1
|
Yuan T, Yang J, Xu D, Li H, Min W, Wang F. UBL7 is indispensable for spermiogenesis through protecting critical factors from excessive degradation by proteasomes. Nat Commun 2025; 16:3803. [PMID: 40268954 PMCID: PMC12019544 DOI: 10.1038/s41467-025-59209-z] [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: 09/03/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
Spermiogenesis is a tightly regulated process to produce mature sperm cells. The ubiquitin-proteasome system (UPS) plays a crucial role in controlling protein half-life and is essential for spermiogenesis. Recently, proteins containing ubiquitin-like domains and ubiquitin-associated domains (UBL-UBA proteins) have emerged as novel regulators within the UPS. In this study, we demonstrate that UBL7, a testis-enriched UBL-UBA protein, is indispensable for sperm formation. Deficiency of UBL7 leads to severe malformations of both the sperm tail and head. Mechanistically, UBL7 interacts with the valosin-containing protein (VCP) complex and proteasomes, and shuttles substrates between them. Notably, UBL7 slows down the degradation rates of substrates involved in endoplasmic reticulum-associated degradation (ERAD) within cells. Through a two-step immunoprecipitation method, we identify several essential factors in spermatids that are protected by UBL7, including factors involved in the development of manchette (such as IFT140), head-tail coupling apparatus (such as SPATA20) and cytoplasmic droplets (such as HK1 and SLC2a3). In summary, our findings highlight UBL7 as a guardian that protects crucial factors from excessive degradation and thereby ensures successful spermiogenesis.
Collapse
Affiliation(s)
- Tianyi Yuan
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- National Institute of Biological Sciences, Beijing, China
| | - Jiajun Yang
- National Institute of Biological Sciences, Beijing, China
- College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Dan Xu
- National Institute of Biological Sciences, Beijing, China
| | - Huiqi Li
- National Institute of Biological Sciences, Beijing, China
| | - Wanping Min
- National Institute of Biological Sciences, Beijing, China
| | - Fengchao Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
- National Institute of Biological Sciences, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China.
| |
Collapse
|
2
|
He J, Lin X, Tan C, Li Y, Su L, Lin G, Tan YQ, Tu C. Molecular insights into sperm head shaping and its role in human male fertility. Hum Reprod Update 2025:dmaf003. [PMID: 40037590 DOI: 10.1093/humupd/dmaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/16/2024] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Sperm head shaping, controlled by the acrosome-acroplaxome-manchette complex, represents a significant morphological change during spermiogenesis and involves numerous proteins expressed in a spatially and temporally specific manner. Defects in sperm head shaping frequently lead to teratozoospermia concomitant with oligozoospermia and asthenozoospermia, but the pathogenic mechanism underlying sperm head shaping, and its role in male infertility, remain poorly understood. OBJECTIVE AND RATIONALE This review aims to summarize the mechanism underlying sperm head shaping, reveal the relationship between gene defects associated with sperm head shaping and male infertility in humans and mice, and explore potential clinical improvements in ICSI treatment. SEARCH METHODS We searched the PubMed database for articles published in English using the keyword 'sperm head shaping' in combination with the following terms: 'acrosome formation', 'proacrosomal vesicles (PAVs)', 'manchette', 'perinuclear theca (PT)', 'chromatin condensation', 'linker of nucleoskeleton and cytoskeleton (LINC) complex', 'histone-to-protamine (HTP) transition', 'male infertility', 'ICSI', and 'artificial oocyte activation (AOA)'. The selected publications until 1 August 2024 were critically summarized, integrated, and thoroughly discussed, and the irrelevant literature were excluded. OUTCOMES A total of 6823 records were retrieved. After careful screening, integrating relevant literature, and excluding articles unrelated to the topic of this review, 240 articles were ultimately included in the analysis. Firstly, we reviewed the important molecular events and structures integral to sperm head shaping, including PAV formation to fusion, acrosome attachment to the nucleus, structure and function of the manchette, PT, chromatin condensation, and HTP transition. Then, we set forth human male infertility associated with sperm head shaping and identified genes related to sperm head shaping resulting in teratozoospermia concomitant with oligozoospermia and asthenozoospermia. Finally, we summarized the outcomes of ICSI in cases of male infertility resulting from mutations in the genes associated with sperm head shaping, as well as the ICSI outcomes through AOA for infertile men with impaired sperm head. WIDER IMPLICATIONS Understanding the molecular mechanisms of sperm head shaping and its relationship with human male infertility holds profound clinical implications, which may contribute to risk prediction, genetic diagnosis, and the potential treatment of human male infertility.
Collapse
Affiliation(s)
- Jiaxin He
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Xinle Lin
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Chen Tan
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yong Li
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Lilan Su
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Ge Lin
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Yue-Qiu Tan
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| | - Chaofeng Tu
- Institute of Reproduction and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Xiangya Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, Hunan, China
| |
Collapse
|
3
|
Bilekova S, Garcia-Colomer B, Cebrian-Serrano A, Schirge S, Krey K, Sterr M, Kurth T, Hauck SM, Lickert H. Inceptor facilitates acrosomal vesicle formation in spermatids and is required for male fertility. Front Cell Dev Biol 2023; 11:1240039. [PMID: 37691832 PMCID: PMC10483240 DOI: 10.3389/fcell.2023.1240039] [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: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Spermatogenesis is a crucial biological process that enables the production of functional sperm, allowing for successful reproduction. Proper germ cell differentiation and maturation require tight regulation of hormonal signals, cellular signaling pathways, and cell biological processes. The acrosome is a lysosome-related organelle at the anterior of the sperm head that contains enzymes and receptors essential for egg-sperm recognition and fusion. Even though several factors crucial for acrosome biogenesis have been discovered, the precise molecular mechanism of pro-acrosomal vesicle formation and fusion is not yet known. In this study, we investigated the role of the insulin inhibitory receptor (inceptor) in acrosome formation. Inceptor is a single-pass transmembrane protein with similarities to mannose-6-phosphate receptors (M6PR). Inceptor knockout male mice are infertile due to malformations in the acrosome and defects in the nuclear shape of spermatozoa. We show that inceptor is expressed in early spermatids and mainly localizes to vesicles between the Golgi apparatus and acrosome. Here we show that inceptor is an essential factor in the intracellular transport of trans-Golgi network-derived vesicles which deliver acrosomal cargo in maturing spermatids. The absence of inceptor results in vesicle-fusion defects, acrosomal malformation, and male infertility. These findings support our hypothesis of inceptor as a universal lysosomal or lysosome-related organelle sorting receptor expressed in several secretory tissues.
Collapse
Affiliation(s)
- Sara Bilekova
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Balma Garcia-Colomer
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Helmholtz Center Munich, Institute for Diabetes and Obesity, Neuherberg, Germany
| | - Alberto Cebrian-Serrano
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Helmholtz Center Munich, Institute for Diabetes and Obesity, Neuherberg, Germany
| | - Silvia Schirge
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karsten Krey
- School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Virology, Technical University of Munich, Munich, Germany
| | - Michael Sterr
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform, Core Facility Electron Microscopy and Histology, Dresden University of Technology, Dresden, Germany
| | - Stefanie M. Hauck
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Munich, Germany
| | - Heiko Lickert
- Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Institute of Diabetes and Regeneration Research, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
4
|
Fang YZ, Jiang L, He Q, Cao J, Yang B. Commentary: Deubiquitination complex platform: a plausible mechanism for regulating the substrate specificity of deubiquitinating enzymes. Acta Pharm Sin B 2023. [PMID: 37521861 PMCID: PMC10372820 DOI: 10.1016/j.apsb.2023.02.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Deubiquitinating enzymes (DUBs) or deubiquitinases facilitate the escape of multiple proteins from ubiquitin‒proteasome degradation and are critical for regulating protein expression levels in vivo. Therefore, dissecting the underlying mechanism of DUB recognition is needed to advance the development of drugs related to DUB signaling pathways. To data, extensive studies on the ubiquitin chain specificity of DUBs have been reported, but substrate protein recognition is still not clearly understood. As a breakthrough, the scaffolding role may be significant to substrate protein selectivity. From this perspective, we systematically characterized the scaffolding proteins and complexes contributing to DUB substrate selectivity. Furthermore, we proposed a deubiquitination complex platform (DCP) as a potentially generic mechanism for DUB substrate recognition based on known examples, which might fill the gaps in the understanding of DUB substrate specificity.
Collapse
|
5
|
Otčenášková T, Macíčková E, Vondráková J, Frolíková M, Komrskova K, Stopková R, Stopka P. Proteomic analysis of the mouse sperm acrosome - towards an understanding of an organelle with diverse functionality. Eur J Cell Biol 2023; 102:151296. [PMID: 36805822 DOI: 10.1016/j.ejcb.2023.151296] [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: 07/16/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
The acrosome located within the mammalian sperm head is essential for successful fertilization, as it enables the sperm to penetrate the extracellular layers of the oocyte and fuse with oolemma. However, the mammalian acrosomal vesicle is no longer considered to contain only hydrolytic enzymes. Using label-free nano-scale liquid chromatography tandem mass spectrometry (nLC-MS/MS) proteomics, we identified a total of 885 proteins in the acrosome isolated from spermatozoa obtained from cauda epididymis of free-living house mice Mus musculus musculus contains a total of 885 proteins. Among these, 334 proteins were significantly enriched in the acrosome thus representing 27.3% of the whole proteome of the intact sperm. Importantly, we have detected a total of nine calycins while eight of them belong to the lipocalin protein family. In mice, lipocalins are involved in multi-level chemical communication between individuals including pheromone transport and odor perception. Using an indirect immunofluorescence assay, we demonstrated that lipocalin 5 (LCN5) is expressed in the mouse germ cells, and after completing spermatogenesis, it remains localized in the sperm acrosome until the last step of the extratesticular maturation, the acrosome reaction. The presence of lipocalins in the acrosome and acrosome-reacted sperm suggests their original role as chelators of organic and potentially toxic compounds resulting from ongoing spermiogenesis. Along with this evidence, detected mitochondrial (e.g., a subunit of the cytochrome c oxidase MTCO1) and proteasomal proteins (subunits of both 20 S core proteasome [PSMA2, PSMBs] and 19 S regulatory particle [PSMDs]) in acrosomes provide further evidence that acrosomes could also function as `waste baskets` after testicular sperm maturation.
Collapse
Affiliation(s)
- Tereza Otčenášková
- Department of Zoology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic.
| | - Eliška Macíčková
- Department of Zoology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic.
| | - Jana Vondráková
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic.
| | - Michaela Frolíková
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic.
| | - Katerina Komrskova
- Department of Zoology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic; Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic.
| | - Romana Stopková
- Department of Zoology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic.
| | - Pavel Stopka
- Department of Zoology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic.
| |
Collapse
|
6
|
The stallion sperm acrosome: Considerations from a research and clinical perspective. Theriogenology 2023; 196:121-149. [PMID: 36413868 DOI: 10.1016/j.theriogenology.2022.11.012] [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: 08/30/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
During the fertilization process, the interaction between the sperm and the oocyte is mediated by a process known as acrosomal exocytosis (AE). Although the role of the sperm acrosome on fertilization has been studied extensively over the last 70 years, little is known about the molecular mechanisms that govern acrosomal function, particularly in species other than mice or humans. Even though subfertility due to acrosomal dysfunction is less common in large animals than in humans, the evaluation of sperm acrosomal function should be considered not only as a complementary but a routine test when individuals are selected for breeding potential. This certainly holds true for stallions, which might display lower levels of fertility in the face of "acceptable" sperm quality parameters determined by conventional sperm assays. Nowadays, the use of high throughput technologies such as flow cytometry or mass spectrometry-based proteomic analysis is commonplace in the research arena. Such techniques can also be implemented in clinical scenarios of males with "idiopathic" subfertility. The current review focuses on the sperm acrosome, with particular emphasis on the stallion. We aim to describe the physiological events that lead to the acrosome formation within the testis, the role of very specific acrosomal proteins during AE, the methods to study the occurrence of AE under in vitro conditions, and the potential use of molecular biology techniques to discover new markers of acrosomal function and subfertility associated with acrosomal dysfunction in stallions.
Collapse
|
7
|
Wang J, Zhou Q, Ding J, Yin T, Ye P, Zhang Y. The Conceivable Functions of Protein Ubiquitination and Deubiquitination in Reproduction. Front Physiol 2022; 13:886261. [PMID: 35910557 PMCID: PMC9326170 DOI: 10.3389/fphys.2022.886261] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/29/2022] [Indexed: 12/02/2022] Open
Abstract
Protein ubiquitination with general existence in virtually all eukaryotic cells serves as a significant post-translational modification of cellular proteins, which leads to the degradation of proteins via the ubiquitin-proteasome system. Deubiquitinating enzymes (DUBs) can reverse the ubiquitination effect by removing the ubiquitin chain from the target protein. Together, these two processes participate in regulating protein stability, function, and localization, thus modulating cell cycle, DNA repair, autophagy, and transcription regulation. Accumulating evidence indicates that the ubiquitination/deubiquitination system regulates reproductive processes, including the cell cycle, oocyte maturation, oocyte-sperm binding, and early embryonic development, primarily by regulating protein stability. This review summarizes the extensive research concerning the role of ubiquitin and DUBs in gametogenesis and early embryonic development, which helps us to understand human pregnancy further.
Collapse
Affiliation(s)
- Jiayu Wang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Qi Zhou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Jinli Ding
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Tian Y, Liu K, Liu R, Qiu Z, Xu Y, Wei W, Xu X, Wang J, Ding H, Li Z, Bian J. Discovery of Potent Small-Molecule USP8 Inhibitors for the Treatment of Breast Cancer through Regulating ERα Expression. J Med Chem 2022; 65:8914-8932. [PMID: 35786929 DOI: 10.1021/acs.jmedchem.2c00013] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ubiquitin-specific protease 8 (USP8), belonging to the deubiquitinase family, has been implicated to be closely related to the occurrence of many malignant tumors, but only a few USP8-targeting inhibitors have been reported to date. In this study, we present virtual screening to discover novel hit candidates that inhibit the catalytic activity of USP8. Exploration of the structure-activity relationship led to the identification of compound DC-U4106, which binds to USP8 with a KD value of 4.7 μM and is selective over USP2 and USP7. Western blotting and immunoprecipitation showed that DC-U4106 could target the ubiquitin pathway and facilitate the degradation of ERα. In a xenograft tumor model, DC-U4106 also significantly inhibited tumor growth with minimal toxicity. Overall, our findings suggest that DC-U4106 is a promising drug candidate and targeting the USP8-ERα complex could be a new approach to treat ER-positive or drug-resistant breast cancer.
Collapse
Affiliation(s)
- Yucheng Tian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Kang Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Ruoyi Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Zhixia Qiu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Yifan Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Wei Wei
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Xi Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Jubo Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Hong Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Material Medical, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai201203, China
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| | - Jinlei Bian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing210009, China
| |
Collapse
|
9
|
Ren M, Xu Y, Phoon CKL, Erdjument-Bromage H, Neubert TA, Rajan S, Hussain MM, Schlame M. Condensed Mitochondria Assemble Into the Acrosomal Matrix During Spermiogenesis. Front Cell Dev Biol 2022; 10:867175. [PMID: 35531097 PMCID: PMC9068883 DOI: 10.3389/fcell.2022.867175] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/07/2022] [Indexed: 12/02/2022] Open
Abstract
Mammalian spermatogenesis is associated with the transient appearance of condensed mitochondria, a singularity of germ cells with unknown function. Using proteomic analysis, respirometry, and electron microscopy with tomography, we studied the development of condensed mitochondria. Condensed mitochondria arose from orthodox mitochondria during meiosis by progressive contraction of the matrix space, which was accompanied by an initial expansion and a subsequent reduction of the surface area of the inner membrane. Compared to orthodox mitochondria, condensed mitochondria respired more actively, had a higher concentration of respiratory enzymes and supercomplexes, and contained more proteins involved in protein import and expression. After the completion of meiosis, the abundance of condensed mitochondria declined, which coincided with the onset of the biogenesis of acrosomes. Immuno-electron microscopy and the analysis of sub-cellular fractions suggested that condensed mitochondria or their fragments were translocated into the lumen of the acrosome. Thus, it seems condensed mitochondria are formed from orthodox mitochondria by extensive transformations in order to support the formation of the acrosomal matrix.
Collapse
Affiliation(s)
- Mindong Ren
- Departments of Anesthesiology, New York, NY, United States
- Department of Cell Biology, New York, NY, United States
| | - Yang Xu
- Departments of Anesthesiology, New York, NY, United States
| | | | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York, NY, United States
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, United States
| | - Thomas A. Neubert
- Department of Cell Biology, New York, NY, United States
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, United States
| | - Sujith Rajan
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, United States
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, United States
| | - Michael Schlame
- Departments of Anesthesiology, New York, NY, United States
- Department of Cell Biology, New York, NY, United States
| |
Collapse
|
10
|
Xiong Y, Yu C, Zhang Q. Ubiquitin-Proteasome System-Regulated Protein Degradation in Spermatogenesis. Cells 2022; 11:1058. [PMID: 35326509 PMCID: PMC8947704 DOI: 10.3390/cells11061058] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis is a prolonged and highly ordered physiological process that produces haploid male germ cells through more than 40 steps and experiences dramatic morphological and cellular transformations. The ubiquitin proteasome system (UPS) plays central roles in the precise control of protein homeostasis to ensure the effectiveness of certain protein groups at a given stage and the inactivation of them after this stage. Many UPS components have been demonstrated to regulate the progression of spermatogenesis at different levels. Especially in recent years, novel testis-specific proteasome isoforms have been identified to be essential and unique for spermatogenesis. In this review, we set out to discuss our current knowledge in functions of diverse USP components in mammalian spermatogenesis through: (1) the composition of proteasome isoforms at each stage of spermatogenesis; (2) the specificity of each proteasome isoform and the associated degradation events; (3) the E3 ubiquitin ligases mediating protein ubiquitination in male germ cells; and (4) the deubiquitinases involved in spermatogenesis and male fertility. Exploring the functions of UPS machineries in spermatogenesis provides a global picture of the proteome dynamics during male germ cell production and shed light on the etiology and pathogenesis of human male infertility.
Collapse
Affiliation(s)
- Yi Xiong
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Rd, Haining 314400, China;
| | - Chao Yu
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Zhejiang University, Sir Run Run Shaw Hospital, 3 East Qing Chun Rd, Hangzhou 310020, China;
- College of Life Sciences, Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, China
| | - Qianting Zhang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Rd, Haining 314400, China;
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
11
|
Carrasquel Martínez G, Aldana A, Carneiro J, Treviño CL, Darszon A. Acrosomal alkalinization occurs during human sperm capacitation. Mol Hum Reprod 2022; 28:6535714. [PMID: 35201340 DOI: 10.1093/molehr/gaac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 01/22/2022] [Indexed: 11/14/2022] Open
Abstract
Mammalian sperm capacitation is a prerequisite for successful fertilization. Capacitation involves biochemical and physiological modifications of sperm as they travel through the female reproductive tract. These modifications prepare the sperm to undergo the acrosome reaction (AR), an acrosome vesicle exocytosis that is necessary for gamete fusion. Capacitation requires an increase in both intracellular calcium ([Ca2+]i) and pH (pHi). Mouse sperm capacitation is accompanied by acrosomal alkalinization and artificial elevation of the acrosome pH (pHa) is sufficient to trigger the AR in mouse and human sperm, but it is unknown if pHa increases naturally during human sperm capacitation. We used single-cell imaging and image-based flow cytometry to evaluate pHa during capacitation and its regulation. We found that pHa progressively increases during capacitation. The V-ATPase, which immunolocalized to the acrosome and equatorial segment, is mainly responsible for the acidity of the acrosome. It is likely that the regulation of V-ATPase is at least in part responsible for the progressive increase in pHa during capacitation. Acrosome alkalinization was dependent on extracellular HCO3- and Ca2+. Inhibition of the HCO3--dependent adenylyl cyclase and protein kinase A induced significant pHa changes. Overall, alkalinization of the acrosome may be a key step in the path towards the AR.
Collapse
Affiliation(s)
- Gabriela Carrasquel Martínez
- Departamento de Genética del Desarrollo y Fisiología Molecular. Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, 62210, México
| | - Andrés Aldana
- Departamento de Genética del Desarrollo y Fisiología Molecular. Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, 62210, México.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jorge Carneiro
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal.,Instituto de Tecnología Química e Biológica António Xavier, Universida de Nova, Oeiras, Portugal
| | - Claudia Lydia Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular. Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, 62210, México
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular. Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, 62210, México
| |
Collapse
|
12
|
Wang M, Zeng L, Su P, Ma L, Zhang M, Zhang YZ. Autophagy: a multifaceted player in the fate of sperm. Hum Reprod Update 2021; 28:200-231. [PMID: 34967891 PMCID: PMC8889000 DOI: 10.1093/humupd/dmab043] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Autophagy is an intracellular catabolic process of degrading and recycling proteins and organelles to modulate various physiological and pathological events, including cell differentiation and development. Emerging data indicate that autophagy is closely associated with male reproduction, especially the biosynthetic and catabolic processes of sperm. Throughout the fate of sperm, a series of highly specialized cellular events occur, involving pre-testicular, testicular and post-testicular events. Nonetheless, the most fundamental question of whether autophagy plays a protective or harmful role in male reproduction, especially in sperm, remains unclear. OBJECTIVE AND RATIONALE We summarize the functional roles of autophagy in the pre-testicular (hypothalamic–pituitary–testis (HPG) axis), testicular (spermatocytogenesis, spermatidogenesis, spermiogenesis, spermiation) and post-testicular (sperm maturation and fertilization) processes according to the timeline of sperm fate. Additionally, critical mechanisms of the action and clinical impacts of autophagy on sperm are identified, laying the foundation for the treatment of male infertility. SEARCH METHODS In this narrative review, the PubMed database was used to search peer-reviewed publications for summarizing the functional roles of autophagy in the fate of sperm using the following terms: ‘autophagy’, ‘sperm’, ‘hypothalamic–pituitary–testis axis’, ‘spermatogenesis’, ‘spermatocytogenesis’, ‘spermatidogenesis’, ‘spermiogenesis’, ‘spermiation’, ‘sperm maturation’, ‘fertilization’, ‘capacitation’ and ‘acrosome’ in combination with autophagy-related proteins. We also performed a bibliographic search for the clinical impact of the autophagy process using the keywords of autophagy inhibitors such as ‘bafilomycin A1’, ‘chloroquine’, ‘hydroxychloroquine’, ‘3-Methyl Adenine (3-MA)’, ‘lucanthone’, ‘wortmannin’ and autophagy activators such as ‘rapamycin’, ‘perifosine’, ‘metformin’ in combination with ‘disease’, ‘treatment’, ‘therapy’, ‘male infertility’ and equivalent terms. In addition, reference lists of primary and review articles were reviewed for additional relevant publications. All relevant publications until August 2021 were critically evaluated and discussed on the basis of relevance, quality and timelines. OUTCOMES (i) In pre-testicular processes, autophagy-related genes are involved in the regulation of the HPG axis; and (ii) in testicular processes, mTORC1, the main gate to autophagy, is crucial for spermatogonia stem cell (SCCs) proliferation, differentiation, meiotic progression, inactivation of sex chromosomes and spermiogenesis. During spermatidogenesis, autophagy maintains haploid round spermatid chromatoid body homeostasis for differentiation. During spermiogenesis, autophagy participates in acrosome biogenesis, flagella assembly, head shaping and the removal of cytoplasm from elongating spermatid. After spermatogenesis, through PDLIM1, autophagy orchestrates apical ectoplasmic specialization and basal ectoplasmic specialization to handle cytoskeleton assembly, governing spermatid movement and release during spermiation. In post-testicular processes, there is no direct evidence that autophagy participates in the process of capacitation. However, autophagy modulates the acrosome reaction, paternal mitochondria elimination and clearance of membranous organelles during fertilization. WIDER IMPLICATIONS Deciphering the roles of autophagy in the entire fate of sperm will provide valuable insights into therapies for diseases, especially male infertility.
Collapse
Affiliation(s)
- Mei Wang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Harvard Reproductive Endocrine Science Center and Reproductive Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| | - Ling Zeng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Ping Su
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Ling Ma
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| | - Ming Zhang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| | - Yuan Zhen Zhang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| |
Collapse
|
13
|
Dunleavy JEM, O'Connor AE, Okuda H, Merriner DJ, O'Bryan MK. KATNB1 is a master regulator of multiple katanin enzymes in male meiosis and haploid germ cell development. Development 2021; 148:273717. [PMID: 34822718 DOI: 10.1242/dev.199922] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Katanin microtubule-severing enzymes are crucial executers of microtubule regulation. Here, we have created an allelic loss-of-function series of the katanin regulatory B-subunit KATNB1 in mice. We reveal that KATNB1 is the master regulator of all katanin enzymatic A-subunits during mammalian spermatogenesis, wherein it is required to maintain katanin A-subunit abundance. Our data shows that complete loss of KATNB1 from germ cells is incompatible with sperm production, and we reveal multiple new spermatogenesis functions for KATNB1, including essential roles in male meiosis, acrosome formation, sperm tail assembly, regulation of both the Sertoli and germ cell cytoskeletons during sperm nuclear remodelling, and maintenance of seminiferous epithelium integrity. Collectively, our findings reveal that katanins are able to differentially regulate almost all key microtubule-based structures during mammalian male germ cell development, through the complexing of one master controller, KATNB1, with a 'toolbox' of neofunctionalised katanin A-subunits.
Collapse
Affiliation(s)
- Jessica E M Dunleavy
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Anne E O'Connor
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Hidenobu Okuda
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia
| | - D Jo Merriner
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Moira K O'Bryan
- School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
14
|
Li Y, Wang Y, Wen Y, Zhang T, Wang X, Jiang C, Zheng R, Zhou F, Chen D, Yang Y, Shen Y. Whole-exome sequencing of a cohort of infertile men reveals novel causative genes in teratozoospermia that are chiefly related to sperm head defects. Hum Reprod 2021; 37:152-177. [PMID: 34791246 DOI: 10.1093/humrep/deab229] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION Can whole-exome sequencing (WES) and in vitro validation studies identify new causative genes associated with teratozoospermia, particularly for sperm head defect? SUMMARY ANSWER We investigated a core group of infertile patients, including 82 cases with unexplained abnormal sperm head and 67 individuals with multiple morphological abnormalities of the sperm flagella (MMAF), and revealed rare and novel deleterious gene variants correlated with morphological abnormalities of the sperm head or tail defects. WHAT IS KNOWN ALREADY Teratozoospermia is one of the most common factors causing male infertility. Owing to high phenotypic variability, currently known genetic causes of teratozoospermia can only explain a rather minor component for patients with anomalous sperm-head shapes, and the agents responsible for atypical sperm head shapes remain largely unknown. STUDY DESIGN, SIZE, DURATION We executed WES analysis of a Chinese cohort of patients (N = 149) with teratozoospermia to identify novel genetic causes particularly for defective sperm head. We also sought to reveal the influence of different abnormalities of sperm morphology on ICSI outcome. PARTICIPANTS/MATERIALS, SETTING, METHODS In this study, a cohort of 149 infertile men (82 with abnormal sperm head and 67 with MMAF) were recruited. We implemented WES on infertile patients and analyzed the negative effects of the mutations of candidate genes on their protein conformations and/or expression. We also investigated the candidate genes' spatiotemporal expression/localization during spermatogenesis in both humans and mice, and explored their interactions with proteins that are known to be involved in sperm development. We also compared the ICSI outcomes of the affected individuals with various aberrations in sperm morphology. MAIN RESULTS AND THE ROLE OF CHANCE We identified rare and deleterious variants of piwi like RNA-mediated gene silencing 4 (PIWIL4: 1/82 patients, 1.21%), coiled-coil and C2 domain containing 1B (CC2D1B: 1/82 patients, 1.21%), cyclin B3 (CCNB3: 1/82 patients, 1.21%), KIAA1210 (KIAA1210: 2/82 patients, 2.43%) and choline phosphotransferase 1 (CHPT1: 1/82 patients, 1.21%), which are novel correlates of morphological abnormalities of the sperm head; functional evidence supports roles for all of these genes in sperm head formation. The mutations of septin 12 (SEPTIN12: 2/82 patients, 2.43%) are suggested to be associated with acrosome defects. We additionally observed novel causative mutations of dynein axonemal heavy chain 2 (DNAH2: 1/67 patients, 1.49%), dynein axonemal heavy chain 10 (DNAH10: 1/67 patients, 1.49%) and dynein axonemal heavy chain 12 (DNAH12: 1/67 patients, 1.49%) in patients with MMAF, and revealed a significantly lower fertilization rate of the abnormal sperm-head group compared to the MMAF group following ICSI. Consequently, our study also suggests that the mutations of PIWIL4 and CC2D1B might be circumvented by ICSI to a degree, and that CHPT1 and KIAA1210 loss-of-function variants might be associated with failed ICSI treatment. LIMITATIONS, REASONS FOR CAUTION In this study, we discovered the relationship between the genotype and phenotype of the novel causative genes of sperm head deformities in humans. However, the molecular mechanism of the relevant genes involved in sperm head development needs to be further illuminated in future research. Furthermore, evidence should be provided using knockout/knock-in mouse models for additional confirmation of the roles of these novel genes in spermatogenesis. WIDER IMPLICATIONS OF THE FINDINGS This cohort study of 149 Chinese infertile men documents novel genetic factors involved in teratozoospermia, particularly in anomalous sperm head formation. For the first time, we suggest that SEPTIN12 is related to human acrosomal hypoplasia, and that CCNB3 is a novel causative gene for globozoospermia in humans. We also uncovered variants in two genes-KIAA1210 and CHPT1associated with acrosomal biogenesis in patients with small or absent acrosomes. Additionally, it is postulated that loss-of-function mutations of PIWIL4 and CC2D1B have a contribution to the abnormal sperm-head formation. Furthermore, we are first to demonstrate the influence of different sperm morphologies on ICSI outcomes and indicates that the abnormal sperm head may play a significant role in fertilization failure. Our findings therefore provide valuable information for the diagnosis of teratozoospermia, particularly with respect to abnormalities of the sperm head. This will allow clinicians to adopt the optimal treatment strategy and to develop personalized medicine directly targeting these effects. STUDY FUNDING/COMPETING INTEREST(S) This work was financed by the West China Second University Hospital of Sichuan University (KS369 and KL042). The authors declare that they do not have any conflicts of interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yaqian Li
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Wang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Yuting Wen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Tao Zhang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Xiaodong Wang
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chuan Jiang
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Zheng
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fan Zhou
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Daijuan Chen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yihong Yang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Ying Shen
- Department of Obstetrics/Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
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: 19] [Impact Index Per Article: 4.8] [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
|
16
|
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: 3.0] [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
|
17
|
Berruti G. Destruction or Reconstruction: A Subtle Liaison between the Proteolytic and Signaling Role of Protein Ubiquitination in Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:215-240. [PMID: 34453739 DOI: 10.1007/978-3-030-77779-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Ubiquitination is one of the most diverse forms of protein post-translational modification that changes the function of the landscape of substrate proteins in response to stimuli, without the need for "de novo" protein synthesis. Ubiquitination is involved in almost all aspects of eukaryotic cell biology, from the best-studied role in promoting the removal of faulty or unnecessary proteins by the way of the ubiquitin proteasome system and autophagy-lysosome pathway to the recruitment of proteins in specific non-proteolytic signaling pathways, as emerged by the more recent discoveries about the protein signature with peculiar types of ubiquitin chains. Spermatogenesis, on its own, is a complex cellular developmental process in which mitosis, meiosis, and cell differentiation coexist so to result in the continuous formation of haploid spermatozoa. Successful spermatogenesis is thus at the same time a mixed result of the precise expression and correct intracellular destination of structural proteins and enzymes, from one hand, and the fine removal by targeted degradation of unfolded or damaged proteins as well as of obsolete, outlived proteins, from the other hand. In this minireview, I will focus on the importance of the ubiquitin system all over the spermatogenic process, discussing both proteolytic and non-proteolytic functions of protein ubiquitination. Alterations in the ubiquitin system have been in fact implicated in pathologies leading to male infertility. Notwithstanding several aspects of the multifaceted world of the ubiquitin system have been clarified, the physiological meaning of the so-called ubiquitin code remains still partially elusive. The studies reviewed in this chapter provide information that could aid the investigators to pursue new promising discoveries in the understanding of human and animal reproductive potential.
Collapse
|
18
|
Cerván-Martín M, Bossini-Castillo L, Rivera-Egea R, Garrido N, Luján S, Romeu G, Santos-Ribeiro S, Castilla JA, Gonzalvo MC, Clavero A, Vicente FJ, Guzmán-Jiménez A, Costa C, Llinares-Burguet I, Khantham C, Burgos M, Barrionuevo FJ, Jiménez R, Sánchez-Curbelo J, López-Rodrigo O, Peraza MF, Pereira-Caetano I, Marques PI, Carvalho F, Barros A, Bassas L, Seixas S, Gonçalves J, Larriba S, Lopes AM, Palomino-Morales RJ, Carmona FD. Evaluation of Male Fertility-Associated Loci in a European Population of Patients with Severe Spermatogenic Impairment. J Pers Med 2020; 11:22. [PMID: 33383876 PMCID: PMC7823507 DOI: 10.3390/jpm11010022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/05/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022] Open
Abstract
Infertility is a growing concern in developed societies. Two extreme phenotypes of male infertility are non-obstructive azoospermia (NOA) and severe oligospermia (SO), which are characterized by severe spermatogenic failure (SpF). We designed a genetic association study comprising 725 Iberian infertile men as a consequence of SpF and 1058 unaffected controls to evaluate whether five single-nucleotide polymorphisms (SNPs), previously associated with reduced fertility in Hutterites, are also involved in the genetic susceptibility to idiopathic SpF and specific clinical entities. A significant difference in the allele frequencies of USP8-rs7174015 was observed under the recessive model between the NOA group and both the control group (p = 0.0226, OR = 1.33) and the SO group (p = 0.0048, OR = 1.78). Other genetic associations for EPSTI1-rs12870438 and PSAT1-rs7867029 with SO and between TUSC1-rs10966811 and testicular sperm extraction (TESE) success in the context of NOA were observed. In silico analysis of functional annotations demonstrated cis-eQTL effects of such SNPs likely due to the modification of binding motif sites for relevant transcription factors of the spermatogenic process. The findings reported here shed light on the molecular mechanisms leading to severe phenotypes of idiopathic male infertility, and may help to better understand the contribution of the common genetic variation to the development of these conditions.
Collapse
Affiliation(s)
- Miriam Cerván-Martín
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
| | - Lara Bossini-Castillo
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
| | - Rocío Rivera-Egea
- Andrology Laboratory and Sperm Bank, IVIRMA Valencia, 46015 Valencia, Spain;
- IVI Foundation, Health Research Institute La Fe, 46026 Valencia, Spain;
| | - Nicolás Garrido
- IVI Foundation, Health Research Institute La Fe, 46026 Valencia, Spain;
- Servicio de Urología, Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (S.L.); (G.R.)
| | - Saturnino Luján
- Servicio de Urología, Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (S.L.); (G.R.)
| | - Gema Romeu
- Servicio de Urología, Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (S.L.); (G.R.)
| | - Samuel Santos-Ribeiro
- IVI-RMA Lisbon, 1800-282 Lisbon, Portugal;
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | | | | | - José A. Castilla
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, 18014 Granada, Spain
- CEIFER Biobanco—NextClinics, 18004 Granada, Spain
| | - M. Carmen Gonzalvo
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, 18014 Granada, Spain
| | - Ana Clavero
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, 18014 Granada, Spain
| | - F. Javier Vicente
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
- UGC de Urología, HU Virgen de las Nieves, 18014 Granada, Spain
| | - Andrea Guzmán-Jiménez
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
| | - Cláudia Costa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), 4200-135 Porto, Portugal; (C.C.); (P.I.M.); (F.C.); (A.B.); (S.S.); (A.M.L.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - Inés Llinares-Burguet
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
| | - Chiranan Khantham
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
| | - Francisco J. Barrionuevo
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
| | - Rafael Jiménez
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
| | - Josvany Sánchez-Curbelo
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, 08025 Barcelona, Spain; (J.S.-C.); (O.L.-R.); (M.F.P.); (L.B.)
| | - Olga López-Rodrigo
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, 08025 Barcelona, Spain; (J.S.-C.); (O.L.-R.); (M.F.P.); (L.B.)
| | - M. Fernanda Peraza
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, 08025 Barcelona, Spain; (J.S.-C.); (O.L.-R.); (M.F.P.); (L.B.)
| | - Iris Pereira-Caetano
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal; (I.P.-C.); (J.G.)
| | - Patricia I. Marques
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), 4200-135 Porto, Portugal; (C.C.); (P.I.M.); (F.C.); (A.B.); (S.S.); (A.M.L.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - Filipa Carvalho
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), 4200-135 Porto, Portugal; (C.C.); (P.I.M.); (F.C.); (A.B.); (S.S.); (A.M.L.)
- Serviço de Genética, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Alberto Barros
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), 4200-135 Porto, Portugal; (C.C.); (P.I.M.); (F.C.); (A.B.); (S.S.); (A.M.L.)
- Serviço de Genética, Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Lluís Bassas
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, 08025 Barcelona, Spain; (J.S.-C.); (O.L.-R.); (M.F.P.); (L.B.)
| | - Susana Seixas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), 4200-135 Porto, Portugal; (C.C.); (P.I.M.); (F.C.); (A.B.); (S.S.); (A.M.L.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - João Gonçalves
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, 1649-016 Lisbon, Portugal; (I.P.-C.); (J.G.)
- ToxOmics—Centro de Toxicogenómica e Saúde Humana, Nova Medical School, 1169-056 Lisbon, Portugal
| | - Sara Larriba
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain;
| | - Alexandra M. Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S), 4200-135 Porto, Portugal; (C.C.); (P.I.M.); (F.C.); (A.B.); (S.S.); (A.M.L.)
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), 4200-135 Porto, Portugal
| | - Rogelio J. Palomino-Morales
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
- Departamento de Bioquímica y Biología Molecular I, Universidad de Granada, 18071 Granada, Spain
| | - F. David Carmona
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, 18016 Granada, Spain; (M.C.-M.); (L.B.-C.); (A.G.-J.); (I.L.-B.); (M.B.); (F.J.B.); (R.J.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain; (J.A.C.); (M.C.G.); (A.C.); (F.J.V.)
| |
Collapse
|
19
|
Pleuger C, Lehti MS, Dunleavy JE, Fietz D, O'Bryan MK. Haploid male germ cells-the Grand Central Station of protein transport. Hum Reprod Update 2020; 26:474-500. [PMID: 32318721 DOI: 10.1093/humupd/dmaa004] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/15/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The precise movement of proteins and vesicles is an essential ability for all eukaryotic cells. Nowhere is this more evident than during the remarkable transformation that occurs in spermiogenesis-the transformation of haploid round spermatids into sperm. These transformations are critically dependent upon both the microtubule and the actin cytoskeleton, and defects in these processes are thought to underpin a significant percentage of human male infertility. OBJECTIVE AND RATIONALE This review is aimed at summarising and synthesising the current state of knowledge around protein/vesicle transport during haploid male germ cell development and identifying knowledge gaps and challenges for future research. To achieve this, we summarise the key discoveries related to protein transport using the mouse as a model system. Where relevant, we anchored these insights to knowledge in the field of human spermiogenesis and the causality of human male infertility. SEARCH METHODS Relevant studies published in English were identified using PubMed using a range of search terms related to the core focus of the review-protein/vesicle transport, intra-flagellar transport, intra-manchette transport, Golgi, acrosome, manchette, axoneme, outer dense fibres and fibrous sheath. Searches were not restricted to a particular time frame or species although the emphasis within the review is on mammalian spermiogenesis. OUTCOMES Spermiogenesis is the final phase of sperm development. It results in the transformation of a round cell into a highly polarised sperm with the capacity for fertility. It is critically dependent on the cytoskeleton and its ability to transport protein complexes and vesicles over long distances and often between distinct cytoplasmic compartments. The development of the acrosome covering the sperm head, the sperm tail within the ciliary lobe, the manchette and its role in sperm head shaping and protein transport into the tail, and the assembly of mitochondria into the mid-piece of sperm, may all be viewed as a series of overlapping and interconnected train tracks. Defects in this redistribution network lead to male infertility characterised by abnormal sperm morphology (teratozoospermia) and/or abnormal sperm motility (asthenozoospermia) and are likely to be causal of, or contribute to, a significant percentage of human male infertility. WIDER IMPLICATIONS A greater understanding of the mechanisms of protein transport in spermiogenesis offers the potential to precisely diagnose cases of male infertility and to forecast implications for children conceived using gametes containing these mutations. The manipulation of these processes will offer opportunities for male-based contraceptive development. Further, as increasingly evidenced in the literature, we believe that the continuous and spatiotemporally restrained nature of spermiogenesis provides an outstanding model system to identify, and de-code, cytoskeletal elements and transport mechanisms of relevance to multiple tissues.
Collapse
Affiliation(s)
- Christiane Pleuger
- School of Biological Sciences, Monash University, Clayton 3800, Australia.,Institute for Veterinary Anatomy, Histology and Embryology, Justus-Liebig University Giessen, Giessen 35392, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Mari S Lehti
- School of Biological Sciences, Monash University, Clayton 3800, Australia.,Institute of Biomedicine, University of Turku, Turku 20520, Finland
| | | | - Daniela Fietz
- Institute for Veterinary Anatomy, Histology and Embryology, Justus-Liebig University Giessen, Giessen 35392, Germany.,Hessian Centre of Reproductive Medicine, Justus Liebig University Giessen, Giessen 35392, Germany
| | - Moira K O'Bryan
- School of Biological Sciences, Monash University, Clayton 3800, Australia
| |
Collapse
|
20
|
Jiao SY, Yang YH, Chen SR. Molecular genetics of infertility: loss-of-function mutations in humans and corresponding knockout/mutated mice. Hum Reprod Update 2020; 27:154-189. [PMID: 33118031 DOI: 10.1093/humupd/dmaa034] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Infertility is a major issue in human reproductive health, affecting an estimated 15% of couples worldwide. Infertility can result from disorders of sex development (DSD) or from reproductive endocrine disorders (REDs) with onset in infancy, early childhood or adolescence. Male infertility, accounting for roughly half of all infertility cases, generally manifests as decreased sperm count (azoospermia or oligozoospermia), attenuated sperm motility (asthenozoospermia) or a higher proportion of morphologically abnormal sperm (teratozoospermia). Female infertility can be divided into several classical types, including, but not limited to, oocyte maturation arrest, premature ovarian insufficiency (POI), fertilization failure and early embryonic arrest. An estimated one half of infertility cases have a genetic component; however, most genetic causes of human infertility are currently uncharacterized. The advent of high-throughput sequencing technologies has greatly facilitated the identification of infertility-associated gene mutations in patients over the past 20 years. OBJECTIVE AND RATIONALE This review aims to conduct a narrative review of the genetic causes of human infertility. Loss-of-function mutation discoveries related to human infertility are summarized and further illustrated in tables. Corresponding knockout/mutated animal models of causative genes for infertility are also introduced. SEARCH METHODS A search of the PubMed database was performed to identify relevant studies published in English. The term 'mutation' was combined with a range of search terms related to the core focus of the review: infertility, DSD, REDs, azoospermia or oligozoospermia, asthenozoospermia, multiple morphological abnormalities of the sperm flagella (MMAF), primary ciliary dyskinesia (PCD), acephalic spermatozoa syndrome (ASS), globozoospermia, teratozoospermia, acrosome, oocyte maturation arrest, POI, zona pellucida, fertilization defects and early embryonic arrest. OUTCOMES Our search generated ∼2000 records. Overall, 350 articles were included in the final review. For genetic investigation of human infertility, the traditional candidate gene approach is proceeding slowly, whereas high-throughput sequencing technologies in larger cohorts of individuals is identifying an increasing number of causative genes linked to human infertility. This review provides a wide panel of gene mutations in several typical forms of human infertility, including DSD, REDs, male infertility (oligozoospermia, MMAF, PCD, ASS and globozoospermia) and female infertility (oocyte maturation arrest, POI, fertilization failure and early embryonic arrest). The causative genes, their identified mutations, mutation rate, studied population and their corresponding knockout/mutated mice of non-obstructive azoospermia, MMAF, ASS, globozoospermia, oocyte maturation arrest, POI, fertilization failure and early embryonic arrest are further illustrated by tables. In this review, we suggest that (i) our current knowledge of infertility is largely obtained from knockout mouse models; (ii) larger cohorts of clinical cases with distinct clinical characteristics need to be recruited in future studies; (iii) the whole picture of genetic causes of human infertility relies on both the identification of more mutations for distinct types of infertility and the integration of known mutation information; (iv) knockout/mutated animal models are needed to show whether the phenotypes of genetically altered animals are consistent with findings in human infertile patients carrying a deleterious mutation of the homologous gene; and (v) the molecular mechanisms underlying human infertility caused by pathogenic mutations are largely unclear in most current studies. WILDER IMPLICATIONS It is important to use our current understanding to identify avenues and priorities for future research in the field of genetic causes of infertility as well as to apply mutation knowledge to risk prediction, genetic diagnosis and potential treatment for human infertility.
Collapse
Affiliation(s)
- Shi-Ya Jiao
- Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| | - Yi-Hong Yang
- Reproduction Medical Center of West China Second University Hospital, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, 610041 Chengdu, China
| | - Su-Ren Chen
- Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| |
Collapse
|
21
|
Patel PH, Wilkinson EC, Starke EL, McGimsey MR, Blankenship JT, Barbee SA. Vps54 regulates Drosophila neuromuscular junction development and interacts genetically with Rab7 to control composition of the postsynaptic density. Biol Open 2020; 9:bio053421. [PMID: 32747448 PMCID: PMC7473652 DOI: 10.1242/bio.053421] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/22/2020] [Indexed: 01/04/2023] Open
Abstract
Vps54 is a subunit of the Golgi-associated retrograde protein (GARP) complex, which is involved in tethering endosome-derived vesicles to the trans-Golgi network (TGN). In the wobbler mouse, a model for human motor neuron (MN) disease, reduction in the levels of Vps54 causes neurodegeneration. However, it is unclear how disruption of the GARP complex leads to MN dysfunction. To better understand the role of Vps54 in MNs, we have disrupted expression of the Vps54 ortholog in Drosophila and examined the impact on the larval neuromuscular junction (NMJ). Surprisingly, we show that both null mutants and MN-specific knockdown of Vps54 leads to NMJ overgrowth. Reduction of Vps54 partially disrupts localization of the t-SNARE, Syntaxin-16, to the TGN but has no visible impact on endosomal pools. MN-specific knockdown of Vps54 in MNs combined with overexpression of the small GTPases Rab5, Rab7, or Rab11 suppresses the Vps54 NMJ phenotype. Conversely, knockdown of Vps54 combined with overexpression of dominant negative Rab7 causes NMJ and behavioral abnormalities including a decrease in postsynaptic Dlg and GluRIIB levels without any effect on GluRIIA. Taken together, these data suggest that Vps54 controls larval MN axon development and postsynaptic density composition through a mechanism that requires Rab7.
Collapse
Affiliation(s)
- Prajal H Patel
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Emily C Wilkinson
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Emily L Starke
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Malea R McGimsey
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - J Todd Blankenship
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
- Molecular and Cellular Biophysics Program, University of Denver, Denver, CO 80210, USA
| | - Scott A Barbee
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
- Molecular and Cellular Biophysics Program, University of Denver, Denver, CO 80210, USA
| |
Collapse
|
22
|
The Role of Deubiquitinating Enzymes in the Various Forms of Autophagy. Int J Mol Sci 2020; 21:ijms21124196. [PMID: 32545524 PMCID: PMC7352190 DOI: 10.3390/ijms21124196] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
Deubiquitinating enzymes (DUBs) have an essential role in several cell biological processes via removing the various ubiquitin patterns as posttranslational modification forms from the target proteins. These enzymes also contribute to the normal cytoplasmic ubiquitin pool during the recycling of this molecule. Autophagy, a summary name of the lysosome dependent self-degradative processes, is necessary for maintaining normal cellular homeostatic equilibrium. Numerous forms of autophagy are known depending on how the cellular self-material is delivered into the lysosomal lumen. In this review we focus on the colorful role of DUBs in autophagic processes and discuss the mechanistic contribution of these molecules to normal cellular homeostasis via the possible regulation forms of autophagic mechanisms.
Collapse
|
23
|
Avidor-Reiss T, Zhang Z, Li XZ. Editorial: Sperm Differentiation and Spermatozoa Function: Mechanisms, Diagnostics, and Treatment. Front Cell Dev Biol 2020; 8:219. [PMID: 32318570 PMCID: PMC7154170 DOI: 10.3389/fcell.2020.00219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Tomer Avidor-Reiss
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States.,Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, MI, United States.,Department of Obstetrics & Gynecology, Wayne State University, Detroit, MI, United States
| | - Xin Zhiguo Li
- Center for RNA Biology: From Genome to Therapeutics, Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, United States.,Department of Urology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
24
|
Ubiquitin-specific protease 8 (USP8/UBPy): a prototypic multidomain deubiquitinating enzyme with pleiotropic functions. Biochem Soc Trans 2020; 47:1867-1879. [PMID: 31845722 PMCID: PMC6925526 DOI: 10.1042/bst20190527] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 01/07/2023]
Abstract
Protein modification by ubiquitin is one of the most versatile posttranslational regulations and counteracted by almost 100 deubiquitinating enzymes (DUBs). USP8 was originally identified as a growth regulated ubiquitin-specific protease and is like many other DUBs characterized by its multidomain architecture. Besides the catalytic domain, specific protein-protein interaction modules were characterized which contribute to USP8 substrate recruitment, regulation and targeting to distinct protein complexes. Studies in mice and humans impressively showed the physiological relevance and non-redundant function of USP8 within the context of the whole organism. USP8 knockout (KO) mice exhibit early embryonic lethality while induced deletion in adult animals rapidly causes lethal liver failure. Furthermore, T-cell specific ablation disturbs T-cell development and function resulting in fatal autoimmune inflammatory bowel disease. In human patients, somatic mutations in USP8 were identified as the underlying cause of adrenocorticotropic hormone (ACTH) releasing pituitary adenomas causing Cushing's disease (CD). Here we provide an overview of the versatile molecular, cellular and pathology associated function and regulation of USP8 which appears to depend on specific protein binding partners, substrates and the cellular context.
Collapse
|
25
|
Da Costa R, Bordessoules M, Guilleman M, Carmignac V, Lhussiez V, Courot H, Bataille A, Chlémaire A, Bruno C, Fauque P, Thauvin C, Faivre L, Duplomb L. Vps13b is required for acrosome biogenesis through functions in Golgi dynamic and membrane trafficking. Cell Mol Life Sci 2020; 77:511-529. [PMID: 31218450 PMCID: PMC11104845 DOI: 10.1007/s00018-019-03192-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 01/03/2023]
Abstract
The sperm acrosome is a lysosome-related organelle that develops using membrane trafficking from the Golgi apparatus as well as the endolysosomal compartment. How vesicular trafficking is regulated in spermatids to form the acrosome remains to be elucidated. VPS13B, a RAB6-interactor, was recently shown involved in endomembrane trafficking. Here, we report the generation of the first Vps13b-knockout mouse model and show that male mutant mice are infertile due to oligoasthenoteratozoospermia. This phenotype was explained by a failure of Vps13b deficient spermatids to form an acrosome. In wild-type spermatids, immunostaining of Vps13b and Rab6 revealed that they transiently locate to the acrosomal inner membrane. Spermatids lacking Vps13b did not present with the Golgi structure that characterizes wild-type spermatids and showed abnormal targeting of PNA- and Rab6-positive Golgi-derived vesicles to Eea1- and Lamp2-positive structures. Altogether, our results uncover a function of Vps13b in the regulation of the vesicular transport between Golgi apparatus, acrosome, and endolysosome.
Collapse
Affiliation(s)
- Romain Da Costa
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France.
- FHU TRANSLAD, CHU Dijon, 21000, Dijon, France.
| | - Morgane Bordessoules
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
- FHU TRANSLAD, CHU Dijon, 21000, Dijon, France
| | - Magali Guilleman
- Laboratoire de Biologie de la Reproduction, Hôpital François Mitterrand, Université de Bourgogne, 21000, Dijon, France
| | - Virginie Carmignac
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
- Centre de Référence Maladies Génétique à Expression Cutanée MAGEC-Mosaique, CHU Dijon, Dijon, France
| | - Vincent Lhussiez
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
| | - Hortense Courot
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
| | - Amandine Bataille
- Plateforme d'Imagerie Cellulaire CellImaP/DimaCell, Inserm LNC UMR1231, 21000, Dijon, France
| | - Amandine Chlémaire
- Plateforme d'Imagerie Cellulaire CellImaP/DimaCell, Inserm LNC UMR1231, 21000, Dijon, France
| | - Céline Bruno
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
- Laboratoire de Biologie de la Reproduction, Hôpital François Mitterrand, Université de Bourgogne, 21000, Dijon, France
| | - Patricia Fauque
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
- Laboratoire de Biologie de la Reproduction, Hôpital François Mitterrand, Université de Bourgogne, 21000, Dijon, France
| | - Christel Thauvin
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
- FHU TRANSLAD, CHU Dijon, 21000, Dijon, France
- Centre de Référence Déficiences Intellectuelles de Causes Rares, CHU Dijon, 21000, Dijon, France
| | - Laurence Faivre
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
- FHU TRANSLAD, CHU Dijon, 21000, Dijon, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Dijon, 21000, Dijon, France
| | - Laurence Duplomb
- Inserm, UMR1231, Equipe GAD, Bâtiment B3, Université de Bourgogne Franche Comté, 15 boulevard du Maréchal de Lattre de Tassigny, 21000, Dijon Cedex, France
- FHU TRANSLAD, CHU Dijon, 21000, Dijon, France
| |
Collapse
|
26
|
Ma J, Williams J, Eastwood D, Lin S, Qian X, Fang Q, Cope D, Yuan Z, Cao L, An J. High-dose Propofol Anesthesia Reduces the Occurrence of Postoperative Cognitive Dysfunction via Maintaining Cytoskeleton. Neuroscience 2019; 421:136-143. [PMID: 31682819 DOI: 10.1016/j.neuroscience.2019.09.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/15/2022]
Abstract
Postoperative cognitive dysfunction (POCD) is a common postoperative complication observed in patients following. Here we tested the molecular mechanisms of memory loss in hippocampus of rat POCD model. We found that high-dose propofol anesthesia significantly alleviated spatial memory loss. The proteomes and transcriptomes in hippocampus showed that hippocampal cytoskeleton related pathways were abnormal in low group while not in high group. The protein assays confirmed that hippocampal actin cytoskeleton was depolymerized in low group while maintained in high group. This study confirms that high-dose propofol anesthesia could mitigate the development of POCD and provides evidences for actin cytoskeleton associated with this syndrome.
Collapse
Affiliation(s)
- Jun Ma
- Department of Anesthesiology, Pain Medicine & CCM, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China; Laboratory of Medical Cell Biology, College of Translational Medicine, China Medical University, Shenyang, China
| | - John Williams
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Derek Eastwood
- Murrayfield Hospital Wirral, Royal College of Anaesthetists, London, UK
| | - Siyu Lin
- Department of Anesthesiology, Pain Medicine & CCM, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Qian
- Department of Anesthesiology, Pain Medicine & CCM, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Qiwu Fang
- Department of Anesthesiology, Pain Medicine & CCM, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Doris Cope
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zengqiang Yuan
- Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Liu Cao
- Laboratory of Medical Cell Biology, College of Translational Medicine, China Medical University, Shenyang, China
| | - Jianxiong An
- Department of Anesthesiology, Pain Medicine & CCM, Aviation General Hospital of China Medical University & Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
27
|
Khawar MB, Gao H, Li W. Mechanism of Acrosome Biogenesis in Mammals. Front Cell Dev Biol 2019; 7:195. [PMID: 31620437 PMCID: PMC6759486 DOI: 10.3389/fcell.2019.00195] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/29/2019] [Indexed: 11/13/2022] Open
Abstract
During sexual reproduction, two haploid gametes fuse to form the zygote, and the acrosome is essential to this fusion process (fertilization) in animals. The acrosome is a special kind of organelle with a cap-like structure that covers the anterior portion of the head of the spermatozoon. The acrosome is derived from the Golgi apparatus and contains digestive enzymes. With the progress of our understanding of acrosome biogenesis, a number of models have been proposed to address the origin of the acrosome. The acrosome has been regarded as a lysosome-related organelle, and it has been proposed to have originated from the lysosome or the autolysosome. Our review will provide a brief historical overview and highlight recent findings on acrosome biogenesis in mammals.
Collapse
Affiliation(s)
- Muhammad Babar Khawar
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
28
|
Modarres P, Tavalaee M, Ghaedi K, Nasr-Esfahani MH. An Overview of The Globozoospermia as A Multigenic Identified Syndrome. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2018; 12:273-277. [PMID: 30291685 PMCID: PMC6186287 DOI: 10.22074/ijfs.2019.5561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 05/16/2018] [Indexed: 11/15/2022]
Abstract
Acrosome plays an integral role during fertilization and its absence in individuals with globozoospermia leads to
failure of in vitro fertilization (IVF) and oocyte activation post-intracytoplasmic sperm injection (ICSI). A variety
of processes, organelles and structures are involved in acrosome biogenesis including, trans-golgi network (TGN),
acroplaxome and cellular trafficking. This review aims to explain roles of related signals and molecules involved in
this process and also describe how their absence in form of mutation, deletion and knockout model may lead to phe-
nomenon referred to globozoospermia.
Collapse
Affiliation(s)
- Parastoo Modarres
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Marziyeh Tavalaee
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.,Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.Electronic Address:
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.Electronic Address:.,Isfahan Fertility and Infertility Center, Isfahan, Iran
| |
Collapse
|
29
|
Qu C, Kunkalla K, Vaghefi A, Frederiksen JK, Liu Y, Chapman JR, Blonska M, Bernal-Mizrachi L, Alderuccio JP, Lossos IS, Landgraf R, Vega F. Smoothened stabilizes and protects TRAF6 from degradation: A novel non-canonical role of smoothened with implications in lymphoma biology. Cancer Lett 2018; 436:149-158. [PMID: 30165192 DOI: 10.1016/j.canlet.2018.08.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022]
Abstract
Tumor necrosis factor receptor-associated factor 6 (TRAF6), an (K63) E3-ligase, plays a role in many biological processes and its activity is relevant in diffuse large B cell lymphoma (DLBCL) biology. Although molecules that trigger TRAF6 activation have been defined, those that stabilize TRAF6 and/or enhance TRAF6 function remain largely unclear. We found that TRAF6 amplifies pAKT signaling in DLBCL. Moreover, TRAF6 activation and stabilization of its ubiquitination profile are facilitated by smoothened (SMO), signal transducer of canonical Hedgehog signaling. Here, we report that SMO is needed to facilitate and maintain TRAF6-dependent elevated pAKT levels, and that the SMO/TRAF6 axis contributes to doxorubicin resistance in DLBCL. Mechanistically, we found that SMO, through its C-terminal tail, stabilizes and protects TRAF6 from degradation, an effect mediated by ubiquitin-specific protease-8. Moreover, this functional link between SMO and TRAF6 is reflected in DLBCL patients where high expression of both molecules correlates with poor prognosis. In summary, our study reveals a novel cell survival mechanism in which SMO stabilizes and protects TRAF6 from degradation. The axis SMO/TRAF6/AKT is highly relevant in the biology of DLBCL and is involved in doxorubicin resistance.
Collapse
Affiliation(s)
- Changju Qu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology; Institute of Blood and Marrow Transplantation; Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China
| | - Kranthi Kunkalla
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, University of Miami/Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Amineh Vaghefi
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, University of Miami/Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - John K Frederiksen
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, University of Miami/Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Yadong Liu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Jiangsu Institute of Hematology; Institute of Blood and Marrow Transplantation; Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215000, China
| | - Jennifer R Chapman
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, University of Miami/Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Marzenna Blonska
- Division of Hematology-Oncology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Leon Bernal-Mizrachi
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Georgia
| | - Juan Pablo Alderuccio
- Division of Hematology-Oncology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Izidore S Lossos
- Division of Hematology-Oncology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Ralf Landgraf
- Department of Biochemistry and Molecular Biology, University of Miami/Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Francisco Vega
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, University of Miami/Sylvester Comprehensive Cancer Center, Miami, FL, USA; Division of Hematology-Oncology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
| |
Collapse
|
30
|
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: 2.9] [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]
|
31
|
Horner DS, Pasini ME, Beltrame M, Mastrodonato V, Morelli E, Vaccari T. ESCRT genes and regulation of developmental signaling. Semin Cell Dev Biol 2017; 74:29-39. [PMID: 28847745 DOI: 10.1016/j.semcdb.2017.08.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/06/2017] [Accepted: 08/18/2017] [Indexed: 11/30/2022]
Abstract
ESCRT (Endosomal Sorting Complex Required for Transport) proteins have been shown to control an increasing number of membrane-associated processes. Some of these, and prominently regulation of receptor trafficking, profoundly shape signal transduction. Evidence in fungi, plants and multiple animal models support the emerging concept that ESCRTs are main actors in coordination of signaling with the changes in cells and tissues occurring during development and homeostasis. Consistent with their pleiotropic function, ESCRTs are regulated in multiple ways to tailor signaling to developmental and homeostatic needs. ESCRT activity is crucial to correct execution of developmental programs, especially at key transitions, allowing eukaryotes to thrive and preventing appearance of congenital defects.
Collapse
Affiliation(s)
- David S Horner
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Maria E Pasini
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Monica Beltrame
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy
| | - Valeria Mastrodonato
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Elena Morelli
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milano, Italy
| | - Thomas Vaccari
- Dipartimento di Bioscienze, Universita' degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy; IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milano, Italy.
| |
Collapse
|
32
|
Gioria M, Pasini ME, Berruti G. Dynamic of contribution of UBPy-sorted cargo to acrosome biogenesis: effects of its derailment in a mouse model of globozoospermia, the infertile Vps54 (L967Q) mutant. Cell Tissue Res 2017; 369:413-427. [DOI: 10.1007/s00441-017-2592-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/21/2017] [Indexed: 12/14/2022]
|
33
|
Berruti G. Towards defining an ‘origin’—The case for the mammalian acrosome. Semin Cell Dev Biol 2016; 59:46-53. [DOI: 10.1016/j.semcdb.2016.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 01/19/2023]
|
34
|
Belmonte SA, Mayorga LS, Tomes CN. The Molecules of Sperm Exocytosis. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2016; 220:71-92. [PMID: 27194350 DOI: 10.1007/978-3-319-30567-7_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Exocytosis is a fundamental process used by eukaryotic cells to release biological compounds and to insert lipids and proteins in the plasma membrane. Specialized secretory cells undergo regulated exocytosis in response to physiological signals. Sperm exocytosis or acrosome reaction (AR) is essentially a regulated secretion with special characteristics. We will focus here on some of these unique features, covering the topology, kinetics, and molecular mechanisms that prepare, drive, and regulate membrane fusion during the AR. Last, we will compare acrosomal release with exocytosis in other model systems.
Collapse
Affiliation(s)
- Silvia A Belmonte
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, IHEM-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Mendoza, Argentina
| | - Luis S Mayorga
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, IHEM-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Mendoza, Argentina
| | - Claudia N Tomes
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, IHEM-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Mendoza, Argentina.
| |
Collapse
|
35
|
Fári K, Takács S, Ungár D, Sinka R. The role of acroblast formation during Drosophila spermatogenesis. Biol Open 2016; 5:1102-10. [PMID: 27481842 PMCID: PMC5004609 DOI: 10.1242/bio.018275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Protein recycling is important for maintaining homeostasis of the Golgi and its cisternae. The Vps54 (Scat) protein, a subunit of the GARP tethering complex, is a central factor in retrograde transport to the trans-Golgi. We found the scat1 mutant to be male sterile in Drosophila with individualization problems occurring during spermatogenesis. Another typically observed phenotype was the abnormal nuclear structure in elongated mutant cysts. When examining the structure and function of the Golgi, a failure in acrosome formation and endosome-Golgi vesicular transport were found in the scat1 mutant. This acrosome formation defect was due to a fault in the trans-Golgi side of the acroblast ribbon. When testing a mutation in a second retrograde transport protein, Fws, a subunit of the conserved oligomeric Golgi (COG) tethering complex, the acroblast structure, was again disrupted. fwsP caused a similar, albeit milder, acrosome and sperm individualization phenotype as the scat1 mutant. In the case of fwsP the cis side of the acroblast ribbon was dispersed, in-line with the intra-Golgi retrograde function of COG. Our results highlight the importance of an intact acroblast for acrosome formation, nuclear elongation and therefore sperm maturation. Moreover, these results suggest the importance of retrograde tethering complexes in the formation of a functional Golgi ribbon. Summary: This study demonstrates that retrograde tethering complexes are necessary to form a functional acroblast, which is essential for normal nuclear elongation and acrosome formation during Drosophila spermatogenesis.
Collapse
Affiliation(s)
- Karolina Fári
- Department of Genetics, University of Szeged, Szeged 6726, Hungary
| | - Sándor Takács
- Department of Genetics, University of Szeged, Szeged 6726, Hungary
| | - Dániel Ungár
- Department of Biology, University of York, York YO10 5DD, UK
| | - Rita Sinka
- Department of Genetics, University of Szeged, Szeged 6726, Hungary
| |
Collapse
|
36
|
Chandrasekaran AP, Suresh B, Kim HH, Kim KS, Ramakrishna S. Concise Review: Fate Determination of Stem Cells by Deubiquitinating Enzymes. Stem Cells 2016; 35:9-16. [PMID: 27341175 DOI: 10.1002/stem.2446] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/05/2016] [Indexed: 12/22/2022]
Abstract
Post-translational modification by ubiquitin molecules is a key regulatory process for stem cell fate determination. Ubiquitination and deubiquitination are the major cellular processes used to balance the protein turnover of several transcription factors that regulate stem cell differentiation. Deubiquitinating enzymes (DUBs), which facilitate the processing of ubiquitin, significantly influence stem cell fate choices. Specifically, DUBs play a critical regulatory role during development by directing the production of new specialized cells. This review focuses on the regulatory role of DUBs in various cellular processes, including stem cell pluripotency and differentiation, adult stem cell signaling, cellular reprogramming, spermatogenesis, and oogenesis. Specifically, the identification of interactions of DUBs with core transcription factors has provided new insight into the role of DUBs in regulating stem cell fate determination. Thus, DUBs have emerged as key pharmacologic targets in the search to develop highly specific agents to treat various illnesses. Stem Cells 2017;35:9-16.
Collapse
Affiliation(s)
| | - Bharathi Suresh
- Department of Pharmacology and Brain Korea 21 plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology and Brain Korea 21 plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea.,College of Medicine, Hanyang University, Seoul, South Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea.,College of Medicine, Hanyang University, Seoul, South Korea
| |
Collapse
|
37
|
Brown C, Szpryngiel S, Kuang G, Srivastava V, Ye W, McKee LS, Tu Y, Mäler L, Bulone V. Structural and functional characterization of the microtubule interacting and trafficking domains of two oomycete chitin synthases. FEBS J 2016; 283:3072-88. [PMID: 27363606 DOI: 10.1111/febs.13794] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 05/31/2016] [Accepted: 06/28/2016] [Indexed: 12/18/2022]
Abstract
UNLABELLED Chitin synthases (Chs) are responsible for the synthesis of chitin, a key structural cell wall polysaccharide in many organisms. They are essential for growth in certain oomycete species, some of which are pathogenic to diverse higher organisms. Recently, a microtubule interacting and trafficking (MIT) domain, which is not found in any fungal Chs, has been identified in some oomycete Chs proteins. Based on experimental data relating to the binding specificity of other eukaryotic MIT domains, there was speculation that this domain may be involved in the intracellular trafficking of Chs proteins. However, there is currently no evidence for this or any other function for the MIT domain in these enzymes. To attempt to elucidate their function, MIT domains from two Chs enzymes from the oomycete Saprolegnia monoica were cloned, expressed, and characterized. Both were shown to interact strongly with the plasma membrane component, phosphatidic acid, and to have additional putative interactions with proteins thought to be involved in protein transport and localization. Aiding our understanding of these data, the structure of the first MIT domain from a carbohydrate-active enzyme (MIT1) was solved by NMR, and a model structure of a second MIT domain (MIT2) was built by homology modeling. Our results suggest a potential function for these MIT domains in the intracellular transport and/or regulation of Chs enzymes in the oomycetes. DATABASE Structural data are available in the Biological Magnetic Resonance Bank (BMRB) database under the accession number 19987 and the PDB database under the accession number 2MPK.
Collapse
Affiliation(s)
- Christian Brown
- Division of Glycoscience, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
| | - Scarlett Szpryngiel
- Department of Biochemistry and Biophysics, The Arrhenius Laboratory, Stockholm University, Sweden
| | - Guanglin Kuang
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Vaibhav Srivastava
- Division of Glycoscience, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
| | - Weihua Ye
- Department of Biochemistry and Biophysics, The Arrhenius Laboratory, Stockholm University, Sweden
| | - Lauren S McKee
- Division of Glycoscience, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden
| | - Yaoquan Tu
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Lena Mäler
- Department of Biochemistry and Biophysics, The Arrhenius Laboratory, Stockholm University, Sweden
| | - Vincent Bulone
- Division of Glycoscience, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Centre, Stockholm, Sweden.,ARC Centre of Excellence in Plant Cell Walls, School of Agriculture, Food and Wine, The University of Adelaide, Waite Campus, Urrbrae, South Australia, Australia
| |
Collapse
|
38
|
Ma TJ, Zhang XJ, Ding XP, Chen HH, Zhang YW, Ding M. Association of single nucleotide polymorphisms in UBR2 gene with idiopathic aspermia or oligospermia in Sichuan, China. Andrologia 2016; 48:1253-1260. [PMID: 26940145 DOI: 10.1111/and.12569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2016] [Indexed: 02/05/2023] Open
Abstract
The associations between three single nucleotide polymorphisms (SNPs; rs3749897, rs16895863 and rs373341) of UBR2 gene and idiopathic aspermia or oligospermia were investigated in this study by a case-control experiment with 149 fertile and 316 infertile men, including 244 patients with idiopathic aspermia and 72 patients with severe oligospermia. The time-of-flight mass spectrometry (Sequenom MassARRAY® system) was used in this study. A significant difference between the oligospermia men (oligospermia group) and the fertile men (control group) was observed in this research (odds ratio [OR]: 2.764; 95% CI: 95% confidence interval [CI]: 1.171-6.525; P = 0.017), which could indicate that the combined AT-TC-CC genotype in the UBR2 gene (rs16895863, rs373341, rs3749897 respectively) is a possible risk of idiopathic oligospermia for men in Sichuan, China.
Collapse
Affiliation(s)
- T-J Ma
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, School of Life Science, Institute of Medical Genetics, Sichuan University, Chengdu, China.,Bio-resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chengdu, China.,Biotechnology Academy of Nanchuan, Chongqing, China
| | - X-J Zhang
- Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing, China
| | - X-P Ding
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, School of Life Science, Institute of Medical Genetics, Sichuan University, Chengdu, China.,Bio-resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chengdu, China.,Biotechnology Academy of Nanchuan, Chongqing, China
| | - H-H Chen
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, School of Life Science, Institute of Medical Genetics, Sichuan University, Chengdu, China.,Bio-resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chengdu, China.,Biotechnology Academy of Nanchuan, Chongqing, China
| | - Y-W Zhang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, School of Life Science, Institute of Medical Genetics, Sichuan University, Chengdu, China.,Bio-resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chengdu, China.,Biotechnology Academy of Nanchuan, Chongqing, China
| | - M Ding
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, School of Life Science, Institute of Medical Genetics, Sichuan University, Chengdu, China.,Bio-resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chengdu, China.,Biotechnology Academy of Nanchuan, Chongqing, China
| |
Collapse
|
39
|
Intasqui P, Camargo M, Antoniassi MP, Cedenho AP, Carvalho VM, Cardozo KHM, Zylbersztejn DS, Bertolla RP. Association between the seminal plasma proteome and sperm functional traits. Fertil Steril 2016; 105:617-628. [DOI: 10.1016/j.fertnstert.2015.11.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/07/2015] [Accepted: 11/02/2015] [Indexed: 01/11/2023]
|
40
|
Kuang G, Liang L, Brown C, Wang Q, Bulone V, Tu Y. Insight into the adsorption profiles of the Saprolegnia monoica chitin synthase MIT domain on POPA and POPC membranes by molecular dynamics simulation studies. Phys Chem Chem Phys 2016; 18:5281-90. [DOI: 10.1039/c5cp05391a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The binding mode and binding free energy of the Saprolegnia monoica chitin synthase MIT domain with the POPA membrane have been studied by molecular simulation methods.
Collapse
Affiliation(s)
- Guanglin Kuang
- Division of Theoretical Chemistry and Biology
- School of Biotechnology
- Royal Institute of Technology (KTH)
- AlbaNova University Center
- 106 91 Stockholm
| | - Lijun Liang
- Division of Theoretical Chemistry and Biology
- School of Biotechnology
- Royal Institute of Technology (KTH)
- AlbaNova University Center
- 106 91 Stockholm
| | - Christian Brown
- Division of Glycoscience
- School of Biotechnology
- Royal Institute of Technology (KTH)
- AlbaNova University Center
- 106 91 Stockholm
| | - Qi Wang
- Department of Chemistry and Soft Matter
- Zhejiang University
- Hangzhou 310027
- China
| | - Vincent Bulone
- Division of Glycoscience
- School of Biotechnology
- Royal Institute of Technology (KTH)
- AlbaNova University Center
- 106 91 Stockholm
| | - Yaoquan Tu
- Division of Theoretical Chemistry and Biology
- School of Biotechnology
- Royal Institute of Technology (KTH)
- AlbaNova University Center
- 106 91 Stockholm
| |
Collapse
|
41
|
Suresh B, Lee J, Hong SH, Kim KS, Ramakrishna S. The role of deubiquitinating enzymes in spermatogenesis. Cell Mol Life Sci 2015; 72:4711-20. [PMID: 26350476 PMCID: PMC11113867 DOI: 10.1007/s00018-015-2030-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/10/2015] [Accepted: 08/24/2015] [Indexed: 12/11/2022]
Abstract
Spermatogenesis is a complex process through which spermatogonial stem cells undergo mitosis, meiosis, and cell differentiation to generate mature spermatozoa. During this process, male germ cells experience several translational modifications. One of the major post-translational modifications in eukaryotes is the ubiquitination of proteins, which targets proteins for degradation; this enables control of the expression of enzymes and structural proteins during spermatogenesis. It has become apparent that ubiquitination plays a key role in regulating every stage of spermatogenesis starting from gonocytes to differentiated spermatids. It is understood that, where there is ubiquitination, deubiquitination by deubiquitinating enzymes (DUBs) also exists to counterbalance the ubiquitination process in a reversible manner. Normal spermatogenesis is dependent on the balanced actions of ubiquitination and deubiquitination. This review highlights the current knowledge of the role of DUBs and their essential regulatory contribution to spermatogenesis, especially during progression into meiotic phase, acrosome biogenesis, quality sperm production, and apoptosis of germ cells.
Collapse
Affiliation(s)
- Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seongdong-gu, Seoul, South Korea
| | - Junwon Lee
- Department of Physiology and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seongdong-gu, Seoul, South Korea.
- College of Medicine, Hanyang University, Seoul, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seongdong-gu, Seoul, South Korea.
- College of Medicine, Hanyang University, Seoul, South Korea.
| |
Collapse
|
42
|
Pocognoni CA, Berberián MV, Mayorga LS. ESCRT (Endosomal Sorting Complex Required for Transport) Machinery Is Essential for Acrosomal Exocytosis in Human Sperm1. Biol Reprod 2015; 93:124. [DOI: 10.1095/biolreprod.115.132001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/09/2015] [Indexed: 12/31/2022] Open
|
43
|
Molecular Effects of Polymorphism in the 3'UTR of Unc-5 homolog C Associated with Conception Rate in Holsteins. PLoS One 2015; 10:e0131283. [PMID: 26147436 PMCID: PMC4493121 DOI: 10.1371/journal.pone.0131283] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 06/01/2015] [Indexed: 11/20/2022] Open
Abstract
Conception rates among dairy cows in Japan have declined in recent decades. To enhance our understanding of the genes involved in conception rates, we conducted a genome-wide association study (GWAS) using 822 Holsteins and identified a single-nucleotide polymorphism (SNP) associated with conception rate: A+169G in the 3’ untranslated region (UTR) of unc-5 homolog C (UNC5C). Cows with higher conception rates carried the A polymorphism in the UNC5C 3’UTR. Luciferase assays and quantitative analysis of allele ratios revealed that UNC5C transcripts with the A polymorphism were expressed at higher levels than those carrying the G polymorphism. UNC5C transmits either pro- or anti-apoptotic signals depending on the availability of its ligand, Netrin-1. UNC5C expression is negatively regulated by reproductive homeobox X-linked 5 (Rhox5), and the Rhox5 locus is methylated by G9a methyltransferase. G9a-knockout mice have previously been demonstrated to be subfertile, and we found that UNC5C, G9a, and Netrin-1 expression levels increased from the 4-cell stage to the blastocyst stage in fertilized murine embryos, whereas Rhox5 expression decreased. Repression of UNC5C, G9a, or Netrin-1 or forced expression of Rhox5 in the anterior nucleus stage inhibited development to the blastocyst stage, suggesting that cows carrying the G polymorphism in UNC5C might have lower conception rates because of the poor development of preimplantation embryos. This study provides novel insights into the role of UNC5C during embryonic development.
Collapse
|
44
|
Berruti G, Paiardi C. USP8/UBPy-regulated sorting and the development of sperm acrosome: the recruitment of MET. Reproduction 2015; 149:633-44. [DOI: 10.1530/rep-14-0671] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/05/2015] [Indexed: 01/12/2023]
Abstract
The acrosome is a peculiar vacuole that at fertilization undergoes the acrosome reaction (AR), an event unique in the sperm life. Contents released promote sperm penetration through oocyte's investments; membranous components are involved in sperm–egg interaction/fusion. Therefore, both constituents play a role in fertilization. The biogenesis of this vacuole, however, has not been clarified yet; recently, it has been proposed as a novel lysosome-related organelle (LRO). Our research focuses on the involvement of the endosomal pathway in acrosomogenesis starting from the early phases. The trafficking sorted by USP8/UBPy, an endosomal regulator recently described as a compelling candidate for male fertility gene, was investigated in comparison to that of SP56, a marker of the biosynthetic pathway. Mouse spermatids were double/triple immunolabeled and examined by confocal microscopy. The contribution of the vesicular traffic assisted by the cortical microtubule array was also evaluated in nocodazole-treated spermatids. USP8/UBPy-sorted cargo contributes early to acrosomogenesis and its trafficking is microtubule mediated. It was identified, through co-immunoprecipitation/co-immunolocalization assays, that the membrane receptor MET, described herein for the first time in spermatids, as an USP8/UBPy-target substrate is delivered to the acrosome. MET and USP8/UBPy still colocalize in epididymal spermatozoa. Following the AR, MET and USP8/UBPy show a distinct fate. MET, in particular, translocates at the PAS, the post acrosomal segment known to harbor sperm-borne factors involved in oocyte activation. Overall, our results support the concept of the acrosome as a LRO and provide evidence for the identification of MET as a tyrosine kinase receptor that may play a role in fertilization.
Collapse
|
45
|
Sato Y, Tajima A, Tsunematsu K, Nozawa S, Yoshiike M, Koh E, Kanaya J, Namiki M, Matsumiya K, Tsujimura A, Komatsu K, Itoh N, Eguchi J, Imoto I, Yamauchi A, Iwamoto T. An association study of four candidate loci for human male fertility traits with male infertility. Hum Reprod 2015; 30:1510-4. [PMID: 25908656 DOI: 10.1093/humrep/dev088] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/25/2015] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Are the four candidate loci (rs7867029, rs7174015, rs12870438 and rs724078) for human male fertility traits, identified in a genome-wide association study (GWAS) of a Hutterite population in the USA, associated with male infertility in a Japanese population? SUMMARY ANSWER rs7867029, rs7174015 and rs12870438 are significantly associated with the risk of male infertility in a Japanese population. WHAT IS KNOWN ALREADY Recently, a GWAS of a Hutterite population in the USA revealed that 41 single-nucleotide polymorphisms (SNPs) were significantly correlated with family size or birth rate. Of these, four SNPs (rs7867029, rs7174015, rs12870438 and rs724078) were found to be associated with semen parameters in ethnically diverse men from Chicago. STUDY DESIGN, SIZE, DURATION This is a case-control association study in a total of 917 Japanese subjects, including 791 fertile men, 76 patients with azoospermia and 50 patients with oligozoospermia. PARTICIPANTS/MATERIALS, SETTING, METHODS Azoospermia was diagnosed on the basis of semen analysis (the absence of sperm in ejaculate), serum hormone levels and physical examinations. Oligozoospermia was defined as a sperm concentration of <20 × 10(6)/ml. We excluded patients with any known cause of infertility (i.e. obstructive azoospermia, varicocele, cryptorchidism, hypogonadotropic hypogonadism, karyotype abnormalities or complete deletion of AZF a, b or c). The SNPs rs7867029, rs7174015, rs12870438 and rs724078 were genotyped using DNA from peripheral blood samples and either restriction fragment length polymorphism PCR or TaqMan probes. Genetic associations between the four SNPs and male infertility were assessed using a logistic regression analysis under three different comparative models (additive, recessive or dominant). MAIN RESULTS AND THE ROLE OF CHANCE The genotypes of all four SNPs were in Hardy-Weinberg equilibrium in the fertile controls. The SNPs rs7867029 and rs7174015 are associated with oligozoospermia [rs7867029: odds ratio (OR) = 1.70, 95% confidence interval (CI) = 1.07-2.68, P = 0.024 (log-additive); rs7174015: OR = 6.52, 95% CI = 1.57-27.10, P = 0.0099 (dominant)] and rs12870438 is associated with azoospermia (OR = 10.90, 95% CI = 2.67-44.60, P = 0.00087 (recessive)] and oligozoospermia [OR = 8.54, 95% CI = 1.52-47.90, P = 0.015 (recessive)]. The association between rs7174015 and oligozoospermia under a dominant model and between rs12870438 and azoospermia under additive and recessive models remained after correction for multiple testing. There were no associations between rs724078 and azoospermia or oligozoospermia. LIMITATIONS, REASONS FOR CAUTION Even though the sample size of case subjects was not very large, we found that three SNPs were associated with the risk of male infertility in a Japanese population. WIDER IMPLICATIONS OF THE FINDINGS The three infertility-associated SNPs may be contributing to a quantitative reduction in spermatogenesis. STUDY FUNDING/COMPETING INTERESTS This study was supported in part by the Ministry of Health and Welfare of Japan (1013201) (to T.I.), Grant-in-Aids for Scientific Research (C) (23510242) (to A.Ta.) from the Japan Society for the Promotion of Science, the European Union (BMH4-CT96-0314) (to T. I.) and the Takeda Science Foundation (to A.Ta.). None of the authors has any competing interests to declare.
Collapse
Affiliation(s)
- Youichi Sato
- Department of Pharmaceutical Information Science, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8505, Japan
| | - Atsushi Tajima
- Department of Human Genetics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8640, Japan
| | - Kouki Tsunematsu
- Department of Pharmaceutical Information Science, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8505, Japan
| | - Shiari Nozawa
- Department of Urology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Miki Yoshiike
- Department of Urology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Eitetsue Koh
- Department of Urology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan
| | - Jiro Kanaya
- Department of Urology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan
| | - Mikio Namiki
- Department of Urology, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan
| | - Kiyomi Matsumiya
- Department of Urology, Osaka Police Hospital, Osaka 543-0035, Japan
| | - Akira Tsujimura
- Department of Urology, Graduate School of Medicine, Faculty of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Kiyoshi Komatsu
- Department of Urology, Harasanshinkai Hospital, Fukuoka 812-0033, Japan
| | - Naoki Itoh
- Department of Urology, Sapporo Medical University, Sapporo 060-8543, Japan
| | - Jiro Eguchi
- Department of Urology, Nagasaki University, Nagasaki 852-8523, Japan
| | - Issei Imoto
- Department of Human Genetics, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Aiko Yamauchi
- Department of Pharmaceutical Information Science, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8505, Japan
| | - Teruaki Iwamoto
- Department of Urology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan Center for Infertility and IVF, International University of Health and Welfare Hospital, Nasushiobara 329-2763, Japan
| |
Collapse
|
46
|
Da Ros M, Hirvonen N, Olotu O, Toppari J, Kotaja N. Retromer vesicles interact with RNA granules in haploid male germ cells. Mol Cell Endocrinol 2015; 401:73-83. [PMID: 25486514 DOI: 10.1016/j.mce.2014.11.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/21/2014] [Accepted: 11/28/2014] [Indexed: 10/24/2022]
Abstract
Spermatozoa are produced during spermatogenesis as a result of mitotic proliferation, meiosis and cellular differentiation. Postmeiotic spermatids are exceptional cells given their haploid genome and remarkable sperm-specific structural transformations to compact and reshape the nucleus and to construct the flagellum and acrosome. These processes require delicate coordination and active communication between distinct cellular compartments. In this study, we elucidated the interplay between the haploid RNA regulation and the vesicular transport system. We identified a novel interaction between VPS26A/VPS35-containing retromer vesicles and the chromatoid body (CB), which is a large ribonucleoprotein (RNP) granule unique to haploid male germ cells. VPS26A/VPS35-positive vesicles were shown to be involved in the endosomal pathway, as well as in acrosomal formation that is dependent on the Golgi complex-derived vesicular trafficking. While the exact role of the retromer vesicles in the CB function remains unclear, our results suggest a direct functional link between vesicle transport and CB-mediated RNA regulation.
Collapse
Affiliation(s)
- Matteo Da Ros
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku FIN-20520, Finland
| | - Noora Hirvonen
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku FIN-20520, Finland
| | - Opeyemi Olotu
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku FIN-20520, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku FIN-20520, Finland; Department of Pediatrics, University of Turku, Turku FIN-20520, Finland
| | - Noora Kotaja
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku FIN-20520, Finland.
| |
Collapse
|
47
|
Ceriani M, Amigoni L, D'Aloia A, Berruti G, Martegani E. The deubiquitinating enzyme UBPy/USP8 interacts with TrkA and inhibits neuronal differentiation in PC12 cells. Exp Cell Res 2015; 333:49-59. [PMID: 25662281 DOI: 10.1016/j.yexcr.2015.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/20/2015] [Accepted: 01/28/2015] [Indexed: 11/19/2022]
Abstract
The tropomyosin-related kinase (Trk) family of receptor tyrosine kinases controls synaptic function, plasticity and sustains differentiation, morphology, and neuronal cell survival. Understanding Trk receptors down-regulation and recycling is a crucial step to point out sympathetic and sensory neuron function and survival. PC12 cells derived from pheochromocytoma of the rat adrenal medulla have been widely used as a model system for studies of neuronal differentiation as they respond to nerve growth factor (NGF) with a dramatic change in phenotype and acquire a number of properties characteristic of sympathetic neurons. In this study we demonstrated that in PC12 cells the TrkA receptor interacts with the deubiquitinating enzyme USP8/UBPy in a NGF-dependent manner and that it is deubiquitinated in vivo and in vitro by USP8. USP8 overexpression blocked NGF-induced neurites outgrowth while the overexpression of the catalytically inactive mutant USP8/UBPy(C748A) caused a marked increase of cell differentiation. Localization and biochemical experiments have point out that USP8 and TrkA partially co-localize in endosomes after NGF stimulation. Finally we have studied the role played by USP8 on TrkA turnover; using specific siRNA for USP8 we found that USP8 knockdown increases TrkA half-life, suggesting that the deubiquitinating activity of USP8 promotes TrkA degradation.
Collapse
Affiliation(s)
- Michela Ceriani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy.
| | - Loredana Amigoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy.
| | - Alessia D'Aloia
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy.
| | - Giovanna Berruti
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milan, Italy.
| | - Enzo Martegani
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy.
| |
Collapse
|
48
|
Integrated miRNA and mRNA expression profiling to identify mRNA targets of dysregulated miRNAs in non-obstructive azoospermia. Sci Rep 2015; 5:7922. [PMID: 25628250 PMCID: PMC4310093 DOI: 10.1038/srep07922] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 12/10/2014] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to identify mRNA targets of dysregulated miRNAs through the integrated analysis of miRNA and mRNA expression profiling in men with normal versus impaired spermatogenesis. The expression of mRNAs and miRNAs in testicular tissues obtained from males with non-obstructive azoospermia (NOA, n = 4) or obstructive azoospermia (OA, n = 3) with normal spermatogenesis was analyzed using microarray technology. Some of the most interesting results were validated by real time PCR using samples from the same cohort. Ninety-three miRNAs and 4172 mRNAs were differentially expressed in the NOA and normozoospermic OA patients. In addition to confirming that significantly dysregulated genes and miRNAs play pivotal roles in NOA, promising correlation signatures of these miRNA/mRNA pairs were discovered in this study. The functional classification of the miRNA/mRNA pairs revealed that differentially expressed genes were most frequently associated with spermatogenesis, the cell meiosis, the cell cycle, and the development of secondary male sexual characteristics. This is the first systematic profiling of both mRNA and miRNA in testicular tissues of patients with NOA and OA. Our results indicate that the phenotypic status of NOA is characterized by the dysfunction of normal spermatogenesis when compared with OA or normozoospermic males.
Collapse
|
49
|
de Boer P, de Vries M, Ramos L. A mutation study of sperm head shape and motility in the mouse: lessons for the clinic. Andrology 2014; 3:174-202. [PMID: 25511638 DOI: 10.1111/andr.300] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 09/19/2014] [Accepted: 09/24/2014] [Indexed: 12/11/2022]
Abstract
Mouse mutants that show effects on sperm head shape, the sperm tail (flagellum), and motility were analysed in a systematic way. This was achieved by grouping mutations in the following classes: manchette, acrosome, Sertoli cell contact, chromatin remodelling, and mutations involved in complex regulations such as protein (de)phosphorylation and RNA stability, and flagellum/motility mutations. For all mutant phenotypes, flagellum function (motility) was affected. Head shape, including the nucleus, was also affected in spermatozoa of most mouse models, though with considerable variation. For the mutants that were categorized in the flagellum/motility group, generally normal head shapes were found, even when the flagellum did not develop or only poorly so. Most mutants are sterile, an occasional one semi-sterile. For completeness, the influence of the sex chromosomes on sperm phenotype is included. Functionally, the genes involved can be categorized as regulators of spermiogenesis. When extrapolating these data to human sperm samples, in vivo selection for motility would be the tool for weeding out the products of suboptimal spermiogenesis and epididymal sperm maturation. The striking dependency of motility on proper sperm head development is not easy to understand, but likely is of evolutionary benefit. Also, sperm competition after mating can never act against the long-term multi-generation interest of genetic integrity. Hence, it is plausible to suggest that short-term haplophase fitness i.e., motility, is developmentally integrated with proper nucleus maturation, including genetic integrity to protect multi-generation fitness. We hypothesize that, when the prime defect is in flagellum formation, apparently a feedback loop was not necessary as head morphogenesis in these mutants is mostly normal. Extrapolating to human-assisted reproductive techniques practice, this analysis would supply the arguments for the development of tools to select for motility as a continuous (non-discrete) parameter.
Collapse
Affiliation(s)
- P de Boer
- Department of Obstetrics and Gynaecology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | | |
Collapse
|
50
|
Berruti G, Paiardi C. Acrosome biogenesis: Revisiting old questions to yield new insights. SPERMATOGENESIS 2014; 1:95-98. [PMID: 22319656 DOI: 10.4161/spmg.1.2.16820] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 06/07/2011] [Indexed: 01/23/2023]
Abstract
The acrosome is a unique membranous organelle located over the anterior part of the sperm nucleus that is highly conserved throughout evolution. This acidic vacuole contains a number of hydrolytic enzymes that, when secreted, help the sperm penetrate the egg's coats. Although acrosome biogenesis is an important aspect of spermiogenesis, the molecular mechanism(s) that regulates this event remains unknown. Active trafficking from the Golgi apparatus is involved in acrosome formation, but experimental evidence indicates that trafficking of vesicles out of the Golgi also occurs during acrosomogenesis. Unfortunately, this second aspect of acrosome biogenesis remains poorly studied. In this article, we briefly discuss how the biosynthetic and endocytic pathways, assisted by a network of microtubules, tethering factors, motor proteins and small GTPases, relate and connect to give rise to the sperm-specific vacuole, with a particular emphasis placed on the endosomal compartment. It is hoped that this information will be useful to engage more studies on acrosome biogenesis by focusing attention towards suggested directions.
Collapse
Affiliation(s)
- Giovanna Berruti
- Department of Biology; Laboratory of Cellular and Molecular Biology of Reproduction; University of Milan; Milan, Italy
| | | |
Collapse
|