015; 156:2254-68. [PMID: 25781564 DOI: 10.1210/en.2014-1555] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activin production and signaling must be strictly regulated for normal testis development and function. Inhibins are potent activin inhibitors; mice lacking the inhibin-α gene (Inha-/- mice) cannot make inhibin and consequently have highly elevated activin and FSH serum concentrations and excessive activin signaling, resulting in somatic gonadal tumors and infertility. Dose-dependent effects of activin in testicular biology have been widely reported; hence, we hypothesized that male mice lacking one copy of the Inha gene would produce less inhibin and have an abnormal reproductive phenotype. To test this, we compared hormone concentrations, testis development, and sperm production in Inha+/+ and Inha+/- mice. Serum and testicular inhibin-α concentrations in adult Inha+/- mice were approximately 33% lower than wild type, whereas activin A, activin B, FSH, LH, and T were normal. Sixteen-day-old Inha+/- mice had a mixed phenotype, with tubules containing extensive germ cell depletion juxtaposed to tubules with advanced Sertoli and germ cell development. This abnormal phenotype resolved by day 28. By 8 weeks, Inha+/- testes were 11% larger than wild type and supported 44% greater daily sperm production. By 26 weeks of age, Inha+/- testes had distinct abnormalities. Although still fertile, Inha+/- mice had a 27% reduction in spermatogenic efficiency, a greater proportion of S-phase Sertoli cells and lower Leydig cell CYP11A1 expression. This study is the first to identify an intratesticular role for inhibin/inhibin-α subunit, demonstrating that a threshold level of this protein is required for normal testis development and to sustain adult somatic testicular cell function.
Collapse
Affiliation(s)
- Catherine Itman
- Priority Research Centres for Reproductive Science (C.I., A.B., J.M., S.E.) and Chemical Biology (C.I.), School of Environmental and Life Sciences, Faculty of Science and Information Technology, University of Newcastle, Callaghan, New South Wales 2308, Australia; Departments of Anatomy and Developmental Biology (H.G., Q.L., K.L.L.) and Biochemistry and Molecular Biology (S.C.M., K.L.L.) and Monash Micro Imaging (A.J.F.), Monash University, Clayton, Victoria 3800, Australia; and Faculty of Medicine, Nursing, and Health Sciences (J.C.G.D.), Department of Medicine, Monash Medical Centre, and Monash Institute of Medical Research-Prince Henry's Institute of Medical Research (M.P.H.), Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Saucedo L, Buffa GN, Rosso M, Guillardoy T, Góngora A, Munuce MJ, Vazquez-Levin MH, Marín-Briggiler C. Fibroblast Growth Factor Receptors (FGFRs) in Human Sperm: Expression, Functionality and Involvement in Motility Regulation. PLoS One 2015; 10:e0127297. [PMID: 25970615 PMCID: PMC4430232 DOI: 10.1371/journal.pone.0127297] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/13/2015] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factors receptors (FGFRs) have been widely characterized in somatic cells, but there is scarce evidence of their expression and function in mammalian gametes. The objective of the present study was to evaluate the expression of FGFRs in human male germ cells, to determine sperm FGFR activation by the FGF2 ligand and their participation in the regulation of sperm motility. The expression of FGFR1, 2, 3 and 4 mRNAs and proteins in human testis and localization of these receptors in germ cells of the seminiferous epithelium was demonstrated. In ejaculated sperm, FGFRs were localized to the acrosomal region and flagellum. Sperm exposure to FGF2 caused an increase in flagellar FGFR phosphorylation and activation of extracellular signal-regulated kinase (ERK) and protein kinase B (PKB or Akt) signaling pathways. Incubation with FGF2 led to a significant increase in the percentage of total and progressive sperm motility, as well as in sperm kinematics. All responses were prevented by sperm preincubation with BGJ398, a specific inhibitor of FGFR tyrosine kinase activity. In addition to confirming the expression of FGFRs in germ cells of the human testis, our study describes for the first time the presence, localization and functionality of human sperm FGFRs, and provides evidence of the beneficial effect of FGF2 upon sperm motility.
Collapse
Affiliation(s)
- Lucía Saucedo
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Gabriela N. Buffa
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Marina Rosso
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Tomás Guillardoy
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Adrian Góngora
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - María J. Munuce
- Laboratorio de Medicina Reproductiva, Area de Bioquímica Clínica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
- Reprolab, Sanatorio Británico de Rosario, Rosario, Santa Fe, Argentina
| | - Mónica H. Vazquez-Levin
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
| | - Clara Marín-Briggiler
- Instituto de Biología y Medicina Experimental (IBYME), CONICET-FIBYME, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
28
|
Vazquez-Levin MH, Marín-Briggiler CI, Caballero JN, Veiga MF. Epithelial and neural cadherin expression in the mammalian reproductive tract and gametes and their participation in fertilization-related events. Dev Biol 2015; 401:2-16. [DOI: 10.1016/j.ydbio.2014.12.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/23/2014] [Accepted: 12/28/2014] [Indexed: 01/10/2023]
|
29
|
Liu Y, DeBoer K, de Kretser DM, O’Donnell L, O’Connor AE, Merriner DJ, Okuda H, Whittle B, Jans DA, Efthymiadis A, McLachlan RI, Ormandy CJ, Goodnow CC, Jamsai D, O’Bryan MK. LRGUK-1 is required for basal body and manchette function during spermatogenesis and male fertility. PLoS Genet 2015; 11:e1005090. [PMID: 25781171 PMCID: PMC4363142 DOI: 10.1371/journal.pgen.1005090] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/23/2015] [Indexed: 12/23/2022] Open
Abstract
Male infertility affects at least 5% of reproductive age males. The most common pathology is a complex presentation of decreased sperm output and abnormal sperm shape and motility referred to as oligoasthenoteratospermia (OAT). For the majority of OAT men a precise diagnosis cannot be provided. Here we demonstrate that leucine-rich repeats and guanylate kinase-domain containing isoform 1 (LRGUK-1) is required for multiple aspects of sperm assembly, including acrosome attachment, sperm head shaping and the initiation of the axoneme growth to form the core of the sperm tail. Specifically, LRGUK-1 is required for basal body attachment to the plasma membrane, the appropriate formation of the sub-distal appendages, the extension of axoneme microtubules and for microtubule movement and organisation within the manchette. Manchette dysfunction leads to abnormal sperm head shaping. Several of these functions may be achieved in association with the LRGUK-1 binding partner HOOK2. Collectively, these data establish LRGUK-1 as a major determinant of microtubule structure within the male germ line.
Collapse
Affiliation(s)
- Yan Liu
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
| | - Kathleen DeBoer
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
| | - David M. de Kretser
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
| | - Liza O’Donnell
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
- MIMR-PHI Institute of Medical Research, Monash Medical Centre, Clayton, Australia
| | - Anne E. O’Connor
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
| | - D. Jo Merriner
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
| | - Hidenobu Okuda
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Belinda Whittle
- Australian Phenomics Facility, The Australian National University, Canberra, Australia
| | - David A. Jans
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Athina Efthymiadis
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Robert I. McLachlan
- MIMR-PHI Institute of Medical Research, Monash Medical Centre, Clayton, Australia
| | - Christopher J. Ormandy
- The Garvan Institute of Medical Research and St. Vincent’s Hospital Clinical School, UNSW Australia, Sydney, Australia
| | - Chris C. Goodnow
- Australian Phenomics Facility, The Australian National University, Canberra, Australia
| | - Duangporn Jamsai
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
| | - Moira K. O’Bryan
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Australia
- * E-mail:
| |
Collapse
|
30
|
Hasegawa K, Saga Y. FGF8-FGFR1 signaling acts as a niche factor for maintaining undifferentiated spermatogonia in the mouse. Biol Reprod 2014; 91:145. [PMID: 25359900 DOI: 10.1095/biolreprod.114.121012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In mammalian testes, spermatogonial stem cells (SSCs) maintain spermatogenesis over a long period of time by undergoing self-renewal and differentiation. SSCs are among the most primitive of spermatogenic cells (undifferentiated spermatogonia), and their activities are strictly regulated by extrinsic niche factors. However, the factors that constitute a testicular niche remain poorly understood. In this study, we demonstrate that fibroblast growth factor (FGF) signaling maintains undifferentiated spermatogonia through activating ERK1/2 signaling in vivo. Undifferentiated spermatogonia comprise GFRA1(+) and NANOS3(+) subpopulations, which are likely to undergo self-renewal and enter the differentiation pathway, respectively. In the testis, Fgfr1 was expressed in the entire population of undifferentiated spermatogonia, and deleting FGFR1 in spermatogenic cells partially inactivated ERK1/2 and resulted in reduced numbers of both GFRA1(+) and NANOS3(+) cells. In addition, Fgf8 was expressed in spermatogenic cells, and loss- and gain-of-function models of FGF8 demonstrated that FGF8 positively regulated the numbers of undifferentiated spermatogonia through FGFR1, particularly among NANOS3(+) cells. Finally we show a possible involvement of FGF signaling in the reversion from NANOS3(+) into GFRA1(+) undifferentiated spermatogonia. Taken together, our data suggest that FGF signaling is an important component of the testicular niche and has a unique function for maintaining undifferentiated spermatogonia.
Collapse
Affiliation(s)
- Kazuteru Hasegawa
- Division of Mammalian Development, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Yumiko Saga
- Division of Mammalian Development, National Institute of Genetics, Mishima, Shizuoka, Japan Department of Genetics, Sokendai, Mishima, Shizuoka, Japan
| |
Collapse
|
31
|
Aitken RJ, Nixon B. Sperm capacitation: a distant landscape glimpsed but unexplored. Mol Hum Reprod 2013; 19:785-93. [DOI: 10.1093/molehr/gat067] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
32
|
Colley SM, Wintle L, Searles R, Russell V, Firman RC, Smith S, DeBoer K, Merriner DJ, Genevieve B, Bentel JM, Stuckey BGA, Phillips MR, Simmons LW, de Kretser DM, O'Bryan MK, Leedman PJ. Loss of the nuclear receptor corepressor SLIRP compromises male fertility. PLoS One 2013; 8:e70700. [PMID: 23976951 PMCID: PMC3744554 DOI: 10.1371/journal.pone.0070700] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 06/20/2013] [Indexed: 11/24/2022] Open
Abstract
Nuclear receptors (NRs) and their coregulators play fundamental roles in initiating and directing gene expression influencing mammalian reproduction, development and metabolism. SRA stem Loop Interacting RNA-binding Protein (SLIRP) is a Steroid receptor RNA Activator (SRA) RNA-binding protein that is a potent repressor of NR activity. SLIRP is present in complexes associated with NR target genes in the nucleus; however, it is also abundant in mitochondria where it affects mitochondrial mRNA transcription and energy turnover. In further characterisation studies, we observed SLIRP protein in the testis where its localization pattern changes from mitochondrial in diploid cells to peri-acrosomal and the tail in mature sperm. To investigate the in vivo effects of SLIRP, we generated a SLIRP knockout (KO) mouse. This animal is viable, but sub-fertile. Specifically, when homozygous KO males are crossed with wild type (WT) females the resultant average litter size is reduced by approximately one third compared with those produced by WT males and females. Further, SLIRP KO mice produced significantly fewer progressively motile sperm than WT animals. Electron microscopy identified disruption of the mid-piece/annulus junction in homozygous KO sperm and altered mitochondrial morphology. In sum, our data implicates SLIRP in regulating male fertility, wherein its loss results in asthenozoospermia associated with compromised sperm structure and mitochondrial morphology.
Collapse
Affiliation(s)
- Shane M. Colley
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
| | - Larissa Wintle
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
| | | | - Victoria Russell
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
| | - Renee C. Firman
- Centre for Evolutionary Biology, School of Animal Biology, The University of Western Australia, Crawley, Australia
| | - Stephanie Smith
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Kathleen DeBoer
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - D. Jo Merriner
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Ben Genevieve
- Keogh Institute for Medical Research, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Jacqueline M. Bentel
- Anatomical Pathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Australia
| | - Bronwyn G. A. Stuckey
- Keogh Institute for Medical Research, Sir Charles Gairdner Hospital, Nedlands, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Australia
| | - Michael R. Phillips
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Australia
| | - Leigh W. Simmons
- Centre for Evolutionary Biology, School of Animal Biology, The University of Western Australia, Crawley, Australia
| | - David M. de Kretser
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Moira K. O'Bryan
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Peter J. Leedman
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Australia
- * E-mail:
| |
Collapse
|
33
|
Sagare-Patil V, Vernekar M, Galvankar M, Modi D. Progesterone utilizes the PI3K-AKT pathway in human spermatozoa to regulate motility and hyperactivation but not acrosome reaction. Mol Cell Endocrinol 2013; 374:82-91. [PMID: 23623968 DOI: 10.1016/j.mce.2013.04.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/19/2013] [Accepted: 04/09/2013] [Indexed: 01/26/2023]
Abstract
Progesterone is a physiologic regulator of sperm hyperactivation and acrosome reaction and it does so by activating a range of kinases present in the spermatozoa. In the present study, the involvement of the AKT- phosphatidylinositol 3-kinase (PI3K) signaling pathway in mediating progesterone response in human spermatozoa was investigated. In capacitated spermatozoa, progesterone transiently and concentration dependently lead to phosphorylation of AKT at both Thr 308 and Ser 473 in the tail region. This phosphorylation was inhibited by the PI3K inhibitor wortmannin, suggesting that progesterone leads to activation of PI3K-AKT pathway. The activation of AKT in response to progesterone is calcium dependent and the CatSper channel inhibitor mibefradil significantly reduced progesterone mediated AKT phosphorylation. Preincubation of spermatozoa with wortmannin inhibited the progesterone mediated increase in tyrosine phosphorylation and also attenuated the increase in number of motile, progressively motile and hyperactive spermatozoa but not the number of acrosome reacted spermatozoa. These observations imply that progesterone via CatSper activates the PI3K-AKT pathway required for motility and hyperactivation but not for acrosome reaction.
Collapse
Affiliation(s)
- Vrushali Sagare-Patil
- Molecular and Cellular Biology Laboratory, National Institute for Research in Reproductive Health, Indian Council of Medical Research, JM Street, Parel, Mumbai 400 012, India
| | | | | | | |
Collapse
|
34
|
De Amicis F, Perrotta I, Santoro M, Guido C, Morelli C, Cesario MG, Bruno R, Aquila S. Human Sperm Anatomy: Different Expression and Localization of Phosphatidylinositol 3-Kinase in Normal and Varicocele Human Spermatozoa. Ultrastruct Pathol 2013; 37:176-82. [DOI: 10.3109/01913123.2013.763881] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Zheng G, Dahl JA, Niu Y, Fedorcsak P, Huang CM, Li CJ, Vågbø CB, Shi Y, Wang WL, Song SH, Lu Z, Bosmans RPG, Dai Q, Hao YJ, Yang X, Zhao WM, Tong WM, Wang XJ, Bogdan F, Furu K, Fu Y, Jia G, Zhao X, Liu J, Krokan HE, Klungland A, Yang YG, He C. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell 2012. [PMID: 23177736 DOI: 10.1016/j.molcel.2012.10.015] [Citation(s) in RCA: 2632] [Impact Index Per Article: 202.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
N(6)-methyladenosine (m(6)A) is the most prevalent internal modification of messenger RNA (mRNA) in higher eukaryotes. Here we report ALKBH5 as another mammalian demethylase that oxidatively reverses m(6)A in mRNA in vitro and in vivo. This demethylation activity of ALKBH5 significantly affects mRNA export and RNA metabolism as well as the assembly of mRNA processing factors in nuclear speckles. Alkbh5-deficient male mice have increased m(6)A in mRNA and are characterized by impaired fertility resulting from apoptosis that affects meiotic metaphase-stage spermatocytes. In accordance with this defect, we have identified in mouse testes 1,551 differentially expressed genes that cover broad functional categories and include spermatogenesis-related mRNAs involved in the p53 functional interaction network. The discovery of this RNA demethylase strongly suggests that the reversible m(6)A modification has fundamental and broad functions in mammalian cells.
Collapse
Affiliation(s)
- Guanqun Zheng
- Department of Chemistry, Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Protein-tyrosine kinase signaling in the biological functions associated with sperm. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:181560. [PMID: 23209895 PMCID: PMC3503396 DOI: 10.1155/2012/181560] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 05/17/2012] [Accepted: 05/31/2012] [Indexed: 01/07/2023]
Abstract
In sexual reproduction, two gamete cells (i.e., egg and sperm) fuse (fertilization) to create a newborn with a genetic identity distinct from those of the parents. In the course of these developmental processes, a variety of signal transduction events occur simultaneously in each of the two gametes, as well as in the fertilized egg/zygote/early embryo. In particular, a growing body of knowledge suggests that the tyrosine kinase Src and/or other protein-tyrosine kinases are important elements that facilitate successful implementation of the aforementioned processes in many animal species. In this paper, we summarize recent findings on the roles of protein-tyrosine phosphorylation in many sperm-related processes (from spermatogenesis to epididymal maturation, capacitation, acrosomal exocytosis, and fertilization).
Collapse
|
37
|
RAB-like 2 has an essential role in male fertility, sperm intra-flagellar transport, and tail assembly. PLoS Genet 2012; 8:e1002969. [PMID: 23055941 PMCID: PMC3464206 DOI: 10.1371/journal.pgen.1002969] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 08/08/2012] [Indexed: 12/05/2022] Open
Abstract
A significant percentage of young men are infertile and, for the majority, the underlying cause remains unknown. Male infertility is, however, frequently associated with defective sperm motility, wherein the sperm tail is a modified flagella/cilia. Conversely, a greater understanding of essential mechanisms involved in tail formation may offer contraceptive opportunities, or more broadly, therapeutic strategies for global cilia defects. Here we have identified Rab-like 2 (RABL2) as an essential requirement for sperm tail assembly and function. RABL2 is a member of a poorly characterized clade of the RAS GTPase superfamily. RABL2 is highly enriched within developing male germ cells, where it localizes to the mid-piece of the sperm tail. Lesser amounts of Rabl2 mRNA were observed in other tissues containing motile cilia. Using a co-immunoprecipitation approach and RABL2 affinity columns followed by immunochemistry, we demonstrated that within developing haploid germ cells RABL2 interacts with intra-flagella transport (IFT) proteins and delivers a specific set of effector (cargo) proteins, including key members of the glycolytic pathway, to the sperm tail. RABL2 binding to effector proteins is regulated by GTP. Perturbed RABL2 function, as exemplified by the Mot mouse line that contains a mutation in a critical protein–protein interaction domain, results in male sterility characterized by reduced sperm output, and sperm with aberrant motility and short tails. Our data demonstrate a novel function for the RABL protein family, an essential role for RABL2 in male fertility and a previously uncharacterised mechanism for protein delivery to the flagellum. A greater understanding of the mechanism of male fertility is essential in order to address the medical needs of the 1 in 20 men of reproductive age who are infertile. Conversely, there remains a critical need for additional contraceptive options, including those that target male gametes. Towards the aim of filling these knowledge gaps, we have used random mutagenesis to produce the Mot mouse line and to identify RABL2 as an essential regulator of male fertility. Mice carrying a mutant Rabl2 gene are sterile as a consequence of severely compromised sperm motility. Using biochemical approaches we have revealed that RABL2 binds to components of the intraflagellar transport machinery and have identified a number of RABL2 binding (effector) proteins. The presence of the Mot mutation in RABL2 leads to a significantly compromised ability to deliver binding proteins into the sperm tail. RABL2 is predominantly produced in male germ cells; however, lower levels are notably produced in organs that contain motile cilia (hair like structures involved in fluid/cell movement), thus raising the possibility that RABL2 may be involved in a broader set of human diseases collectively known as primary cilia dyskinesia.
Collapse
|
38
|
Luna C, Colás C, Pérez-Pé R, Cebrián-Pérez JA, Muiño-Blanco T. A Novel Epidermal Growth Factor-Dependent Extracellular Signal-Regulated MAP Kinase Cascade Involved in Sperm Functionality in Sheep1. Biol Reprod 2012; 87:93. [DOI: 10.1095/biolreprod.112.100073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
39
|
An essential role for katanin p80 and microtubule severing in male gamete production. PLoS Genet 2012; 8:e1002698. [PMID: 22654669 PMCID: PMC3359970 DOI: 10.1371/journal.pgen.1002698] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 03/20/2012] [Indexed: 12/02/2022] Open
Abstract
Katanin is an evolutionarily conserved microtubule-severing complex implicated in multiple aspects of microtubule dynamics. Katanin consists of a p60 severing enzyme and a p80 regulatory subunit. The p80 subunit is thought to regulate complex targeting and severing activity, but its precise role remains elusive. In lower-order species, the katanin complex has been shown to modulate mitotic and female meiotic spindle dynamics and flagella development. The in vivo function of katanin p80 in mammals is unknown. Here we show that katanin p80 is essential for male fertility. Specifically, through an analysis of a mouse loss-of-function allele (the Taily line), we demonstrate that katanin p80, most likely in association with p60, has an essential role in male meiotic spindle assembly and dissolution and the removal of midbody microtubules and, thus, cytokinesis. Katanin p80 also controls the formation, function, and dissolution of a microtubule structure intimately involved in defining sperm head shaping and sperm tail formation, the manchette, and plays a role in the formation of axoneme microtubules. Perturbed katanin p80 function, as evidenced in the Taily mouse, results in male sterility characterized by decreased sperm production, sperm with abnormal head shape, and a virtual absence of progressive motility. Collectively these data demonstrate that katanin p80 serves an essential and evolutionarily conserved role in several aspects of male germ cell development. Microtubules are critical components of cells, acting as a “scaffold” for the movement of organelles and proteins within the cytoplasm. The control of microtubule length, number, and movement is essential for many cellular processes, including division, architecture, and migration. We have defined the role of the microtubule severing protein katanin p80 in male germ cell development. Male mice carrying a point mutation in the p80 gene are sterile as a consequence of low numbers of sperm, abnormal sperm morphology, and poor motility (ability to “swim”). We show that this mutation is associated with defects in microtubule structures involved in the division of immature sperm cells, in structures that shape the sperm head, and in the sperm tail, which is essential for sperm movement in the female reproductive tract. This study is the first to show that katanin p80, via its effects on microtubule dynamics within the testis, is required for male fertility.
Collapse
|
40
|
Elo T, Sipilä P, Valve E, Kujala P, Toppari J, Poutanen M, Härkönen P. Fibroblast Growth Factor 8b Causes Progressive Stromal and Epithelial Changes in the Epididymis and Degeneration of the Seminiferous Epithelium in the Testis of Transgenic Mice1. Biol Reprod 2012; 86:157, 1-12. [DOI: 10.1095/biolreprod.111.097352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
41
|
The neural cell adhesion molecule promotes FGFR-dependent phosphorylation and membrane targeting of the exocyst complex to induce exocytosis in growth cones. J Neurosci 2011; 31:3522-35. [PMID: 21389209 DOI: 10.1523/jneurosci.3109-10.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The exocyst complex is an essential regulator of polarized exocytosis involved in morphogenesis of neurons. We show that this complex binds to the intracellular domain of the neural cell adhesion molecule (NCAM). NCAM promotes FGF receptor-mediated phosphorylation of two tyrosine residues in the sec8 subunit of the exocyst complex and is required for efficient recruitment of the exocyst complex to growth cones. NCAM at the surface of growth cones induces Ca(2+)-dependent vesicle exocytosis, which is blocked by an inhibitor of L-type voltage-dependent Ca(2+) channels and tetanus toxin. Preferential exocytosis in growth cones underlying neurite outgrowth is inhibited in NCAM-deficient neurons as well as in neurons transfected with phosphorylation-deficient sec8 and dominant-negative peptides derived from the intracellular domain of NCAM. Thus, we reveal a novel role for a cell adhesion molecule in that it regulates addition of the new membrane to the cell surface of growth cones in developing neurons.
Collapse
|
42
|
Cysteine-rich secretory protein 4 is an inhibitor of transient receptor potential M8 with a role in establishing sperm function. Proc Natl Acad Sci U S A 2011; 108:7034-9. [PMID: 21482758 DOI: 10.1073/pnas.1015935108] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The cysteine-rich secretory proteins (CRISPs) are a group of four proteins in the mouse that are expressed abundantly in the male reproductive tract, and to a lesser extent in other tissues. Analysis of reptile CRISPs and mouse CRISP2 has shown that CRISPs can regulate cellular homeostasis via ion channels. With the exception of the ability of CRISP2 to regulate ryanodine receptors, the in vivo targets of mammalian CRISPs function are unknown. In this study, we have characterized the ion channel regulatory activity of epididymal CRISP4 using electrophysiology, cell assays, and mouse models. Through patch-clamping of testicular sperm, the CRISP4 CRISP domain was shown to inhibit the transient receptor potential (TRP) ion channel TRPM8. These data were confirmed using a stably transfected CHO cell line. TRPM8 is a major cold receptor in the body, but is found in other tissues, including the testis and on the tail and head of mouse and human sperm. Functional assays using sperm from wild-type mice showed that TRPM8 activation significantly reduced the number of sperm undergoing the progesterone-induced acrosome reaction following capacitation, and that this response was reversed by the coaddition of CRISP4. In accordance, sperm from Crisp4 null mice had a compromised ability to undergo to the progesterone-induced acrosome reaction. Collectively, these data identify CRISP4 as an endogenous regulator of TRPM8 with a role in normal sperm function.
Collapse
|
43
|
Singh SR, Burnicka-Turek O, Chauhan C, Hou SX. Spermatogonial stem cells, infertility and testicular cancer. J Cell Mol Med 2011; 15:468-83. [PMID: 21155977 PMCID: PMC3064728 DOI: 10.1111/j.1582-4934.2010.01242.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 10/04/2010] [Indexed: 02/06/2023] Open
Abstract
The spermatogonial stem cells (SSCs) are responsible for the transmission of genetic information from an individual to the next generation. SSCs play critical roles in understanding the basic reproductive biology of gametes and treatments of human infertility. SSCs not only maintain normal spermatogenesis, but also sustain fertility by critically balancing both SSC self-renewal and differentiation. This self-renewal and differentiation in turn is tightly regulated by a combination of intrinsic gene expression within the SSC as well as the extrinsic gene signals from the niche. Increased SSCs self-renewal at the expense of differentiation result in germ cell tumours, on the other hand, higher differentiation at the expense of self-renewal can result in male sterility. Testicular germ cell cancers are the most frequent cancers among young men in industrialized countries. However, understanding the pathogenesis of testis cancer has been difficult because it is formed during foetal development. Recent studies suggest that SSCs can be reprogrammed to become embryonic stem (ES)-like cells to acquire pluripotency. In the present review, we summarize the recent developments in SSCs biology and role of SSC in testicular cancer. We believe that studying the biology of SSCs will not only provide better understanding of stem cell regulation in the testis, but eventually will also be a novel target for male infertility and testicular cancers.
Collapse
Affiliation(s)
- Shree Ram Singh
- Mouse Cancer Genetics Program, National Institutes of Health, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | | | | | | |
Collapse
|
44
|
Martinović-Weigelt D, Wang RL, Villeneuve DL, Bencic DC, Lazorchak J, Ankley GT. Gene expression profiling of the androgen receptor antagonists flutamide and vinclozolin in zebrafish (Danio rerio) gonads. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2011; 101:447-458. [PMID: 21126777 DOI: 10.1016/j.aquatox.2010.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 09/25/2010] [Accepted: 10/09/2010] [Indexed: 05/30/2023]
Abstract
The studies presented in this manuscript focus on characterization of transcriptomic responses to anti-androgens in zebrafish (Danio rerio). Research on the effects of anti-androgens in fish has been characterized by a heavy reliance on apical endpoints, and molecular mechanisms of action (MOA) of anti-androgens remain poorly elucidated. In the present study, we examined effects of a short term exposure (24-96h) to the androgen receptor antagonists flutamide (FLU) and vinclozolin (VZ) on gene expression in gonads of sexually mature zebrafish, using commercially available zebrafish oligonucleotide microarrays (4×44K platform). We found that VZ and FLU potentially impact reproductive processes via multiple pathways related to steroidogenesis, spermatogenesis, and fertilization. Observed changes in gene expression often were shared by VZ and FLU, as demonstrated by overlap in differentially-expressed genes and enrichment of several common key pathways including: (1) integrin and actin signaling, (2) nuclear receptor 5A1 signaling, (3) fibroblast growth factor receptor signaling, (4) polyamine synthesis, and (5) androgen synthesis. This information should prove useful to elucidating specific mechanisms of reproductive effects of anti-androgens in fish, as well as developing biomarkers for this important class of endocrine-active chemicals.
Collapse
Affiliation(s)
- Dalma Martinović-Weigelt
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, 6201 Congdon Blvd., Duluth, MN 55804, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Hu Y, Yu H, Shaw G, Pask AJ, Renfree MB. Kallmann syndrome 1 gene is expressed in the marsupial gonad. Biol Reprod 2010; 84:595-603. [PMID: 21123819 DOI: 10.1095/biolreprod.110.087437] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kallmann syndrome is characterized by hypogonadotrophic hypogonadism and anosmia. The syndrome can be caused by mutations in several genes, but the X-linked form is caused by mutation in the Kallmann syndrome 1 (KAL1). KAL1 plays a critical role in gonadotropin-releasing hormone (GnRH) neuronal migration that is essential for the normal development of the hypothalamic-pituitary-gonadal axis. Interestingly, KAL1 appears to be missing from the rodent X, and no orthologue has been detected as yet. We investigated KAL1 during development and in adults of an Australian marsupial, the tammar wallaby, Macropus eugenii. Marsupial KAL1 maps to an autosome within a group of genes that was added as a block to the X chromosome in eutherian evolution. KAL1 expression was widespread in embryonic and adult tissues. In the adult testis, tammar KAL1 mRNA and protein were detected in the germ cells at specific stages of differentiation. In the adult testis, the protein encoded by KAL1, anosmin-1, was restricted to the round spermatids and elongated spermatids. In the adult ovary, anosmin-1 was not only detected in the oocytes but was also localized in the granulosa cells throughout folliculogenesis. This is the first examination of KAL1 mRNA and protein localization in adult mammalian gonads. The protein localization suggests that KAL1 participates in gametogenesis not only through the development of the hypothalamic-pituitary-gonadal axis by activation of GnRH neuronal migration, but also directly within the gonads themselves. Because KAL1 is autosomal in marsupials but is X-linked in eutherians, its conserved involvement in gametogenesis supports the hypothesis that reproduction-related genes were actively recruited to the eutherian X chromosome.
Collapse
Affiliation(s)
- Yanqiu Hu
- ARC Centre of Excellence for Kangaroo Genomics, The University of Melbourne, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
46
|
|
47
|
Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 5: intercellular junctions and contacts between germs cells and Sertoli cells and their regulatory interactions, testicular cholesterol, and genes/proteins associated with more than one germ cell generation. Microsc Res Tech 2010; 73:409-94. [PMID: 19941291 DOI: 10.1002/jemt.20786] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the testis, cell adhesion and junctional molecules permit specific interactions and intracellular communication between germ and Sertoli cells and apposed Sertoli cells. Among the many adhesion family of proteins, NCAM, nectin and nectin-like, catenins, and cadherens will be discussed, along with gap junctions between germ and Sertoli cells and the many members of the connexin family. The blood-testis barrier separates the haploid spermatids from blood borne elements. In the barrier, the intercellular junctions consist of many proteins such as occludin, tricellulin, and claudins. Changes in the expression of cell adhesion molecules are also an essential part of the mechanism that allows germ cells to move from the basal compartment of the seminiferous tubule to the adluminal compartment thus crossing the blood-testis barrier and well-defined proteins have been shown to assist in this process. Several structural components show interactions between germ cells to Sertoli cells such as the ectoplasmic specialization which are more closely related to Sertoli cells and tubulobulbar complexes that are processes of elongating spermatids embedded into Sertoli cells. Germ cells also modify several Sertoli functions and this also appears to be the case for residual bodies. Cholesterol plays a significant role during spermatogenesis and is essential for germ cell development. Lastly, we list genes/proteins that are expressed not only in any one specific generation of germ cells but across more than one generation.
Collapse
Affiliation(s)
- Louis Hermo
- Faculty of Medicine, Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada H3A 2B2.
| | | | | | | |
Collapse
|
48
|
Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 3: developmental changes in spermatid flagellum and cytoplasmic droplet and interaction of sperm with the zona pellucida and egg plasma membrane. Microsc Res Tech 2010; 73:320-63. [PMID: 19941287 DOI: 10.1002/jemt.20784] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Spermiogenesis constitutes the steps involved in the metamorphosis of spermatids into spermatozoa. It involves modification of several organelles in addition to the formation of several structures including the flagellum and cytoplasmic droplet. The flagellum is composed of a neck region and middle, principal, and end pieces. The axoneme composed of nine outer microtubular doublets circularly arranged to form a cylinder around a central pair of microtubules is present throughout the flagellum. The middle and principal pieces each contain specific components such as the mitochondrial sheath and fibrous sheath, respectively, while outer dense fibers are common to both. A plethora of proteins are constituents of each of these structures, with each playing key roles in functions related to the fertility of spermatozoa. At the end of spermiogenesis, a portion of spermatid cytoplasm remains associated with the released spermatozoa, referred to as the cytoplasmic droplet. The latter has as its main feature Golgi saccules, which appear to modify the plasma membrane of spermatozoa as they move down the epididymal duct and hence may be partly involved in male gamete maturation. The end product of spermatogenesis is highly streamlined and motile spermatozoa having a condensed nucleus equipped with an acrosome. Spermatozoa move through the female reproductive tract and eventually penetrate the zona pellucida and bind to the egg plasma membrane. Many proteins have been implicated in the process of fertilization as well as a plethora of proteins involved in the development of spermatids and sperm, and these are high lighted in this review.
Collapse
Affiliation(s)
- Louis Hermo
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada H3A 2B2.
| | | | | | | |
Collapse
|
49
|
Nixon B, Bielanowicz A, Anderson AL, Walsh A, Hall T, Mccloghry A, Aitken RJ. Elucidation of the signaling pathways that underpin capacitation-associated surface phosphotyrosine expression in mouse spermatozoa. J Cell Physiol 2010; 224:71-83. [PMID: 20232304 DOI: 10.1002/jcp.22090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent studies from within our laboratory have demonstrated a causal relationship between capacitation-associated surface phosphotyrosine expression and the ability of mouse spermatozoa to recognize the oocyte and engage in sperm-zona pellucida interaction. In the studies described herein we have sought to investigate the signaling pathways that underpin the tyrosine phosphorylation of sperm surface protein targets and validate the physiological significance of these pathways in relation to sperm-zona pellucida adhesion. Through selective pharmacological inhibition we have demonstrated that surface phosphotyrosine expression is unlikely to be mediated by the canonical cAMP-dependent protein kinase A (PKA) signaling cascade that has been most widely studied in relation to sperm capacitation. Rather, it appears to be primarily driven by the extracellular signal-regulated kinase (ERK) module of the mitogen-activated protein kinase (MAPK) pathway. Consistent with this notion, the main components of the ERK module (RAS, RAF1, MEK, and ERK1/2) were localized to the periacrosomal region of the head of mature mouse spermatozoa and their phosphorylation status within this region of the cell was positively modulated by capacitation. Furthermore, inhibition of several elements of this pathway suppressed sperm surface phosphotyrosine expression and induced a concomitant reduction sperm-zona pellucida interaction. Collectively, these data highlight a previously unappreciated role of the ERK module in the modification of the sperm surface during capacitation to render these cells functionally competent to engage in the process of fertilization.
Collapse
Affiliation(s)
- Brett Nixon
- Reproductive Science Group, School of Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.
| | | | | | | | | | | | | |
Collapse
|
50
|
Gibbs GM, Lo JCY, Nixon B, Jamsai D, O'Connor AE, Rijal S, Sanchez-Partida LG, Hearn MTW, Bianco DM, O'Bryan MK. Glioma pathogenesis-related 1-like 1 is testis enriched, dynamically modified, and redistributed during male germ cell maturation and has a potential role in sperm-oocyte binding. Endocrinology 2010; 151:2331-42. [PMID: 20219979 DOI: 10.1210/en.2009-1255] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The glioma pathogenesis-related 1 (GLIPR1) family consists of three genes [GLIPR1, GLIPR1-like 1 (GLIPR1L1), and GLIPR1-like 2 (GLIPR1L2)] and forms a distinct subgroup within the cysteine-rich secretory protein (CRISP), antigen 5, and pathogenesis-related 1 (CAP) superfamily. CAP superfamily proteins are found in phyla ranging from plants to humans and, based largely on expression and limited functional studies, are hypothesized to have roles in carcinogenesis, immunity, cell adhesion, and male fertility. Specifically data from a number of systems suggests that sequences within the C-terminal CAP domain of CAP proteins have the ability to promote cell-cell adhesion. Herein we cloned mouse Glipr1l1 and have shown it has a testis-enriched expression profile. GLIPR1L1 is posttranslationally modified by N-linked glycosylation during spermatogenesis and ultimately becomes localized to the connecting piece of elongated spermatids and sperm. After sperm capacitation, however, GLIPR1L1 is also localized to the anterior regions of the sperm head. Zona pellucida binding assays indicate that GLIPR1L1 has a role in the binding of sperm to the zona pellucida surrounding the oocyte. These data suggest that, along with other members of the CAP superfamily and several other proteins, GLIPR1L1 is involved in the binding of sperm to the oocyte complex. Collectively these data further strengthen the role of CAP domain-containing proteins in cellular adhesion and propose a mechanism whereby CAP proteins show overlapping functional significance during fertilization.
Collapse
Affiliation(s)
- Gerard M Gibbs
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Monash University, Victoria 3800, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|