1
|
Dai CL, Yin XY, Peng ZY, Lin H, Zhang P, Liu G, Li WN. Novel variants of FSIP2 and SPEF2 cause varying degrees of spermatozoa damage in MMAF patients and favorable ART outcomes. J Assist Reprod Genet 2025; 42:977-989. [PMID: 39753944 PMCID: PMC11950586 DOI: 10.1007/s10815-024-03378-1] [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: 05/17/2024] [Accepted: 12/18/2024] [Indexed: 03/28/2025] Open
Abstract
PURPOSE This study identified novel variants of the FSIP2 and SPEF2 genes in multiple morphological abnormalities of the sperm flagella (MMAF) patients and to investigate the potential effect of variations on male infertility and assisted reproductive outcomes. METHODS Whole-exome sequencing was performed in 106 Chinese MMAF patients. The discovered variants were evaluated in silico and confirmed by Sanger sequencing. A mini-gene assay and immunofluorescence staining were used to determine the effects on mRNA and protein. Assisted reproductive technology (ART) based on intracytoplasmic sperm injection (ICSI) and in vitro fertilization (IVF) was used for MMAF patients carrying novel variants. RESULTS Biallelic variants in FSIP2 or SPEF2 involving nineteen novel variations were found in eleven MMAF patients. These variations included fourteen missense variants, two nonsense variants, two frameshift variants, and a splicing variant. The FSIP2 protein was markedly reduced or mislocalized to the spermatozoa head. Two novel missense variants of SPEF2 reduced cell diameter. Eleven MMAF couples had 12 ICSI cycles and 2 IVF cycles. The 2PN fertilization rate, good-quality embryos rate, and clinical pregnancy rate were 80.1% (133/166), 74.4% (99/133), and 45.7% (16/35). Four of them have seven babies born. CONCLUSION Our work revealed that missense variations of FSIP2 or SPEF2 might cause a milder spermatozoa damage. The infertility caused by FSIP2 and SPEF2 variants can be mitigated through ICSI or even IVF. The results of this study presented signs of the correlation of phenotype/genotype for the FSIP2 and SPEF2, which might provide a reference for clinical genetic and fertility consultation.
Collapse
Affiliation(s)
- Cong-Ling Dai
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Hunan Guangxiu Hi-Tech Life Technology Co., Ltd, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Xin-Yu Yin
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Zi-Yan Peng
- Laboratory of Zebrafish Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hao Lin
- School of Medicine, Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Pan Zhang
- School of Medicine, Hunan Guangxiu Hospital, Hunan Normal University, Changsha, China
| | - Gang Liu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Wei-Na Li
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China.
- Hunan Guangxiu Hi-Tech Life Technology Co., Ltd, Changsha, China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive & Genetic Hospital of CITIC-Xiangya, Changsha, China.
| |
Collapse
|
2
|
Goli RC, Mahar K, Manohar PS, Chishi KG, Prabhu IG, Choudhary S, Rathi P, Chinnareddyvari CS, Haritha P, Metta M, Shetkar M, Kumar A, N D CP, Vidyasagar, Sukhija N, Kanaka KK. Insights from homozygous signatures of cervus nippon revealed genetic architecture for components of fitness. Mamm Genome 2024; 35:657-672. [PMID: 39191871 DOI: 10.1007/s00335-024-10064-1] [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: 05/07/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
This study investigates the genomic landscape of Sika deer populations, emphasizing the detection and characterization of runs of homozygosity (ROH) and their contribution towards components of fitness. Using 85,001 high-confidence SNPs, the investigation into ROH distribution unveiled nuanced patterns of autozygosity across individuals especially in 2 out of the 8 farms, exhibiting elevated ROH levels and mean genome coverage under ROH segments. The prevalence of shorter ROH segments (0.5-4 Mb) suggests historical relatedness and potential selective pressures within these populations. Intriguingly, despite observed variations in ROH profiles, the overall genomic inbreeding coefficient (FROH) remained relatively low across all farms, indicating a discernible degree of genetic exchange and effective mitigation of inbreeding within the studied Sika deer populations. Consensus ROH (cROH) were found to harbor genes for important functions viz., EGFLAM gene which is involved in the vision function of the eye, SKP2 gene which regulates cell cycle, CAPSL involved in adipogenesis, SPEF2 which is essential for sperm flagellar assembly, DCLK3 involved in the heat stress. This first ever study on ROH in Sika deer, to shed light on the adaptive role of genes in these homozygous regions. The insights garnered from this study have broader implications in the management of genetic diversity in this vulnerable species.
Collapse
Affiliation(s)
- Rangasai Chandra Goli
- ICAR-National Dairy Research Institute, Karnal, Haryana, India
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Karan Mahar
- ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Peela Sai Manohar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, India
| | - Kiyevi G Chishi
- ICAR-National Dairy Research Institute, Karnal, Haryana, India
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | | | - Sonu Choudhary
- ICAR-National Dairy Research Institute, Karnal, Haryana, India
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Pallavi Rathi
- ICAR-National Dairy Research Institute, Karnal, Haryana, India
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Chandana Sree Chinnareddyvari
- ICAR-National Dairy Research Institute, Karnal, Haryana, India
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Pala Haritha
- ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Muralidhar Metta
- College of Veterinary Science, SVVU, Garividi, Andhra Pradesh, India
| | - Mahantesh Shetkar
- College of Veterinary Sciences and Animal Husbandry, DUVASU, Mathura, Uttar Pradesh, India
| | - Amit Kumar
- ICAR- Indian Institute of Agricultural Biotechnology, Ranchi, Jharkhand, India
| | - Chethan Patil N D
- Department of Agricultural Economics & Extension, Lovely Professional University, Punjab, India
| | - Vidyasagar
- Veterinary College, KVAFSU, Bidar, Karnataka, India
| | - Nidhi Sukhija
- CSB-Central Tasar Research and Training Institute, Ranchi, Jharkhand, India.
| | - K K Kanaka
- ICAR- Indian Institute of Agricultural Biotechnology, Ranchi, Jharkhand, India
| |
Collapse
|
3
|
Ahmed Z, Liu M, Huang S, Xu D. CATSPER2 and SPEF2 are potential molecular markers for boar sperm quality: a population association study. J Assist Reprod Genet 2024; 41:3105-3117. [PMID: 39312031 PMCID: PMC11621296 DOI: 10.1007/s10815-024-03252-0] [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: 05/16/2024] [Accepted: 09/05/2024] [Indexed: 12/06/2024] Open
Abstract
PURPOSE This study investigates the role of cation channel sperm associated 2 (CATSPER2) and sperm flagella 2 (SPEF2) genes in boar spermatogenesis, focusing on their association with sperm quality traits in boars. METHODS Utilizing targeted next-generation sequencing, we identified and genotyped two polymorphisms in CATSPER2 (rs341636020G > A, rs326912346G > T) and three variants in SPEF2 (rs320839956A > G, rs334209514C > A, rs325319860C > T) across three boar breeds (Duroc, n = 181; Landrace, n = 87; Large White, n = 52). RESULTS Our results confirmed the presence of the specified single nucleotide polymorphisms (SNPs), adhering to association study criteria. In CATSPER2, significant associations were detected between rs341636020G > A and sperm curvilinear velocity (VCL) in Duroc and Landrace boars, and between rs326912346G > T and straight velocity (VSL) in Duroc and Large White boars. For SPEF2, rs320839956A > G was significantly linked to sperm viability in Duroc and Landrace and to sperm concentration (SCON) in Large White boars. Additionally, rs334209514C > A and rs325319860C > T showed significant associations with SCON and VCL respectively, in Doruc and Landrace boars. CONCLUSIONS Overall, our findings suggest that CATSPER2 and SPEF2 SNPs significantly impact boar sperm quality traits. These genetic markers have the potential to enhance boar fertility through selective breeding programs, contributing to the optimization of reproductive performance in pigs.
Collapse
Affiliation(s)
- Zulfiqar Ahmed
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China
| | - Min Liu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China
| | - Shuntao Huang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China
| | - Dequan Xu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, and Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, Hubie, People's Republic of China.
| |
Collapse
|
4
|
Yang J, Wang DF, Huang JH, Zhu QH, Luo LY, Lu R, Xie XL, Salehian-Dehkordi H, Esmailizadeh A, Liu GE, Li MH. Structural variant landscapes reveal convergent signatures of evolution in sheep and goats. Genome Biol 2024; 25:148. [PMID: 38845023 PMCID: PMC11155191 DOI: 10.1186/s13059-024-03288-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/21/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Sheep and goats have undergone domestication and improvement to produce similar phenotypes, which have been greatly impacted by structural variants (SVs). Here, we report a high-quality chromosome-level reference genome of Asiatic mouflon, and implement a comprehensive analysis of SVs in 897 genomes of worldwide wild and domestic populations of sheep and goats to reveal genetic signatures underlying convergent evolution. RESULTS We characterize the SV landscapes in terms of genetic diversity, chromosomal distribution and their links with genes, QTLs and transposable elements, and examine their impacts on regulatory elements. We identify several novel SVs and annotate corresponding genes (e.g., BMPR1B, BMPR2, RALYL, COL21A1, and LRP1B) associated with important production traits such as fertility, meat and milk production, and wool/hair fineness. We detect signatures of selection involving the parallel evolution of orthologous SV-associated genes during domestication, local environmental adaptation, and improvement. In particular, we find that fecundity traits experienced convergent selection targeting the gene BMPR1B, with the DEL00067921 deletion explaining ~10.4% of the phenotypic variation observed in goats. CONCLUSIONS Our results provide new insights into the convergent evolution of SVs and serve as a rich resource for the future improvement of sheep, goats, and related livestock.
Collapse
Affiliation(s)
- Ji Yang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dong-Feng Wang
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences (UCAS), Beijing, 100049, China
| | - Jia-Hui Huang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Qiang-Hui Zhu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences (UCAS), Beijing, 100049, China
| | - Ling-Yun Luo
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ran Lu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xing-Long Xie
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences (UCAS), Beijing, 100049, China
| | - Hosein Salehian-Dehkordi
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences (UCAS), Beijing, 100049, China
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, 76169-133, Iran
| | - George E Liu
- Animal Genomics and Improvement Laboratory, BARC, USDA-ARS, Beltsville, MD, 20705, USA
| | - Meng-Hua Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China.
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
5
|
Lu W, Li Y, Meng L, Tan C, Nie H, Zhang Q, Song Y, Zhang H, Tan YQ, Tu C, Guo H, Wu L, Du J. Novel SPEF2 variants cause male infertility and likely primary ciliary dyskinesia. J Assist Reprod Genet 2024; 41:1485-1498. [PMID: 38568462 PMCID: PMC11224184 DOI: 10.1007/s10815-024-03106-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/20/2024] [Indexed: 07/05/2024] Open
Abstract
PURPOSE This study aimed to identify the genetic causes of male infertility and primary ciliary dyskinesia (PCD)/PCD-like phenotypes in three unrelated Han Chinese families. METHODS We conducted whole-exome sequencing of three patients with male infertility and PCD/PCD-like phenotypes from three unrelated Chinese families. Ultrastructural and immunostaining analyses of patient spermatozoa and respiratory cilia and in vitro analyses were performed to analyze the effects of SPEF2 variants. Intracytoplasmic sperm injection (ICSI) was administered to three affected patients. RESULTS We identified four novel SPEF2 variants, including one novel homozygous splicing site variant [NC_000005.10(NM_024867.4): c.4447 + 1G > A] of the SPEF2 gene in family 1, novel compound heterozygous nonsense variants [NC_000005.10(NM_024867.4): c.1339C > T (p.R447*) and NC_000005.10(NM_024867.4): c.1645G > T (p.E549*)] in family 2, and one novel homozygous missense variant [NC_000005.10(NM_024867.4): c.2524G > A (p.D842N)] in family 3. All the patients presented with male infertility and PCD/likely PCD. All variants were present at very low levels in public databases, predicted to be deleterious in silico prediction tools, and were further confirmed deleterious by in vitro analyses. Ultrastructural analyses of the spermatozoa of the patients revealed the absence of the central pair complex in the sperm flagella. Immunostaining of the spermatozoa and respiratory cilia of the patients validated the pathogenicity of the SPEF2 variants. All patients carrying SPEF2 variants underwent one ICSI cycle and delivered healthy infants. CONCLUSION Our study reported four novel pathogenic variants of SPEF2 in three male patients with infertility and PCD/PCD-like phenotypes, which not only extend the spectrum of SPEF2 mutations but also provide information for genetic counseling and treatment of such conditions.
Collapse
Affiliation(s)
- Wenqing Lu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yong Li
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lanlan Meng
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Chen Tan
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Hongchuan Nie
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Qianjun Zhang
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yuying Song
- Changsha Maternal and Child Health Care Hospital, Changsha, China
| | - Huan Zhang
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Chaofeng Tu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Haichun Guo
- Changsha Maternal and Child Health Care Hospital, Changsha, China.
| | - Longxiang Wu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China.
| |
Collapse
|
6
|
Tian S, Tu C, He X, Meng L, Wang J, Tang S, Gao Y, Liu C, Wu H, Zhou Y, Lv M, Lin G, Jin L, Cao Y, Tang D, Zhang F, Tan YQ. Biallelic mutations in CFAP54 cause male infertility with severe MMAF and NOA. J Med Genet 2023; 60:827-834. [PMID: 36593121 DOI: 10.1136/jmg-2022-108887] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND Spermatogenic impairments can lead to male infertility by different pathological conditions, such as multiple morphological abnormalities of the sperm flagella (MMAF) and non-obstructive azoospermia (NOA). Genetic factors are involved in impaired spermatogenesis. METHODS AND RESULTS Here, we performed genetic analyses through whole-exome sequencing in a cohort of 334 Han Chinese probands with severe MMAF or NOA. Biallelic variants of CFAP54 were identified in three unrelated men, including one homozygous frameshift variant (c.3317del, p.Phe1106Serfs*19) and two compound heterozygous variants (c.878G>A, p.Arg293His; c.955C>T, p.Arg319Cys and c.4885C>T, p.Arg1629Cys; c.937G>A, p.Gly313Arg). All of the identified variants were absent or extremely rare in the public human genome databases and predicted to be damaging by bioinformatic tools. The men harbouring CFAP54 mutations exhibited abnormal sperm morphology, reduced sperm concentration and motility in ejaculated semen. Significant axoneme disorganisation and other ultrastructure abnormities were also detected inside the sperm cells from men harbouring CFAP54 mutations. Furthermore, immunofluorescence assays showed remarkably reduced staining of four flagellar assembly-associated proteins (IFT20, IFT52, IFT122 and SPEF2) in the spermatozoa of CFAP54-deficient men. Notably, favourable clinical pregnancy outcomes were achieved with sperm from men carrying CFAP54 mutations after intracytoplasmic sperm injection treatment. CONCLUSION Our genetic analyses and experimental observations revealed that biallelic deleterious mutations of CFAP54 can induce severe MMAF and NOA in humans.
Collapse
Affiliation(s)
- Shixiong Tian
- Institute of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Chaofeng Tu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, China
| | - Lanlan Meng
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Jiaxiong Wang
- Center for Reproduction and Genetics, State Key Laboratory of Reproductive Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Shuyan Tang
- Institute of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yang Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
| | - Chunyu Liu
- Institute of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, China
| | - Yiling Zhou
- Institute of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Li Jin
- Institute of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, China
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, China
| | - Feng Zhang
- Institute of Metabolism and Integrative Biology, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| |
Collapse
|
7
|
Yu H, Shi X, Shao Z, Geng H, Guo S, Li K, Gu M, Xu C, Gao Y, Tan Q, Duan Z, Wu H, Hua R, Guo R, Wei Z, Zhou P, Cao Y, He X, Li L, Zhang X, Lv M. Novel HYDIN variants associated with male infertility in two Chinese families. Front Endocrinol (Lausanne) 2023; 14:1118841. [PMID: 36742411 PMCID: PMC9889981 DOI: 10.3389/fendo.2023.1118841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Infertility is a major disease affecting human life and health, among which male factors account for about half. Asthenoteratozoospermia accounts for the majority of male infertility. High-throughput sequencing techniques have identified numerous variants in genes responsible for asthenoteratozoospermia; however, its etiology still needs to be studied. METHOD In this study, we performed whole-exome sequencing on samples from 375 patients with asthenoteratozoospermia and identified two HYDIN compound heterozygous variants, a primary ciliary dyskinesia (PCD)-associated gene, in two unrelated subjects. H&E staining, SEM were employed to analyze the varies on sperm of patients, further, TEM was employed to determine the ultrastructure defects. And westernblot and immunostaining were chose to evaluate the variation of structural protein. ICSI was applied to assist the mutational patient to achieve offspring. RESULT We identified two HYDIN compound heterozygous variants. Patient AY078 had novel compound heterozygous splice variants (c.5969-2A>G, c.6316+1G>A), altering the consensus splice acceptor site of HYDIN. He was diagnosed with male infertility and PCD, presenting with decreased sperm progressive motility and morphological abnormalities, and bronchial dilatation in the inferior lobe. Compared to the fertile control, HYDIN levels, acrosome and centrosome markers (ACTL7A, ACROSIN, PLCζ1, and Centrin1), and flagella components (TOMM20, SEPT4, SPEF2, SPAG6, and RSPHs) were significantly reduced in HYDIN-deficient patients. Using intracytoplasmic sperm injection (ICSI), the patient successfully achieved clinical pregnancy. AY079 had deleterious compound heterozygous missense variants, c.9507C>G (p. Asn3169Lys) and c.14081G>A (p. Arg4694His), presenting with infertility; however, semen samples and PCD examination were unavailable. DISCUSSION Our findings provide the first evidence that the loss of HYDIN function causes asthenoteratozoospermia presenting with various defects in the flagella structure and the disassembly of the acrosome and neck. Additionally, ICSI could rescue this failure of insemination caused by immobile and malformed sperm induced by HYDIN deficiency.
Collapse
Affiliation(s)
- Hui Yu
- Department of Obstetrics and Gynecology, Fuyang Hospital of Anhui Medical University, Fuyang, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiao Shi
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhongmei Shao
- Department of Obstetrics and Gynecology, Fuyang Hospital of Anhui Medical University, Fuyang, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hao Geng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Senzhao Guo
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Kuokuo Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Meng Gu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Chuan Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Yang Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Qing Tan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Provincial Human Sperm Bank, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zongliu Duan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Rong Hua
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Rui Guo
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
| | - Liang Li
- Department of Obstetrics and Gynecology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Xiaoping Zhang
- Department of Obstetrics and Gynecology, Fuyang Hospital of Anhui Medical University, Fuyang, China
| | - Mingrong Lv
- Department of Obstetrics and Gynecology, Fuyang Hospital of Anhui Medical University, Fuyang, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People’s Republic of China, Hefei, China
- *Correspondence: Mingrong Lv,
| |
Collapse
|
8
|
Wang J, Wang W, Shen L, Zheng A, Meng Q, Li H, Yang S. Clinical detection, diagnosis and treatment of morphological abnormalities of sperm flagella: A review of literature. Front Genet 2022; 13:1034951. [PMID: 36425067 PMCID: PMC9679630 DOI: 10.3389/fgene.2022.1034951] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/28/2022] [Indexed: 11/12/2023] Open
Abstract
Sperm carries male genetic information, and flagella help move the sperm to reach oocytes. When the ultrastructure of the flagella is abnormal, the sperm is unable to reach the oocyte and achieve insemination. Multiple morphological abnormalities of sperm flagella (MMAF) is a relatively rare idiopathic condition that is mainly characterized by multiple defects in sperm flagella. In the last decade, with the development of high-throughput DNA sequencing approaches, many genes have been revealed to be related to MMAF. However, the differences in sperm phenotypes and reproductive outcomes in many cases are attributed to different pathogenic genes or different pathogenic mutations in the same gene. Here, we will review information about the various phenotypes resulting from different pathogenic genes, including sperm ultrastructure and encoding proteins with their location and functions as well as assisted reproductive technology (ART) outcomes. We will share our clinical detection and diagnosis experience to provide additional clinical views and broaden the understanding of this disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shenmin Yang
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
9
|
Tapia R, Hecht GA. Spef1/CLAMP binds microtubules and actin-based structures and regulates cell migration and epithelia cell polarity. Ann N Y Acad Sci 2022; 1515:97-104. [PMID: 35710871 PMCID: PMC9796845 DOI: 10.1111/nyas.14845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
During migration, cells invade, repair, and create barriers leading to the formation of new cellular contacts in target tissues. Cell migration requires many proteins that collectively form the cytoskeleton. The main cytoskeletal elements are actin filaments, microtubules (MTs), and intermediate filaments. These structures work in concert with a large number of accessory proteins that contribute in a variety of ways to regulate filament assembly and turnover, to alter the configuration or arrangement of filaments by bundling or crosslinking, to link the cytoskeleton to other structures in the cell, such as membranes and junctions, and to transport cargo along the filaments. Sperm flagella protein-1 (Spef1), also designated calponin homology and microtubules-associated protein (CLAMP), is a multifunctional protein that interacts with cytoskeletal structures, including MTs, actin filaments, and focal adhesions in epithelia. In this review, we outline Spef1/CLAMP structure and expression in several cellular models. The function of Spef1/CLAMP in flagellar and ciliary motility, MT-binding and stability, regulation of planar cell polarity, and potential contribution to the maintenance of actin-based structures, such as lamellipodia and filopodia during cell migration, are also discussed.
Collapse
Affiliation(s)
- Rocio Tapia
- Division of Gastroenterology and Nutrition, Department of MedicineLoyola University Chicago, Loyola University Medical CenterMaywoodIllinoisUSA
| | - Gail A. Hecht
- Division of Gastroenterology and Nutrition, Department of MedicineLoyola University Chicago, Loyola University Medical CenterMaywoodIllinoisUSA
- Department of Microbiology and ImmunologyLoyola University Chicago, Loyola University Medical CenterMaywoodIllinoisUSA
| |
Collapse
|
10
|
Teves ME, Roldan ERS. Sperm bauplan and function and underlying processes of sperm formation and selection. Physiol Rev 2022; 102:7-60. [PMID: 33880962 PMCID: PMC8812575 DOI: 10.1152/physrev.00009.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
The spermatozoon is a highly differentiated and polarized cell, with two main structures: the head, containing a haploid nucleus and the acrosomal exocytotic granule, and the flagellum, which generates energy and propels the cell; both structures are connected by the neck. The sperm's main aim is to participate in fertilization, thus activating development. Despite this common bauplan and function, there is an enormous diversity in structure and performance of sperm cells. For example, mammalian spermatozoa may exhibit several head patterns and overall sperm lengths ranging from ∼30 to 350 µm. Mechanisms of transport in the female tract, preparation for fertilization, and recognition of and interaction with the oocyte also show considerable variation. There has been much interest in understanding the origin of this diversity, both in evolutionary terms and in relation to mechanisms underlying sperm differentiation in the testis. Here, relationships between sperm bauplan and function are examined at two levels: first, by analyzing the selective forces that drive changes in sperm structure and physiology to understand the adaptive values of this variation and impact on male reproductive success and second, by examining cellular and molecular mechanisms of sperm formation in the testis that may explain how differentiation can give rise to such a wide array of sperm forms and functions.
Collapse
Affiliation(s)
- Maria Eugenia Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia
| | - Eduardo R S Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales (CSIC), Madrid, Spain
| |
Collapse
|
11
|
Yogo K. Molecular basis of the morphogenesis of sperm head and tail in mice. Reprod Med Biol 2022; 21:e12466. [PMID: 35619659 PMCID: PMC9126569 DOI: 10.1002/rmb2.12466] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022] Open
Abstract
Background The spermatozoon has a complex molecular apparatus necessary for fertilization in its head and flagellum. Recently, numerous genes that are needed to construct the molecular apparatus of spermatozoa have been identified through the analysis of genetically modified mice. Methods Based on the literature information, the molecular basis of the morphogenesis of sperm heads and flagella in mice was summarized. Main findings (Results) The molecular mechanisms of vesicular trafficking and intraflagellar transport in acrosome and flagellum formation were listed. With the development of cryo‐electron tomography and mass spectrometry techniques, the details of the axonemal structure are becoming clearer. The fine structure and the proteins needed to form the central apparatus, outer and inner dynein arms, nexin‐dynein regulatory complex, and radial spokes were described. The important components of the formation of the mitochondrial sheath, fibrous sheath, outer dense fiber, and the annulus were also described. The similarities and differences between sperm flagella and Chlamydomonas flagella/somatic cell cilia were also discussed. Conclusion The molecular mechanism of formation of the sperm head and flagellum has been clarified using the mouse as a model. These studies will help to better understand the diversity of sperm morphology and the causes of male infertility.
Collapse
Affiliation(s)
- Keiichiro Yogo
- Department of Applied Life Sciences Faculty of Agriculture Shizuoka University Shizuoka Japan
| |
Collapse
|
12
|
Search for Associations of FSHR, INHA, INHAB, PRL, TNP2 and SPEF2 Genes Polymorphisms with Semen Quality in Russian Holstein Bulls (Pilot Study). Animals (Basel) 2021; 11:ani11102882. [PMID: 34679903 PMCID: PMC8532936 DOI: 10.3390/ani11102882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 11/16/2022] Open
Abstract
The aim of the study was to search for new mutations in the previously studied gene loci of follicle-stimulating hormone receptor (FSHR), inhibin α (INHA), inhibin β A (INHAB), prolactin (PRL), transition protein 2 (TNP2), and sperm flagella 2 (SPEF2) by sequencing, as well as the search for associations of previously identified mutations at these loci with fresh semen quality in Russian Holstein bulls. Phenotypic data from 189 bulls was collected. Data was analyzed for most bulls for three years of semen collection. The maximum value of each semen quality indicator (doublet ejaculate volume, sperm concentration, progressive motility and total number of spermatozoa) were selected. SNPs were identified in the FSHR, INHA, INHAB, TNP2, SPEF2 genes. The PRL gene did not have polymorphism. Significant (p < 0.05) associations of polymorphisms in the FSHR gene with double ejaculate volume, concentration and total number of spermatozoa were identified. Polymorphism in the INHA gene was significantly associated (p < 0.05) with sperm concentration. Polymorphism in the INHAB gene was significantly associated (p < 0.05) with doublet ejaculate volume and total number of spermatozoa. Polymorphisms in the TNP2 and SPEF2 genes did not have significant associations with semen quality. The SNPs studied in our pilot work may be considered as candidate genetic markers in the selection of bulls.
Collapse
|
13
|
Ratnapriya R, Acar İE, Geerlings MJ, Branham K, Kwong A, Saksens NTM, Pauper M, Corominas J, Kwicklis M, Zipprer D, Starostik MR, Othman M, Yashar B, Abecasis GR, Chew EY, Ferrington DA, Hoyng CB, Swaroop A, den Hollander AI. Family-based exome sequencing identifies rare coding variants in age-related macular degeneration. Hum Mol Genet 2021; 29:2022-2034. [PMID: 32246154 PMCID: PMC7390936 DOI: 10.1093/hmg/ddaa057] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/18/2020] [Accepted: 03/26/2020] [Indexed: 12/24/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified 52 independent variants at 34 genetic loci that are associated with age-related macular degeneration (AMD), the most common cause of incurable vision loss in the elderly worldwide. However, causal genes at the majority of these loci remain unknown. In this study, we performed whole exome sequencing of 264 individuals from 63 multiplex families with AMD and analyzed the data for rare protein-altering variants in candidate target genes at AMD-associated loci. Rare coding variants were identified in the CFH, PUS7, RXFP2, PHF12 and TACC2 genes in three or more families. In addition, we detected rare coding variants in the C9, SPEF2 and BCAR1 genes, which were previously suggested as likely causative genes at respective AMD susceptibility loci. Identification of rare variants in the CFH and C9 genes in our study validated previous reports of rare variants in complement pathway genes in AMD. We then extended our exome-wide analysis and identified rare protein-altering variants in 13 genes outside the AMD-GWAS loci in three or more families. Two of these genes, SCN10A and KIR2DL4, are of interest because variants in these genes also showed association with AMD in case-control cohorts, albeit not at the level of genome-wide significance. Our study presents the first large-scale, exome-wide analysis of rare variants in AMD. Further independent replications and molecular investigation of candidate target genes, reported here, would assist in gaining novel insights into mechanisms underlying AMD pathogenesis.
Collapse
Affiliation(s)
- Rinki Ratnapriya
- Neurobiology, Neurodegeneration and Repair Laboratory (NNRL), National Eye Institute, Bethesda, MD 20892, USA.,Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - İlhan E Acar
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500, The Netherlands
| | - Maartje J Geerlings
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500, The Netherlands
| | - Kari Branham
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Alan Kwong
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicole T M Saksens
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500, The Netherlands
| | - Marc Pauper
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500, The Netherlands
| | - Jordi Corominas
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500, The Netherlands
| | - Madeline Kwicklis
- Neurobiology, Neurodegeneration and Repair Laboratory (NNRL), National Eye Institute, Bethesda, MD 20892, USA
| | - David Zipprer
- Neurobiology, Neurodegeneration and Repair Laboratory (NNRL), National Eye Institute, Bethesda, MD 20892, USA
| | - Margaret R Starostik
- Neurobiology, Neurodegeneration and Repair Laboratory (NNRL), National Eye Institute, Bethesda, MD 20892, USA
| | - Mohammad Othman
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Beverly Yashar
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Goncalo R Abecasis
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily Y Chew
- Neurobiology, Neurodegeneration and Repair Laboratory (NNRL), National Eye Institute, Bethesda, MD 20892, USA
| | - Deborah A Ferrington
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carel B Hoyng
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500, The Netherlands
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory (NNRL), National Eye Institute, Bethesda, MD 20892, USA
| | - Anneke I den Hollander
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen 6500, The Netherlands
| |
Collapse
|
14
|
Wu B, Gao H, Liu C, Li W. The coupling apparatus of the sperm head and tail†. Biol Reprod 2021; 102:988-998. [PMID: 31995163 DOI: 10.1093/biolre/ioaa016] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/05/2019] [Accepted: 01/26/2020] [Indexed: 12/23/2022] Open
Abstract
A strong sperm head-tail coupling apparatus (HTCA) is needed to ensure the integrity of spermatozoa during their fierce competition to fertilize the egg. A lot of HTCA-specific components have evolved to strengthen the attachment of the tail to the implantation fossa at the sperm head. Defects in HTCA formation lead to acephalic spermatozoa syndrome and pathologies of some male infertility. Recent studies have provided insights into the pathogenic molecular mechanisms of acephalic spermatozoa syndrome. Here, we summarize the proteins involved in sperm neck development and focus on their roles in the formation of HTCA. In addition, we discuss the fine structures of the sperm neck in different species from an evolutionary view, highlighting the potential conservative mechanism of HTCA formation.
Collapse
Affiliation(s)
- Bingbing Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
15
|
Wang WL, Tu CF, Tan YQ. Insight on multiple morphological abnormalities of sperm flagella in male infertility: what is new? Asian J Androl 2021; 22:236-245. [PMID: 31210147 PMCID: PMC7275805 DOI: 10.4103/aja.aja_53_19] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The syndrome of multiple morphological abnormalities of the sperm flagella (MMAF) is a specific kind of asthenoteratozoospermia with a mosaic of flagellar morphological abnormalities (absent, short, bent, coiled, and irregular flagella). MMAF was proposed in 2014 and has attracted increasing attention; however, it has not been clearly understood. In this review, we elucidate the definition of MMAF from a systematical view, the difference between MMAF and other conditions with asthenoteratozoospermia or asthenozoospermia (such as primary mitochondrial sheath defects and primary ciliary dyskinesia), the knowledge regarding its etiological mechanism and related genetic findings, and the clinical significance of MMAF for intracytoplasmic sperm injection and genetic counseling. This review provides the basic knowledge for MMAF and puts forward some suggestions for further investigations.
Collapse
Affiliation(s)
- Wei-Li Wang
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Chao-Feng Tu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha 410078, China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha 410078, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410078, China.,National Engineering and Research Center of Human Stem Cell, Changsha 410078, China
| |
Collapse
|
16
|
Composition and function of the C1b/C1f region in the ciliary central apparatus. Sci Rep 2021; 11:11760. [PMID: 34083607 PMCID: PMC8175508 DOI: 10.1038/s41598-021-90996-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/19/2021] [Indexed: 02/04/2023] Open
Abstract
Motile cilia are ultrastructurally complex cell organelles with the ability to actively move. The highly conserved central apparatus of motile 9 × 2 + 2 cilia is composed of two microtubules and several large microtubule-bound projections, including the C1b/C1f supercomplex. The composition and function of C1b/C1f subunits has only recently started to emerge. We show that in the model ciliate Tetrahymena thermophila, C1b/C1f contains several evolutionarily conserved proteins: Spef2A, Cfap69, Cfap246/LRGUK, Adgb/androglobin, and a ciliate-specific protein Tt170/TTHERM_00205170. Deletion of genes encoding either Spef2A or Cfap69 led to a loss of the entire C1b projection and resulted in an abnormal vortex motion of cilia. Loss of either Cfap246 or Adgb caused only minor alterations in ciliary motility. Comparative analyses of wild-type and C1b-deficient mutant ciliomes revealed that the levels of subunits forming the adjacent C2b projection but not C1d projection are greatly reduced, indicating that C1b stabilizes C2b. Moreover, the levels of several IFT and BBS proteins, HSP70, and enzymes that catalyze the final steps of the glycolytic pathway: enolase ENO1 and pyruvate kinase PYK1, are also reduced in the C1b-less mutants.
Collapse
|
17
|
Central Apparatus, the Molecular Kickstarter of Ciliary and Flagellar Nanomachines. Int J Mol Sci 2021; 22:ijms22063013. [PMID: 33809498 PMCID: PMC7999657 DOI: 10.3390/ijms22063013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
Motile cilia and homologous organelles, the flagella, are an early evolutionarily invention, enabling primitive eukaryotic cells to survive and reproduce. In animals, cilia have undergone functional and structural speciation giving raise to typical motile cilia, motile nodal cilia, and sensory immotile cilia. In contrast to other cilia types, typical motile cilia are able to beat in complex, two-phase movements. Moreover, they contain many additional structures, including central apparatus, composed of two single microtubules connected by a bridge-like structure and assembling numerous complexes called projections. A growing body of evidence supports the important role of the central apparatus in the generation and regulation of the motile cilia movement. Here we review data concerning the central apparatus structure, protein composition, and the significance of its components in ciliary beating regulation.
Collapse
|
18
|
Aprea I, Raidt J, Höben IM, Loges NT, Nöthe-Menchen T, Pennekamp P, Olbrich H, Kaiser T, Biebach L, Tüttelmann F, Horvath J, Schubert M, Krallmann C, Kliesch S, Omran H. Defects in the cytoplasmic assembly of axonemal dynein arms cause morphological abnormalities and dysmotility in sperm cells leading to male infertility. PLoS Genet 2021; 17:e1009306. [PMID: 33635866 PMCID: PMC7909641 DOI: 10.1371/journal.pgen.1009306] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
Axonemal protein complexes, such as outer (ODA) and inner (IDA) dynein arms, are responsible for the generation and regulation of flagellar and ciliary beating. Studies in various ciliated model organisms have shown that axonemal dynein arms are first assembled in the cell cytoplasm and then delivered into axonemes during ciliogenesis. In humans, mutations in genes encoding for factors involved in this process cause structural and functional defects of motile cilia in various organs such as the airways and result in the hereditary disorder primary ciliary dyskinesia (PCD). Despite extensive knowledge about the cytoplasmic assembly of axonemal dynein arms in respiratory cilia, this process is still poorly understood in sperm flagella. To better define its clinical relevance on sperm structure and function, and thus male fertility, further investigations are required. Here we report the fertility status in different axonemal dynein preassembly mutant males (DNAAF2/ KTU, DNAAF4/ DYX1C1, DNAAF6/ PIH1D3, DNAAF7/ZMYND10, CFAP300/C11orf70 and LRRC6). Besides andrological examinations, we functionally and structurally analyzed sperm flagella of affected individuals by high-speed video- and transmission electron microscopy as well as systematically compared the composition of dynein arms in sperm flagella and respiratory cilia by immunofluorescence microscopy. Furthermore, we analyzed the flagellar length in dynein preassembly mutant sperm. We found that the process of axonemal dynein preassembly is also critical in sperm, by identifying defects of ODAs and IDAs in dysmotile sperm of these individuals. Interestingly, these mutant sperm consistently show a complete loss of ODAs, while some respiratory cilia from the same individual can retain ODAs in the proximal ciliary compartment. This agrees with reports of solely one distinct ODA type in sperm, compared to two different ODA types in proximal and distal respiratory ciliary axonemes. Consistent with observations in model organisms, we also determined a significant reduction of sperm flagellar length in these individuals. These findings are relevant to subsequent studies on the function and composition of sperm flagella in PCD patients and non-syndromic infertile males. Our study contributes to a better understanding of the fertility status in PCD-affected males and should help guide genetic and andrological counselling for affected males and their families. Impaired male fertility is a major issue and affects several men worldwide. Patients may present with reduced number or complete absence of sperm in the ejaculate, as well as functional and/or morphological sperm defects compromising sperm motility. Despite several diagnostic efforts, the underlying causes of these defects often remain unknown („idiopathic“). The beating of sperm flagella as well as motile cilia, such as those of the respiratory tract, is driven by dynein-based motor protein complexes, namely outer and inner dynein arms. In motile cilia these protein complexes are known to be first assembled in the cytoplasm and then delivered into the cilium. In sperm, this process is still poorly understood. Here we analyze sperm cells of male individuals with mutations in distinct genes encoding factors involved in the preassembly of these motor protein complexes. Consistent with defects in their respiratory ciliated cells, these individuals also demonstrate defects in sperm flagella that cause male infertility due to immotile sperm, with a reduction of flagellar length. Our results strengthen the assumption that the preassembly process of outer and inner dynein arms is clinically relevant also in sperm and provide knowledge that should guide genetic and andrological counselling for a subgroup of men with idiopathic infertility.
Collapse
Affiliation(s)
- Isabella Aprea
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Johanna Raidt
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Inga Marlena Höben
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Niki Tomas Loges
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Tabea Nöthe-Menchen
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Heike Olbrich
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Thomas Kaiser
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Luisa Biebach
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Muenster, Muenster, Germany
| | - Judit Horvath
- Institute of Human Genetics, University Hospital Muenster, Muenster, Germany
| | - Maria Schubert
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Claudia Krallmann
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Muenster, Muenster, Germany
| | - Sabine Kliesch
- Institute of Human Genetics, University Hospital Muenster, Muenster, Germany
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
- * E-mail:
| |
Collapse
|
19
|
Liu X, Wu Z, Li J, Bao H, Wu C. Genome-Wide Association Study and Transcriptome Differential Expression Analysis of the Feather Rate in Shouguang Chickens. Front Genet 2021; 11:613078. [PMID: 33414812 PMCID: PMC7783405 DOI: 10.3389/fgene.2020.613078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/02/2020] [Indexed: 12/01/2022] Open
Abstract
The feather rate phenotype in chicks, including early-feathering and late-feathering phenotypes, are widely used as a sexing system in the poultry industry. The objective of this study was to obtain candidate genes associated with the feather rate in Shouguang chickens. In the present study, we collected 56 blood samples and 12 hair follicle samples of flight feathers from female Shouguang chickens. Then we identified the chromosome region associated with the feather rate by genome-wide association analysis (GWAS). We also performed RNA sequencing and analyzed differentially expressed genes between the early-feathering and late-feathering phenotypes using HISAT2, StringTie, and DESeq2. We identified a genomic region of 10.0–13.0 Mb of chromosome Z, which is statistically associated with the feather rate of Shouguang chickens at one-day old. After RNA sequencing analysis, 342 differentially expressed known genes between the early-feathering (EF) and late-feathering (LF) phenotypes were screened out, which were involved in epithelial cell differentiation, intermediate filament organization, protein serine kinase activity, peptidyl-serine phosphorylation, retinoic acid binding, and so on. The sperm flagellar 2 gene (SPEF2) and prolactin receptor (PRLR) gene were the only two overlapping genes between the results of GWAS and differential expression analysis, which implies that SPEF2 and PRLR are possible candidate genes for the formation of the chicken feathering phenotype in the present study. Our findings help to elucidate the molecular mechanism of the feather rate in chicks.
Collapse
Affiliation(s)
- Xiayi Liu
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhou Wu
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Animal Breeding and Genomics, Wageningen University & Research, Wageningen, Netherlands
| | - Junying Li
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Haigang Bao
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Changxin Wu
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
20
|
Gao Y, Jian L, Lu W, Xue Y, Machaty Z, Luo H. Vitamin E can promote spermatogenesis by regulating the expression of proteins associated with the plasma membranes and protamine biosynthesis. Gene 2021; 773:145364. [PMID: 33359122 DOI: 10.1016/j.gene.2020.145364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/31/2020] [Accepted: 12/08/2020] [Indexed: 11/26/2022]
Abstract
Vitamin E is generally believed to promote the production of ovine sperm mainly through its antioxidant effect. Our previous studies have shown that some non-antioxidant genes may also be key in mediating this process. The objective of this study was to identify key candidate proteins that were differentially expressed in response to a treatment with Vitamin E. Prepubertal ovine testicular cells were isolated and divided into two groups. They were either treated with 800 μM Vitamin E (based on our previous results) or used as a non-treated control. After 24 h, all the cells were harvested for proteomic analysis. We found 115 differentially expressed proteins, 4 of which were up-regulated and 111 were down-regulated. A GO term enrichment analysis identified 127 Biological Process, 63 Cell Component and 26 Molecular Function terms that were enriched. Within those terms, 13, 11 and 26 terms were significantly enriched, respectively. Terms related to membrane and enzyme activity including the inner acrosomal membrane, signal peptidase complex, cysteine-type endopeptidase activity, etc., were also markedly enriched, while none of the KEGG pathways were enriched. We found that many of the differentially expressed proteins, such as CD46 (membrane cofactor protein), FLNA (Filamin A), DYSF (Dysferlin), IFT20 (Intraflagellar transport 20), SPCS1 (Signal peptidase complex subunit 1) and SPCS3 (Signal peptidase complex subunit 3) were related to the acrosomal and plasma membranes. A parallel reaction monitoring (PRM) analysis verified that Vitamin E improved spermatogenesis by regulating the expression of FLNA, SPCS3, YBX3 and RARS, proteins that are associated with the plasma membranes and protamine biosynthesis of the spermatozoa.
Collapse
Affiliation(s)
- Yuefeng Gao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Luyang Jian
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Wei Lu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Ying Xue
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Zoltan Machaty
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA.
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
21
|
Serrano M, Ramón M, Calvo JH, Jiménez MÁ, Freire F, Vázquez JM, Arranz JJ. Genome-wide association studies for sperm traits in Assaf sheep breed. Animal 2020; 15:100065. [PMID: 33573944 DOI: 10.1016/j.animal.2020.100065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Sperm quality traits routinely collected by artificial insemination (AI) center for rams progeny test are related with the capacity to produce sperm doses for AI and, in more or less grade, with males' fertility. Low-quality ejaculates are unuseful to perform AI sperm doses, which suppose high economic loses for the AI center. Moreover, sperm quality traits have low heritability values which make traditional genetic selection little efficient to its improvement. In this work, a genome-wide association study (GWAS) was conducted by using sperm quality traits data and 50 K Affymetrix custom chip genotypes of 429 rams of Assaf breed from OVIGEN AI centre. Furthermore, 47 of these rams were also genotyped with the Illumina HD Ovine BeadChip, and therefore HD genotypes were imputed for all rams with phenotype data. Previous to the GWAS, a linear regression model was fitted including sperm traits as dependent variables; the flock of origin, date of sperm collection, and jump number as fixed effects; rams age at collection in months as covariate; and ram permanent effect as random. Pseudo-phenotypes obtained from this model were used as input for GWAS. Associations at the chromosome-wise level (FDR 10%) of 76 single-nucleotide polymorphisms (SNPs) in 4 chromosomes for ejaculate concentration (CON), 20 SNPs in 3 chromosomes for ejaculate volume (VOL), 32 SNPs in 1 chromosome for ejaculate number of spermatozoa (SPZ), and 23 SNPs for spermatozoa mass motility (MOT) in 17 chromosomes were found. Only SNPs associated with MOT overcame the genome-wide significance level. Some candidate genes for sperm traits variability were SLC9C1 (OAR1), TSN (OAR2), and FUT10 (OAR26) for MOT;. DOCK2, CPLANE1, SPEF2, and RAI14 (OAR16) for CON; SCAPER and PSMA4 (OAR18) for VOL; and PARM1 and LOC101110593 (OAR6) for SPZ. SNPs associated with sperm traits were not found to be correlated with milk production genetic variation; however, the high frequencies of some SNPs with negative effect over sperm traits found in animals at the top milk yield estimated breeding values (EBVs) ranking would allow to exert some selective presure to improve rams sperm performances. Effects and frequencies of some of the SNPs detected over sperm quality traits make these variants good candidates to be used in marker-assisted selection to improve sperm characteristics of Assaf rams and AI center efficiency to produce sperm doses.
Collapse
Affiliation(s)
- M Serrano
- Departamento de Mejora Genética Animal, INIA, 28040 Madrid, Spain.
| | - M Ramón
- IRIAF-CERSYRA, Valdepeñas 13300, Ciudad Real, Spain
| | - J H Calvo
- Unidad de Tecnología en Producción Animal, CITA, 59059 Zaragoza, Spain; ARAID, 50004 Zaragoza, Spain
| | - M Á Jiménez
- Departamento de Mejora Genética Animal, INIA, 28040 Madrid, Spain
| | - F Freire
- OVIGEN, Granja Florencia s/n, Ctra. Villalazán-Peleagonzalo, 49800 Toro, Zamora, Spain
| | - J M Vázquez
- OVIGEN, Granja Florencia s/n, Ctra. Villalazán-Peleagonzalo, 49800 Toro, Zamora, Spain
| | - J J Arranz
- Departamento de Producción Animal, Universidad de León, 24007 León, Spain
| |
Collapse
|
22
|
Pleuger C, Lehti MS, Dunleavy JE, Fietz D, O'Bryan MK. Haploid male germ cells-the Grand Central Station of protein transport. Hum Reprod Update 2020; 26:474-500. [PMID: 32318721 DOI: 10.1093/humupd/dmaa004] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/15/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The precise movement of proteins and vesicles is an essential ability for all eukaryotic cells. Nowhere is this more evident than during the remarkable transformation that occurs in spermiogenesis-the transformation of haploid round spermatids into sperm. These transformations are critically dependent upon both the microtubule and the actin cytoskeleton, and defects in these processes are thought to underpin a significant percentage of human male infertility. OBJECTIVE AND RATIONALE This review is aimed at summarising and synthesising the current state of knowledge around protein/vesicle transport during haploid male germ cell development and identifying knowledge gaps and challenges for future research. To achieve this, we summarise the key discoveries related to protein transport using the mouse as a model system. Where relevant, we anchored these insights to knowledge in the field of human spermiogenesis and the causality of human male infertility. SEARCH METHODS Relevant studies published in English were identified using PubMed using a range of search terms related to the core focus of the review-protein/vesicle transport, intra-flagellar transport, intra-manchette transport, Golgi, acrosome, manchette, axoneme, outer dense fibres and fibrous sheath. Searches were not restricted to a particular time frame or species although the emphasis within the review is on mammalian spermiogenesis. OUTCOMES Spermiogenesis is the final phase of sperm development. It results in the transformation of a round cell into a highly polarised sperm with the capacity for fertility. It is critically dependent on the cytoskeleton and its ability to transport protein complexes and vesicles over long distances and often between distinct cytoplasmic compartments. The development of the acrosome covering the sperm head, the sperm tail within the ciliary lobe, the manchette and its role in sperm head shaping and protein transport into the tail, and the assembly of mitochondria into the mid-piece of sperm, may all be viewed as a series of overlapping and interconnected train tracks. Defects in this redistribution network lead to male infertility characterised by abnormal sperm morphology (teratozoospermia) and/or abnormal sperm motility (asthenozoospermia) and are likely to be causal of, or contribute to, a significant percentage of human male infertility. WIDER IMPLICATIONS A greater understanding of the mechanisms of protein transport in spermiogenesis offers the potential to precisely diagnose cases of male infertility and to forecast implications for children conceived using gametes containing these mutations. The manipulation of these processes will offer opportunities for male-based contraceptive development. Further, as increasingly evidenced in the literature, we believe that the continuous and spatiotemporally restrained nature of spermiogenesis provides an outstanding model system to identify, and de-code, cytoskeletal elements and transport mechanisms of relevance to multiple tissues.
Collapse
Affiliation(s)
- Christiane Pleuger
- School of Biological Sciences, Monash University, Clayton 3800, Australia.,Institute for Veterinary Anatomy, Histology and Embryology, Justus-Liebig University Giessen, Giessen 35392, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Mari S Lehti
- School of Biological Sciences, Monash University, Clayton 3800, Australia.,Institute of Biomedicine, University of Turku, Turku 20520, Finland
| | | | - Daniela Fietz
- Institute for Veterinary Anatomy, Histology and Embryology, Justus-Liebig University Giessen, Giessen 35392, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Moira K O'Bryan
- School of Biological Sciences, Monash University, Clayton 3800, Australia
| |
Collapse
|
23
|
Bi-allelic Loss-of-function Variants in CFAP58 Cause Flagellar Axoneme and Mitochondrial Sheath Defects and Asthenoteratozoospermia in Humans and Mice. Am J Hum Genet 2020; 107:514-526. [PMID: 32791035 DOI: 10.1016/j.ajhg.2020.07.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/15/2020] [Indexed: 01/08/2023] Open
Abstract
Multiple morphological abnormalities of the sperm flagella (MMAF) is a severe form of asthenoteratozoospermia. Although recent studies have revealed several MMAF-associated genes and demonstrated MMAF to be a genetically heterogeneous disease, at least one-third of the cases are still not well understood for their etiology. Here, we identified bi-allelic loss-of-function variants in CFAP58 by using whole-exome sequencing in five (5.6%) unrelated individuals from a cohort of 90 MMAF-affected Chinese men. Each of the men harboring bi-allelic CFAP58 variants presented typical MMAF phenotypes. Transmission electron microscopy demonstrated striking flagellar defects with axonemal and mitochondrial sheath malformations. CFAP58 is predominantly expressed in the testis and encodes a cilia- and flagella-associated protein. Immunofluorescence assays showed that CFAP58 localized at the entire flagella of control sperm and predominantly concentrated in the mid-piece. Immunoblotting and immunofluorescence assays showed that the abundances of axoneme ultrastructure markers SPAG6 and SPEF2 and a mitochondrial sheath protein, HSP60, were significantly reduced in the spermatozoa from men harboring bi-allelic CFAP58 variants. We generated Cfap58-knockout mice via CRISPR/Cas9 technology. The male mice were infertile and presented with severe flagellar defects, consistent with the sperm phenotypes in MMAF-affected men. Overall, our findings in humans and mice strongly suggest that CFAP58 plays a vital role in sperm flagellogenesis and demonstrate that bi-allelic loss-of-function variants in CFAP58 can cause axoneme and peri-axoneme malformations leading to male infertility. This study provides crucial insights for understanding and counseling of MMAF-associated asthenoteratozoospermia.
Collapse
|
24
|
Cindrić S, Dougherty GW, Olbrich H, Hjeij R, Loges NT, Amirav I, Philipsen MC, Marthin JK, Nielsen KG, Sutharsan S, Raidt J, Werner C, Pennekamp P, Dworniczak B, Omran H. SPEF2- and HYDIN-Mutant Cilia Lack the Central Pair-associated Protein SPEF2, Aiding Primary Ciliary Dyskinesia Diagnostics. Am J Respir Cell Mol Biol 2020; 62:382-396. [PMID: 31545650 DOI: 10.1165/rcmb.2019-0086oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Primary ciliary dyskinesia (PCD) is a genetically heterogeneous chronic destructive airway disease. PCD is traditionally diagnosed by nasal nitric oxide measurement, analysis of ciliary beating, transmission electron microscopy (TEM), and/or genetic testing. In most genetic PCD variants, laterality defects can occur. However, it is difficult to establish a diagnosis in individuals with PCD and central pair (CP) defects, and alternative strategies are required because of very subtle ciliary beating abnormalities, a normal ciliary ultrastructure, and normal situs composition. Mutations in HYDIN are known to cause CP defects, but the genetic analysis of HYDIN variants is confounded by the pseudogene HYDIN2, which is almost identical in terms of intron/exon structure. We have previously shown that several types of PCD can be diagnosed via immunofluorescence (IF) microscopy analyses. Here, using IF microscopy, we demonstrated that in individuals with PCD and CP defects, the CP-associated protein SPEF2 is absent in HYDIN-mutant cells, revealing its dependence on functional HYDIN. Next, we performed IF analyses of SPEF2 in respiratory cells from 189 individuals with suspected PCD and situs solitus. Forty-one of the 189 individuals had undetectable SPEF2 and were subjected to a genetic analysis, which revealed one novel loss-of-function mutation in SPEF2 and three reported and 13 novel HYDIN mutations in 15 individuals. The remaining 25 individuals are good candidates for new, as-yet uncharacterized PCD variants that affect the CP apparatus. SPEF2 mutations have been associated with male infertility but have not previously been identified to cause PCD. We identified a mutation of SPEF2 that is causative for PCD with a CP defect. We conclude that SPEF2 IF analyses can facilitate the detection of CP defects and evaluation of the pathogenicity of HYDIN variants, thus aiding the molecular diagnosis of CP defects.
Collapse
Affiliation(s)
- Sandra Cindrić
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Gerard W Dougherty
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Heike Olbrich
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Rim Hjeij
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Niki Tomas Loges
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Israel Amirav
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Maria C Philipsen
- Danish PCD Centre, ERN Accredited, Pediatric Pulmonary Service, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - June K Marthin
- Danish PCD Centre, ERN Accredited, Pediatric Pulmonary Service, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kim G Nielsen
- Danish PCD Centre, ERN Accredited, Pediatric Pulmonary Service, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sivagurunathan Sutharsan
- Department of Pulmonary Medicine, Ruhrlandklinik, Universitätsmedizin Essen, Essen, Germany; and
| | - Johanna Raidt
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Claudius Werner
- Department of Pediatrics, HELIOS Hospital Schwerin, Schwerin, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Bernd Dworniczak
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| | - Heymut Omran
- Department of General Pediatrics, University Children's Hospital Muenster, Muenster, Germany
| |
Collapse
|
25
|
Sironen A, Shoemark A, Patel M, Loebinger MR, Mitchison HM. Sperm defects in primary ciliary dyskinesia and related causes of male infertility. Cell Mol Life Sci 2020; 77:2029-2048. [PMID: 31781811 PMCID: PMC7256033 DOI: 10.1007/s00018-019-03389-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 01/22/2023]
Abstract
The core axoneme structure of both the motile cilium and sperm tail has the same ultrastructural 9 + 2 microtubular arrangement. Thus, it can be expected that genetic defects in motile cilia also have an effect on sperm tail formation. However, recent studies in human patients, animal models and model organisms have indicated that there are differences in components of specific structures within the cilia and sperm tail axonemes. Primary ciliary dyskinesia (PCD) is a genetic disease with symptoms caused by malfunction of motile cilia such as chronic nasal discharge, ear, nose and chest infections and pulmonary disease (bronchiectasis). Half of the patients also have situs inversus and in many cases male infertility has been reported. PCD genes have a role in motile cilia biogenesis, structure and function. To date mutations in over 40 genes have been identified cause PCD, but the exact effect of these mutations on spermatogenesis is poorly understood. Furthermore, mutations in several additional axonemal genes have recently been identified to cause a sperm-specific phenotype, termed multiple morphological abnormalities of the sperm flagella (MMAF). In this review, we discuss the association of PCD genes and other axonemal genes with male infertility, drawing particular attention to possible differences between their functions in motile cilia and sperm tails.
Collapse
Affiliation(s)
- Anu Sironen
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.
| | - Amelia Shoemark
- Department of Paediatrics, Royal Brompton Hospital, London, UK
- School of Medicine, University of Dundee, Dundee, UK
| | - Mitali Patel
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Michael R Loebinger
- Host Defence Unit, Royal Brompton and Harefield NHS Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Hannah M Mitchison
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| |
Collapse
|
26
|
Teves ME, Roldan ERS, Krapf D, Strauss III JF, Bhagat V, Sapao P. Sperm Differentiation: The Role of Trafficking of Proteins. Int J Mol Sci 2020; 21:E3702. [PMID: 32456358 PMCID: PMC7279445 DOI: 10.3390/ijms21103702] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/10/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
Sperm differentiation encompasses a complex sequence of morphological changes that takes place in the seminiferous epithelium. In this process, haploid round spermatids undergo substantial structural and functional alterations, resulting in highly polarized sperm. Hallmark changes during the differentiation process include the formation of new organelles, chromatin condensation and nuclear shaping, elimination of residual cytoplasm, and assembly of the sperm flagella. To achieve these transformations, spermatids have unique mechanisms for protein trafficking that operate in a coordinated fashion. Microtubules and filaments of actin are the main tracks used to facilitate the transport mechanisms, assisted by motor and non-motor proteins, for delivery of vesicular and non-vesicular cargos to specific sites. This review integrates recent findings regarding the role of protein trafficking in sperm differentiation. Although a complete characterization of the interactome of proteins involved in these temporal and spatial processes is not yet known, we propose a model based on the current literature as a framework for future investigations.
Collapse
Affiliation(s)
- Maria E. Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond VA 23298, USA;
| | - Eduardo R. S. Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales (CSIC), 28006-Madrid, Spain
| | - Diego Krapf
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Jerome F. Strauss III
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond VA 23298, USA;
| | - Virali Bhagat
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond VA 23298, USA;
| | - Paulene Sapao
- Department of Chemistry, Virginia Commonwealth University, Richmond VA, 23298, USA;
| |
Collapse
|
27
|
Qu W, Yuan S, Quan C, Huang Q, Zhou Q, Yap Y, Shi L, Zhang D, Guest T, Li W, Yee SP, Zhang L, Cazin C, Hess RA, Ray PF, Kherraf ZE, Zhang Z. The essential role of intraflagellar transport protein IFT81 in male mice spermiogenesis and fertility. Am J Physiol Cell Physiol 2020; 318:C1092-C1106. [PMID: 32233951 DOI: 10.1152/ajpcell.00450.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intraflagellar transport (IFT) is an evolutionarily conserved mechanism that is indispensable for the formation and maintenance of cilia and flagella; however, the implications and functions of IFT81 remain unknown. In this study, we disrupted IFT81 expression in male germ cells starting from the spermatocyte stage. As a result, homozygous mutant males were completely infertile and displayed abnormal sperm parameters. In addition to oligozoospermia, spermatozoa presented dysmorphic and nonfunctional flagella. Histological examination of testes from homozygous mutant mice revealed abnormal spermiogenesis associated with sloughing of germ cells and the presence of numerous multinucleated giant germ cells (symblasts) in the lumen of seminiferous tubules and epididymis. Moreover, only few elongated spermatids and spermatozoa were seen in analyzed cross sections. Transmission electron microscopy showed a complete disorganization of the axoneme and para-axonemal structures such as the mitochondrial sheath, fibrous sheath, and outer dense fibers. In addition, numerous vesicles that contain unassembled microtubules were observed within developing spermatids. Acrosome structure analysis showed normal appearance, thus excluding a crucial role of IFT81 in acrosome biogenesis. These observations showed that IFT81 is an important member of the IFT process during spermatogenesis and that its absence is associated with abnormal flagellum formation leading to male infertility. The expression levels of several IFT components in testes, including IFT20, IFT25, IFT27, IFT57, IFT74, and IFT88, but not IFT140, were significantly reduced in homozygous mutant mice. Overall, our study demonstrates that IFT81 plays an essential role during spermatogenesis by modulating the assembly and elongation of the sperm flagella.
Collapse
Affiliation(s)
- Wei Qu
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Shuo Yuan
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Chao Quan
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Qian Huang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Qi Zhou
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Yitian Yap
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Lin Shi
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - David Zhang
- College of William & Mary, Williamsburg, Virginia
| | - Tamia Guest
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Ling Zhang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Caroline Cazin
- Team Genetic, Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5309, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble Alpes, Grenoble, France
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois
| | - Pierre F Ray
- Team Genetic, Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5309, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble Alpes, Grenoble, France
| | - Zine-Eddine Kherraf
- Team Genetic, Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Université Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5309, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble Alpes, Grenoble, France
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, Michigan.,Department of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan
| |
Collapse
|
28
|
Touré A, Martinez G, Kherraf ZE, Cazin C, Beurois J, Arnoult C, Ray PF, Coutton C. The genetic architecture of morphological abnormalities of the sperm tail. Hum Genet 2020; 140:21-42. [PMID: 31950240 DOI: 10.1007/s00439-020-02113-x] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/06/2020] [Indexed: 12/29/2022]
Abstract
Spermatozoa contain highly specialized structural features reflecting unique functions required for fertilization. Among them, the flagellum is a sperm-specific organelle required to generate the motility, which is essential to reach the egg. The flagellum integrity is, therefore, critical for normal sperm function and flagellum defects consistently lead to male infertility due to reduced or absent sperm motility defined as asthenozoospermia. Multiple morphological abnormalities of the flagella (MMAF), also called short tails, is among the most severe forms of sperm flagellum defects responsible for male infertility and is characterized by the presence in the ejaculate of spermatozoa being short, coiled, absent and of irregular caliber. Recent studies have demonstrated that MMAF is genetically heterogeneous which is consistent with the large number of proteins (over one thousand) localized in the human sperm flagella. In the past 5 years, genomic investigation of the MMAF phenotype allowed the identification of 18 genes whose mutations induce MMAF and infertility. Here we will review information about those genes including their expression pattern, the features of the encoded proteins together with their localization within the different flagellar protein complexes (axonemal or peri-axonemal) and their potential functions. We will categorize the identified MMAF genes following the protein complexes, functions or biological processes they may be associated with, based on the current knowledge in the field.
Collapse
Affiliation(s)
- Aminata Touré
- Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France.,INSERM U1016, Institut Cochin, 75014, Paris, France.,Centre National de La Recherche Scientifique UMR8104, 75014, Paris, France
| | - Guillaume Martinez
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Univ. Grenoble Alpes, 38000, Grenoble, France.,CHU Grenoble Alpes, UM de Génétique Chromosomique, 38000, Grenoble, France
| | - Zine-Eddine Kherraf
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Univ. Grenoble Alpes, 38000, Grenoble, France.,CHU Grenoble Alpes, UM GI-DPI, 38000, Grenoble, France
| | - Caroline Cazin
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Univ. Grenoble Alpes, 38000, Grenoble, France
| | - Julie Beurois
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Univ. Grenoble Alpes, 38000, Grenoble, France
| | - Christophe Arnoult
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Univ. Grenoble Alpes, 38000, Grenoble, France
| | - Pierre F Ray
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Univ. Grenoble Alpes, 38000, Grenoble, France.,CHU Grenoble Alpes, UM GI-DPI, 38000, Grenoble, France
| | - Charles Coutton
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Team Genetics Epigenetics and Therapies of Infertility, Univ. Grenoble Alpes, 38000, Grenoble, France. .,CHU Grenoble Alpes, UM de Génétique Chromosomique, 38000, Grenoble, France.
| |
Collapse
|
29
|
Zhang L, Zhen J, Huang Q, Liu H, Li W, Zhang S, Min J, Li Y, Shi L, Woods J, Chen X, Shi Y, Liu Y, Hess RA, Song S, Zhang Z. Mouse spermatogenesis-associated protein 1 (SPATA1), an IFT20 binding partner, is an acrosomal protein. Dev Dyn 2020; 249:543-555. [PMID: 31816150 DOI: 10.1002/dvdy.141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Intraflagellar transport is a motor-driven trafficking system that is required for the formation of cilia. Intraflagellar transport protein 20 (IFT20) is a master regulator for the control of spermatogenesis and male fertility in mice. However, the mechanism of how IFT20 regulates spermatogenesis is unknown. RESULTS Spermatogenesis associated 1 (SPATA1) was identified to be a major potential binding partner of IFT20 by a yeast two-hybrid screening. The interaction between SPATA1 and IFT20 was examined by direct yeast two-hybrid, co-localization, and co-immunoprecipitation assays. SPATA1 is highly abundant in the mouse testis, and is also expressed in the heart and kidney. During the first wave of spermatogenesis, SPATA1 is detectable at postnatal day 24 and its expression is increased at day 30 and 35. Immunofluorescence staining of mouse testis sections and epididymal sperm demonstrated that SPATA1 is localized mainly in the acrosome of developing spermatids but not in epididymal sperm. IFT20 is also present in the acrosome area of round spermatids. In conditional Ift20 knockout mice, testicular expression level and acrosomal localization of SPATA1 are not changed. CONCLUSIONS SPATA1 is an IFT20 binding protein and may provide a docking site for IFT20 complex binding to the acrosome area.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Jingkai Zhen
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Qian Huang
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Hong Liu
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Shiyang Zhang
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Jie Min
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yuhong Li
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Lin Shi
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - James Woods
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Xuequn Chen
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Yuqin Shi
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yunhao Liu
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois
| | - Shizhen Song
- Department of Occupational and Environmental Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan, China
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, Michigan.,Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan
| |
Collapse
|
30
|
Wang Z, Shi Y, Ma S, Huang Q, Yap YT, Shi L, Zhang S, Zhou T, Li W, Hu B, Zhang L, Krawetz SA, Pazour GJ, Hess RA, Zhang Z. Abnormal fertility, acrosome formation, IFT20 expression and localization in conditional Gmap210 knockout mice. Am J Physiol Cell Physiol 2020; 318:C174-C190. [PMID: 31577511 PMCID: PMC6985835 DOI: 10.1152/ajpcell.00517.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 01/06/2023]
Abstract
GMAP210 (TRIP11) is a cis-Golgi network-associated protein and a Golgi membrane receptor for IFT20, an intraflagellar transport component essential for male fertility and spermiogenesis in mice. To investigate the role of GMAP210 in male fertility and spermatogenesis, floxed Gmap210 mice were bred with Stra8-iCre mice so that the Gmap210 gene is disrupted in spermatocytes and spermatids in this study. The Gmap210flox/flox: Stra8-iCre mutant mice showed no gross abnormalities and survived to adulthood. In adult males, testis and body weights showed no difference between controls and mutant mice. Low-magnification histological examination of the testes revealed normal seminiferous tubule structure, but sperm counts and fertility were significantly reduced in mutant mice compared with controls. Higher resolution examination of the mutant seminiferous epithelium showed that nearly all sperm had more oblong, abnormally shaped heads, while the sperm tails appeared to have normal morphology. Electron microscopy also revealed abnormally shaped sperm heads but normal axoneme core structure; some sperm showed membrane defects in the midpiece. In mutant mice, expression levels of IFT20 and other selective acrosomal proteins were significantly reduced, and their localization was also affected. Peanut-lectin, an acrosome maker, was almost absent in the spermatids and epididymal sperm. Mitochondrion staining was highly concentrated in the heads of sperm, suggesting that the midpieces were coiling around or aggregating near the heads. Defects in acrosome biogenesis were further confirmed by electron microscopy. Collectively, our findings suggest that GMAP210 is essential for acrosome biogenesis, normal mitochondrial sheath formation, and male fertility, and it determines expression levels and acrosomal localization of IFT20 and other acrosomal proteins.
Collapse
Affiliation(s)
- Zhenyu Wang
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Yuqin Shi
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
| | - Suheng Ma
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Qian Huang
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Yi Tian Yap
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Lin Shi
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Shiyang Zhang
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Ting Zhou
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, Michigan
| | - Bo Hu
- Department of Neurology, Wayne State University, Detroit, Michigan
| | - Ling Zhang
- School of Medicine, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China
| | - Stephen A Krawetz
- Department of Obstetrics/Gynecology and Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, Michigan
- Department of Obstetrics/Gynecology and Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan
| |
Collapse
|
31
|
Chen M, Yan H, Wang K, Cui Y, Chen R, Liu J, Zhu H, Qu L, Pan C. Goat SPEF2: Expression profile, indel variants identification and association analysis with litter size. Theriogenology 2019; 139:147-155. [DOI: 10.1016/j.theriogenology.2019.08.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/04/2019] [Accepted: 08/04/2019] [Indexed: 01/04/2023]
|
32
|
Nakamura N, Sloper DT, Del Valle PL. Gene expression profiling of cultured mouse testis fragments treated with ethinylestradiol. J Toxicol Sci 2019; 44:667-679. [PMID: 31588058 DOI: 10.2131/jts.44.667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The assessment of xenobiotic-induced testicular toxicity is important in drug development. Nonetheless, in vitro models to test drugs and chemicals that may cause testicular toxicity are lacking, requiring the continued use of animal models for those studies. We previously evaluated an in vitro mouse testis organ culture system using ethinylestradiol (EE), a well-studied testicular toxicant, and demonstrated a dose-dependent relationship between adverse effects to germ cell differentiation and increasing EE concentrations. However, we terminated that study after 20 days of culture due to oxygen deficiency during germ cell differentiation. Therefore, in the current study, we aimed to identify gene(s) with potential for supporting the histopathological evaluations of testicular toxicity using in vitro testis organ culture system. We cultured testis fragments obtained from mice at postnatal day (PND) 5 in α-Minimal Essential Medium containing 40 mg/mL AlbuMAX™ I and treated them with 0.01 or 1 nM EE on day 1 of culture. On day 20, we collected testis fragments for RNA sequencing analysis and quantitative polymerase chain reaction (qPCR). We found that phospholipase C, zeta 1 and testis-specific serine kinase 4 genes, that are involved in spermatogenesis and predominantly expressed in the testis, were significantly reduced in testis fragments treated with the highest concentration of EE. Also, cytochrome P450, family 26, subfamily b, polypeptide 1 (Cyp26b1) and interleukin 16 (Il16) were up-regulated in the highest EE-treated groups. Further studies are needed to confirm the variations of these gene expression using other testicular toxicants.
Collapse
Affiliation(s)
- Noriko Nakamura
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration, USA
| | - Daniel T Sloper
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration, USA
| | - Pedro L Del Valle
- Center for Drug Evaluation and Research, Food and Drug Administration, USA
| |
Collapse
|
33
|
Moye AR, Bedoni N, Cunningham JG, Sanzhaeva U, Tucker ES, Mathers P, Peter VG, Quinodoz M, Paris LP, Coutinho-Santos L, Camacho P, Purcell MG, Winkelmann AC, Foster JA, Pugacheva EN, Rivolta C, Ramamurthy V. Mutations in ARL2BP, a protein required for ciliary microtubule structure, cause syndromic male infertility in humans and mice. PLoS Genet 2019; 15:e1008315. [PMID: 31425546 PMCID: PMC6715254 DOI: 10.1371/journal.pgen.1008315] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 08/29/2019] [Accepted: 07/17/2019] [Indexed: 12/30/2022] Open
Abstract
Cilia are evolutionarily conserved hair-like structures with a wide spectrum of key biological roles, and their dysfunction has been linked to a growing class of genetic disorders, known collectively as ciliopathies. Many strides have been made towards deciphering the molecular causes for these diseases, which have in turn expanded the understanding of cilia and their functional roles. One recently-identified ciliary gene is ARL2BP, encoding the ADP-Ribosylation Factor Like 2 Binding Protein. In this study, we have identified multiple ciliopathy phenotypes associated with mutations in ARL2BP in human patients and in a mouse knockout model. Our research demonstrates that spermiogenesis is impaired, resulting in abnormally shaped heads, shortened and mis-assembled sperm tails, as well as in loss of axonemal doublets. Additional phenotypes in the mouse included enlarged ventricles of the brain and situs inversus. Mouse embryonic fibroblasts derived from knockout animals revealed delayed depolymerization of primary cilia. Our results suggest that ARL2BP is required for the structural maintenance of cilia as well as of the sperm flagellum, and that its deficiency leads to syndromic ciliopathy. The flagellated tails of sperm cells require a stringent developmental process that is essential for motility and fertility. The components that comprise the sperm tail assemble in regulated steps with protein processing, transport, and structural assembly dependent on each other for sperm tail maturity. In this work, we have identified ARL2BP, a previously retinal-associated protein, to be essential for sperm tail development and assembly. We show that without functional ARL2BP in humans or mice, sperm tails fail to develop, starting with the assembly of the core microtubular structure within the tail. Loss of ARL2BP also effects other ciliated cells, indicating a unique role for ARL2BP in ciliary microtubule formation. This research on ARL2BP provides further understanding on the links between vision and fertility. This work also demonstrates how genomic studies for human patients and murine models can coincide to provide greater insight into disease.
Collapse
Affiliation(s)
- Abigail R. Moye
- Department of Ophthalmology, West Virginia University, Morgantown, United States of America
- Department of Biochemistry, West Virginia University, Morgantown, United States of America
| | - Nicola Bedoni
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Jessica G. Cunningham
- Rockefeller Neurosciences Institute, West Virginia University, Morgantown, WV, United States of America
| | - Urikhan Sanzhaeva
- Department of Biochemistry, West Virginia University, Morgantown, United States of America
| | - Eric S. Tucker
- Rockefeller Neurosciences Institute, West Virginia University, Morgantown, WV, United States of America
| | - Peter Mathers
- Department of Ophthalmology, West Virginia University, Morgantown, United States of America
- Department of Biochemistry, West Virginia University, Morgantown, United States of America
- Rockefeller Neurosciences Institute, West Virginia University, Morgantown, WV, United States of America
| | - Virginie G. Peter
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Mathieu Quinodoz
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Liliana P. Paris
- Department of Ophthalmology, Instituto de Oftalmologia Dr Gama Pinto, Lisbon, Portugal
| | - Luísa Coutinho-Santos
- Department of Ophthalmology, Instituto de Oftalmologia Dr Gama Pinto, Lisbon, Portugal
| | - Pedro Camacho
- Department of Ophthalmology, Instituto de Oftalmologia Dr Gama Pinto, Lisbon, Portugal
| | - Madeleine G. Purcell
- Department of Biology, Randolph-Macon College, Ashland, VA, United States of America
| | - Abbie C. Winkelmann
- Department of Biology, Randolph-Macon College, Ashland, VA, United States of America
| | - James A. Foster
- Department of Biology, Randolph-Macon College, Ashland, VA, United States of America
| | - Elena N. Pugacheva
- Department of Biochemistry, West Virginia University, Morgantown, United States of America
| | - Carlo Rivolta
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
- Clinical Research Center, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- Department of Ophthalmology, University Hospital Basel, Switzerland
- * E-mail: (CR); (VR)
| | - Visvanathan Ramamurthy
- Department of Ophthalmology, West Virginia University, Morgantown, United States of America
- Department of Biochemistry, West Virginia University, Morgantown, United States of America
- Rockefeller Neurosciences Institute, West Virginia University, Morgantown, WV, United States of America
- * E-mail: (CR); (VR)
| |
Collapse
|
34
|
Liu Y, Zhang L, Li W, Li Y, Liu J, Zhang S, Pin G, Song S, Ray PF, Arnoult C, Cho C, Garcia-Reyes B, Knippschild U, Strauss JF, Zhang Z. The sperm-associated antigen 6 interactome and its role in spermatogenesis. Reproduction 2019; 158:181-197. [PMID: 31146259 PMCID: PMC7368494 DOI: 10.1530/rep-18-0522] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
Abstract
Mammalian SPAG6, the orthologue of Chlamydomonas reinhardtii PF16, is a component of the central apparatus of the '9 + 2' axoneme that controls ciliary/flagellar motility, including sperm motility. Recent studies revealed that SPAG6 has functions beyond its role in the central apparatus. Hence, we reexamined the role of SPAG6 in male fertility. In wild-type mice, SPAG6 was present in cytoplasmic vesicles in spermatocytes, the acrosome of round and elongating spermatids and the manchette of elongating spermatids. Spag6-deficient testes showed abnormal spermatogenesis, with abnormalities in male germ cell morphology consistent with the multi-compartment pattern of SPAG6 localization. The armadillo repeat domain of mouse SPAG6 was used as a bait in a yeast two-hybrid screen, and several proteins with diverse functions appeared multiple times, including Snapin, SPINK2 and COPS5. Snapin has a similar localization to SPAG6 in male germ cells, and SPINK2, a key protein in acrosome biogenesis, was dramatically reduced in Spag6-deficient mice which have defective acrosomes. SPAG16L, another SPAG6-binding partner, lost its localization to the manchette in Spag6-deficient mice. Our findings demonstrate that SPAG6 is a multi-functional protein that not only regulates sperm motility, but also plays roles in spermatogenesis in multiple cellular compartments involving multiple protein partners.
Collapse
Affiliation(s)
- Yunhao Liu
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Ling Zhang
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, MI, 48201
| | - Yuhong Li
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Junpin Liu
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Shiyang Zhang
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Guanglun Pin
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Shizhen Song
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Pierre F Ray
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Christophe Arnoult
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Chunghee Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Balbina Garcia-Reyes
- Department of General and Visceral Surgery, Ulm University, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Jerome F. Strauss
- Department of Obstetrics/Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, MI, 48201
- Department of Obstetrics/Gynecology, Wayne State University, Detroit, MI, 48201
| |
Collapse
|
35
|
Sha Y, Liu W, Wei X, Zhu X, Luo X, Liang L, Guo T. Biallelic mutations in Sperm flagellum 2 cause human multiple morphological abnormalities of the sperm flagella (MMAF) phenotype. Clin Genet 2019; 96:385-393. [PMID: 31278745 DOI: 10.1111/cge.13602] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 01/05/2023]
Abstract
Male patients with multiple morphological abnormalities of the sperm flagella (MMAF) are infertile and exhibit absent, short, coiled, bent and/or irregular sperm flagella. Mutations in the SPEF2 gene reduce sperm motility and cause sperm tail defects in animal models and humans. In the present study, we performed a genetic analysis on an MMAF patient and identified novel biallelic mutations in the SPEF2 gene. The biallelic mutations were confirmed by Sanger sequencing and in silico analysis revealed that, these variations were deleterious. The expression of truncated SPEF2 protein was reduced significantly in the patient's spermatozoa. The spermatozoa harbored biallelic mutations and showed severe ultrastructural defects in the axoneme and mitochondrial sheath. Our data suggest that biallelic mutations in SPEF2 can cause severe sperm flagellum defects, thus providing a novel candidate genetic pathogen for the human MMAF phenotype.
Collapse
Affiliation(s)
- Yanwei Sha
- Department of Andrology, United Diagnostic and Research Center for Clinical Genetics, School of Public Health & Women and Children's Hospital, Xiamen University, Xiamen, Fujian, China
| | - Wensheng Liu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xiaoli Wei
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xingshen Zhu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xiangmin Luo
- Reproductive Medicine Center, the Second Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lei Liang
- Department of Andrology, United Diagnostic and Research Center for Clinical Genetics, School of Public Health & Women and Children's Hospital, Xiamen University, Xiamen, Fujian, China
| | - Tonghang Guo
- Reproductive and Genetic Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
36
|
Liu W, Sha Y, Li Y, Mei L, Lin S, Huang X, Lu J, Ding L, Kong S, Lu Z. Loss-of-function mutations in SPEF2 cause multiple morphological abnormalities of the sperm flagella (MMAF). J Med Genet 2019; 56:678-684. [PMID: 31151990 DOI: 10.1136/jmedgenet-2018-105952] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/10/2019] [Accepted: 05/02/2019] [Indexed: 11/04/2022]
Abstract
BACKGROUND Multiple morphological abnormalities of the sperm flagella (MMAF) is a kind of severe teratozoospermia. Patients with the MMAF phenotype are infertile and present aberrant spermatozoa with absent, short, coiled, bent and/or irregular flagella. Mutations in several genes can explain approximately 30%-50% of MMAF cases and more genetic pathogenies need to be explored. SPEF2 was previously demonstrated to play an essential role in sperm tail development in mice and pig. Dysfunctional mutations in SPEF2 impair sperm motility and cause a short-tail phenotype in both animal models. OBJECTIVE Based on 42 patients with severe infertility and MMAF phenotype, we explored the new genetic cause of human MMAF phenotype. METHODS AND RESULTS By screening gene variants in 42 patients with MMAF using whole exome sequencing, we identified the c. 12delC, c. 1745-2A > G, c. 4102 G > T and c. 4323dupA mutations in the SPEF2 gene from two patients. Both of these mutations are rare and potentially deleterious. Transmission electron microscope (TEM) analysis showed a disrupted axonemal structure with mitochondrial sheath defects in the patients' spermatozoa. The SPEF2 protein level was significantly decreased in the spermatozoa of the patients revealed by Western blot (WB) and immunofluorescence (IF) analyses. CONCLUSION Our experimental findings indicate that loss-of-function mutations in the SPEF2 gene can cause the MMAF phenotype in human.
Collapse
Affiliation(s)
- Wensheng Liu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| | - Yanwei Sha
- Department of Reproductive Medicine, Xiamen Maternity and Child Care Hospital, Xiamen, China
| | - Yang Li
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| | - Libin Mei
- Department of Reproductive Medicine, Xiamen Maternity and Child Care Hospital, Xiamen, China
| | - Shaobin Lin
- Department of Reproductive Medicine, Xiamen Maternity and Child Care Hospital, Xiamen, China
| | - Xianjing Huang
- Department of Reproductive Medicine, Xiamen Maternity and Child Care Hospital, Xiamen, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Lu Ding
- Department of Reproductive Medicine, Xiamen Maternity and Child Care Hospital, Xiamen, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Zhongxian Lu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| |
Collapse
|
37
|
Liu C, Lv M, He X, Zhu Y, Amiri-Yekta A, Li W, Wu H, Kherraf ZE, Liu W, Zhang J, Tan Q, Tang S, Zhu YJ, Zhong Y, Li C, Tian S, Zhang Z, Jin L, Ray P, Zhang F, Cao Y. Homozygous mutations in SPEF2 induce multiple morphological abnormalities of the sperm flagella and male infertility. J Med Genet 2019; 57:31-37. [PMID: 31048344 DOI: 10.1136/jmedgenet-2019-106011] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/21/2019] [Accepted: 04/02/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Male infertility due to multiple morphological abnormalities of the sperm flagella (MMAF) is a genetically heterogeneous disorder. Previous studies revealed several MMAF-associated genes, which account for approximately 60% of human MMAF cases. The pathogenic mechanisms of MMAF remain to be illuminated. METHODS AND RESULTS We conducted genetic analyses using whole-exome sequencing in 50 Han Chinese probands with MMAF. Two homozygous stop-gain variants (c.910C>T (p.Arg304*) and c.3400delA (p.Ile1134Serfs*13)) of the SPEF2 (sperm flagellar 2) gene were identified in two unrelated consanguineous families. Consistently, an Iranian subject from another cohort also carried a homozygous SPEF2 stop-gain variant (c.3240delT (p.Phe1080Leufs*2)). All these variants affected the long SPEF2 transcripts that are expressed in the testis and encode the IFT20 (intraflagellar transport 20) binding domain, important for sperm tail development. Notably, previous animal studies reported spontaneous mutations of SPEF2 causing sperm tail defects in bulls and pigs. Our further functional studies using immunofluorescence assays showed the absence or a remarkably reduced staining of SPEF2 and of the MMAF-associated CFAP69 protein in the spermatozoa from SPEF2-affected subjects. CONCLUSIONS We identified SPEF2 as a novel gene for human MMAF across the populations. Functional analyses suggested that the deficiency of SPEF2 in the mutated subjects could alter the localisation of other axonemal proteins.
Collapse
Affiliation(s)
- Chunyu Liu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Yong Zhu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Amir Amiri-Yekta
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble, UM GI-DPI, Grenoble, France.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture, and Research, Tehran, Iran
| | - Weiyu Li
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Zine-Eddine Kherraf
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble, UM GI-DPI, Grenoble, France
| | - Wangjie Liu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingjing Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Qing Tan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Shuyan Tang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yong-Jun Zhu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yading Zhong
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Caihua Li
- Genesky Biotechnologies Inc, Shanghai, China
| | - Shixiong Tian
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Li Jin
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Pierre Ray
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,Centre Hospitalier Universitaire de Grenoble, UM GI-DPI, Grenoble, France
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China .,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China .,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| |
Collapse
|
38
|
Liu H, Li W, Zhang Y, Zhang Z, Shang X, Zhang L, Zhang S, Li Y, Somoza AV, Delpi B, Gerton GL, Foster JA, Hess RA, Pazour GJ, Zhang Z. IFT25, an intraflagellar transporter protein dispensable for ciliogenesis in somatic cells, is essential for sperm flagella formation. Biol Reprod 2018; 96:993-1006. [PMID: 28430876 DOI: 10.1093/biolre/iox029] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/13/2017] [Indexed: 12/25/2022] Open
Abstract
Intraflagellar transport (IFT) is a conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella. However, IFT25, a component of the IFT complex, is not required for the formation of cilia in somatic tissues. In mice, the gene is highly expressed in the testis, and its expression is upregulated during the final phase when sperm flagella are formed. To investigate the role of IFT25 in sperm flagella formation, the gene was specifically disrupted in male germ cells. All homozygous knockout mice survived to adulthood and did not show any gross abnormalities. However, all homozygous knockout males were completely infertile. Sperm numbers were reduced and these sperm were completely immotile. Multiple morphological abnormalities were observed in sperm, including round heads, short and bent tails, with some tails showing branched flagella and others with frequent abnormal thicknesses, as well as swollen tips of the tail. Transmission electron microscopy revealed that flagellar accessory structures, including the fibrous sheath and outer dense fibers, were disorganized, and most sperm had also lost the "9+2" microtubule structure. In the testis, IFT25 forms a complex with other IFT proteins. In Ift25 knockout testes, IFT27, an IFT25 binding partner, was missing, and IFT20 and IFT81 levels were also reduced. Our findings suggest that IFT25, although not necessary for the formation of cilia in somatic cells, is indispensable for sperm flagellum formation and male fertility in mice.
Collapse
Affiliation(s)
- Hong Liu
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Wei Li
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yong Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Dermatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengang Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuejun Shang
- Department of Andrology, Jinling Hospital, Nanjing University, School of Medicine, Nanjing, China
| | - Ling Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Shiyang Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yanwei Li
- Department of Computer Science, Wellesley College, Wellesley, Massachusetts, USA
| | - Andres V Somoza
- Department of Humanities and Sciences, Honor College, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Brandon Delpi
- Department of Biology, Randolph-Macon College, Ashland, Virginia, USA
| | - George L Gerton
- Center for Research on Reproduction and Women's Health Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James A Foster
- Department of Biology, Randolph-Macon College, Ashland, Virginia, USA
| | - Rex A Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, USA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zhibing Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
39
|
Dong FN, Amiri-Yekta A, Martinez G, Saut A, Tek J, Stouvenel L, Lorès P, Karaouzène T, Thierry-Mieg N, Satre V, Brouillet S, Daneshipour A, Hosseini SH, Bonhivers M, Gourabi H, Dulioust E, Arnoult C, Touré A, Ray PF, Zhao H, Coutton C. Absence of CFAP69 Causes Male Infertility due to Multiple Morphological Abnormalities of the Flagella in Human and Mouse. Am J Hum Genet 2018; 102:636-648. [PMID: 29606301 DOI: 10.1016/j.ajhg.2018.03.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/05/2018] [Indexed: 10/17/2022] Open
Abstract
The multiple morphological abnormalities of the flagella (MMAF) phenotype is among the most severe forms of sperm defects responsible for male infertility. The phenotype is characterized by the presence in the ejaculate of immotile spermatozoa with severe flagellar abnormalities including flagella being short, coiled, absent, and of irregular caliber. Recent studies have demonstrated that MMAF is genetically heterogeneous, and genes thus far associated with MMAF account for only one-third of cases. Here we report the identification of homozygous truncating mutations (one stop-gain and one splicing variant) in CFAP69 of two unrelated individuals by whole-exome sequencing of a cohort of 78 infertile men with MMAF. CFAP69 encodes an evolutionarily conserved protein found at high levels in the testis. Immunostaining experiments in sperm from fertile control individuals showed that CFAP69 localized to the midpiece of the flagellum, and the absence of CFAP69 was confirmed in both individuals carrying CFPA69 mutations. Additionally, we found that sperm from a Cfap69 knockout mouse model recapitulated the MMAF phenotype. Ultrastructural analysis of testicular sperm from the knockout mice showed severe disruption of flagellum structure, but histological analysis of testes from these mice revealed the presence of all stages of the seminiferous epithelium, indicating that the overall progression of spermatogenesis is preserved and that the sperm defects likely arise during spermiogenesis. Together, our data indicate that CFAP69 is necessary for flagellum assembly/stability and that in both humans and mice, biallelic truncating mutations in CFAP69 cause autosomal-recessive MMAF and primary male infertility.
Collapse
|
40
|
Shi Y, Liu W, He M. Proteome and Transcriptome Analysis of Ovary, Intersex Gonads, and Testis Reveals Potential Key Sex Reversal/Differentiation Genes and Mechanism in Scallop Chlamys nobilis. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2018; 20:220-245. [PMID: 29546597 DOI: 10.1007/s10126-018-9800-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 02/19/2018] [Indexed: 06/08/2023]
Abstract
Bivalve mollusks exhibit hermaphroditism and sex reversal/differentiation. Studies generally focus on transcriptional profiling and specific genes related to sex determination and differentiation. Few studies on sex reversal/differentiation have been reported. A combination analysis of gonad proteomics and transcriptomics was conducted on Chlamys nobilis to provide a systematic understanding of sex reversal/differentiation in bivalves. We obtained 4258 unique peptides and 93,731 unigenes with good correlation between messenger RNA and protein levels. Candidate genes in sex reversal/differentiation were found: 15 genes differentially expressed between sexes were identified and 12 had obvious sexual functions. Three novel genes (foxl2, β-catenin, and sry) were expressed highly in intersex individuals and were likely involved in the control of gonadal sex in C. nobilis. High expression of foxl2 or β-catenin may inhibit sry and activate 5-HT receptor and vitellogenin to maintain female development. High expression of sry may inhibit foxl2 and β-catenin and activate dmrt2, fem-1, sfp2, sa6, Amy-1, APCP4, and PLK to maintain male function. High expression of sry, foxl2, and β-catenin in C. nobilis may be involved in promoting and maintaining sex reversal/differentiation. The downstream regulator may not be dimorphic expressed genes, but genes expressed in intersex individuals, males and females. Different expression patterns of sex-related genes and gonadal histological characteristics suggested that C. nobilis may change its sex from male to female. These findings suggest highly conserved sex reversal/differentiation with diverged regulatory pathways during C. nobilis evolution. This study provides valuable genetic resources for understanding sex reversal/differentiation (intersex) mechanisms and pathways underlying bivalve reproductive regulation.
Collapse
Affiliation(s)
- Yu Shi
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
| | - Wenguang Liu
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
| | - Maoxian He
- CAS Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China.
- Key Laboratory of Marine Bio-resources Sustainable Utilization, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China.
| |
Collapse
|
41
|
Lehti MS, Henriksson H, Rummukainen P, Wang F, Uusitalo-Kylmälä L, Kiviranta R, Heino TJ, Kotaja N, Sironen A. Cilia-related protein SPEF2 regulates osteoblast differentiation. Sci Rep 2018; 8:859. [PMID: 29339787 PMCID: PMC5770417 DOI: 10.1038/s41598-018-19204-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 12/22/2017] [Indexed: 11/09/2022] Open
Abstract
Sperm flagellar protein 2 (SPEF2) is essential for motile cilia, and lack of SPEF2 function causes male infertility and primary ciliary dyskinesia. Cilia are pointing out from the cell surface and are involved in signal transduction from extracellular matrix, fluid flow and motility. It has been shown that cilia and cilia-related genes play essential role in commitment and differentiation of chondrocytes and osteoblasts during bone formation. Here we show that SPEF2 is expressed in bone and cartilage. The analysis of a Spef2 knockout (KO) mouse model revealed hydrocephalus, growth retardation and death prior to five weeks of age. To further elucidate the causes of growth retardation we analyzed the bone structure and possible effects of SPEF2 depletion on bone formation. In Spef2 KO mice, long bones (tibia and femur) were shorter compared to wild type, and X-ray analysis revealed reduced bone mineral content. Furthermore, we showed that the in vitro differentiation of osteoblasts isolated from Spef2 KO animals was compromised. In conclusion, this study reveals a novel function for SPEF2 in bone formation through regulation of osteoblast differentiation and bone growth.
Collapse
Affiliation(s)
- Mari S Lehti
- Natural Resources Institute Finland (Luke), Green Technology, FI-31600, Jokioinen, Finland.,Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Henna Henriksson
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Petri Rummukainen
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Fan Wang
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | | | - Riku Kiviranta
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland.,Department of Endocrinology, Division of Medicine, University of Turku and Turku University Hospital, FI-20520, Turku, Finland
| | - Terhi J Heino
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Noora Kotaja
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Anu Sironen
- Natural Resources Institute Finland (Luke), Green Technology, FI-31600, Jokioinen, Finland.
| |
Collapse
|
42
|
Martins LR, Bung RK, Koch S, Richter K, Schwarzmüller L, Terhardt D, Kurtulmus B, Niehrs C, Rouhi A, Lohmann I, Pereira G, Fröhling S, Glimm H, Scholl C. Stk33 is required for spermatid differentiation and male fertility in mice. Dev Biol 2018; 433:84-93. [DOI: 10.1016/j.ydbio.2017.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 11/29/2022]
|
43
|
Lehti MS, Sironen A. Formation and function of sperm tail structures in association with sperm motility defects†. Biol Reprod 2017; 97:522-536. [DOI: 10.1093/biolre/iox096] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022] Open
|
44
|
Lehti MS, Zhang FP, Kotaja N, Sironen A. SPEF2 functions in microtubule-mediated transport in elongating spermatids to ensure proper male germ cell differentiation. Development 2017; 144:2683-2693. [PMID: 28619825 DOI: 10.1242/dev.152108] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/11/2017] [Indexed: 11/20/2022]
Abstract
Sperm differentiation requires specific protein transport for correct sperm tail formation and head shaping. A transient microtubular structure, the manchette, appears around the differentiating spermatid head and serves as a platform for protein transport to the growing tail. Sperm flagellar 2 (SPEF2) is known to be essential for sperm tail development. In this study we investigated the function of SPEF2 during spermatogenesis using a male germ cell-specific Spef2 knockout mouse model. In addition to defects in sperm tail development, we observed a duplication of the basal body and failure in manchette migration resulting in an abnormal head shape. We identified cytoplasmic dynein 1 and GOLGA3 as novel interaction partners for SPEF2. SPEF2 and dynein 1 colocalize in the manchette and the inhibition of dynein 1 disrupts the localization of SPEF2 to the manchette. Furthermore, the transport of a known SPEF2-binding protein, IFT20, from the Golgi complex to the manchette was delayed in the absence of SPEF2. These data indicate a possible novel role of SPEF2 as a linker protein for dynein 1-mediated cargo transport along microtubules.
Collapse
Affiliation(s)
- Mari S Lehti
- Natural Resources Institute Finland (Luke), Green Technology, FI-31600 Jokioinen, Finland.,Department of Physiology, Institute of Biomedicine, University of Turku, FI-20520 Turku, Finland
| | - Fu-Ping Zhang
- Department of Physiology, Institute of Biomedicine, University of Turku, FI-20520 Turku, Finland.,Turku Center for Disease Modeling, University of Turku, FI-20520 Turku, Finland
| | - Noora Kotaja
- Department of Physiology, Institute of Biomedicine, University of Turku, FI-20520 Turku, Finland
| | - Anu Sironen
- Natural Resources Institute Finland (Luke), Green Technology, FI-31600 Jokioinen, Finland
| |
Collapse
|
45
|
de Simoni Gouveia JJ, Paiva SR, McManus CM, Caetano AR, Kijas JW, Facó O, Azevedo HC, de Araujo AM, de Souza CJH, Yamagishi MEB, Carneiro PLS, Braga Lôbo RN, de Oliveira SMP, da Silva MVG. Genome-wide search for signatures of selection in three major Brazilian locally adapted sheep breeds. Livest Sci 2017. [DOI: 10.1016/j.livsci.2017.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
46
|
The impact of RABL2B gene (rs144944885) on human male infertility in patients with oligoasthenoteratozoospermia and immotile short tail sperm defects. J Assist Reprod Genet 2017; 34:505-510. [PMID: 28138870 DOI: 10.1007/s10815-016-0863-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/19/2016] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Male infertility is a multifactorial disorder with impressively genetic basis; besides, sperm abnormalities are the cause of numerous cases of male infertility. In this study, we evaluated the genetic variants in exons 4 and 5 and their intron-exon boundaries in RABL2B gene in infertile men with oligoasthenoteratozoospermia (OAT) and immotile short tail sperm (ISTS) defects to define if there is any association between these variants and human male infertility. METHODS To this purpose, DNA was extracted from peripheral blood and after PCR reaction and sequencing, the results of sequenced segments were analyzed. In the present study, 30 infertile men with ISTS defect and 30 oligoasthenoteratozoospermic infertile men were recruited. All men were of Iranian origin and it took 3 years to collect patient's samples with ISTS defect. RESULTS As a result, the 50776482 delC intronic variant (rs144944885) was identified in five patients with oligoasthenoteratozoospermia defect and one patient with ISTS defect in heterozygote form. This variant was not identified in controls. The allelic frequency of the 50776482 delC variant was significantly statistically higher in oligoasthenoteratozoospermic infertile men (p < 0.05). Bioinformatics studies suggested that the 50776482 delC allele would modify the splicing of RABL2B pre-mRNA. In addition, we identified a new genetic variant in RABL2B gene. CONCLUSIONS According to the present study, 50776482 delC allele in the RABL2B gene could be a risk factor in Iranian infertile men with oligoasthenoteratozoospermia defect, but more genetic studies are required to understand the accurate role of this variant in pathogenesis of human male infertility.
Collapse
|
47
|
Teves ME, Nagarkatti-Gude DR, Zhang Z, Strauss JF. Mammalian axoneme central pair complex proteins: Broader roles revealed by gene knockout phenotypes. Cytoskeleton (Hoboken) 2016; 73:3-22. [PMID: 26785425 DOI: 10.1002/cm.21271] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 11/22/2015] [Accepted: 12/24/2015] [Indexed: 01/09/2023]
Abstract
The axoneme genes, their encoded proteins, their functions and the structures they form are largely conserved across species. Much of our knowledge of the function and structure of axoneme proteins in cilia and flagella is derived from studies on model organisms like the green algae, Chlamydomonas reinhardtii. The core structure of cilia and flagella is the axoneme, which in most motile cilia and flagella contains a 9 + 2 configuration of microtubules. The two central microtubules are the scaffold of the central pair complex (CPC). Mutations that disrupt CPC genes in Chlamydomonas and other model organisms result in defects in assembly, stability and function of the axoneme, leading to flagellar motility defects. However, targeted mutations generated in mice in the orthologous CPC genes have revealed significant differences in phenotypes of mutants compared to Chlamydomonas. Here we review observations that support the concept of cell-type specific roles for the CPC genes in mice, and an expanded repertoire of functions for the products of these genes in cilia, including non-motile cilia, and other microtubule-associated cellular functions.
Collapse
Affiliation(s)
- Maria E Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia
| | - David R Nagarkatti-Gude
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Zhibing Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
48
|
Zhang Z, Li W, Zhang Y, Zhang L, Teves ME, Liu H, Strauss JF, Pazour GJ, Foster JA, Hess RA, Zhang Z. Intraflagellar transport protein IFT20 is essential for male fertility and spermiogenesis in mice. Mol Biol Cell 2016; 27:mbc.E16-05-0318. [PMID: 27682589 PMCID: PMC5170554 DOI: 10.1091/mbc.e16-05-0318] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/06/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022] Open
Abstract
Intraflagellar transport (IFT) is a conserved mechanism thought to be essential for the assembly and maintenance of cilia and flagella. However, little is known about its role in mammalian sperm flagella formation. To fill this gap, we disrupted the Ift20 gene in male germ cells. Homozygous mutant mice were infertile with significantly reduced sperm counts and motility. In addition, abnormally shaped elongating spermatid heads and bulbous round spermatids were found in the lumen of the seminiferous tubules. Electron microscopy revealed increased cytoplasmic vesicles, fiber-like structures, abnormal accumulation of mitochondria and a decrease in mature lysosomes. The few developed sperm had disrupted axonemes and some retained cytoplasmic lobe components on the flagella. ODF2 and SPAG16L, two sperm flagella proteins failed to be incorporated into sperm tails of the mutant mice, and in the germ cells, both were assembled into complexes with lighter density in the absence of IFT20. Disrupting IFT20 did not significantly change expression levels of IFT88, a component of IFT-B complex, and IFT140, a component of IFT-A complex. Even though the expression level of an autophagy core protein that associates with IFT20, ATG16, was reduced in the testis of the Ift20 mutant mice, expression levels of other major autophagy markers, including LC3 and ubiquitin were not changed. Our studies suggest that IFT20 is essential for male fertility and spermiogenesis in mice, and its major function is to transport cargo proteins for sperm flagella formation. It also appears to be involved in removing excess cytoplasmic components.
Collapse
Affiliation(s)
- Zhengang Zhang
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China, 430030 Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Wei Li
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Yong Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298 Department of Dermatology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China, 430030
| | - Ling Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298 School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Maria E Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Hong Liu
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298 School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - James A Foster
- Department of Biology, Randolph-Macon College, Ashland, VA 23005
| | - Rex A Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, 2001 S. Lincoln, Urbana, IL 61802-6199
| | - Zhibing Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| |
Collapse
|
49
|
Abstract
Most motile and all non-motile (also known as primary) eukaryotic cilia possess microtubule-based axonemes that are assembled at the cell surface to form hair-like or more elaborate compartments endowed with motility and/or signaling functions. Such compartmentalized ciliogenesis depends on the core intraflagellar transport (IFT) machinery and the associated Bardet-Biedl syndrome complex (BBSome) for dynamic delivery of ciliary components. The transition zone (TZ), an ultrastructurally complex barrier or 'gate' at the base of cilia, also contributes to the formation of compartmentalized cilia. Yet, some ciliated protists do not have IFT components and, like some metazoan spermatozoa, use IFT-independent mechanisms to build axonemes exposed to the cytosol. Moreover, various ciliated protists lack TZ components, whereas Drosophila sperm surprisingly requires the activity of dynamically localized TZ proteins for cytosolic ciliogenesis. Here, we discuss the various ways eukaryotes use IFT and/or TZ proteins to generate the wide assortment of compartmentalized and cytosolic cilia observed in nature. Consideration of the different ciliogenesis pathways allows us to propose how three types of cytosol-exposed cilia (primary, secondary and tertiary), including cilia found in the human sperm proximal segment, are likely generated by evolutionary derivations of compartmentalized ciliogenesis.
Collapse
|
50
|
Transcriptome analysis of male and female mature gonads of Japanese scallop Patinopecten yessonsis. Genes Genomics 2016. [DOI: 10.1007/s13258-016-0449-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|