1
|
Zhou D, Wu H, Wang L, Wang X, Tang S, Zhou Y, Wang J, Wu B, Tang J, Zhou X, Tian S, Liu S, Lv M, He X, Jin L, Shi H, Zhang F, Cao Y, Liu C. Deficiency of MFSD6L, an acrosome membrane protein, causes oligoasthenoteratozoospermia in humans and mice. J Genet Genomics 2024; 51:1007-1019. [PMID: 38909778 DOI: 10.1016/j.jgg.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/25/2024]
Abstract
Oligoasthenoteratozoospermia is an important factor affecting male fertility and has been found to be associated with genetic factors. However, there are still a proportion of oligoasthenoteratozoospermia cases that cannot be explained by known pathogenic genetic variants. Here, we perform genetic analyses and identify bi-allelic loss-of-function variants of MFSD6L from an oligoasthenoteratozoospermia-affected family. Mfsd6l knock-out male mice also present male subfertility with reduced sperm concentration, motility, and deformed acrosomes. Further mechanistic analyses reveal that MFSD6L, as an acrosome membrane protein, plays an important role in the formation of acrosome by interacting with the inner acrosomal membrane protein SPACA1. Moreover, poor embryonic development is consistently observed after intracytoplasmic sperm injection treatment using spermatozoa from the MFSD6L-deficient man and male mice. Collectively, our findings reveal that MFSD6L is required for the anchoring of sperm acrosome and head shaping. The deficiency of MFSD6L affects male fertility and causes oligoasthenoteratozoospermia in humans and mice.
Collapse
Affiliation(s)
- Dapeng Zhou
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China; Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Lingbo Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, China
| | - Xuemei Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Shuyan Tang
- Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Yiling Zhou
- Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Jiaxiong Wang
- State Key Laboratory of Reproductive Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, China; Suzhou Municipal Hospital, Suzhou, Jiangsu 215002, China
| | - Bangguo Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, China
| | - Jianan Tang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Xuehai Zhou
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Shixiong Tian
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institute of Reproduction and Development, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, China
| | - Shuang Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China; Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China
| | - Huijuan Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Pharmacy, Fudan University, Shanghai 200237, China
| | - Feng Zhang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 200438, China; Institute of Medical Genetics and Genomics, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Embryo Original Diseases, Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui 230032, China.
| | - Chunyu Liu
- Shanghai Key Laboratory of Embryo Original Diseases, Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
2
|
Kumaresan A, Yadav P, Sinha MK, Nag P, John Peter ESK, Mishra JS, Kumar S. Male infertility and perfluoroalkyl and poly-fluoroalkyl substances: evidence for alterations in phosphorylation of proteins and fertility-related functional attributes in bull spermatozoa†. Biol Reprod 2024; 111:723-739. [PMID: 38847481 PMCID: PMC11402523 DOI: 10.1093/biolre/ioae089] [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: 02/09/2024] [Revised: 05/13/2024] [Accepted: 06/05/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Perfluoroalkyl and poly-fluoroalkyl substances (PFAS) are pervasive environmental pollutants and potential threats to reproductive health. Epidemiological studies have established an association between PFAS and male infertility, but the underlying mechanisms are unclear. OBJECTIVES Investigate the effect of perfluorooctane sulfonic acid (PFOS), the most prevalent and representative PFAS, on bull sperm protein phosphorylation and function. METHODS We exposed bull sperm to PFOS at 10 (average population exposure) and 100 μM (high-exposure scenario), and analyzed global proteomic and phosphoproteomic analysis by TMT labeling and Nano LC-MS/MS. We also measured sperm fertility functions by flow cytometry. RESULTS PFOS at 10-μM altered sperm proteins linked to spermatogenesis and chromatin condensation, while at 100 μM, PFOS affected proteins associated with motility and fertility. We detected 299 phosphopeptides from 116 proteins, with 45 exhibiting differential expression between control and PFOS groups. PFOS dysregulated phosphorylation of key proteins (ACRBP, PRKAR2A, RAB2B, SPAG8, TUBB4B, ZPBP, and C2CD6) involved in sperm capacitation, acrosome reaction, sperm-egg interaction, and fertilization. PFOS also affected phosphorylation of other proteins (AQP7, HSBP9, IL4I1, PRKAR1A, and CCT8L2) related to sperm stress resistance and cryotolerance. Notably, four proteins (PRM1, ACRBP, TSSK1B, and CFAP45) exhibited differential regulation at both proteomic and phosphoproteomic levels. Flow cytometric analysis confirmed that PFOS increased protein phosphorylation in sperm and also decreased sperm motility, viability, calcium, and mitochondrial membrane potential and increased mitochondrial ROS in a dose-dependent manner. CONCLUSIONS This study demonstrates that PFOS exposure negatively affects phosphorylation of proteins vital for bull sperm function and fertilization.
Collapse
Affiliation(s)
- Arumugam Kumaresan
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- Theriogenology Laboratory, Southern Regional Station of ICAR National Dairy Research Institute, Bengaluru, Karnataka 560030, India
| | - Pankaj Yadav
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Manish Kumar Sinha
- Theriogenology Laboratory, Southern Regional Station of ICAR National Dairy Research Institute, Bengaluru, Karnataka 560030, India
| | - Pradeep Nag
- Department of Animal Sciences, University of Missouri, Columbia, WI 65211, USA
| | - Ebenezer Samuel King John Peter
- Theriogenology Laboratory, Southern Regional Station of ICAR National Dairy Research Institute, Bengaluru, Karnataka 560030, India
| | - Jay S Mishra
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, WI 53706, USA
| |
Collapse
|
3
|
Fujisawa Y, Kikuchi S, Kuba F, Oishi K, Murayama S, Sugiyama T, Tokito R, Ueno H, Kashiwabara S, Yumura Y, Kurihara Y. Ectopic expression of the mitochondrial protein COXFA4L3 in human sperm acrosome and its potential application in the selection of male infertility treatments. Reprod Med Biol 2024; 23:e12602. [PMID: 39478730 PMCID: PMC11522028 DOI: 10.1002/rmb2.12602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 11/02/2024] Open
Abstract
Purpose Spermatogenesis requires a large amount of energy, which is primarily produced by the mitochondrial electron transfer chain. Mitochondrial dysfunction affects male infertility, suggesting a relationship between the electron transfer chain and male infertility. COXFA4L3 (C15ORF48) is an emerging subunit protein of cytochrome oxidase specifically expressed in germ cells during spermatogenesis, and it may be involved in male infertility. Therefore, to investigate whether COXFA4L3 could be a marker of mitochondrial dysfunction in the sperm, this study examined the protein expression and localization profile of COXFA4L3 in the sperm of male patients with infertility. Methods Twenty-seven semen samples from a male infertility clinic at the Reproductive Center of Yokohama City University Medical Center were used to analyze sperm quality parameters and the expression and localization of energy production-related proteins. These data were compared with the outcomes of infertility treatment. Results The expression levels of COXFA4L3 varied significantly between samples. Furthermore, COXFA4L3 was ectopically localized to the acrosome. Conclusions Ectopic expression of COXFA4L3 and PNA-stained acrosomes may be useful parameters for fertility treatment selection. Assessing the acrosomal localization of COXFA4L3 will expedite pregnancy treatment planning.
Collapse
Affiliation(s)
- Yusuke Fujisawa
- Graduate School of Engineering ScienceYokohama National UniversityKanagawaJapan
| | - Sayaka Kikuchi
- Life Science Laboratory, Technology and Development DivisionKanto Chemical Co., Inc.KanagawaJapan
| | - Fujino Kuba
- Graduate School of Engineering ScienceYokohama National UniversityKanagawaJapan
| | - Kosei Oishi
- Graduate School of Engineering ScienceYokohama National UniversityKanagawaJapan
| | - Soushi Murayama
- Graduate School of Engineering ScienceYokohama National UniversityKanagawaJapan
| | - Tomoya Sugiyama
- Graduate School of Engineering ScienceYokohama National UniversityKanagawaJapan
| | - Reiji Tokito
- Graduate School of Engineering ScienceYokohama National UniversityKanagawaJapan
| | - Hiroe Ueno
- Department of Urology, Reproduction CenterYokohama City University Medical CenterKanagawaJapan
| | | | - Yasushi Yumura
- Department of Urology, Reproduction CenterYokohama City University Medical CenterKanagawaJapan
| | - Yasuyuki Kurihara
- Laboratory of Molecular Biology, Faculty of Engineering ScienceYokohama National UniversityKanagawaJapan
| |
Collapse
|
4
|
Zhang M, Chiozzi RZ, Bromfield EG, Heck AJR, Helms JB, Gadella BM. Characterization of acrosin and acrosin binding protein as novel CRISP2 interacting proteins in boar spermatozoa. Andrology 2023; 11:1460-1471. [PMID: 36815564 PMCID: PMC10947329 DOI: 10.1111/andr.13413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/07/2023] [Accepted: 02/12/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND Previously, we reported that cysteine-rich secretory protein 2 is involved in high molecular weight complexes in boar spermatozoa. These cysteine-rich secretory protein 2protein complexes are formed at the last phase of sperm formation in the testis and play a role in sperm shaping and functioning. OBJECTIVES This study aimed to identify cysteine-rich secretory protein 2 interacting partners. These binding partner interactions were investigated under different conditions, namely, non-capacitating conditions, after the induction of in vitro sperm capacitation and subsequently during an ionophore A23187-induced acrosome reaction. MATERIALS AND METHODS The incubated pig sperm samples were subjected to protein extraction. Extracted proteins were subjected to blue native gel electrophoresis and native immunoblots. Immunoreactive gel bands were excised and subjected to liquid chromatography-mass spectrometry (LC-MS) analysis for protein identification. Protein extracts were also subjected to CRISP2 immunoprecipitation and analyzed by LC-MS for protein identification. The most prominent cystein-rich secretory protein 2 interacting proteins that appeared in both independent LC-MS analyses were studied with a functional in situ proximity interaction assay to validate their property to interact with cystein-rich secretory protein 2 in pig sperm. RESULTS Blue native gel electrophoresis and native immunoblots revealed that cystein-rich secretory protein 2 was present within a ∼150 kDa protein complex under all three conditions. Interrogation of cystein-rich secretory-protein 2-immunoreactive bands from blue native gels as well as cystein-rich secretory protein 2 immunoprecipitated products using mass spectrometry consistently revealed that, beyond cystein-rich secretory protein 2, acrosin and acrosin binding protein were among the most abundant interacting proteins and did interact under all three conditions. Co-immunoprecipitation and immunoblotting indicated that cystein-rich secretory protein 2 interacted with pro-acrosin (∼53 kDa) and Aacrosin binding protein under all three conditions and additionally to acrosin (∼35 kDa) after capacitation and the acrosome reaction. The colocalization of these interacting proteins with cystein-rich secretory protein 2 was assessed via in situ proximity ligation assays. The colocalization signal of cystein-rich secretory protein 2 and acrosin in the acrosome seemed dispersed after capacitation but was consistently present in the sperm tail under all conditions. The fluorescent foci of cystein-rich secretory protein 2 and acrsin binding protein colocalization appeared to be redistributed within the sperm head from the anterior acrosome to the post-acrosomal sheath region upon capacitation. DISCUSSION AND CONCLUSION These results suggest that CRISP2 may act as a scaffold for protein complex formation and dissociation to ensure the correct positioning of proteins required for the acrosome reaction and zona pellucida penetration.
Collapse
Affiliation(s)
- Min Zhang
- Department of Biomolecular Health SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and ProteomicsBijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Netherlands Proteomics CentreUtrechtThe Netherlands
| | - Elizabeth G Bromfield
- Department of Biomolecular Health SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life Sciences, Discipline of Biological Sciences, University of NewcastleCallaghanNew South WalesAustralia
| | - Albert JR Heck
- Biomolecular Mass Spectrometry and ProteomicsBijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
- Netherlands Proteomics CentreUtrechtThe Netherlands
| | - J Bernd Helms
- Department of Biomolecular Health SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
| | - Bart M Gadella
- Department of Biomolecular Health SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
5
|
Moreno RD. Human globozoospermia-related genes and their role in acrosome biogenesis. WIREs Mech Dis 2023; 15:e1589. [PMID: 36493758 DOI: 10.1002/wsbm.1589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 12/13/2022]
Abstract
The mammalian acrosome is a secretory vesicle attached to the sperm nucleus whose fusion with the overlying plasma membrane is required to achieve fertilization. Acrosome biogenesis starts during meiosis, but it lasts through the entire process of haploid cell differentiation (spermiogenesis). Acrosome biogenesis is a stepwise process that involves membrane traffic from the Golgi apparatus, but it also seems that the lysosome/endosome system participates in this process. Defective sperm head morphology is accompanied by defective acrosome shape and function, and patients with these characteristics are infertile or subfertile. The most extreme case of acrosome biogenesis failure is globozoospermia syndrome, which is primarily characterized by the presence of round-headed spermatozoa without acrosomes with cytoskeleton defects around the nucleus and infertility. Several genes participating in acrosome biogenesis have been uncovered using genetic deletions in mice, but only a few of them have been found to be deleted or modified in patients with globozoospermia. Understanding acrosome biogenesis is crucial to uncovering the molecular basis of male infertility and developing new diagnostic tools and assisted reproductive technologies that may help infertile patients through more effective treatment techniques. This article is categorized under: Reproductive System Diseases > Environmental Factors Infectious Diseases > Stem Cells and Development Reproductive System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Ricardo D Moreno
- Departmento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
6
|
Yu K, Xiao K, Sun QQ, Liu RF, Huang LF, Zhang PF, Xu HY, Lu YQ, Fu Q. Comparative proteomic analysis of seminal plasma exosomes in buffalo with high and low sperm motility. BMC Genomics 2023; 24:8. [PMID: 36624393 PMCID: PMC9830767 DOI: 10.1186/s12864-022-09106-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Exosomes are nanosized membranous vesicles secreted by various types of cells, which facilitate intercellular communication by transporting bioactive compounds. Exosomes are abundant in biological fluids including semen, and their protein composition and the potential of seminal plasma exosomes (SPEs) as fertility biomarkers were elucidated in humans, however, little information is available regarding buffalo (Bubalus bubalis). Here, we examined protein correlation between spermatozoa, seminal plasma (SP), and SPEs, and we compared and analyzed protein differences between high-motility (H-motility) and low-motility (L-motility) SPEs in buffalo. RESULTS SPEs were concentrated and purified by ultracentrifugation combined with sucrose density gradient centrifugation, followed by verification using western blotting, nanoparticle tracking analysis, and transmission electron microscopy. Protein composition in spermatozoa, SP and SPEs, and protein difference in H- and L-motility SPEs were identified by LC-MS/MS proteomic analysis and were functionally analyzed through comprehensive bioinformatics. Many SPEs proteins originated from spermatozoa and SP, and nearly one third were also present in spermatozoa and SP. A series of proteins associated with reproductive processes including sperm capacitation, spermatid differentiation, fertilization, sperm-egg recognition, membrane fusion, and acrosome reaction were integrated in a functional network. Comparative proteomic analyses showed 119 down-regulated and 41 up-regulated proteins in L-motility SPEs, compared with H-motility SPEs. Gene Ontology (GO) enrichment of differentially expressed proteins (DEPs) showed that most differential proteins were located in sperm and vesicles, with activities of hydrolase and metalloproteinase, and were involved in sperm-egg recognition, fertilization, single fertilization, and sperm-zona pellucida binding processes, etc. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that differential proteins were mainly involved in the PPRP signaling pathway, calcium signaling pathway, and cAMP signaling pathway, among others. Furthermore, 6 proteins associated with reproduction were validated by parallel reaction monitoring analysis. CONCLUSION This study provides a comprehensive description of the seminal plasma exosome proteome and may be of use for further screening of biomarkers associated with male infertility.
Collapse
Affiliation(s)
- Kai Yu
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Kai Xiao
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Qin-qiang Sun
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Run-feng Liu
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Liang-feng Huang
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Peng-fei Zhang
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Hui-yan Xu
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Yang-qing Lu
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China ,grid.256609.e0000 0001 2254 5798College of Animal Science and Technology, Guangxi University, Nanning, 530004 China
| | - Qiang Fu
- grid.256609.e0000 0001 2254 5798State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530004 China
| |
Collapse
|
7
|
Shen C, Xiong W, Li C, Ge H, Shen Y, Tang L, Zhang H, Lu S, Fei J, Wang Z. Testis-specific serine protease PRSS54 regulates acrosomal granule localization and sperm head morphogenesis in mice. Biol Reprod 2022; 107:1139-1154. [PMID: 35863763 DOI: 10.1093/biolre/ioac146] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/20/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Serine proteases (PRSS) constitute nearly one-third of all proteases, and many of them have been identified to be testis-specific and play significant roles during sperm development and male reproduction. PRSS54 is one of the testis-specific PRSS in mouse and human but its physiological function remains largely unclear. In the present study, we demonstrate in detail that PRSS54 exists not only in testis but also in mature sperm, exhibiting a change in protein size from 50 kDa in testis to 42 kDa in sperm. Loss of PRSS54 in mice results in male subfertility, acrosome deformation, defective sperm-zona penetration, and phenotypes of male subfertility and acrosome deformation can be rescued by Prss54 transgene. Ultrastructure analyses by transmission electronic microscopy further reveal various morphological abnormalities of Prss54-/- spermatids during spermiogenesis, including unfused vacuoles in acrosome, detachment and eccentrical localization of the acrosomal granules, and asymmetrical elongation of the nucleus. Subcellular localization of PRSS54 display that it appears in the acrosomal granule at the early phase of acrosome biogenesis, then extends along the inner acrosomal membrane, and ultimately presents in the acrosome region of the mature sperm. PRSS54 interacts with acrosomal proteins ZPBP1, ZPBP2, ACRBP and ZP3R, and loss of PRSS54 affects the distribution of these proteins in testis and sperm, although their protein levels are largely unaffected. Moreover, Prss54-/- sperm are more sensitive to acrosome reaction inducers. These data indicate that PRSS54 is an acrosomal protein and plays an important role in regulating acrosome biogenesis, sperm function and male fertility.
Collapse
Affiliation(s)
- Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenfeng Xiong
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chaojie Li
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haoyang Ge
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lingyun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongxin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shunyuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian Fei
- Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai, 201203, China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.,Shanghai Engineering and Technology Research Center for Model Animals, Shanghai Model Organisms Center, Inc., Shanghai, 201203, China
| |
Collapse
|
8
|
Yamauchi Y, Matsumura T, Bakse J, Holmlund H, Blanchet G, Carrot E, Ikawa M, Ward MA. Loss of mouse Y chromosome gene Zfy1 and Zfy2 leads to spermatogenesis impairment, sperm defects, and infertility. Biol Reprod 2022; 106:1312-1326. [PMID: 35293998 PMCID: PMC9199016 DOI: 10.1093/biolre/ioac057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/17/2022] [Accepted: 03/11/2022] [Indexed: 11/14/2022] Open
Abstract
Using mice with Y chromosome deficiencies and supplementing Zfy transgenes, we, and others, have previously shown that the loss of Y chromosome Zfy1 and Zfy2 genes is associated with infertility and spermiogenic defects and that the addition of Zfy transgenes rescues these defects. In these past studies, the absence of Zfy was linked to the loss of other Y chromosome genes, which might have contributed to spermiogenic phenotypes. Here, we used CRISPR/Cas9 to specifically remove open reading frame of Zfy1, Zfy2, or both Zfy1 and Zfy2, and generated Zfy knockout (KO) and double knockout (DKO) mice. Zfy1 KO and Zfy2 KO mice were both fertile, but the latter had decreased litters size and sperm number, and sperm headshape abnormalities. Zfy DKO males were infertile and displayed severe spermatogenesis defects. Postmeiotic arrest largely prevented production of sperm and the few sperm that were produced all displayed gross headshape abnormalities and structural defects within head and tail. Infertility of Zfy DKO mice could be overcome by injection of spermatids or sperm directly to oocytes, and the resulting male offspring had the same spermiogenic phenotype as their fathers. The study is the first describing detailed phenotypic characterization of mice with the complete Zfy gene loss. It provides evidence supporting that the presence of at least one Zfy homolog is essential for male fertility and development of normal sperm functional in unassisted fertilization. The data also show that while the loss of Zfy1 is benign, the loss of Zfy2 is mildly detrimental for spermatogenesis.
Collapse
Affiliation(s)
- Yasuhiro Yamauchi
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Takafumi Matsumura
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Jackson Bakse
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Hayden Holmlund
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Genevieve Blanchet
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Emmaelle Carrot
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Monika A Ward
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| |
Collapse
|
9
|
Fu J, Yingying Ge, Qingmei Zhang, Lin Y, Liu C, Nong W, Luo X, Xiao S, Xie X, Luo B. Immunohistochemistry Study of OY-TES-1 Location in Fetal and Adult Human Tissues. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7052830. [PMID: 35463688 PMCID: PMC9020931 DOI: 10.1155/2022/7052830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022]
Abstract
OY-TES-1 is reportedly involved in carcinogenesis and spermatogenesis. However, the tissue distribution of OY-TES-1 in the normal human body remains elusive. This study detected OY-TES-1 expression in human fetal and adult normal tissues by immunohistochemistry. We identified a general principle of OY-TES-1 expression. The expression of OY-TES-1 was found in neurons, smooth muscle cells, and cardiac muscle cells from both fetuses and adults. The connective tissue showed no specific staining throughout the fetal and adult samples. With OY-TES-1-positive staining of the epithelium irregular, OY-TES-1 was strongly expressed in the epithelium of the skin and bladder, as well as hepatocytes, pancreatic islets, and acinous cells during the fetal stage but was not detected in the postnatal period. In contrast to the epithelium of blood vessels, the fetal and adult central hepatic vein and glomeruli showed negative expression of the OY-TES-1 protein. Sex-dimorphism was observed in the distribution of OY-TES-1 in male and female germ cells. Collectively, our results indicate that OY-TES-1 is a member of the cancer-testis antigen and autoantigen, with tissue-specific and period-specific expression patterns, revealing potential contributions of OY-TES-1 to the diagnosis and therapeutic treatment for neoplasms and infertility.
Collapse
Affiliation(s)
- Jun Fu
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Yingying Ge
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Qingmei Zhang
- Guangxi Colleges and Universities Key Laboratory Research of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Yongda Lin
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Chang Liu
- Department of Neurosurgery, First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Weixia Nong
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Xin Luo
- Department of Histology & Embryology, School of Pre-clinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Shaowen Xiao
- Department of Neurosurgery, First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Xiaoxun Xie
- Guangxi Colleges and Universities Key Laboratory Research of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| | - Bin Luo
- Guangxi Colleges and Universities Key Laboratory Research of Preclinical Medicine, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
10
|
Kortleve D, Coelho RM, Hammerl D, Debets R. Cancer germline antigens and tumor-agnostic CD8+ T cell evasion. Trends Immunol 2022; 43:391-403. [DOI: 10.1016/j.it.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/31/2022]
|
11
|
Proteomic Analysis of Intracellular and Membrane-Associated Fractions of Canine (Canis lupus familiaris) Epididymal Spermatozoa and Sperm Structure Separation. Animals (Basel) 2022; 12:ani12060772. [PMID: 35327169 PMCID: PMC8944539 DOI: 10.3390/ani12060772] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Epididymal spermatozoa have great potential in current dog reproductive technologies. In the case of azoospermia or when the male dies, the recovery of epididymal spermatozoa opens new possibilities for reproduction. It is of great importance to analyze the quality of the sperm in such cases. Proteomic studies contribute to explaining the role of proteins at various stages of epididymal sperm maturation and offer potential opportunities to use them as markers of sperm quality. The present study showed, for the first time, mass spectrometry and bioinformatic analysis of intracellular and membrane-associated proteins of canine epididymal spermatozoa. Additionally, sonication was used for the separation of dog epididymal sperm morphological elements (heads, tails and acrosomes). The results revealed the presence of differentially abundant proteins in both sperm protein fractions significant for sperm function and fertilizing ability. It was also shown that these proteins participate in important sperm metabolic pathways, which may suggest their potential as sperm quality biomarkers. Abstract This study was provided for proteomic analysis of intracellular and membrane-associated fractions of canine (Canis lupus familiaris) epididymal spermatozoa and additionally to find optimal sonication parameters for the epididymal sperm morphological structure separation and sperm protein isolation. Sperm samples were collected from 15 dogs. Sperm protein fractions: intracellular (SIPs) and membrane-associated (SMAPs) were isolated. After sonication, sperm morphology was evaluated using Spermac Stain™. The sperm protein fractions were analyzed using gel electrophoresis (SDS-PAGE) and nanoliquid chromatography coupled to quadrupole time-of-flight mass spectrometry (NanoLC-Q-TOF/MS). UniProt database-supported identification resulted in 42 proteins identified in the SIPs and 153 proteins in the SMAPs. Differentially abundant proteins (DAPs) were found in SIPs and SMAPs. Based on a gene ontology analysis, the dominant molecular functions of SIPs were catalytic activity (50%) and binding (28%). Hydrolase activity (33%) and transferase activity (21%) functions were dominant for SMAPs. Bioinformatic analysis of SIPs and SMAPs showed their participation in important metabolic pathways in epididymal sperm, which may suggest their potential as sperm quality biomarkers. The use of sonication 150 W, 10 min, may be recommended for the separation of dog epididymal sperm heads, tails, acrosomes and the protein isolation.
Collapse
|
12
|
Wu D, Khan FA, Huo L, Sun F, Huang C. Alternative splicing and MicroRNA: epigenetic mystique in male reproduction. RNA Biol 2022; 19:162-175. [PMID: 35067179 PMCID: PMC8786336 DOI: 10.1080/15476286.2021.2024033] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Infertility is rarely life threatening, however, it poses a serious global health issue posing far-reaching socio-economic impacts affecting 12–15% of couples worldwide where male factor accounts for 70%. Functional spermatogenesis which is the result of several concerted coordinated events to produce sperms is at the core of male fertility, Alternative splicing and microRNA (miRNA) mediated RNA silencing (RNAi) constitute two conserved post-transcriptional gene (re)programming machinery across species. The former by diversifying transcriptome signature and the latter by repressing target mRNA activity orchestrate a spectrum of testicular events, and their dysfunctions has several implications in male infertility. This review recapitulates the knowledge of these mechanistic events in regulation of spermatogenesis and testicular homoeostasis. In addition, miRNA payload in sperm, vulnerable to paternal inputs, including unhealthy diet, infection and trauma, creates epigenetic memory to initiate intergenerational phenotype. Naive zygote injection of sperm miRNAs from stressed father recapitulates phenotypes of offspring of stressed father. The epigenetic inheritance of paternal pathologies through miRNA could be a tantalizing avenue to better appreciate ‘Paternal Origins of Health and Disease’ and the power of tiny sperm.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, China
| | - Faheem Ahmed Khan
- Laboratory of Molecular Biology and Genomics, Department of Zoology, Faculty of Science, University of Central Punjab, Lahore, Pakistan
| | - Lijun Huo
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, China
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
13
|
A novel posttranslational modification of histone, H3 S-sulfhydration, is down-regulated in asthenozoospermic sperm. J Assist Reprod Genet 2021; 38:3175-3193. [PMID: 34664184 PMCID: PMC8666411 DOI: 10.1007/s10815-021-02314-x] [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: 06/29/2021] [Accepted: 09/07/2021] [Indexed: 11/14/2022] Open
Abstract
Oxidative stress is one of the major causes leading to male infertility including asthenozoospermia. Hydrogen sulfide (H2S) has been widely recognized to be a potent antioxidant whose role is partially implemented by protein S-sulfhydration. However, protein S-sulfhydration has not been reported in germ cells. Therefore, we investigated whether asthenozoospermia could be associated with sperm protein S-sulfhydration. S-sulfhydrated proteins in human sperm were enriched via biotin-switch assay and analyzed using LC-MS/MS spectrometry. Two hundred forty-four S-sulfhydrated proteins were identified. Importantly, we validated that sperm histones H3.1 and H3.3 were the S-sulfhydrated proteins. Their S-sulfhydrated amino acid residue was Cysteine111. Abundances of S-sulfhydrated H3 (sH3) and S-sulfhydrated H3.3 (sH3.3) were significantly down-regulated in asthenozoospermic sperm, compared with the fertile controls, and were significantly correlated with progressive motility. Retinoic acid (RA) up-regulated level of sH3.3 in primary round spermatids and the C18-4 cells (a mouse spermatogonial stem cell line). Overexpression of the mutant H3.3 (Cysteine111 was replaced with serine) affected expression of 759 genes and raised growth rate of C18-4 cells. For the first time, S-sulfhydration H3 and H3.3 were demonstrated in the present study. Our results highlight that aberrant S-sulfhydration of H3 is a new pathophysiological basis in male infertility.
Collapse
|
14
|
Xiong W, Shen C, Wang Z. The molecular mechanisms underlying acrosome biogenesis elucidated by gene-manipulated mice. Biol Reprod 2021; 105:789-807. [PMID: 34131698 DOI: 10.1093/biolre/ioab117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 02/05/2023] Open
Abstract
Sexual reproduction requires the fusion of two gametes in a multistep and multifactorial process termed fertilization. One of the main steps that ensures successful fertilization is acrosome reaction. The acrosome, a special kind of organelle with a cap-like structure that covers the anterior portion of sperm head, plays a key role in the process. Acrosome biogenesis begins with the initial stage of spermatid development, and it is typically divided into four successive phases: the Golgi phase, cap phase, acrosome phase, and maturation phase. The run smoothly of above processes needs an active and specific coordination between the all kinds of organelles (endoplasmic reticulum, trans-golgi network and nucleus) and cytoplasmic structures (acroplaxome and manchette). During the past two decades, an increasingly genes have been discovered to be involved in modulating acrosome formation. Most of these proteins interact with each other and show a complicated molecular regulatory mechanism to facilitate the occurrence of this event. This Review focuses on the progresses of studying acrosome biogenesis using gene-manipulated mice and highlights an emerging molecular basis of mammalian acrosome formation.
Collapse
Affiliation(s)
- Wenfeng Xiong
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui-Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Kato Y, Kumar S, Lessard C, Bailey JL. ACRBP (Sp32) is involved in priming sperm for the acrosome reaction and the binding of sperm to the zona pellucida in a porcine model. PLoS One 2021; 16:e0251973. [PMID: 34086710 PMCID: PMC8177411 DOI: 10.1371/journal.pone.0251973] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/07/2021] [Indexed: 11/17/2022] Open
Abstract
In boar sperm, we have previously shown that capacitation is associated with the appearance of the p32 tyrosine phosphoprotein complex. The principal tyrosine phosphoprotein involved in this complex is the acrosin-binding protein (ACRBP), which regulates the autoconversion of proacrosin to intermediate forms of acrosin in both boar and mouse sperm. However, the complete biological role of ACRBP has not yet been elucidated. In this study, we tested the hypothesis that tyrosine phophorylation and the presence of the ACRBP in the sperm head are largely necessary to induce capacitation, the acrosome reaction (AR) and sperm-zona pellucida (ZP) binding, all of which are necessary steps for fertilization. In vitro fertilization (IVF) was performed using matured porcine oocytes and pre-capacitated boar sperm cultured with anti-phosphotyrosine antibodies or antibodies against ACRBP. Anti-ACRBP antibodies reduced capacitation and spontaneous AR (P<0.05). Sperm-ZP binding declined in the presence of anti-phosphotyrosine or anti-ACRBP antibodies. The localisation of anti-ACRBP antibodies on the sperm head, reduced the ability of the sperm to undergo the AR in response to solubilized ZP or by inhibiting the sarco/endoplasmic reticulum Ca2+-ATPase. These results support our hypothesis that tyrosine phosphorylated proteins and ACRBP are present upon the sperm surface in order to participate in sperm-ZP binding, and that ACRBP upon the surface of the sperm head facilitates capacitation and the AR in the porcine.
Collapse
Affiliation(s)
- Yoku Kato
- Département des sciences animales, Centre de recherche en reproduction, développement et santé intergénérationnelle, Université Laval, Québec, Canada
| | - Satheesh Kumar
- Département des sciences animales, Centre de recherche en reproduction, développement et santé intergénérationnelle, Université Laval, Québec, Canada
| | - Christian Lessard
- Département des sciences animales, Centre de recherche en reproduction, développement et santé intergénérationnelle, Université Laval, Québec, Canada
| | - Janice L Bailey
- Département des sciences animales, Centre de recherche en reproduction, développement et santé intergénérationnelle, Université Laval, Québec, Canada
| |
Collapse
|
16
|
Liu Q, Guo Q, Guo W, Song S, Wang N, Chen X, Sun A, Yan L, Qiao J. Loss of CEP70 function affects acrosome biogenesis and flagella formation during spermiogenesis. Cell Death Dis 2021; 12:478. [PMID: 33980814 PMCID: PMC8116340 DOI: 10.1038/s41419-021-03755-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/03/2023]
Abstract
The spermatogenesis process is complex and delicate, and any error in a step may cause spermatogenesis arrest and even male infertility. According to our previous transcriptomic data, CEP70 is highly expressed throughout various stages of human spermatogenesis, especially during the meiosis and deformation stages. CEP70 is present in sperm tails and that it exists in centrosomes as revealed by human centrosome proteomics. However, the specific mechanism of this protein in spermatogenesis is still unknown. In this study, we found a heterozygous site of the same mutation on CEP70 through mutation screening of patients with clinical azoospermia. To further verify, we deleted CEP70 in mice and found that it caused abnormal spermatogenesis, leading to male sterility. We found that the knockout of CEP70 did not affect the prophase of meiosis I, but led to male germ-cell apoptosis and abnormal spermiogenesis. By transmission electron microscopy (TEM) and scanning electron microscopy (SEM) analysis, we found that the deletion of CEP70 resulted in the abnormal formation of flagella and acrosomes during spermiogenesis. Tandem mass tag (TMT)-labeled quantitative proteomic analysis revealed that the absence of CEP70 led to a significant decrease in the proteins associated with the formation of the flagella, head, and acrosome of sperm, and the microtubule cytoskeleton. Taken together, our results show that CEP70 is essential for acrosome biogenesis and flagella formation during spermiogenesis.
Collapse
Affiliation(s)
- Qiang Liu
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Qianying Guo
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Wei Guo
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Shi Song
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Nan Wang
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Xi Chen
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Andi Sun
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Liying Yan
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China ,grid.411642.40000 0004 0605 3760Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China ,grid.506261.60000 0001 0706 7839Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Choi H, Wang Z, Dean J. Sperm acrosome overgrowth and infertility in mice lacking chromosome 18 pachytene piRNA. PLoS Genet 2021; 17:e1009485. [PMID: 33831001 PMCID: PMC8057611 DOI: 10.1371/journal.pgen.1009485] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/20/2021] [Accepted: 03/12/2021] [Indexed: 01/08/2023] Open
Abstract
piRNAs are small non-coding RNAs required to maintain genome integrity and preserve RNA homeostasis during male gametogenesis. In murine adult testes, the highest levels of piRNAs are present in the pachytene stage of meiosis, but their mode of action and function remain incompletely understood. We previously reported that BTBD18 binds to 50 pachytene piRNA-producing loci. Here we show that spermatozoa in gene-edited mice lacking a BTBD18 targeted pachytene piRNA cluster on Chr18 have severe sperm head dysmorphology, poor motility, impaired acrosome exocytosis, zona pellucida penetration and are sterile. The mutant phenotype arises from aberrant formation of proacrosomal vesicles, distortion of the trans-Golgi network, and up-regulation of GOLGA2 transcripts and protein associated with acrosome dysgenesis. Collectively, our findings reveal central role of pachytene piRNAs in controlling spermiogenesis and male fertility.
Collapse
Affiliation(s)
- Heejin Choi
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, United States of America
| | - Zhengpin Wang
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, United States of America
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
18
|
Zakrzewski P, Lenartowska M, Buss F. Diverse functions of myosin VI in spermiogenesis. Histochem Cell Biol 2021; 155:323-340. [PMID: 33386429 PMCID: PMC8021524 DOI: 10.1007/s00418-020-01954-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Spermiogenesis is the final stage of spermatogenesis, a differentiation process during which unpolarized spermatids undergo excessive remodeling that results in the formation of sperm. The actin cytoskeleton and associated actin-binding proteins play crucial roles during this process regulating organelle or vesicle delivery/segregation and forming unique testicular structures involved in spermatid remodeling. In addition, several myosin motor proteins including MYO6 generate force and movement during sperm differentiation. MYO6 is highly unusual as it moves towards the minus end of actin filaments in the opposite direction to other myosin motors. This specialized feature of MYO6 may explain the many proposed functions of this myosin in a wide array of cellular processes in animal cells, including endocytosis, secretion, stabilization of the Golgi complex, and regulation of actin dynamics. These diverse roles of MYO6 are mediated by a range of specialized cargo-adaptor proteins that link this myosin to distinct cellular compartments and processes. During sperm development in a number of different organisms, MYO6 carries out pivotal functions. In Drosophila, the MYO6 ortholog regulates actin reorganization during spermatid individualization and male KO flies are sterile. In C. elegans, the MYO6 ortholog mediates asymmetric segregation of cytosolic material and spermatid budding through cytokinesis, whereas in mice, this myosin regulates assembly of highly specialized actin-rich structures and formation of membrane compartments to allow the formation of fully differentiated sperm. In this review, we will present an overview and compare the diverse function of MYO6 in the specialized adaptations of spermiogenesis in flies, worms, and mammals.
Collapse
Affiliation(s)
- Przemysław Zakrzewski
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| | - Marta Lenartowska
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Folma Buss
- Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
19
|
Zheng B. Expression and clinical importance of a newly discovered alternative splice variant of the gene for acrosin binding protein found in human brain tumors. ASIAN BIOMED 2020; 14:243-252. [PMID: 37551308 PMCID: PMC10373398 DOI: 10.1515/abm-2020-0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Background Acrosin binding protein (ACRBP) is a member of the cancer-testis antigen (CTA) family. Normally, ACRBP mRNA is expressed only in seminiferous tubules, while abnormally it is expressed in various types of cancers in tumor tissues, such as brain tumor. Objectives To determine the expression and clinical impact of a newly discovered splice variant of ACRBP in brain tumor. Methods Total RNA was extracted and reverse transcribed from 92 tumor specimens and 3 cell lines. Primers were designed to determine the expression of the new splice variant in all the samples. Quantitative real-time PCR (qPCR) was conducted for samples positive in reverse transcriptase-PCR. Association of the expression of ACRBP with the clinicopathological features of the various brain tumors was assessed statistically. Results The primers identified a newly discovered splice variant of ACRBP named ACRBP-V5a. The proportions of samples of the various brain tumor types positive for the ACRBP-V5a splicing variant were as follows: astrocytoma 10/33 (30%), glioblastoma 10/30 (33%), medulloblastoma 14/29 (48%), all tumors 34/92 (37%). Although we did not find a significant difference in the proportions of samples of various types of brain tumor tissues positive for the new splice variant (P > 0.05), levels of expression of the ACRBP-V5a splice variant were significantly different for tumor grade (P = 0.01) and tumor type (P = 0.02). Conclusions A newly discovered splice variant, ACRBP-V5a, is present in brain tumor. The new splicing variant may have discriminative value and potential importance in molecular-targeted therapy for brain tumors.
Collapse
Affiliation(s)
- Baolong Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi530021, China
| |
Collapse
|
20
|
Umaoka A, Takeuchi H, Mizutani K, Seo N, Matsushima Y, Habe K, Hagimori K, Yamaguchi Y, Ikeda T, Yamanaka K. Skin Inflammation and Testicular Function: Dermatitis Causes Male Infertility via Skin-Derived Cytokines. Biomedicines 2020; 8:biomedicines8090293. [PMID: 32825298 PMCID: PMC7555019 DOI: 10.3390/biomedicines8090293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 11/16/2022] Open
Abstract
The medical comorbidities including skin diseases are associated with male infertility. The most common cause of male infertility is the inability of testes to produce sperm; however, the influence of persistent dermatitis on testicular function has not been elucidated so far. We investigated the relationship between skin inflammation and impaired sperm production using a spontaneous dermatitis mouse model. We examined the breeding records of dermatitis mice and their wild-type littermates. Sperm count, motility, and viability were analyzed by direct microscopic observation and flow cytometry. In addition, testis and epididymis were histologically examined. Finally, sperm viability was evaluated in another dermatitis mouse model and in wild-type mice in which inflammatory cytokines were intraperitoneally administered. Compared to wild-type littermate mice, the number of children born was lower in mice with dermatitis. The body weight and testis size were decreased age-dependently. In the skin disease group, the sperm count and movement ratio were clearly decreased, and reduced sperm viability was observed. Histological examination revealed the detachment of Sertoli cells and reduced spermatogenesis. The fibrosis of epididymal stroma was severe, and it might affect defective sperm maturation in the epididymis. In addition, this phenomena was reproduced by a hapten applied dermatitis mouse model and the intraperitoneal administration of inflammatory cytokines. Once the skin is inflamed, inflammatory cytokines are produced and released, which may affect testicular and sperm function. Additional studies are needed to determine the relationship between male infertility and severe dermatitis in human.
Collapse
Affiliation(s)
- Ai Umaoka
- Department of Dermatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (A.U.); (K.H.)
| | - Hiroki Takeuchi
- Obstetrics and Gynecology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (H.T.); (T.I.)
| | - Kento Mizutani
- Department of Dermatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (A.U.); (K.H.)
| | - Naohiro Seo
- Immuno-Gene Therapy, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan;
| | - Yoshiaki Matsushima
- Department of Dermatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (A.U.); (K.H.)
| | - Koji Habe
- Department of Dermatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (A.U.); (K.H.)
| | - Kohei Hagimori
- Medicines Development Unit Japan, Eli Lilly Japan K.K., 5-1-28 Isogamidori, Chuo-ku, Kobe, Hyogo 651-0086, Japan;
| | - Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama 236-0027, Japan;
| | - Tomoaki Ikeda
- Obstetrics and Gynecology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (H.T.); (T.I.)
| | - Keiichi Yamanaka
- Department of Dermatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (A.U.); (K.H.)
- Correspondence: ; Tel.: +81-59-231-5025; Fax: +81-59-231-5206
| |
Collapse
|
21
|
Zakrzewski P, Rędowicz MJ, Buss F, Lenartowska M. Loss of myosin VI expression affects acrosome/acroplaxome complex morphology during mouse spermiogenesis†. Biol Reprod 2020; 103:521-533. [PMID: 32412041 PMCID: PMC7442776 DOI: 10.1093/biolre/ioaa071] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/24/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
During spermiogenesis in mammals, actin filaments and a variety of actin-binding proteins are involved in the formation and function of highly specialized testis-specific structures. Actin-based motor proteins, such as myosin Va and VIIa, play a key role in this complex process of spermatid transformation into mature sperm. We have previously demonstrated that myosin VI (MYO6) is also expressed in mouse testes. It is present in actin-rich structures important for spermatid development, including one of the earliest events in spermiogenesis—acrosome formation. Here, we demonstrate using immunofluorescence, cytochemical, and ultrastructural approaches that MYO6 is involved in maintaining the structural integrity of these specialized actin-rich structures during acrosome biogenesis in mouse. We show that MYO6 together with its binding partner TOM1/L2 is present at/around the spermatid Golgi complex and the nascent acrosome. Depletion of MYO6 in Snell’s waltzer mice causes structural disruptions of the Golgi complex and affects the acrosomal granule positioning within the developing acrosome. In summary, our results suggest that MYO6 plays an anchoring role during the acrosome biogenesis mainly by tethering of different cargo/membranes to highly specialized actin-related structures.
Collapse
Affiliation(s)
- Przemysław Zakrzewski
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Folma Buss
- Cambridge Institute for Medical Research, The Keith Peters Building, University of Cambridge, Cambridge, UK
| | - Marta Lenartowska
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland.,Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| |
Collapse
|
22
|
Crapster JA, Rack PG, Hellmann ZJ, Le AD, Adams CM, Leib RD, Elias JE, Perrino J, Behr B, Li Y, Lin J, Zeng H, Chen JK. HIPK4 is essential for murine spermiogenesis. eLife 2020; 9:e50209. [PMID: 32163033 PMCID: PMC7067585 DOI: 10.7554/elife.50209] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/23/2020] [Indexed: 12/19/2022] Open
Abstract
Mammalian spermiogenesis is a remarkable cellular transformation, during which round spermatids elongate into chromatin-condensed spermatozoa. The signaling pathways that coordinate this process are not well understood, and we demonstrate here that homeodomain-interacting protein kinase 4 (HIPK4) is essential for spermiogenesis and male fertility in mice. HIPK4 is predominantly expressed in round and early elongating spermatids, and Hipk4 knockout males are sterile, exhibiting phenotypes consistent with oligoasthenoteratozoospermia. Hipk4 mutant sperm have reduced oocyte binding and are incompetent for in vitro fertilization, but they can still produce viable offspring via intracytoplasmic sperm injection. Optical and electron microscopy of HIPK4-null male germ cells reveals defects in the filamentous actin (F-actin)-scaffolded acroplaxome during spermatid elongation and abnormal head morphologies in mature spermatozoa. We further observe that HIPK4 overexpression induces branched F-actin structures in cultured fibroblasts and that HIPK4 deficiency alters the subcellular distribution of an F-actin capping protein in the testis, supporting a role for this kinase in cytoskeleton remodeling. Our findings establish HIPK4 as an essential regulator of sperm head shaping and potential target for male contraception.
Collapse
Affiliation(s)
- J Aaron Crapster
- Department of Chemical and Systems Biology, Stanford University School of MedicineStanfordUnited States
| | - Paul G Rack
- Department of Chemical and Systems Biology, Stanford University School of MedicineStanfordUnited States
| | - Zane J Hellmann
- Department of Chemical and Systems Biology, Stanford University School of MedicineStanfordUnited States
| | - Austen D Le
- Department of Chemical and Systems Biology, Stanford University School of MedicineStanfordUnited States
| | - Christopher M Adams
- Stanford University Mass Spectrometry, Stanford UniversityStanfordUnited States
| | - Ryan D Leib
- Stanford University Mass Spectrometry, Stanford UniversityStanfordUnited States
| | - Joshua E Elias
- Chan Zuckerberg Biohub, Stanford UniversityStanfordUnited States
| | - John Perrino
- Cell Science Imaging Facility, Stanford University School of MedicineStanfordUnited States
| | - Barry Behr
- Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, Stanford University School of MedicineStanfordUnited States
| | - Yanfeng Li
- Transgenic, Knockout, and Tumor Model Center, Stanford University School of MedicineStanfordUnited States
| | - Jennifer Lin
- Transgenic, Knockout, and Tumor Model Center, Stanford University School of MedicineStanfordUnited States
| | - Hong Zeng
- Transgenic, Knockout, and Tumor Model Center, Stanford University School of MedicineStanfordUnited States
| | - James K Chen
- Department of Chemical and Systems Biology, Stanford University School of MedicineStanfordUnited States
- Department of Developmental Biology, Stanford University School of MedicineStanfordUnited States
- Department of Chemistry, Stanford UniversityStanfordUnited States
| |
Collapse
|
23
|
Cannarella R, Condorelli RA, Mongioì LM, La Vignera S, Calogero AE. Molecular Biology of Spermatogenesis: Novel Targets of Apparently Idiopathic Male Infertility. Int J Mol Sci 2020; 21:E1728. [PMID: 32138324 PMCID: PMC7084762 DOI: 10.3390/ijms21051728] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
Male infertility affects half of infertile couples and, currently, a relevant percentage of cases of male infertility is considered as idiopathic. Although the male contribution to human fertilization has traditionally been restricted to sperm DNA, current evidence suggest that a relevant number of sperm transcripts and proteins are involved in acrosome reactions, sperm‒oocyte fusion and, once released into the oocyte, embryo growth and development. The aim of this review is to provide updated and comprehensive insight into the molecular biology of spermatogenesis, including evidence on spermatogenetic failure and underlining the role of the sperm-carried molecular factors involved in oocyte fertilization and embryo growth. This represents the first step in the identification of new possible diagnostic and, possibly, therapeutic markers in the field of apparently idiopathic male infertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (L.M.M.); (A.E.C.)
| | | | | | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (R.A.C.); (L.M.M.); (A.E.C.)
| | | |
Collapse
|
24
|
Kulichová K, Kumar V, Steinbachová L, Klodová B, Timofejeva L, Juříček M, Honys D, Hafidh S. PRP8A and PRP8B spliceosome subunits act co-ordinately to control pollen tube attraction in Arabidopsis. Development 2020; 147:dev.186742. [DOI: 10.1242/dev.186742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/14/2020] [Indexed: 02/04/2023]
Abstract
Precise guided pollen tube growth by the female gametophyte is a pre-requisite for successful sexual reproduction in flowering plants. Cysteine-rich proteins (CRPs) secreted from the embryo sac are known pollen tube attractants perceived by pollen tube receptor-like kinases (RLK's). How pre-mRNA splicing facilitates this cell-to-cell communication is not understood. Here, we report novel function of Pre-mRNA PROCESSING factor 8 paralogs, PRP8A and PRP8B, as regulators of pollen tube attraction. Double mutant prp8a prp8b ovules cannot attract pollen tubes, and prp8a prp8b pollen tubes fail in sensing ovules attraction signals. Only 3% of ovule-expressed genes were misregulated in prp8a prp8b. Combination of RNA-seq and MYB98/LURE1.2-YFP reporter revealed the expression of MYB98, LUREs and 49 other CRPs were downregulated suggesting loss of synergid cell fate. Differential Exon usage (DEU) and Intron-retention (IR) analysis revealed autoregulation of PPR8A/PRP8B splicing. In vivo, PRP8A coimmunoprecipitates with splicing enhancer AtSF3A1, suggesting involvement of PRP8A in 3′-splice site selection. Our data hint that PRP8A/PRP8B module exhibit spliceosome-autoregulation to facilitate pollen tube attraction via transcriptional regulation of MYB98, CRPs and LURE pollen tube attractants.
Collapse
Affiliation(s)
- Katarína Kulichová
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
| | - Vinod Kumar
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
| | - Lenka Steinbachová
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
| | - Božena Klodová
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
| | - Ljudmilla Timofejeva
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
| | - Miloslav Juříček
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
| | - David Honys
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
- Department of Plant Experimental Biology, Faculty of Science, Charles University, Viničná 5, 128 44, Prague 2, Czech Republic
| | - Said Hafidh
- Laboratory of Pollen Biology, Institute of Experimental Botany, Czech Academy of Science, Rozvojová 263, 165 02 Prague 6, Czech Republic
| |
Collapse
|
25
|
Zhou B, Wei C, Khan MA, Chen H, Fu J. Characterization and molecular cloning of novel isoforms of human spermatogenesis associated gene SPATA3. Mol Biol Rep 2019; 46:3827-3834. [PMID: 31006096 DOI: 10.1007/s11033-019-04825-4] [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: 02/18/2019] [Accepted: 04/13/2019] [Indexed: 12/20/2022]
Abstract
This study aimed to clone and characterize novel isoforms of the human SPATA3 gene. The isoforms of SPATA3 gene was cloned into pGMT vector using human testis cDNA as template, and Sanger sequencing was performed. Their characterizations and tissue-specific expression profiles were analyzed. The two novel isoforms were successfully cloned and deposited into GenBank as MG029442 (AYP71042) and MG029443 (AYP71043) respectively. Isoforms SPATA3-I1 and SPATA3-I2 were found with higher identity, where only 7 amino acids missed at N-terminus in SPATA3-I2, whereas SPATA3-I3 and SPATA3-I4 had more C-terminus deletion but in SPATA3-I3 no amino acid missed at N-terminus. Importantly, we found the characterization of QQPSPESTP domain with two repeats for isoforms SPATA3-I1 and SPATA3-I4, whereas three repeats for isoforms SPATA3-I1 and SPATA3-I2. The SPATA3 family of genes is orthologous conserved; the similar core PEST domain was also revealed with variable repeats, indicating that this domain may pay roles in the spermatogenesis and male development differently. Furthermore, RNA-seq data indicated that the SPATA3 gene is only expressed in testis. This further suggests that SPATA3 plays potential roles only in male development, spermatogenesis or spermatogenesis cell apoptosis. Thus, in this study we cloned the two novel isoforms of SPATA3, SPATA3-I3 and SPATA3-I4, and found interesting characteristic PEST domain (QQPSPESTP) conserved in different isoforms as well as in different species. SPATA3 is an essential gene and may functions in male reproductive system, specifically in spermatogenesis.
Collapse
Affiliation(s)
- Baixu Zhou
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, 3-319, Zhongshan Rd, Luzhou, 646000, Sichuan, China.,Department of Gynecology and Obstetrics, Guangzhou Women and Children's Hospital, Guangzhou, Guangdong, China
| | - Chunli Wei
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, 3-319, Zhongshan Rd, Luzhou, 646000, Sichuan, China
| | - Md Asaduzzaman Khan
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, 3-319, Zhongshan Rd, Luzhou, 646000, Sichuan, China
| | - Hanchun Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, 3-319, Zhongshan Rd, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
26
|
Nagashima K, Usui T, Baba T. Behavior of ACRBP-deficient mouse sperm in the female reproductive tract. J Reprod Dev 2019; 65:97-102. [PMID: 30606959 PMCID: PMC6473115 DOI: 10.1262/jrd.2018-137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Gene-knockout mice lacking ACRBP, a proacrosin-binding protein localized in the acrosome of sperm, have been shown to exhibit male subfertility, owing to abnormal formation of the acrosome.
In this study, to elucidate the mechanism contributing to the subfertility phenotype, we examined the behavior of ACRBP-deficient mouse sperm in the female reproductive tract. When sperm
that had migrated into the uterus and oviduct after mating were counted, the number of ACRBP-deficient sperm was noticeably smaller in the oviduct of mice post mating. However,
ACRBP-deficient sperm recovered from the oviduct possessed morphologically normal head shape and retained normal motility. Importantly, ACRBP-deficient sperm displayed a marked reduction in
the ability to successfully gain access to unfertilized oocytes. These data suggest that male subfertility of ACRBP-deficient mice may be attributed to incompleteness of the acrosome
reaction rather than impairment in sperm migration from the uterus to the oviduct.
Collapse
Affiliation(s)
- Kiyoshi Nagashima
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki 305-8577, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Tomoyuki Usui
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Tadashi Baba
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki 305-8577, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
27
|
Haseeb A, Tarique I, Bai X, Yang P, Ali Vistro W, Huang Y, Ali Fazllani S, Ahmed Z, Chen Q. Inhibition of autophagy impairs acrosome and mitochondrial crista formation during spermiogenesis in turtle: Ultrastructural evidence. Micron 2019; 121:84-89. [PMID: 30953869 DOI: 10.1016/j.micron.2019.03.006] [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: 02/07/2019] [Revised: 03/14/2019] [Accepted: 03/31/2019] [Indexed: 12/11/2022]
Abstract
Autophagy is a subcellular process that is extensively involved in spermiogenesis. In this study, we observed ultrastructural malformation of acrosome and mitochondrial cristae during the spermiogenesis of Chinese soft-shelled turtle due to the inhibition of autophagy. Autophagy was blocked with 3-MA, and the inhibition of autophagy was confirmed through western blot analysis. The morphological abnormalities of acrosomes and mitochondria were observed under transmission electron microscopy (TEM). In the early spermiogenesis (Golgi and cap phases), damaged macrovesicle was observed, and its proper expansion over the nucleus failed to be form a normal acrosomal cap. As spermiogenesis proceeded, the malformation of the acrosome in spermatids became more severe. In the late spermiogenesis (acrosomal and maturation phases), defective acrosome with damaged acrosomal membrane that was detached from the nucleus was observed. Along with malformed acrosome, elongation failed nucleus having oval or round shaped morphology was also observed. Moreover, morphological damage to the mitochondrial cristae was observed. Lacuna formation, half and complete loss of cristae were observed in the mitochondria of developing spermatids. We proposed that autophagy is required for normal formation of the acrosome and mitochondrial cristae during turtle spermiogenesis.
Collapse
Affiliation(s)
- Abdul Haseeb
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu Province, 210095, China; Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Azad Kashmir, Pakistan
| | - Imran Tarique
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu Province, 210095, China
| | - Xuebing Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu Province, 210095, China
| | - Ping Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu Province, 210095, China
| | - Waseem Ali Vistro
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu Province, 210095, China
| | - Yufei Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu Province, 210095, China
| | | | - Zulfqur Ahmed
- Faculty of Veterinary and Animal Sciences, University of Poonch Rawalakot, Azad Kashmir, Pakistan
| | - Qiusheng Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu Province, 210095, China.
| |
Collapse
|
28
|
Hirota T, Blakeley P, Sangrithi MN, Mahadevaiah SK, Encheva V, Snijders AP, ElInati E, Ojarikre OA, de Rooij DG, Niakan KK, Turner JMA. SETDB1 Links the Meiotic DNA Damage Response to Sex Chromosome Silencing in Mice. Dev Cell 2018; 47:645-659.e6. [PMID: 30393076 PMCID: PMC6286383 DOI: 10.1016/j.devcel.2018.10.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 08/15/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022]
Abstract
Meiotic synapsis and recombination ensure correct homologous segregation and genetic diversity. Asynapsed homologs are transcriptionally inactivated by meiotic silencing, which serves a surveillance function and in males drives meiotic sex chromosome inactivation. Silencing depends on the DNA damage response (DDR) network, but how DDR proteins engage repressive chromatin marks is unknown. We identify the histone H3-lysine-9 methyltransferase SETDB1 as the bridge linking the DDR to silencing in male mice. At the onset of silencing, X chromosome H3K9 trimethylation (H3K9me3) enrichment is downstream of DDR factors. Without Setdb1, the X chromosome accrues DDR proteins but not H3K9me3. Consequently, sex chromosome remodeling and silencing fail, causing germ cell apoptosis. Our data implicate TRIM28 in linking the DDR to SETDB1 and uncover additional factors with putative meiotic XY-silencing functions. Furthermore, we show that SETDB1 imposes timely expression of meiotic and post-meiotic genes. Setdb1 thus unites the DDR network, asynapsis, and meiotic chromosome silencing.
Collapse
Affiliation(s)
- Takayuki Hirota
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Paul Blakeley
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Mahesh N Sangrithi
- KK Women's and Children's Hospital, Department of Reproductive Medicine, Singapore 229899, Singapore; Duke-NUS Graduate Medical School, Singapore 119077, Singapore
| | | | - Vesela Encheva
- Mass Spectrometry Science Technology Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Ambrosius P Snijders
- Mass Spectrometry Science Technology Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Elias ElInati
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Obah A Ojarikre
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Dirk G de Rooij
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands; Center for Reproductive Medicine, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Kathy K Niakan
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
29
|
Tomizawa SI, Kobayashi Y, Shirakawa T, Watanabe K, Mizoguchi K, Hoshi I, Nakajima K, Nakabayashi J, Singh S, Dahl A, Alexopoulou D, Seki M, Suzuki Y, Royo H, Peters AHFM, Anastassiadis K, Stewart AF, Ohbo K. Kmt2b conveys monovalent and bivalent H3K4me3 in mouse spermatogonial stem cells at germline and embryonic promoters. Development 2018; 145:145/23/dev169102. [PMID: 30504434 DOI: 10.1242/dev.169102] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022]
Abstract
The mammalian male germline is sustained by a pool of spermatogonial stem cells (SSCs) that can transmit both genetic and epigenetic information to offspring. However, the mechanisms underlying epigenetic transmission remain unclear. The histone methyltransferase Kmt2b is highly expressed in SSCs and is required for the SSC-to-progenitor transition. At the stem-cell stage, Kmt2b catalyzes H3K4me3 at bivalent H3K27me3-marked promoters as well as at promoters of a new class of genes lacking H3K27me3, which we call monovalent. Monovalent genes are mainly activated in late spermatogenesis, whereas most bivalent genes are mainly not expressed until embryonic development. These data suggest that SSCs are epigenetically primed by Kmt2b in two distinguishable ways for the upregulation of gene expression both during the spermatogenic program and through the male germline into the embryo. Because Kmt2b is also the major H3K4 methyltransferase for bivalent promoters in embryonic stem cells, we also propose that Kmt2b has the capacity to prime stem cells epigenetically.
Collapse
Affiliation(s)
- Shin-Ichi Tomizawa
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Yuki Kobayashi
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Takayuki Shirakawa
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Kumiko Watanabe
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Keita Mizoguchi
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Ikue Hoshi
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Kuniko Nakajima
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Jun Nakabayashi
- Bioinformatics Laboratory, Advanced Medical Research Center, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Sukhdeep Singh
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Andreas Dahl
- Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Dimitra Alexopoulou
- Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Hélène Royo
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics, 4056 Basel, Switzerland
| | - Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.,Faculty of Sciences, University of Basel, 4058 Basel, Switzerland
| | - Konstantinos Anastassiadis
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - A Francis Stewart
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Kazuyuki Ohbo
- Department of Histology and Cell Biology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
30
|
Goldammer G, Neumann A, Strauch M, Müller-McNicoll M, Heyd F, Preußner M. Characterization of cis-acting elements that control oscillating alternative splicing. RNA Biol 2018; 15:1081-1092. [PMID: 30200840 DOI: 10.1080/15476286.2018.1502587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Alternative splicing (AS) in response to changing external conditions often requires alterations in the ability of sequence-specific RNA-binding proteins to bind to cis-acting sequences in their target pre-mRNA. While daily oscillations in AS events have been described in several organisms, cis-acting sequences that control time of the day-dependent AS remain largely elusive. Here we define cis-regulatory RNA elements that control body-temperature driven rhythmic AS using the mouse U2af26 gene as a model system. We identify a complex network of cis-regulatory sequences that regulate AS of U2af26, and show that the activity of two enhancer elements is necessary for oscillating AS. A minigene comprising these U2af26 regions recapitulates rhythmic splicing of the endogenous gene, which is controlled through temperature-regulated SR protein phosphorylation. Mutagenesis of the minigene delineates the cis-acting enhancer element for SRSF2 within exon 6 to single nucleotide resolution and reveals that the combined activity of SRSF2 and SRSF7 is required for oscillating U2af26 AS. By combining RNA-Seq with an siRNA screen and individual-nucleotide resolution cross-linking and immunoprecipitation (iCLIP), we identify a complex network of SR proteins that globally controls temperature-dependent rhythmic AS, with the direction of splicing depending on the position of the cis-acting elements. Together, we provide detailed insights into the sequence requirements that allow trans-acting factors to generate daily rhythms in AS.
Collapse
Affiliation(s)
- Gesine Goldammer
- a Laboratory of RNA Biochemistry , Freie Universität Berlin, Institute of Chemistry and Biochemistry , Berlin , Germany
| | - Alexander Neumann
- a Laboratory of RNA Biochemistry , Freie Universität Berlin, Institute of Chemistry and Biochemistry , Berlin , Germany
| | - Miriam Strauch
- a Laboratory of RNA Biochemistry , Freie Universität Berlin, Institute of Chemistry and Biochemistry , Berlin , Germany
| | - Michaela Müller-McNicoll
- b Cluster of Excellence Macromolecular Complexes, Institute of Cell Biology and Neuroscience , Goethe University Frankfurt , Frankfurt am Main , Germany
| | - Florian Heyd
- a Laboratory of RNA Biochemistry , Freie Universität Berlin, Institute of Chemistry and Biochemistry , Berlin , Germany
| | - Marco Preußner
- a Laboratory of RNA Biochemistry , Freie Universität Berlin, Institute of Chemistry and Biochemistry , Berlin , Germany
| |
Collapse
|
31
|
Wei YL, Yang WX. The acroframosome-acroplaxome-manchette axis may function in sperm head shaping and male fertility. Gene 2018; 660:28-40. [DOI: 10.1016/j.gene.2018.03.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 12/27/2022]
|
32
|
Zuo X, Rong B, Li L, Lv R, Lan F, Tong MH. The histone methyltransferase SETD2 is required for expression of acrosin-binding protein 1 and protamines and essential for spermiogenesis in mice. J Biol Chem 2018; 293:9188-9197. [PMID: 29716999 DOI: 10.1074/jbc.ra118.002851] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/26/2018] [Indexed: 11/06/2022] Open
Abstract
Spermatogenesis is precisely controlled by complex gene expression programs and involves epigenetic reprogramming, including histone modification and DNA methylation. SET domain-containing 2 (SETD2) is the predominant histone methyltransferase catalyzing the trimethylation of histone H3 lysine 36 (H3K36me3) and plays key roles in embryonic stem cell differentiation and somatic cell development. However, its role in male germ cell development remains elusive. Here, we demonstrate an essential role of Setd2 for spermiogenesis, the final stage of spermatogenesis. Using RNA-seq, we found that, in postnatal mouse testes, Setd2 mRNA levels dramatically increase in 14-day-old mice. Using a germ cell-specific Setd2 knockout mouse model, we also found that targeted Setd2 knockout in germ cells causes aberrant spermiogenesis with acrosomal malformation before step 8 of the round-spermatid stage, resulting in complete infertility. Furthermore, we noted that the Setd2 deficiency results in complete loss of H3K36me3 and significantly decreases expression of thousands of genes, including those encoding acrosin-binding protein 1 (Acrbp1) and protamines, required for spermatogenesis. Our findings thus reveal a previously unappreciated role of the SETD2-dependent H3K36me3 modification in spermiogenesis and provide clues to the molecular mechanisms in epigenetic disorders underlying male infertility.
Collapse
Affiliation(s)
- Xiaoli Zuo
- From the State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Bowen Rong
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Epigenetics, Shanghai Ministry of Education, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China, and
| | - Li Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Ruitu Lv
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Epigenetics, Shanghai Ministry of Education, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China, and
| | - Fei Lan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Epigenetics, Shanghai Ministry of Education, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China, and
| | - Ming-Han Tong
- From the State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China,
| |
Collapse
|
33
|
Taloni A, Font-Clos F, Guidetti L, Milan S, Ascagni M, Vasco C, Pasini ME, Gioria MR, Ciusani E, Zapperi S, La Porta CAM. Probing spermiogenesis: a digital strategy for mouse acrosome classification. Sci Rep 2017. [PMID: 28623263 PMCID: PMC5473909 DOI: 10.1038/s41598-017-03867-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Classification of morphological features in biological samples is usually performed by a trained eye but the increasing amount of available digital images calls for semi-automatic classification techniques. Here we explore this possibility in the context of acrosome morphological analysis during spermiogenesis. Our method combines feature extraction from three dimensional reconstruction of confocal images with principal component analysis and machine learning. The method could be particularly useful in cases where the amount of data does not allow for a direct inspection by trained eye.
Collapse
Affiliation(s)
- Alessandro Taloni
- Center for Complexity and Biosystems University of Milano, via Celoria 16, 20133, Milano, Italy.,Department of Physics, University of Milano, Via Celoria 16, 20133, Milano, Italy.,CNR-Consiglio Nazionale delle Ricerche, ISC, Via dei Taurini 19, 00185, Roma, Italy
| | | | - Luca Guidetti
- Center for Complexity and Biosystems University of Milano, via Celoria 16, 20133, Milano, Italy.,Department of Environmental Science and Policy, University of Milano, via Celoria 26, 20133, Milano, Italy
| | - Simone Milan
- Center for Complexity and Biosystems University of Milano, via Celoria 16, 20133, Milano, Italy.,ISI Foundation, Via Chisola 5, 10126, Torino, Italy.,Department of Environmental Science and Policy, University of Milano, via Celoria 26, 20133, Milano, Italy
| | - Miriam Ascagni
- Department of Biosciences University of Milano, via Celoria 26, 20133, Milano, Italy
| | - Chiara Vasco
- Istituto Neurologico Carlo Besta, Via Celoria, 11, 20133, Milano, Italy
| | - Maria Enrica Pasini
- Department of Biosciences University of Milano, via Celoria 26, 20133, Milano, Italy
| | - Maria Rosa Gioria
- Department of Biosciences University of Milano, via Celoria 26, 20133, Milano, Italy
| | - Emilio Ciusani
- Istituto Neurologico Carlo Besta, Via Celoria, 11, 20133, Milano, Italy
| | - Stefano Zapperi
- Center for Complexity and Biosystems University of Milano, via Celoria 16, 20133, Milano, Italy.,Department of Physics, University of Milano, Via Celoria 16, 20133, Milano, Italy.,ISI Foundation, Via Chisola 5, 10126, Torino, Italy.,Department of Applied Physics, Aalto University, P.O. Box 11100, FIN-00076, Aalto, Espoo, Finland.,CNR-Consiglio Nazionale delle Ricerche, ICMATE, Via Roberto Cozzi 53, 20125, Milano, Italy
| | - Caterina A M La Porta
- Center for Complexity and Biosystems University of Milano, via Celoria 16, 20133, Milano, Italy. .,Department of Environmental Science and Policy, University of Milano, via Celoria 26, 20133, Milano, Italy.
| |
Collapse
|
34
|
Jiang L, Li T, Zhang X, Zhang B, Yu C, Li Y, Fan S, Jiang X, Khan T, Hao Q, Xu P, Nadano D, Huleihel M, Lunenfeld E, Wang PJ, Zhang Y, Shi Q. RPL10L Is Required for Male Meiotic Division by Compensating for RPL10 during Meiotic Sex Chromosome Inactivation in Mice. Curr Biol 2017; 27:1498-1505.e6. [DOI: 10.1016/j.cub.2017.04.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/05/2017] [Accepted: 04/11/2017] [Indexed: 10/19/2022]
|
35
|
Gallego-Paez LM, Bordone MC, Leote AC, Saraiva-Agostinho N, Ascensão-Ferreira M, Barbosa-Morais NL. Alternative splicing: the pledge, the turn, and the prestige : The key role of alternative splicing in human biological systems. Hum Genet 2017; 136:1015-1042. [PMID: 28374191 PMCID: PMC5602094 DOI: 10.1007/s00439-017-1790-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/25/2017] [Indexed: 02/06/2023]
Abstract
Alternative pre-mRNA splicing is a tightly controlled process conducted by the spliceosome, with the assistance of several regulators, resulting in the expression of different transcript isoforms from the same gene and increasing both transcriptome and proteome complexity. The differences between alternative isoforms may be subtle but enough to change the function or localization of the translated proteins. A fine control of the isoform balance is, therefore, needed throughout developmental stages and adult tissues or physiological conditions and it does not come as a surprise that several diseases are caused by its deregulation. In this review, we aim to bring the splicing machinery on stage and raise the curtain on its mechanisms and regulation throughout several systems and tissues of the human body, from neurodevelopment to the interactions with the human microbiome. We discuss, on one hand, the essential role of alternative splicing in assuring tissue function, diversity, and swiftness of response in these systems or tissues, and on the other hand, what goes wrong when its regulatory mechanisms fail. We also focus on the possibilities that splicing modulation therapies open for the future of personalized medicine, along with the leading techniques in this field. The final act of the spliceosome, however, is yet to be fully revealed, as more knowledge is needed regarding the complex regulatory network that coordinates alternative splicing and how its dysfunction leads to disease.
Collapse
Affiliation(s)
- L M Gallego-Paez
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - M C Bordone
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - A C Leote
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - N Saraiva-Agostinho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - M Ascensão-Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - N L Barbosa-Morais
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
36
|
The control of male fertility by spermatid-specific factors: searching for contraceptive targets from spermatozoon's head to tail. Cell Death Dis 2016; 7:e2472. [PMID: 27831554 PMCID: PMC5260884 DOI: 10.1038/cddis.2016.344] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 09/18/2016] [Accepted: 09/26/2016] [Indexed: 02/06/2023]
Abstract
Male infertility due to abnormal spermatozoa has been reported in both animals and humans, but its pathogenic causes, including genetic abnormalities, remain largely unknown. On the other hand, contraceptive options for men are limited, and a specific, reversible and safe method of male contraception has been a long-standing quest in medicine. Some progress has recently been made in exploring the effects of spermatid-specifical genetic factors in controlling male fertility. A comprehensive search of PubMed for articles and reviews published in English before July 2016 was carried out using the search terms 'spermiogenesis failure', 'globozoospermia', 'spermatid-specific', 'acrosome', 'infertile', 'manchette', 'sperm connecting piece', 'sperm annulus', 'sperm ADAMs', 'flagellar abnormalities', 'sperm motility loss', 'sperm ion exchanger' and 'contraceptive targets'. Importantly, we have opted to focus on articles regarding spermatid-specific factors. Genetic studies to define the structure and physiology of sperm have shown that spermatozoa appear to be one of the most promising contraceptive targets. Here we summarize how these spermatid-specific factors regulate spermiogenesis and categorize them according to their localization and function from spermatid head to tail (e.g., acrosome, manchette, head-tail conjunction, annulus, principal piece of tail). In addition, we emphatically introduce small-molecule contraceptives, such as BRDT and PPP3CC/PPP3R2, which are currently being developed to target spermatogenic-specific proteins. We suggest that blocking the differentiation of haploid germ cells, which rarely affects early spermatogenic cell types and the testicular microenvironment, is a better choice than spermatogenic-specific proteins. The studies described here provide valuable information regarding the genetic and molecular defects causing male mouse infertility to improve our understanding of the importance of spermatid-specific factors in controlling fertility. Although a male contraceptive 'pill' is still many years away, research into the production of new small-molecule contraceptives targeting spermatid-specific proteins is the right avenue.
Collapse
|