1
|
Antinozzi C, Di Luigi L, Sireno L, Caporossi D, Dimauro I, Sgrò P. Protective Role of Physical Activity and Antioxidant Systems During Spermatogenesis. Biomolecules 2025; 15:478. [PMID: 40305156 PMCID: PMC12024800 DOI: 10.3390/biom15040478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Oxidative stress is a significant factor that contributes to male infertility and sperm dysfunction. In this condition, an increase in ROS production exceeds the body's antioxidant defenses, resulting in a decline in spermatozoa quality and fertilizing capacity. Furthermore, excessive ROS production has been linked to the promotion of genomic damage, lipid peroxidation, inflammation, altered enzyme activity, and ultimately, irreversible alterations, cell death, and a decline in seminal parameters associated with male infertility. It is established that physical activity (PA), acting on inflammatory parameters and improving antioxidant defense, can alleviate the negative effects caused by free radicals, offering numerous health benefits and positively influencing sperm quality. The objective of this review is to highlight the mechanisms of ROS production, the physiological and pathophysiological roles of ROS in relation to the male reproductive system, and recent knowledge on the impact of some protocols of PA on these systems and the molecular mechanisms involved.
Collapse
Affiliation(s)
- Cristina Antinozzi
- Endocrinology Unit, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy (P.S.)
| | - Luigi Di Luigi
- Endocrinology Unit, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy (P.S.)
| | - Laura Sireno
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro De Bosis 15, 00135 Rome, Italy; (L.S.); (I.D.)
| | - Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro De Bosis 15, 00135 Rome, Italy; (L.S.); (I.D.)
| | - Ivan Dimauro
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro De Bosis 15, 00135 Rome, Italy; (L.S.); (I.D.)
| | - Paolo Sgrò
- Endocrinology Unit, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy (P.S.)
| |
Collapse
|
2
|
Lykins J, Moschitto MJ, Zhou Y, Filippova EV, Le HV, Tomita T, Fox BA, Bzik DJ, Su C, Rajagopala SV, Flores K, Spano F, Woods S, Roberts CW, Hua C, El Bissati K, Wheeler KM, Dovgin S, Muench SP, McPhillie M, Fishwick CW, Anderson WF, Lee PJ, Hickman M, Weiss LM, Dubey JP, Lorenzi HA, Silverman RB, McLeod RL. From TgO/GABA-AT, GABA, and T-263 Mutant to Conception of Toxoplasma. iScience 2024; 27:108477. [PMID: 38205261 PMCID: PMC10776954 DOI: 10.1016/j.isci.2023.108477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 04/28/2023] [Accepted: 11/13/2023] [Indexed: 01/12/2024] Open
Abstract
Toxoplasma gondii causes morbidity, mortality, and disseminates widely via cat sexual stages. Here, we find T. gondii ornithine aminotransferase (OAT) is conserved across phyla. We solve TgO/GABA-AT structures with bound inactivators at 1.55 Å and identify an inactivator selective for TgO/GABA-AT over human OAT and GABA-AT. However, abrogating TgO/GABA-AT genetically does not diminish replication, virulence, cyst-formation, or eliminate cat's oocyst shedding. Increased sporozoite/merozoite TgO/GABA-AT expression led to our study of a mutagenized clone with oocyst formation blocked, arresting after forming male and female gametes, with "Rosetta stone"-like mutations in genes expressed in merozoites. Mutations are similar to those in organisms from plants to mammals, causing defects in conception and zygote formation, affecting merozoite capacitation, pH/ionicity/sodium-GABA concentrations, drawing attention to cyclic AMP/PKA, and genes enhancing energy or substrate formation in TgO/GABA-AT-related-pathways. These candidates potentially influence merozoite's capacity to make gametes that fuse to become zygotes, thereby contaminating environments and causing disease.
Collapse
Affiliation(s)
- Joseph Lykins
- Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Matthew J. Moschitto
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL 60208-3113, USA
| | - Ying Zhou
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Ekaterina V. Filippova
- Center for Structural Genomics of Infectious Diseases and the Department of Biochemistry and Molecular Genetics, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hoang V. Le
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL 60208-3113, USA
| | - Tadakimi Tomita
- Division of Parasitology, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Barbara A. Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - David J. Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Chunlei Su
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | - Seesandra V. Rajagopala
- Department of Infectious Diseases, The J. Craig Venter Institute, 9704 Medical Center Drive, Rockville, MD 20850, USA
| | - Kristin Flores
- Center for Structural Genomics of Infectious Diseases and the Department of Biochemistry and Molecular Genetics, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Furio Spano
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow Scotland, UK
| | - Craig W. Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow Scotland, UK
| | - Cong Hua
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Kamal El Bissati
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Kelsey M. Wheeler
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah Dovgin
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
| | - Stephen P. Muench
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, The University of Leeds, Leeds, West York LS2 9JT, UK
| | - Martin McPhillie
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Colin W.G. Fishwick
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Wayne F. Anderson
- Center for Structural Genomics of Infectious Diseases and the Department of Biochemistry and Molecular Genetics, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Patricia J. Lee
- Division of Experimental Therapeutics, Military Malaria Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Mark Hickman
- Division of Experimental Therapeutics, Military Malaria Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Louis M. Weiss
- Division of Parasitology, Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jitender P. Dubey
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Hernan A. Lorenzi
- Department of Infectious Diseases, The J. Craig Venter Institute, 9704 Medical Center Drive, Rockville, MD 20850, USA
| | - Richard B. Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, and Center for Developmental Therapeutics, Northwestern University, Evanston, IL 60208-3113, USA
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Rima L. McLeod
- Department of Ophthalmology and Visual Sciences, The University of Chicago, Chicago, IL 60637, USA
- Department of Pediatrics (Infectious Diseases), Institute of Genomics, Genetics, and Systems Biology, Global Health Center, Toxoplasmosis Center, CHeSS, The College, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
3
|
Duma-Pauta JM, Juárez-López NO, Gutiérrez-Pérez O, Córdova-Izquierdo A, Vigueras-Villaseñor RM, Juárez-Mosqueda MDL. Cryopreservation, in addition to protein tyrosine phosphorylation, alters the distribution of phosphatidyl inositol bisphosphate and the localization of cytoskeletal and signaling proteins (gelsolin, tyrosine kinase c-SRC and phospholipase C-ζ) in the perinuclear theca of boar sperm. Cryobiology 2023; 113:104589. [PMID: 37778407 DOI: 10.1016/j.cryobiol.2023.104589] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
Cryopreservation of boar spermatozoa affects the perinuclear theca (PT) and involves several proteins and molecules that play important roles during capacitation and the acrosomal reaction. The objective of the present study was to evaluate whether the deleterious effects of cryopreservation in addition to protein tyrosine phosphorylation are accompanied by changes in the distribution of phosphatidyl inositol bisphosphate (PIP2) and the localization of cytoskeletal and signaling proteins in the perinuclear theca of cryopreserved boar spermatozoa. For this purpose, by immunocytochemistry (IC) the changes in localization of phosphorylated proteins in tyrosine residues, gelsolin, c-SRC kinase and PLC-ζ, as well as in the distribution of phosphatidyl inositol bisphosphate were analyzed in thawed spermatozoa (T) non capacitated (NC), capacitated (C) and in those with acrosomal reaction (AR) and compared with fresh spermatozoa (F) under the same physiological status. Western blotting (WB) and co-immunoprecipitation were performed to confirm the presence of these proteins in PT and to determine the interaction between these molecules. IC showed that immunostaining for phosphorylated proteins significantly increased in the acrosomal region and flagellum in TNC spermatozoa (p < 0.05). The proportion of cells displaying immunolabeling for gelsolin in the acrosomal region decreased after capacitation in cryopreserved spermatozoa; the same change was found (p < 0.05) in the proportion of spermatozoa immunoreactive to PIP2 in the sperm head. c-SRC was observed in the equatorial segment and acrosomal region, subdomains that coincide with the site where phosphorylated proteins were detected. PLC-ζ immunolocalization in fresh spermatozoa underwent changes after capacitation and acrosomal reaction, with a significant increase in the equatorial segment and post-acrosomal region in cryopreserved spermatozoa (p < 0.05). WB analysis indicated the presence of gelsolin, c-SRC and PLC-ζ in PT; besides, we confirmed that gelsolin co-immunoprecipitated with c-SRC and PLC-ζ, which changes according to the physiological state of spermatozoa. As a conclusion, cryopreservation together with increased immunodetection of tyrosine phosphorylated proteins decreases the detection of PIP2 and alters the immunolocalization patterns of gelsolin, c-SRC and PLC-ζ in the PT in boar spermatozoa.
Collapse
Affiliation(s)
- José Mauricio Duma-Pauta
- Universidad Nacional Autónoma de México, Departamento de Morfología, Facultad de Medicina Veterinaria y Zootecnia, Ciudad Universitaria No. 3000, Ciudad de México, CP, 04510, Mexico; Universidad de Cuenca, Laboratorio de Biotecnología de la Reproducción Animal, Facultad de Ciencias Agropecuarias, Avda. 12 de octubre, EC101205, Cuenca, Ecuador.
| | - Noé Orlando Juárez-López
- Universidad Nacional Autónoma de México, Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Ciudad Universitaria No. 3000, Ciudad deMéxico, CP, 04510, Mexico.
| | - Oscar Gutiérrez-Pérez
- Universidad Nacional Autónoma de México, Centro de Enseñanaza de Investigación y Extensión en Producción Porcina, Facultad de Medicina Veterinaria y Zootecnia, Ciudad Universitaria No. 3000, Ciudad de México, CP. 04510, México.
| | - Alejandro Córdova-Izquierdo
- Universidad Autónoma Metropolitana Unidad Xochimilco, Departamento de Agricultura y Producción Animal, Calzada del hueso 1100, Ciudad de México, CP, 04960, Mexico.
| | - Rosa María Vigueras-Villaseñor
- Instituto Nacional de Pediatría, Subdirección de Medicina Experimental, Av. Insurgentes Sur 3700-C, Ciudad de México, CP, 04530, Mexico
| | - María de Lourdes Juárez-Mosqueda
- Universidad Nacional Autónoma de México, Departamento de Morfología, Facultad de Medicina Veterinaria y Zootecnia, Ciudad Universitaria No. 3000, Ciudad de México, CP, 04510, Mexico.
| |
Collapse
|
4
|
Hashemi Karoii D, Azizi H, Skutella T. Altered G-Protein Transduction Protein Gene Expression in the Testis of Infertile Patients with Nonobstructive Azoospermia. DNA Cell Biol 2023; 42:617-637. [PMID: 37610843 DOI: 10.1089/dna.2023.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Recent studies have shown that several members of the G-protein-coupled receptors (GPCR) superfamily play crucial roles in the maintenance of ion-water homeostasis of the sperm and Sertoli cells, development of the germ cells, formation of the blood barrier, and maturation of sperm. The GPCR, guanyl-nucleotide exchange factor, membrane traffic protein, and small GTPase genes were analyzed by microarray and bioinformatics (3513 sperm and Sertoli cell genes). In the microarray analyses of three human cases with different nonobstructive azoospermia sperm, the expression of GOLGA8IP, OR2AT4, PHKA1, A2M, OR56A1, SEMA3G, LRRC17, APP, ARHGAP33, RABGEF1, NPY2R, GHRHR, LTB4R2, GRIK5, OR6K6, NAPG, OR6C65, VPS35, FPR3, and ARL4A was upregulated, while expression of MARS, SIRPG, OGFR, GPR150, LRRK1, and NGEF was downregulated. There was an increase in GBP3, GBP3, TNF, TGFB3, and CLTC expression in the Sertoli cells of three human cases with NOA, whereas expression of PAQR4, RRAGD, RAC2, SERPINB8, IRPB1, MRGPRF, RASA2, SIRPG, RGS2, RAP2A, RAB2B, ARL17, SERINC4, XIAP, DENND4C, ANKRA2, CSTA, STX18, and SNAP23 were downregulated. A combined analysis of Enrich Shiny Gene Ontology (GO), STRING, and Cytoscape was used to predict proteins' molecular interactions and then to recognize master pathways. Functional enrichment analysis showed that the biological process (BP), regulation of protein metabolic process, regulation of small GTPase-mediated signal transduction were significantly expressed in up-/downregulated differentially expressed genes (DEGs) in sperm. In molecular function (MF) experiments of DEGs that were up-/downregulated, it was found that GPCR activity, guanyl ribonucleotide binding, GTPase activity and nucleoside-triphosphatase activity were overexpressed. An analysis of GO enrichment findings of Sertoli cells showed BP and MF to be common DEGs. When these gene mutations have been validated, they can be used to create new GPCR antagonists or agonists that are receptor-selective.
Collapse
Affiliation(s)
- Danial Hashemi Karoii
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Thomas Skutella
- Medical Faculty, Institute for Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Bollwein H, Malama E. Review: Evaluation of bull fertility. Functional and molecular approaches. Animal 2023; 17 Suppl 1:100795. [PMID: 37567681 DOI: 10.1016/j.animal.2023.100795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 08/13/2023] Open
Abstract
With the term "assisted reproduction technologies" in modern cattle farming, one could imply the collection of techniques that aim at the optimal use of bovine gametes to produce animals of high genetic value in a time- and cost-efficient manner. The accurate characterisation of sperm quality plays a critical role for the efficiency of several assisted reproduction-related procedures, such as sperm processing, in vitro embryo production and artificial insemination. Bull fertility is ultimately a collective projection of the ability of a series of ejaculates to endure sperm processing stress, and achieve fertilisation of the oocyte and production of a viable and well-developing embryo. In this concept, the assessment of sperm functional and molecular characteristics is key to bull fertility diagnostics and prognostics. Among others, functional features linked to sperm plasma membrane, acrosome and DNA integrity are usually assessed as a measure of the ability of sperm to express the phenotypes that will allow them to maintain their homeostasis and orchestrate-in a strict temporal manner-the course of events that will enable the delivery of their genetic content to the oocyte upon fertilisation. Nevertheless, measures of sperm functionality are not always adequate indicators of bull fertility. Nowadays, advancements in the field of molecular biology have facilitated the profiling of several biomolecules in male gametes. The molecular profiling of bovine sperm offers a deeper insight into the mechanisms underlying sperm physiology and, thus, can reveal novel candidate markers for bull fertility prognosis. In this review, the importance of three organelles (the nucleus, the plasma membrane and the acrosome) for the characterisation of sperm fertilising capacity and bull fertility is discussed at functional and molecular levels. In particular, information about sperm head morphometry, chromatin structure, viability as well as the ability of sperm to capacitate and undergo the acrosome reaction are presented in relation to the cryotolerance of male gametes and bull fertility. Finally, major spermatozoal coding and non-coding RNAs, and proteins that are involved in the above-mentioned aspects of sperm functionality are also summarised.
Collapse
Affiliation(s)
- H Bollwein
- Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - E Malama
- Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| |
Collapse
|
6
|
The Dual Role of Oxidants in Male (In)fertility: Every ROSe Has a Thorn. Int J Mol Sci 2023; 24:ijms24054994. [PMID: 36902424 PMCID: PMC10002566 DOI: 10.3390/ijms24054994] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The role of oxidative stress (OS) in male infertility as a primary etiology and/or concomitant cause in other situations, such as inflammation, varicocele and gonadotoxin effects, is well documented. While reactive oxygen species (ROS) are implicated in many important roles, from spermatogenesis to fertilization, epigenetic mechanisms which are transmissible to offspring have also recently been described. The present review is focused on the dual aspects of ROS, which are regulated by a delicate equilibrium with antioxidants due to the special frailty of spermatozoa, in continuum from physiological condition to OS. When the ROS production is excessive, OS ensues and is amplified by a chain of events leading to damage of lipids, proteins and DNA, ultimately causing infertility and/or precocious pregnancy termination. After a description of positive ROS actions and of vulnerability of spermatozoa due to specific maturative and structural characteristics, we linger on the total antioxidant capacity (TAC) of seminal plasma, which is a measure of non-enzymatic non-proteic antioxidants, due to its importance as a biomarker of the redox status of semen; the therapeutic implications of these mechanism play a key role in the personalized approach to male infertility.
Collapse
|
7
|
Skerrett-Byrne DA, Anderson AL, Bromfield EG, Bernstein IR, Mulhall JE, Schjenken JE, Dun MD, Humphrey SJ, Nixon B. Global profiling of the proteomic changes associated with the post-testicular maturation of mouse spermatozoa. Cell Rep 2022; 41:111655. [DOI: 10.1016/j.celrep.2022.111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 06/15/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022] Open
|
8
|
Zhang R, Liang C, Guo X, Bao P, Pei J, Wu F, Yin M, Chu M, Yan P. Quantitative phosphoproteomics analyses reveal the regulatory mechanisms related to frozen-thawed sperm capacitation and acrosome reaction in yak (Bos grunniens). Front Physiol 2022; 13:1013082. [PMID: 36277216 PMCID: PMC9583833 DOI: 10.3389/fphys.2022.1013082] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Mammalian spermatozoa are not mature after ejaculation and must undergo additional functional and structural changes within female reproductive tracts to achieve subsequent fertilization, including both capacitation and acrosome reaction (AR), which are dominated by post-translational modifications (PTMs), especially phosphorylation. However, the mechanism of protein phosphorylation during frozen-thawed sperm capacitation and AR has not been well studied. In this study, the phosphoproteomics approach was employed based on tandem mass tag (TMT) labeling combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS) strategy to analyze frozen-thawed sperm in Ashidan yak under three sequential conditions (density gradient centrifugation-based purification, incubation in the capacitation medium and induction of AR processes by the calcium ionophore A23187 treatment). The identification of 1,377 proteins with 5,509 phosphorylation sites revealed changes in phosphorylation levels of sperm-specific proteins involved in regulation of spermatogenesis, sperm motility, energy metabolism, cilium movement, capacitation and AR. Some phosphorylated proteins, such as AKAP3, AKAP4, SPA17, PDMD11, CABYR, PRKAR1A, and PRKAR2A were found to regulate yak sperm capacitation and AR though the cAMP/PKA signaling pathway cascades. Notably, the phosphorylation level of SPA17 at Y156 increased in capacitated sperm, suggesting that it is also a novel functional protein besides AKAPs during sperm capacitation. Furthermore, the results of this study suggested that the phosphorylation of PRKAR1A and PRKAR2A, and the dephosphorylation of CABYR both play key regulatory role in yak sperm AR process. Protein-protein interaction analysis revealed that differentially phosphorylated proteins (AKAP3, AKAP4, FSIP2, PSMD11, CABYR, and TPPP2) related to capacitation and AR process played a key role in protein kinase A binding, sperm motility, reproductive process, cytoskeleton and sperm flagella function. Taken together, these data provide not only a solid foundation for further exploring phosphoproteome of sperm in yak, but an efficient way to identify sperm fertility-related marker phosphorylated proteins.
Collapse
Affiliation(s)
- Renzheng Zhang
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Chunnian Liang
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xian Guo
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengjia Bao
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jie Pei
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Fude Wu
- Yak Breeding and Extension Service Center in in Qinghai Province, Xining, China
| | - Mancai Yin
- Yak Breeding and Extension Service Center in in Qinghai Province, Xining, China
| | - Min Chu
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- *Correspondence: Min Chu, ; Ping Yan,
| | - Ping Yan
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- *Correspondence: Min Chu, ; Ping Yan,
| |
Collapse
|
9
|
Xie Y, Xu Z, Wu C, Zhou C, Zhang X, Gu T, Yang J, Yang H, Zheng E, Xu Z, Cai G, Li Z, Liu D, Wu Z, Hong L. Extracellular vesicle-encapsulated miR-21-5p in seminal plasma prevents sperm capacitation via Vinculin inhibition. Theriogenology 2022; 193:103-113. [PMID: 36156422 DOI: 10.1016/j.theriogenology.2022.09.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 07/11/2022] [Accepted: 09/10/2022] [Indexed: 10/31/2022]
Abstract
To penetrate the zona pellucida before sperm-egg binding, sperm must undergo highly time-controlled capacitation and acrosome reaction in the female reproductive tract. Our previous study demonstrated that miR-21-5p is the most abundant miRNA in boar seminal plasma (SP)-derived extracellular vesicles (EVs) and can target Vinculin (VCL) gene, which may participate in boar sperm capacitation. Thus, this study aims to explore the potential role of miR-21-5p from SP-derived EVs in preventing sperm capacitation and its underlying mechanism. We observed that sperm could incorporate miR-21-5p from SP-derived EVs. The roles of SP-derived EVs miR-21-5p in sperm capacitation were then determined using gain- and loss-of-function analyses. In addition, the expression levels of miR-21-5p, VCL, and VCL protein in liquid-preserved boar sperm following transfection were determined using RT-qPCR and Western blotting. Our results revealed that miR-21-5p overexpression inhibited sperm capacitation and acrosome reaction. Similarly, miR-21-5p expression was significantly lower (P < 0.05) in capacitated sperm than un-capacitated sperm. However, the protein level of VCL was also significantly lower (P < 0.05) in capacitated sperm than un-capacitated sperm. Furthermore, immunofluorescence analysis showed that VCL protein mainly located in sperm head and sperm capacitation was inhibited after treating with VCL protein inhibitor (Chrysin). In conclusion, our study provides reasonable evidence that miR-21-5p expression in SP-derived EVs could prevent sperm capacitation via VCL inhibition.
Collapse
Affiliation(s)
- Yanshe Xie
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zhiqian Xu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Changhua Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Chen Zhou
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | | | - Ting Gu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Jie Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Enqin Zheng
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zheng Xu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Gengyuan Cai
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangzhou, 510642, China
| | - Dewu Liu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China; State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangzhou, 510642, China.
| | - Linjun Hong
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China.
| |
Collapse
|
10
|
Serafini S, O'Flaherty C. Redox Regulation to Modulate Phosphorylation Events in Human Spermatozoa. Antioxid Redox Signal 2022; 37:437-450. [PMID: 34714121 DOI: 10.1089/ars.2021.0117] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Significance: Spermatozoa are complex and compartmentalized cells that undergo capacitation, a series of biochemical and morphological changes to acquire the ability to fertilize oocytes. Reactive oxygen species (ROS) have a prominent dual role in capacitation. At physiological levels, ROS regulate numerous cellular processes, including increases of cyclic adenosine monophosphate, calcium, and activation of phosphorylation events needed for capacitation. On the contrary, at high concentrations that do not impair sperm viability, ROS can cause loss of motility and inhibition of capacitation. Higher ROS concentrations promote oxidation of lipids, proteins, and DNA leading to cell death, and these damages have been associated with male infertility. Critical Issues: When incubated under specific conditions, spermatozoa can produce low and controlled amounts of ROS that are not harmful but instead regulate numerous cellular processes, including the phosphorylation of tyrosine, serine, and threonine residues in critical proteins needed for sperm capacitation. Here, we outline the complex redox signaling in human spermatozoa needed to achieve fertility and the role of ROS as physiological mediators that trigger phosphorylation cascades. Moreover, we illustrate the importance of various phosphoproteins in spermatozoa capacitation, viability, and hyperactive motility. Future Directions: Further studies to elucidate the different phosphorylation players during sperm capacitation and acrosome reaction (the regulated exocytotic event that releases proteolytic enzymes allowing the spermatozoon to penetrate the zona pellucida and fertilize the oocyte) are essential to understand how the spermatozoon acquires the fertilizing ability to fertilize the oocyte. This knowledge will serve to develop novel diagnostic tools and therapy for male infertility. Antioxid. Redox Signal. 37, 437-450.
Collapse
Affiliation(s)
- Steven Serafini
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montréal, Canada
| | - Cristian O'Flaherty
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montréal, Canada.,Urology Division, Department of Surgery, Faculty of Medicine, McGill University, Montréal, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montréal, Canada.,The Research Institute, McGill University Health Centre, Montréal, Canada
| |
Collapse
|
11
|
Proteomic Analysis of the Role of the Adenylyl Cyclase-cAMP Pathway in Red Blood Cell Mechanical Responses. Cells 2022; 11:cells11071250. [PMID: 35406814 PMCID: PMC8997765 DOI: 10.3390/cells11071250] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Red blood cell (RBC) deformability is modulated by the phosphorylation status of the cytoskeletal proteins that regulate the interactions of integral transmembrane complexes. Proteomic studies have revealed that receptor-related signaling molecules and regulatory proteins involved in signaling cascades are present in RBCs. In this study, we investigated the roles of the cAMP signaling mechanism in modulating shear-induced RBC deformability and examined changes in the phosphorylation of the RBC proteome. We implemented the inhibitors of adenylyl cyclase (SQ22536), protein kinase A (H89), and phosphodiesterase (PDE) (pentoxifylline) to whole blood samples, applied 5 Pa shear stress (SS) for 300 s with a capillary tubing system, and evaluated RBC deformability using a LORRCA MaxSis. The inhibition of signaling molecules significantly deteriorated shear-induced RBC deformability (p < 0.05). Capillary SS slightly increased the phosphorylation of RBC cytoskeletal proteins. Tyrosine phosphorylation was significantly elevated by the modulation of the cAMP/PKA pathway (p < 0.05), while serine phosphorylation significantly decreased as a result of the inhibition of PDE (p < 0.05). AC is the core element of this signaling pathway, and PDE works as a negative feedback mechanism that could have potential roles in SS-induced RBC deformability. The cAMP/PKA pathway could regulate RBC deformability during capillary transit by triggering significant alterations in the phosphorylation state of RBCs.
Collapse
|
12
|
Grinshtain E, Shpungin S, Baum M, Nir U, Breitbart H. The Fer tyrosine kinase protects sperm from spontaneous acrosome reaction. Dev Biol 2022; 487:24-33. [DOI: 10.1016/j.ydbio.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 03/23/2022] [Accepted: 04/13/2022] [Indexed: 11/24/2022]
|
13
|
Sansegundo E, Tourmente M, Roldan ERS. Energy Metabolism and Hyperactivation of Spermatozoa from Three Mouse Species under Capacitating Conditions. Cells 2022; 11:220. [PMID: 35053337 PMCID: PMC8773617 DOI: 10.3390/cells11020220] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Mammalian sperm differ widely in sperm morphology, and several explanations have been presented to account for this diversity. Less is known about variation in sperm physiology and cellular processes that can give sperm cells an advantage when competing to fertilize oocytes. Capacitation of spermatozoa, a process essential for mammalian fertilization, correlates with changes in motility that result in a characteristic swimming pattern known as hyperactivation. Previous studies revealed that sperm motility and velocity depend on the amount of ATP available and, therefore, changes in sperm movement occurring during capacitation and hyperactivation may involve changes in sperm bioenergetics. Here, we examine differences in ATP levels of sperm from three mouse species (genus Mus), differing in sperm competition levels, incubated under non-capacitating and capacitating conditions, to analyse relationships between energetics, capacitation, and swimming patterns. We found that, in general terms, the amount of sperm ATP decreased more rapidly under capacitating conditions. This descent was related to the development of a hyperactivated pattern of movement in two species (M. musculus and M. spicilegus) but not in the other (M. spretus), suggesting that, in the latter, temporal dynamics and energetic demands of capacitation and hyperactivation may be decoupled or that the hyperactivation pattern differs. The decrease in ATP levels during capacitation was steeper in species with higher levels of sperm competition than in those with lower levels. Our results suggest that, during capacitation, sperm consume more ATP than under non-capacitating conditions. This higher ATP consumption may be linked to higher velocity and lateral head displacement, which are associated with hyperactivated motility.
Collapse
Affiliation(s)
- Ester Sansegundo
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, Spanish Research Council (CSIC), 28006 Madrid, Spain;
| | - Maximiliano Tourmente
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, Spanish Research Council (CSIC), 28006 Madrid, Spain;
- Centro de Biología Celular y Molecular, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Cordoba X5016GCA, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), Consejo Nacional de Investigaciones Científica y Técnicas (CONICET), Cordoba X5016GCA, Argentina
| | - Eduardo R. S. Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales, Spanish Research Council (CSIC), 28006 Madrid, Spain;
| |
Collapse
|
14
|
Mouse and Fly Sperm Motility Changes Differently under Modelling Microgravity. Curr Issues Mol Biol 2021; 43:590-604. [PMID: 34287235 PMCID: PMC8929020 DOI: 10.3390/cimb43020043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 11/26/2022] Open
Abstract
Sperm motility is essential for the natural fertilization process in most animal species. Despite the fact that evolution took place under conditions of constant gravity, the motility of spermatozoa of insects and mammals under microgravity conditions changes in different ways. In this work, an attempt was made to explain this effect. The sperm motility of the fruit fly Drosophila melanogaster and the mouse was evaluated after exposure to a random positioning machine for 6 h. Sodium fluoride was used to inhibit serine/threonine phosphatases, sodium orthovanadate was used to inhibit tyrosine phosphatases, and 6-(dimethylamino)purine was used to inhibit protein kinases. The results obtained indicate that simulated microgravity leads to an increase in the speed of movement of fly spermatozoa by 30% (p < 0.05), and this effect is blocked by sodium fluoride. In contrast, a 29% (p < 0.05) decrease in the speed of movement of mouse spermatozoa under simulated microgravity is prevented by 6-(dimethylamino)purine. Moreover, after 6 h of exposure, the content of tubulin cytoskeleton and actin proteins remains at the control level in the spermatozoa of flies and mice. However, the content of the actin-binding protein alpha-actinin in fly sperm decreases by 29% (p < 0.05), while in mouse sperm, the relative content of alpha-actinin1 increases by 94% (p < 0.05) and alpha-actinin4 by 121% (p < 0.05) relative to the control, as determined by 6 simulated microgravity tests. It can be assumed that the effect of simulated microgravity on the motility of mammalian spermatozoa is mediated through the regulation of phosphorylation and that of insects through the regulation of dephosphorylation of motor proteins; moreover, the development of a response to changes in external mechanical conditions has a different time scale.
Collapse
|
15
|
Priyadarshana C, Setiawan R, Tajima A, Asano A. Src family kinases-mediated negative regulation of sperm acrosome reaction in chickens (Gallus gallus domesticus). PLoS One 2020; 15:e0241181. [PMID: 33180820 PMCID: PMC7660528 DOI: 10.1371/journal.pone.0241181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/06/2020] [Indexed: 11/19/2022] Open
Abstract
The acrosome reaction (AR) is a strictly-regulated, synchronous exocytosis that is required for sperm to penetrate ova. This all-or-nothing process occurs only once in the sperm lifecycle through a sequence of signaling pathways. Spontaneous, premature AR therefore compromises fertilization potential. Although protein kinase A (PKA) pathways play a central role in AR across species, the signaling network used for AR induction is poorly understood in birds. Mechanistic studies of mammalian sperm AR demonstrate that PKA activity is downstreamly regulated by Src family kinases (SFKs). Using SFK inhibitors, our study shows that in chicken sperm, SFKs play a role in the regulation of PKA activity and spontaneous AR without affecting motility. Furthermore, we examined the nature of SFK phosphorylation using PKA and protein tyrosine phosphatase inhibitors, which demonstrated that unlike in mammals, SFK phosphorylation in birds does not occur downstream of PKA and is primarily regulated by calcium-dependent tyrosine phosphatase activity. Functional characterization of SFKs in chicken sperm showed that SFK activation modulates the membrane potential and plays a role in inhibiting spontaneous AR. Employing biochemical isolation, we also found that membrane rafts are involved in the regulation of SFK phosphorylation. This study demonstrates a unique mechanism for regulating AR induction inherent to avian sperm that ensure fertilization potential despite prolonged storage.
Collapse
Affiliation(s)
- Chathura Priyadarshana
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Rangga Setiawan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Atsushi Tajima
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Atsushi Asano
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
16
|
Li K, Sun P, Wang Y, Gao T, Zheng D, Liu A, Ni Y. Hsp90 interacts with Cdc37, is phosphorylated by PKA/PKC, and regulates Src phosphorylation in human sperm capacitation. Andrology 2020; 9:185-195. [PMID: 32656999 DOI: 10.1111/andr.12862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heat shock protein 90 (Hsp90) signaling pathways participate in protein phosphorylation during sperm capacitation. However, the underlying mechanism is largely unknown. OBJECTIVE The aim of this study was to explore the interaction between Hsp90 and its co-chaperone protein, cell division cycle protein Cdc37 (Cdc37), in human spermatozoa. MATERIALS AND METHODS We examined the effects of H-89 (a protein kinase A [PKA] inhibitor) and Go6983 (a protein kinase C [PKC] inhibitor) on the phosphorylation of serine, threonine, and tyrosine residues in Hsp90; the effect of 17-allylamino-17-demethoxygeldanamycin (17-AAG, Hsp90 inhibitor) on Y416-Src phosphorylation; and the effects of 17-AAG and geldanamycin on threonine phosphorylation during human sperm capacitation. RESULTS Hsp90 co-localized and interacted with Cdc37. During human sperm capacitation, Hsp90 phosphorylation at serine, threonine, and tyrosine residues was inhibited by H-89 and Go6983. In addition, phosphorylation of residue Y416 in the tyrosine kinase Src (its active site) was inhibited by 17-AAG, and the threonine phosphorylation levels of some proteins were decreased by 17-AAG and geldanamycin. DISCUSSION AND CONCLUSION Taken together, our data showed that the interaction of Hsp90 with Cdc37 regulates total protein threonine phosphorylation and Src phosphorylation via its serine, threonine, and tyrosine phosphorylation, which are controlled by PKA and PKC during human sperm capacitation. The results of this study help understand the mechanism underlying Hsp90 regulation of sperm function.
Collapse
Affiliation(s)
- Kun Li
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Peibei Sun
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Yayan Wang
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Tian Gao
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Dongwang Zheng
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Ajuan Liu
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| | - Ya Ni
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences/Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
17
|
A framework for high-resolution phenotyping of candidate male infertility mutants: from human to mouse. Hum Genet 2020; 140:155-182. [PMID: 32248361 DOI: 10.1007/s00439-020-02159-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 03/27/2020] [Indexed: 12/18/2022]
Abstract
Male infertility is a heterogeneous condition of largely unknown etiology that affects at least 7% of men worldwide. Classical genetic approaches and emerging next-generation sequencing studies support genetic variants as a frequent cause of male infertility. Meanwhile, the barriers to transmission of this disease mean that most individual genetic cases will be rare, but because of the large percentage of the genome required for spermatogenesis, the number of distinct causal mutations is potentially large. Identifying bona fide causes of male infertility thus requires advanced filtering techniques to select for high-probability candidates, including the ability to test causality in animal models. The mouse remains the gold standard for defining the genotype-phenotype connection in male fertility. Here, we present a best practice guide consisting of (a) major points to consider when interpreting next-generation sequencing data performed on infertile men, and, (b) a systematic strategy to categorize infertility types and how they relate to human male infertility. Phenotyping infertility in mice can involve investigating the function of multiple cell types across the testis and epididymis, as well as sperm function. These findings will feed into the diagnosis and treatment of male infertility as well as male health broadly.
Collapse
|
18
|
Aitken RJ, Drevet JR. The Importance of Oxidative Stress in Determining the Functionality of Mammalian Spermatozoa: A Two-Edged Sword. Antioxidants (Basel) 2020; 9:antiox9020111. [PMID: 32012712 PMCID: PMC7070991 DOI: 10.3390/antiox9020111] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
This article addresses the importance of oxidative processes in both the generation of functional gametes and the aetiology of defective sperm function. Functionally, sperm capacitation is recognized as a redox-regulated process, wherein a low level of reactive oxygen species (ROS) generation is intimately involved in driving such events as the stimulation of tyrosine phosphorylation, the facilitation of cholesterol efflux and the promotion of cAMP generation. However, the continuous generation of ROS ultimately creates problems for spermatozoa because their unique physical architecture and unusual biochemical composition means that they are vulnerable to oxidative stress. As a consequence, they are heavily dependent on the antioxidant protection afforded by the fluids in the male and female reproductive tracts and, during the precarious process of insemination, seminal plasma. If this antioxidant protection should be compromised for any reason, then the spermatozoa experience pathological oxidative damage. In addition, situations may prevail that cause the spermatozoa to become exposed to high levels of ROS emanating either from other cells in the immediate vicinity (particularly neutrophils) or from the spermatozoa themselves. The environmental and lifestyle factors that promote ROS generation by the spermatozoa are reviewed in this article, as are the techniques that might be used in a diagnostic context to identify patients whose reproductive capacity is under oxidative threat. Understanding the strengths and weaknesses of ROS-monitoring methodologies is critical if we are to effectively identify those patients for whom treatment with antioxidants might be considered a rational management strategy.
Collapse
Affiliation(s)
- Robert J. Aitken
- Priority Research Centre for Reproductive Sciences, Faculty of Science and Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- Correspondence:
| | - Joel R. Drevet
- GReD Institute, INSERM U1103—CNRS UMR6293—Université Clermont Auvergne, Faculty of Medicine, CRBC building, 28 place Henri Dunant, 63001 Clermont-Ferrand, France;
| |
Collapse
|
19
|
Dey S, Brothag C, Vijayaraghavan S. Signaling Enzymes Required for Sperm Maturation and Fertilization in Mammals. Front Cell Dev Biol 2019; 7:341. [PMID: 31921853 PMCID: PMC6930163 DOI: 10.3389/fcell.2019.00341] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022] Open
Abstract
In mammals, motility and fertilizing ability of spermatozoa develop during their passage through the epididymis. After ejaculation, sperm undergo capacitation and hyperactivation in the female reproductive tract - a motility transition that is required for sperm penetration of the egg. Both epididymal initiation of sperm motility and hyperactivation are essential for male fertility. Motility initiation in the epididymis and sperm hyperactivation involve changes in metabolism, cAMP (cyclic adenosine mono-phosphate), calcium and pH acting through protein kinases and phosphatases. Despite this knowledge, we still do not understand, in biochemical terms, how sperm acquire motility in the epididymis and how motility is altered in the female reproductive tract. Recent data show that the sperm specific protein phosphatase PP1γ2, glycogen synthase kinase 3 (GSK3), and the calcium regulated phosphatase calcineurin (PP2B), are involved in epididymal sperm maturation. The protein phosphatase PP1γ2 is present only in testis and sperm in mammals. PP1γ2 has a isoform-specific requirement for normal function of mammalian sperm. Sperm PP1γ2 is regulated by three proteins - inhibitor 2, inhibitor 3 and SDS22. Changes in phosphorylation of these three inhibitors and their binding to PP1γ2 are involved in initiation and activation of sperm motility. The inhibitors are phosphorylated by protein kinases, one of which is GSK3. The isoform GSK3α is essential for epididymal sperm maturation and fertility. Calcium levels dramatically decrease during sperm maturation and initiation of motility suggesting that the calcium activated sperm phosphatase (PP2B) activity also decreases. Loss of PP2B results in male infertility due to impaired sperm maturation in the epididymis. Thus the three signaling enzymes PP1γ2, GSK3, and PP2B along with the documented PKA (protein kinase A) have key roles in sperm maturation and hyperactivation. Significantly, all these four signaling enzymes are present as specific isoforms only in placental mammals, a testimony to their essential roles in the unique aspects of sperm function in mammals. These findings should lead to a better biochemical understanding of the basis of male infertility and should lead to novel approaches to a male contraception and managed reproduction.
Collapse
|
20
|
Alvarez-Palomo B, Barrot-Feixat C, Sarret H, Requena J, Pau M, Vidal-Taboada JM, Oliva R, Ballesca JL, Edel MJ, Mezquita-Pla J. Two novel ligand-independent variants of the VEGFR-1 receptor are expressed in human testis and spermatozoa, one of them with the ability to activate SRC proto-oncogene tyrosine kinases. Oncotarget 2019; 10:5871-5887. [PMID: 31645906 PMCID: PMC6791376 DOI: 10.18632/oncotarget.27232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/24/2019] [Indexed: 11/25/2022] Open
Abstract
The vascular endothelial growth factor receptor 1 (VEGFR-1) family of receptors is preferentially expressed in endothelial cells, with the full-length and mostly the soluble (sVEGFR-1) isoforms being the most expressed ones. Surprisingly, cancer cells (MDA-MB-231) express, instead, alternative intracellular VEGFR-1 variants. We wondered if these variants, that are no longer dependent on ligands for activation, were expressed in a physiological context, specifically in spermatogenic cells, and whether their expression was maintained in spermatozoa and required for human fertility. By interrogating a human library of mature testis cDNA, we characterized two new truncated intracellular variants different from the ones previously described in cancer cells. The new isoforms were transcribed from alternative transcription start sites (aTSS) located respectively in intron-19 (i19VEGFR-1) and intron-28 (i28VEGFR-1) of the VEGFR-1 gene (GenBank accession numbers JF509744 and JF509745) and expressed in mature testis and spermatozoa. In this paper, we describe the characterization of these isoforms by RT-PCR, northern blot, and western blot, their preferential expression in human mature testis and spermatozoa, and the elements that punctuate their proximal promoters and suggest cues for their expression in spermatogenic cells. Mechanistically, we show that i19VEGFR-1 has a strong ability to phosphorylate and activate SRC proto-oncogene non-receptor tyrosine kinases and a significant bias toward a decrease in expression in patients considered infertile by WHO criteria.
Collapse
Affiliation(s)
- Belen Alvarez-Palomo
- Molecular Genetics and Control of Pluripotency Laboratory, Department of Biomedicine, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| | - Carme Barrot-Feixat
- Forensic Genetics Laboratory, Medicine Department, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| | - Helena Sarret
- Molecular Genetics and Control of Pluripotency Laboratory, Department of Biomedicine, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| | - Jordi Requena
- Molecular Genetics and Control of Pluripotency Laboratory, Department of Biomedicine, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| | - Montserrat Pau
- Molecular Genetics Laboratory, Department of Biomedicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| | - Jose-Manuel Vidal-Taboada
- Peripheral Nervous System, Neuroscience Department, Vall d'Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain
| | - Rafael Oliva
- Molecular Biology of Reproduction and Development Laboratory, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Department of Biomedicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain.,Biochemistry and Molecular Genetics Service, Biomedical Diagnostic Centre, Hospital Clinic, Barcelona, Catalonia, Spain
| | - Josep-Lluis Ballesca
- Clinic Institute of Gynaecology, Obstetrics and Neonatology, Hospital Clinic, Barcelona, Catalonia, Spain
| | - Michael J Edel
- Molecular Genetics and Control of Pluripotency Laboratory, Department of Biomedicine, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain.,International Research Fellow, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia.,Senior Research Fellow, University of Western Australia, School of Medicine and Pharmacology, Harry Perkins Research Institute Centre for Cell Therapy and Regenerative Medicine (CCTRM), Perth, Western Australia, Australia
| | - Jovita Mezquita-Pla
- Molecular Genetics and Control of Pluripotency Laboratory, Department of Biomedicine, Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
21
|
Zhou Y, Wu F, Zhang M, Xiong Z, Yin Q, Ru Y, Shi H, Li J, Mao S, Li Y, Cao X, Hu R, Liew CW, Ding Q, Wang X, Zhang Y. EMC10 governs male fertility via maintaining sperm ion balance. J Mol Cell Biol 2019; 10:503-514. [PMID: 29659949 DOI: 10.1093/jmcb/mjy024] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 04/05/2018] [Indexed: 02/01/2023] Open
Abstract
Infertility is a severe public health problem worldwide that prevails up to 15% in reproductive-age couples, and male infertility accounts for half of total infertility. Studies on genetically modified animal models have identified lots of genes involved in the pathogenesis of male infertility. The underlying causes, however, remain largely unclear. In this study, we provide evidence that EMC10, one subunit of endoplasmic reticulum (ER) membrane protein complex (EMC), is required for male fertility. EMC10 is significantly decreased in spermatozoa from patients with asthenozoospermia and positively associated with human sperm motility. Male mice lacking Emc10 gene are completely sterile. Emc10-null spermatozoa exhibit multiple defects including abnormal morphology, decreased motility, impaired capacitation, and impotency of acrosome reaction, thereby which are incapable of fertilizing intact or ZP-free oocytes. However, intracytoplasmic sperm injection could rescue this defect caused by EMC10 deletion. Mechanistically, EMC10 deficiency leads to inactivation of Na/K-ATPase, in turn giving rise to an increased level of intracellular Na+ in spermatozoa, which contributes to decreased sperm motility and abnormal morphology. Other mechanistic investigations demonstrate that the absence of EMC10 results in a reduction of HCO3- entry and subsequent decreases of both cAMP-dependent protein kinase A substrate phosphorylation and protein tyrosine phosphorylation. These data demonstrate that EMC10 is indispensable to male fertility via maintaining sperm ion balance of Na+ and HCO3-, and also suggest that EMC10 is a promising biomarker for male fertility and a potential pharmaceutical target to treat male infertility.
Collapse
Affiliation(s)
- Yuchuan Zhou
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fei Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Man Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zuquan Xiong
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qianqian Yin
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yanfei Ru
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Huijuan Shi
- China National Population and Family Planning Key Laboratory of Contraceptive Drugs and Devices, Shanghai Institute of Planned Parenthood Research, Shanghai, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shanhua Mao
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanliang Li
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinyi Cao
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Renming Hu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chong Wee Liew
- Department of Physiology & Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Qiang Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuanchun Wang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yonglian Zhang
- State Key Laboratory of Molecular Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
22
|
Alonso CAI, Lottero-Leconte R, Luque GM, Vernaz ZJ, Di Siervi N, Gervasi MG, Buffone MG, Davio C, Perez-Martinez S. MRP4-mediated cAMP efflux is essential for mouse spermatozoa capacitation. J Cell Sci 2019; 132:jcs.230565. [PMID: 31253671 DOI: 10.1242/jcs.230565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
Mammalian spermatozoa must undergo biochemical and structural changes to acquire the capacity for fertilization, in a process known as capacitation. Activation of PKA enzymes is essential for capacitation, and thus cAMP levels are tightly regulated during this process. Previously, we demonstrated that during capacitation, bovine spermatozoa extrude cAMP through multidrug resistance-associated protein 4 (MRP4, also known as ABCC4), which regulates intracellular levels of the nucleotide and provides cAMP to the extracellular space. Here, we report the presence of functional MRP4 in murine spermatozoa, since its pharmacological inhibition with MK571 decreased levels of extracellular cAMP. This also produced a sudden increase in PKA activity, with decreased tyrosine phosphorylation at the end of capacitation. Blockade of MRP4 inhibited induction of acrosome reaction, hyperactivation and in vitro fertilization. Moreover, MRP4 inhibition generated an increase in Ca2+ levels mediated by PKA, and depletion of Ca2+ salts from the medium prevented the loss of motility and phosphotyrosine inhibition produced by MK571. These results were supported using spermatozoa from CatSper Ca2+ channel knockout mice. Taken together, these results suggest that cAMP efflux via MRP4 plays an essential role in mouse sperm capacitation.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- C A I Alonso
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO) (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - R Lottero-Leconte
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO) (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - G M Luque
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428ADN, Argentina
| | - Z J Vernaz
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO) (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - N Di Siervi
- Instituto de Investigaciones Farmacológicas (ININFA) (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina
| | - M G Gervasi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, United States
| | - M G Buffone
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428ADN, Argentina
| | - C Davio
- Instituto de Investigaciones Farmacológicas (ININFA) (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina
| | - S Perez-Martinez
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO) (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
23
|
Bathala P, Fereshteh Z, Li K, Al-Dossary AA, Galileo DS, Martin-DeLeon PA. Oviductal extracellular vesicles (oviductosomes, OVS) are conserved in humans: murine OVS play a pivotal role in sperm capacitation and fertility. Mol Hum Reprod 2019; 24:143-157. [PMID: 29370405 DOI: 10.1093/molehr/gay003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/20/2018] [Indexed: 01/12/2023] Open
Abstract
STUDY QUESTIONS Are extracellular vesicles (EVs) in the murine oviduct (oviductosomes, OVS) conserved in humans and do they play a role in the fertility of Pmca4-/- females? SUMMARY ANSWER OVS and their fertility-modulating proteins are conserved in humans, arise via the apocrine pathway, and mediate a compensatory upregulation of PMCA1 (plasma membrane Ca2+-ATPase 1) in Pmca4-/- female mice during proestrus/estrus, to account for their fertility. WHAT IS KNOWN ALREADY Recently murine OVS were identified and shown during proestrus/estrus to express elevated levels of PMCA4 which they can deliver to sperm. PMCA4 is the major Ca2+ efflux pump in murine sperm and Pmca4 deletion leads to loss of sperm motility and male infertility as there is no compensatory upregulation of the remaining Ca2+ pump, PMCA1. Of the four family members of PMCAs (PMCA1-4), PMCA1 and PMCA4 are ubiquitous, and to date there have been no reports of one isoform being upregulated to compensate for another in any organ/tissue. Since Pmca4-/- females are fertile, despite the abundant expression of PMCA4 in wild-type (WT) OVS, we propose that OVS serve a role of packaging and delivering to sperm elevated levels of PMCA1 in Pmca4-/- during proestrus/estrus to compensate for PMCA4's absence. STUDY DESIGN, SIZE, DURATION Fallopian tubes from pre-menopausal women undergoing hysterectomy were used to study EVs in the luminal fluid. Oviducts from sexually mature WT mice were sectioned after perfusion fixation to detect EVs in situ. Oviducts were recovered from WT and Pmca4-/- after hormonally induced estrus and sectioned for PMCA1 immunofluorescence (IF) (detected with confocal microscopy) and hematoxylin and eosin staining. Reproductive tissues, luminal fluids and EVs were recovered after induced estrus and after natural cycling for western blot analysis of PMCA1 and qRT-PCR of Pmca1 to compare expression levels in WT and Pmca4-/-. OVS, uterosomes, and epididymal luminal fluid were included in the comparisons. WT and Pmca4-/- OVS were analyzed for the presence of known PMCA4 partners in sperm and their ability to interact with PMCA1, via co-immunoprecipitation. In vitro uptake of PMCA1 from OVS was analyzed in capacitated and uncapacitated sperm via quantitative western blot analysis, IF localization and flow cytometry. Caudal sperm were also assayed for uptake of tyrosine-phosphorylated proteins which were shown to be present in OVS. Finally, PMCA1 and PMCA4 in OVS and that delivered to sperm were assayed for enzymatic activity. PARTICIPANTS/MATERIALS, SETTING, METHODS Human fallopian tubes were flushed to recover luminal fluid which was processed for OVS via ultracentrifugation. Human OVS were negatively stained for transmission electron microscopy (TEM) and subjected to immunogold labeling, to detect PMCA4. Western analysis was used to detect HSC70 (an EV biomarker), PMCA1 and endothelial nitric oxide synthase (eNOS) which is a fertility-modulating protein delivered to human sperm by prostasomes. Oviducts of sexually mature female mice were sectioned after perfusion fixation for TEM tomography to obtain 3D information and to distinguish cross-sections of EVs from those of microvilli and cilia. Murine tissues, luminal fluids and EVs were assayed for PMCA1 (IF and western blot) or qRT-PCR. PMCA1 levels from western blots were quantified, using band densities and compared in WT and Pmca4-/- after induced estrus and in proestrus/estrus and metestrus/diestrus in cycling females. In vitro uptake of PMCA1 and tyrosine-phosphorylated proteins was quantified with flow cytometry and/or quantitative western blot. Ca2+-ATPase activity in OVS and sperm before and after PMCA1 and PMCA4 uptake was assayed, via the enzymatic hydrolysis rate of ATP. MAIN RESULTS AND THE ROLE OF CHANCE TEM revealed that human oviducts contain EVs (exosomal and microvesicular). These EVs contain PMCA4 (immunolabeling), eNOS and PMCA1 (western blot) in their cargo. TEM tomography showed the murine oviduct with EV-containing blebs which typify the apocrine pathway for EV biogenesis. Western blots revealed that during proestrus/estrus PMCA1 was significantly elevated in the oviductal luminal fluid (OLF) (P = 0.02) and in OVS (P = 0.03) of Pmca4-/-, compared to WT. Further, while PMCA1 levels did not fluctuate in OLF during the cycle in WT, they were significantly (P = 0.02) higher in proestrus/estrus than at metestrus/diestrus in Pmca4-/-. The elevated levels of PMCA1 in proestrus/estrus, which mimics PMCA4 in WT, is OLF/OVS-specific, and is not seen in oviductal tissues, uterosomes or epididymal luminal fluid of Pmca4-/-. However, qRT-PCR revealed significantly elevated levels of Pmca1 transcript in Pmca4-/- oviductal tissues, compared to WT. PMCA1 could be transferred from OVS to sperm and the levels were significantly higher for capacitated vs uncapacitated sperm, as assessed by flow cytometry (P = 0.001) after 3 h co-incubation, quantitative western blot (P < 0.05) and the frequency of immuno-labeled sperm (P < 0.001) after 30 min co-incubation. Tyrosine phosphorylated proteins were discovered in murine OVS and could be delivered to sperm after their co-incubation with OVS, as detected by western, immunofluorescence localization, and flow cytometry. PMCA1 and PMCA4 in OVS were shown to be enzymatically active and this activity increased in sperm after OVS interaction. LARGE SCALE DATA None. LIMITATIONS REASONS FOR CAUTION Although oviductal tissues of WT and Pmca4-/- showed no significant difference in PMCA1 levels, Pmca4-/- levels of OVS/OLF during proestrus/estrus were significantly higher than in WT. We have attributed this enrichment or upregulation of PMCA1 in Pmca4-/- partly to selective packaging in OVS to compensate for the lack of PMCA4. However, in the absence of a difference between WT and Pmca4-/- in the PMCA1 levels in oviductal tissues as a whole, we cannot rule out significantly higher PMCA1 expression in the oviductal epithelium that gives rise to the OVS as significantly higher Pmca1 transcripts were detected in Pmca4-/-. WIDER IMPLICATIONS OF THE FINDINGS Since OVS and fertility-modulating cargo components are conserved in humans, it suggests that murine OVS role in regulating the expression of proteins required for capacitation and fertility is also conserved. Secondly, OVS may explain some of the differences in in vivo and in vitro fertilization for mouse mutants, as seen in mice lacking the gene for FER which is the enzyme required for sperm protein tyrosine phosphorylation. Our observation that murine OVS carry and can modulate sperm protein tyrosine phosphorylation by delivering them to sperm provides an explanation for the in vivo fertility of Fer mutants, not seen in vitro. Finally, our findings have implications for infertility treatment and exosome therapeutics. STUDY FUNDING AND COMPETING INTEREST(S) The work was supported by National Institute of Health (RO3HD073523 and 5P20RR015588) grants to P.A.M.-D. There are no conflicts of interests.
Collapse
Affiliation(s)
- Pradeepthi Bathala
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Zeinab Fereshteh
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Kun Li
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.,Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences, Room 205 B, Building 3, 182 Tian Mu Shan Road, Hangzhou, Zhejiang 310013, China
| | - Amal A Al-Dossary
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.,Department of Biology, College of Medicine, University of Dammam (UOD), PO Box 2435, Dammam 31451, Saudi Arabia
| | - Deni S Galileo
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | | |
Collapse
|
24
|
Matamoros-Volante A, Moreno-Irusta A, Torres-Rodriguez P, Giojalas L, Gervasi MG, Visconti PE, Treviño CL. Semi-automatized segmentation method using image-based flow cytometry to study sperm physiology: the case of capacitation-induced tyrosine phosphorylation. Mol Hum Reprod 2019; 24:64-73. [PMID: 29186618 DOI: 10.1093/molehr/gax062] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/21/2017] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION Is image-based flow cytometry a useful tool to study intracellular events in human sperm such as protein tyrosine phosphorylation or signaling processes? SUMMARY ANSWER Image-based flow cytometry is a powerful tool to study intracellular events in a relevant number of sperm cells, which enables a robust statistical analysis providing spatial resolution in terms of the specific subcellular localization of the labeling. WHAT IS KNOWN ALREADY Sperm capacitation is required for fertilization. During this process, spermatozoa undergo numerous physiological changes, via activation of different signaling pathways, which are not completely understood. Classical approaches for studying sperm physiology include conventional microscopy, flow cytometry and Western blotting. These techniques present disadvantages for obtaining detailed subcellular information of signaling pathways in a relevant number of cells. This work describes a new semi-automatized analysis using image-based flow cytometry which enables the study, at the subcellular and population levels, of different sperm parameters associated with signaling. The increase in protein tyrosine phosphorylation during capacitation is presented as an example. STUDY DESIGN SIZE, DURATION Sperm cells were isolated from seminal plasma by the swim-up technique. We evaluated the intensity and distribution of protein tyrosine phosphorylation in sperm incubated in non-capacitation and capacitation-supporting media for 1 and 18 h under different experimental conditions. We used an antibody against FER kinase and pharmacological inhibitors in an attempt to identify the kinases involved in protein tyrosine phosphorylation during human sperm capacitation. PARTICIPANTS/MATERIALS, SETTING, METHODS Semen samples from normospermic donors were obtained by masturbation after 2-3 days of sexual abstinence. We used the innovative technique image-based flow cytometry and image analysis tools to segment individual images of spermatozoa. We evaluated and quantified the regions of sperm where protein tyrosine phosphorylation takes place at the subcellular level in a large number of cells. We also used immunocytochemistry and Western blot analysis. Independent experiments were performed with semen samples from seven different donors. MAIN RESULTS AND THE ROLE OF CHANCE Using image analysis tools, we developed a completely novel semi-automatic strategy useful for segmenting thousands of individual cell images obtained using image-based flow cytometry. Contrary to immunofluorescence which relies on the analysis of a limited sperm population and also on the observer, image-based flow cytometry allows for unbiased quantification and simultaneous localization of post-translational changes in an extended sperm population. Interestingly, important data can be independently analyzed by looking to the frame of interest. As an example, we evaluated the capacitation-associated increase in tyrosine phosphorylation in sperm incubated in non-capacitation and capacitation-supporting media for 1 and 18 h. As previously reported, protein tyrosine phosphorylation increases in a time-depending manner, but our method revealed that this increase occurs differentially among distinct sperm segments. FER kinase is reported to be the enzyme responsible for the increase in protein tyrosine phosphorylation in mouse sperm. Our Western blot analysis revealed for the first time the presence of this enzyme in human sperm. Using our segmentation strategy, we aimed to quantify the effect of pharmacological inhibition of FER kinase and found a marked reduction of protein tyrosine phosphorylation only in the flagellum, which corresponded to the physical localization of FER in human sperm. Our method provides an alternative strategy to study signaling markers associated with capacitation, such as protein tyrosine phosphorylation, in a fast and quantitative manner. LARGE SCALE DATA None. LIMITATIONS REASONS FOR CAUTION This is an in vitro study performed under controlled conditions. Chemical inhibitors are not completely specific for the intended target; the possibility of side effects cannot be discarded. WIDER IMPLICATIONS OF THE FINDINGS Our results demonstrate that the use of image-based flow cytometry is a very powerful tool to study sperm physiology. A large number of cells can be easily analyzed and information at the subcellular level can be obtained. As the segmentation process works with bright-field images, it can be extended to study expression of other proteins of interest using different antibodies or it can be used in living sperm to study intracellular parameters that can be followed using fluorescent dyes sensitive to the parameter of interest (e.g. pH, Ca2+). Therefore, this a versatile method that can be exploited to study several aspects of sperm physiology. STUDY FUNDING AND COMPETING INTEREST(S) This work was supported DGAPA (IN203116 to C. Treviño), Fronteras-CONACyT No. 71 and Eunice Kennedy Shriver National Institute of Child Health and Human Development NIH (RO1 HD38082) to P.E. Visconti and by a Lalor Foundation fellowship to M.G. Gervasi. A. Matamoros is a student of the Maestría en Ciencias Bioquímicas-UNAM program supported by CONACyT (416400) and DGAPA-UNAM. A. Moreno obtained a scholarship from Red MacroUniversidades and L. Giojalas obtained a schloarhip from CONICET and Universidad Nacional de Cordoba. The authors declare there are not conflicts of interest.
Collapse
Affiliation(s)
- Arturo Matamoros-Volante
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62250, Mexico
| | - Ayelen Moreno-Irusta
- Universidad Nacional de Córdoba (UNC), Facultad de Ciencias Exactas, Físicas y Naturales, Centro de Biología Celular y Molecular, Córdoba, Argentina
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas, Córdoba, Argentina
| | - Paulina Torres-Rodriguez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62250, Mexico
| | - Laura Giojalas
- Universidad Nacional de Córdoba (UNC), Facultad de Ciencias Exactas, Físicas y Naturales, Centro de Biología Celular y Molecular, Córdoba, Argentina
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas, Córdoba, Argentina
| | - María G Gervasi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA
| | - Claudia L Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62250, Mexico
| |
Collapse
|
25
|
Fereshteh Z, Bathala P, Galileo DS, Martin-DeLeon PA. Detection of extracellular vesicles in the mouse vaginal fluid: Their delivery of sperm proteins that stimulate capacitation and modulate fertility. J Cell Physiol 2018; 234:12745-12756. [PMID: 30536802 DOI: 10.1002/jcp.27894] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 11/13/2018] [Indexed: 12/31/2022]
Abstract
Extracellular vesicles (EVs) were isolated by ultracentrifugation of vaginal luminal fluid (VLF) from superovulated mice and identified for the first time using transmission electron microscopy. Characterized by size and biochemical markers (CD9 and HSC70), EVs were shown to be both microvesicular and exosomal and were dubbed as "Vaginosomes" (VGS). Vaginal cross-sections were analyzed to visualize EVs in situ: EVs were present in the lumen and also embedded between squamous epithelial and keratinized cells, consistent with their endogenous origin. Western blots detected Plasma membrane Ca2+ -ATPase 1 (PMCA1) and tyrosine-phosphorylated proteins in the VGS cargo and also in uterosomes. Flow cytometry revealed that following coincubation of caudal sperm and VLF for 30 min, the frequencies of cells with the highest Sperm adhesion molecule 1 (SPAM1), PMCA1/4, and PMCA1 levels increased 16.4-, 8.2-, and 27-fold, respectively; compared with control coincubated in phosphate buffered saline (PBS). Under identical conditions, sperm tyrosine-phosphorylated proteins were elevated ~3.3-fold, after VLF coincubation. Progesterone-induced acrosome reaction (AR) rates were significantly (p < 0.001) elevated in sperm coincubated with VGS for 10-30 min, compared with PBS. Sperm artificially deposited in the vaginas of superovulated females for these periods also showed significant (p < 0.01) increases in AR rates, compared with PBS. Thus in vitro and in vivo, sperm acquire from the vaginal environment factors that induce capacitation, explaining recent findings for their acrosomal status in the isthmus. Overall, VGS appear to deliver higher levels of proteins involved in preventing premature capacitation and AR than those promoting them. Our findings which have implications for humans open the possibility of new approaches to infertility treatment with exosome therapeutics.
Collapse
Affiliation(s)
- Zeinab Fereshteh
- Department of Biological Sciences, University of Delaware, Newark, Delaware.,Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - Pradeepthi Bathala
- Department of Biological Sciences, University of Delaware, Newark, Delaware
| | - Deni S Galileo
- Department of Biological Sciences, University of Delaware, Newark, Delaware
| | | |
Collapse
|
26
|
Alonso CAI, Osycka-Salut CE, Castellano L, Cesari A, Di Siervi N, Mutto A, Johannisson A, Morrell JM, Davio C, Perez-Martinez S. Extracellular cAMP activates molecular signalling pathways associated with sperm capacitation in bovines. Mol Hum Reprod 2018; 23:521-534. [PMID: 28521061 DOI: 10.1093/molehr/gax030] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/17/2017] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Is extracellular cAMP involved in the regulation of signalling pathways in bovine sperm capacitation? SUMMARY ANSWER Extracellular cAMP induces sperm capacitation through the activation of different signalling pathways that involve phospholipase C (PLC), PKC/ERK1-2 signalling and an increase in sperm Ca2+ levels, as well as soluble AC and cAMP/protein kinase A (PKA) signalling. WHAT IS KNOWN ALREADY In order to fertilize the oocyte, ejaculated spermatozoa must undergo a series of changes in the female reproductive tract, known as capacitation. This correlates with a number of membrane and metabolic modifications that include an increased influx of bicarbonate and Ca2+, activation of a soluble adenylyl cyclase (sAC) to produce cAMP, PKA activation, protein tyrosine phosphorylation and the development of hyperactivated motility. We previously reported that cAMP efflux by Multidrug Resistance Protein 4 (MRP4) occurs during sperm capacitation and the pharmacological blockade of this inhibits the process. Moreover, the supplementation of incubation media with cAMP abolishes the inhibition and leads to sperm capacitation, suggesting that extracellular cAMP regulates crucial signalling cascades involved in this process. STUDY DESIGN, SIZE, DURATION Bovine sperm were selected by the wool glass column method, and washed by centrifugation in BSA-Free Tyrode's Albumin Lactate Pyruvate (sp-TALP). Pellets were resuspended then diluted for each treatment. For in vitro capacitation, 10 to 15 × 106 SPZ/ml were incubated in 0.3% BSA sp-TALP at 38.5°C for 45 min under different experimental conditions. To evaluate the role of extracellular cAMP on different events associated with sperm capacitation, 10 nM cAMP was added to the incubation medium as well as different inhibitors of enzymes associated with signalling transduction pathways: U73122 (PLC inhibitor, 10 μM), Gö6983 (PKC inhibitor, 10 μM), PD98059 (ERK-1/2 inhibitor, 30 μM), H89 and KT (PKA inhibitors, 50 μM and 100 nM, respectively), KH7 (sAC inhibitor, 10 μM), BAPTA-AM (intracellular Ca2+ chelator, 50 μM), EGTA (10 μM) and Probenecid (MRPs general inhibitor, 500 μM). In addition, assays for binding to oviductal epithelial cells and IVF were carried out to test the effect of cAMP compared with other known capacitant agents such as heparin (60 μg/ml) and bicarbonate (40 mM). PARTICIPANTS/MATERIALS, SETTING, METHODS Straws of frozen bovine semen (20-25 × 106 spermatozoa/ml) were kindly provided by Las Lilas, CIALE and CIAVT Artificial Insemination Centers. The methods used in this work include western blot, immunohistochemistry, flow cytometry, computer-assisted semen analysis, live imaging of Ca2+ and fluorescence scanning. At least three independent assays with bull samples of proven fertility were carried. MAIN RESULTS AND THE ROLE OF CHANCE In the present study, we elucidate the molecular events induced by extracellular cAMP. Our results showed that external cAMP induces sperm capacitation, depending upon the action of PLC. Downstream, this enzyme increased ERK1-2 activation through PKC and elicited a rise in sperm Ca2+ levels (P < 0.01). Moreover, extracellular cAMP-induced capacitation also depended on the activity of sAC and PKA, and increased tyrosine phosphorylation, indicating that the nucleotide exerts a broad range of responses. In addition, extracellular cAMP-induced sperm hyperactivation and concomitantly increased the proportion of spermatozoa with high mitochondrial activity (P < 0.01). Finally, cAMP increased the in vitro fertilization rate compared to control conditions (P < 0.001). LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION This is an in vitro study performed with bovine cryopreserved spermatozoa. Studies in other species and with fresh samples are needed to extrapolate these data. WIDER IMPLICATIONS OF THE FINDINGS These findings strongly suggest an important role of extracellular cAMP in the regulation of the signalling pathways involved in the acquisition of bull sperm fertilizing capability. The data presented here indicate that not only a rise, but also a regulation of cAMP levels is necessary to ensure sperm fertilizing ability. Thus, exclusion of the nucleotide to the extracellular space might be essential to guarantee the achievement of a cAMP tone, needed for all capacitation-associated events to take place. Moreover, the ability of cAMP to trigger such broad and complex signalling events allows us to hypothesize that cAMP is a self-produced autocrine/paracrine factor, and supports the emerging paradigm that spermatozoa do not compete but, in fact, communicate with each other. A precise understanding of the functional competence of mammalian spermatozoa is essential to generate clinical advances in the treatment of infertility and the development of novel contraceptive strategies. STUDY FUNDING AND COMPETING INTEREST(S) This work was supported by Consejo Nacional de Investigaciones Científicas y Técnicas [PIP0 496 to S.P.-M.], Agencia Nacional de Promoción Científica y Tecológica [PICT 2012-1195 and PICT2014-2325 to S.P.-M., and PICT 2013-2050 to C.D.], Boehringer Ingelheim Funds, and the Swedish Farmers Foundation [SLF-H13300339 to J.M.]. The authors declare there are no conflicts of interests.
Collapse
Affiliation(s)
- Carlos Agustín I Alonso
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas Técnicas, Facultad de Medicina (CONICET-UBA), Paraguay 2155 (C1121ABG), Ciudad de Buenos Aires, Argentina
| | - Claudia E Osycka-Salut
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Instituto Tecnológico de Chascomús, Consejo Nacional de Investigaciones Científicas Técnicas (IIB/UNTECH-CONICET), Universidad Nacional de San Martín, Matheu 3910 (1650), Buenos Aires, Argentina
| | - Luciana Castellano
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas Técnicas, Facultad de Medicina (CONICET-UBA), Paraguay 2155 (C1121ABG), Ciudad de Buenos Aires, Argentina
| | - Andreína Cesari
- Instituto de Investigaciones Biológicas, Universidad Nacional de Mar del Plata (IIB-CONICET-UNMDP), Funes 3250 (7600), Mar del Plata, Argentina
| | - Nicolás Di Siervi
- Instituto de Investigaciones Farmacológicas, Consejo Nacional de Investigaciones Científicas Técnicas, Facultad de Farmacia y Bioquímica (ININFA-UBA-CONICET), Junín 954 (C1113AAD) Ciudad de Buenos Aires, Argentina
| | - Adrián Mutto
- Instituto de Investigaciones Biotecnológicas Dr. Rodolfo A. Ugalde, Instituto Tecnológico de Chascomús, Consejo Nacional de Investigaciones Científicas Técnicas (IIB/UNTECH-CONICET), Universidad Nacional de San Martín, Matheu 3910 (1650), Buenos Aires, Argentina
| | - Anders Johannisson
- Department of Clinical Sciences, Division of Reproduction, Swedish University of Agricultural Sciences (SE-750 07), Uppsala, Sweden
| | - Jane M Morrell
- Department of Clinical Sciences, Division of Reproduction, Swedish University of Agricultural Sciences (SE-750 07), Uppsala, Sweden
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas, Consejo Nacional de Investigaciones Científicas Técnicas, Facultad de Farmacia y Bioquímica (ININFA-UBA-CONICET), Junín 954 (C1113AAD) Ciudad de Buenos Aires, Argentina
| | - Silvina Perez-Martinez
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas Técnicas, Facultad de Medicina (CONICET-UBA), Paraguay 2155 (C1121ABG), Ciudad de Buenos Aires, Argentina
| |
Collapse
|
27
|
Breitbart H, Finkelstein M. Actin cytoskeleton and sperm function. Biochem Biophys Res Commun 2017; 506:372-377. [PMID: 29102633 DOI: 10.1016/j.bbrc.2017.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/01/2017] [Indexed: 11/17/2022]
Abstract
For the acquisition of the ability to fertilize the egg, mammalian spermatozoa should undergo a series of biochemical transformations in the female reproductive tract, collectively called capacitation. The capacitated sperm can undergo the acrosomal exocytosis process near or on the oocyte, which allows the spermatozoon to penetrate and fertilize it. One of the main processes in capacitation involves dynamic cytoskeletal remodeling particularly of actin. Actin polymerization occurs during sperm capacitation and the produced F-actin should be depolymerized prior to the acrosomal exocytosis. In the present review, we describe the mechanisms that regulate F-actin formation during sperm capacitation and the F-actin dispersion prior to the acrosomal exocytosis. During sperm capacitation, the actin severing proteins gelsolin and cofilin are inactive and they undergo activation prior to the acrosomal exocytosis.
Collapse
Affiliation(s)
- Haim Breitbart
- The Mina & Everard Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | | |
Collapse
|
28
|
Aitken RJ. Reactive oxygen species as mediators of sperm capacitation and pathological damage. Mol Reprod Dev 2017; 84:1039-1052. [PMID: 28749007 DOI: 10.1002/mrd.22871] [Citation(s) in RCA: 387] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/23/2017] [Indexed: 12/12/2022]
Abstract
Oxidative stress plays a major role in the life and death of mammalian spermatozoa. These gametes are professional generators of reactive oxygen species (ROS), which appear to derive from three potential sources: sperm mitochondria, cytosolic L-amino acid oxidases, and plasma membrane Nicotinamide adenine dinucleotide phosphate oxidases. The oxidative stress created via these sources appears to play a significant role in driving the physiological changes associated with sperm capacitation through the stimulation of a cyclic adenosine monophosphate/Protein kinase A phosphorylation cascade, including the activation of Extracellular signal regulated kinase-like proteins, massive up-regulation of tyrosine phosphorylation in the sperm tail, as well as the induction of sterol oxidation. When generated in excess, however, ROS can induce lipid peroxidation that, in turn, disrupts membrane characteristics that are critical for the maintenance of sperm function, including the capacity to fertilize an egg. Furthermore, the lipid aldehydes generated as a consequence of lipid peroxidation bind to proteins in the mitochondrial electron transport chain, triggering yet more ROS generation in a self-perpetuating cycle. The high levels of oxidative stress created as a result of this process ultimately damage the DNA in the sperm nucleus; indeed, DNA damage in the male germ line appears to be predominantly induced oxidatively, reflecting the vulnerability of these cells to such stress. Extensive evaluation of antioxidants that protect the spermatozoa against oxidative stress while permitting the normal reduction-oxidation regulation of sperm capacitation is therefore currently being undertaken, and has already proven efficacious in animal models.
Collapse
Affiliation(s)
- Robert J Aitken
- Discipline of Biological Sciences Priority Research Centre in Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
29
|
Lehti MS, Sironen A. Formation and function of sperm tail structures in association with sperm motility defects†. Biol Reprod 2017; 97:522-536. [DOI: 10.1093/biolre/iox096] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022] Open
|
30
|
Posttranslational Modifications in Spermatozoa and Effects on Male Fertility and Sperm Viability. ACTA ACUST UNITED AC 2017; 21:245-256. [DOI: 10.1089/omi.2016.0173] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
31
|
Calpain inhibition prevents flotillin re-ordering and Src family activation during capacitation. Cell Tissue Res 2017; 369:395-412. [DOI: 10.1007/s00441-017-2591-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 02/17/2017] [Indexed: 01/08/2023]
|
32
|
Harayama H, Minami K, Kishida K, Noda T. Protein biomarkers for male artificial insemination subfertility in bovine spermatozoa. Reprod Med Biol 2017; 16:89-98. [PMID: 29259456 PMCID: PMC5661804 DOI: 10.1002/rmb2.12021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/03/2017] [Indexed: 12/31/2022] Open
Abstract
Background Although artificial insemination (AI) technique is an established biotechnology for bovine reproduction, the results of AI (conception rates) have a tendency to decline gradually. To our annoyance, moreover, AI‐subfertile bulls have been occasionally found in the AI centers. To resolve these serious problems, it is necessary to control the sperm quality more strictly by the examinations of sperm molecules. Methods We reviewed a number of recent articles regarding potentials of bovine sperm proteins as the biomarkers for bull AI‐subfertility and also showed our unpublished supplemental data on the bull AI‐subfertility associated proteins. Main findings Bull AI‐subfertility is caused by the deficiency or dysfunctions of various molecules including regulatory proteins of ATP synthesis, acrosomal proteins, nuclear proteins, capacitation‐related proteins and seminal plasma proteins. Conclusion In order to control the bovine sperm quality more strictly by the molecular examinations, it is necessary to select suitable sperm protein biomarkers for the male reproductive problems which happen in the AI centers.
Collapse
Affiliation(s)
- Hiroshi Harayama
- Division of Animal Science Department of Bioresource Science Graduate School of Agricultural Science Kobe University Kobe Japan
| | - Kenta Minami
- Division of Animal Science Department of Bioresource Science Graduate School of Agricultural Science Kobe University Kobe Japan
| | - Kazumi Kishida
- Department of Obstetrics and Gynecology Shiga University of Medical Science Otsu Japan
| | - Taichi Noda
- Research Institute for Microbial Diseases Osaka University Suita Osaka Japan
| |
Collapse
|
33
|
Xing WR, Goodluck H, Zeng C, Mohan S. Role and mechanism of action of leucine-rich repeat kinase 1 in bone. Bone Res 2017; 5:17003. [PMID: 28326224 PMCID: PMC5348726 DOI: 10.1038/boneres.2017.3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/06/2016] [Accepted: 12/13/2016] [Indexed: 12/13/2022] Open
Abstract
Leucine-rich repeat kinase 1 (LRRK1) plays a critical role in regulating cytoskeletal organization, osteoclast activity, and bone resorption with little effect on bone formation parameters. Deficiency of Lrrk1 in mice causes a severe osteopetrosis in the metaphysis of the long bones and vertebrae bones, which makes LRRK1 an attractive alternative drug target for the treatment of osteoporosis and other high-turnover bone diseases. This review summarizes recent advances on the functions of the Lrrk1-related family members, Lrrk1 deficiency-induced skeletal phenotypes, LRRK1 structure–function, potential biological substrates and interacting proteins, and the mechanisms of LRRK1 action in osteoclasts.
Collapse
Affiliation(s)
- Weirong R Xing
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA; Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Helen Goodluck
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center , Loma Linda, CA, USA
| | - Canjun Zeng
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA; Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA; Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
34
|
Singh DK, Deshmukh RK, Narayanan PK, Shivaji S, Siva AB. SRC family kinases in hamster spermatozoa: evidence for the presence of LCK. Reproduction 2017; 153:655-669. [PMID: 28250239 DOI: 10.1530/rep-16-0591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/03/2017] [Accepted: 02/28/2017] [Indexed: 01/16/2023]
Abstract
Sperm capacitation is a prerequisite for successful fertilization. Increase in tyrosine phosphorylation is considered the hallmark of capacitation and attempts to understand its regulation are ongoing. In this regard, we attempted to study the role of SRC family kinases (SFKs) in the hamster sperm functions. Interestingly, we found the presence of the lymphocyte-specific protein tyrosine kinase, LCK, in mammalian spermatozoa and further characterized it in terms of its localization and function. LCK was found in spermatozoa of several species, and its transcript was identified in the hamster testis. Autophosphorylation of LCK at the Y394 residue increased as capacitation progressed, indicating an upregulation of LCK activity during capacitation. Inhibition of LCK (and perhaps the other SFKs) with the use of a specific inhibitor showed a significant decrease in protein tyrosine phosphorylation of several proteins, implying LCK/SFKs as key tyrosine kinase(s) regulating tyrosine phosphorylation during hamster sperm capacitation. Dihydrolipoamide dehydrogenase was identified as a substrate for LCK/SFK. LCK/SFKs inhibition significantly reduced the percentage fertilization (in vitro) but had no effect on sperm motility, hyperactivation and acrosome reaction. In summary, this is the first report on the presence of LCK, an SFK of hematopoietic lineage in spermatozoa besides being the first study on the role of SFKs in the spermatozoa of Syrian hamsters.
Collapse
Affiliation(s)
| | | | | | - Sisinthy Shivaji
- CSIR-Centre for Cellular and Molecular BiologyHyderabad 500007, India
| | | |
Collapse
|
35
|
Tsukamoto M, Hiyama E, Hirotani K, Gotoh T, Inai T, Iida H. Translocation of Tektin 3 to the equatorial segment of heads in bull spermatozoa exposed to dibutyryl cAMP and calyculin A. Mol Reprod Dev 2016; 84:30-43. [PMID: 27883267 DOI: 10.1002/mrd.22763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/17/2016] [Indexed: 11/06/2022]
Abstract
Tektins (TEKTs) are filamentous proteins associated with microtubules in cilia, flagella, basal bodies, and centrioles. Five TEKTs (TEKT1, -2, -3, -4, and -5) have been identified as components of mammalian sperm flagella. We previously reported that TKET1 and -3 are also present in the heads of rodent spermatozoa. The present study clearly demonstrates that TEKT2 is present at the acrosome cap whereas TEKT3 resides just beneath the plasma membrane of the post-acrosomal region of sperm heads in unactivated bull spermatozoa, and builds on the distributional differences of TEKT1, -2, and -3 on sperm heads. We also discovered that hyperactivation of bull spermatozoa by cell-permeable cAMP and calyculin A, a protein phosphatase inhibitor, promoted translocation of TEKT3 from the post-acrosomal region to the equatorial segment in sperm heads, and that TEKT3 accumulated at the equatorial segment is lost upon acrosome reaction. Thus, translocation of TEKT3 to the equatorial segment may be a capacitation- or hyperactivation-associated phenomenon in bull spermatozoa. Mol. Reprod. Dev. 84: 30-43, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mariko Tsukamoto
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Erina Hiyama
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Karen Hirotani
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takafumi Gotoh
- Kuju Agriculture Research Center, Kyushu University, Oita, Japan
| | - Tetsuichiro Inai
- Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Iida
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| |
Collapse
|
36
|
Bergeron A, Hébert A, Guillemette C, Laroche A, Poulin MP, Aragon JP, Leclerc P, Sullivan R, Blondin P, Vigneault C, Richard FJ. Papaverine-sensitive phosphodiesterase activity is measured in bovine spermatozoa. Andrology 2016; 5:169-179. [DOI: 10.1111/andr.12290] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 08/31/2016] [Accepted: 09/04/2016] [Indexed: 01/21/2023]
Affiliation(s)
- A. Bergeron
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département des Sciences Animales; Faculté des Sciences de L'agriculture et de L'alimentation; Université Laval; Québec QC Canada
| | - A. Hébert
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département des Sciences Animales; Faculté des Sciences de L'agriculture et de L'alimentation; Université Laval; Québec QC Canada
| | - C. Guillemette
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département des Sciences Animales; Faculté des Sciences de L'agriculture et de L'alimentation; Université Laval; Québec QC Canada
| | - A. Laroche
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département des Sciences Animales; Faculté des Sciences de L'agriculture et de L'alimentation; Université Laval; Québec QC Canada
| | - M.-P. Poulin
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département des Sciences Animales; Faculté des Sciences de L'agriculture et de L'alimentation; Université Laval; Québec QC Canada
| | - J. P. Aragon
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département des Sciences Animales; Faculté des Sciences de L'agriculture et de L'alimentation; Université Laval; Québec QC Canada
| | - P. Leclerc
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département Obstétrique, Gynécologie et Reproduction; Faculté de Médecine; Université Laval; Québec QC Canada
| | - R. Sullivan
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département Obstétrique, Gynécologie et Reproduction; Faculté de Médecine; Université Laval; Québec QC Canada
| | | | | | - F. J. Richard
- Centre de Recherche en Reproduction; Développement et Santé Intergénérationnelle; Université Laval; Québec QC Canada
- Département des Sciences Animales; Faculté des Sciences de L'agriculture et de L'alimentation; Université Laval; Québec QC Canada
| |
Collapse
|
37
|
c- Src and its role in cystic fibrosis. Eur J Cell Biol 2016; 95:401-413. [DOI: 10.1016/j.ejcb.2016.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 12/15/2022] Open
|
38
|
Goupil S, Maréchal L, El Hajj H, Tremblay MÈ, Richard FJ, Leclerc P. Identification and Localization of the Cyclic Nucleotide Phosphodiesterase 10A in Bovine Testis and Mature Spermatozoa. PLoS One 2016; 11:e0161035. [PMID: 27548062 PMCID: PMC4993467 DOI: 10.1371/journal.pone.0161035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/28/2016] [Indexed: 11/19/2022] Open
Abstract
In mammals, adenosine 3’, 5’-cyclic monophosphate (cAMP) is known to play highly important roles in sperm motility and acrosomal exocytosis. It is known to act through protein phosphorylation via PRKA and through the activation of guanine nucleotide exchange factors like EPAC. Sperm intracellular cAMP levels depend on the activity of adenylyl cyclases, mostly SACY, though transmembrane-containing adenylyl cyclases are also present, and on the activity of cyclic nucleotide phosphodiesterases (PDE) whose role is to degrade cAMP into 5’-AMP. The PDE superfamily is subdivided into 11 families (PDE1 to 11), which act on either cAMP or cGMP, or on both cAMP and cGMP although with different enzymatic properties. PDE10, which is more effective on cAMP than cGMP, has been known for almost 15 years and is mostly studied in the brain where it is associated with neurological disorders. Although a high level of PDE10A gene expression is observed in the testis, information on the identity of the isoforms or on the cell type that express the PDE10 protein is lacking. The objective of this study was to identify the PDE10A isoforms expressed in the testis and germ cells, and to determine the presence and localization of PDE10A in mature spermatozoa. As a sub-objective, since PDE10A transcript variants were reported strictly through analyses of bovine genomic sequence, we also wanted to determine the nucleotide and amino acid sequences by experimental evidence. Using RT-PCR, 5’- and 3’-RACE approaches we clearly show that PDE10A transcript variants X3 and X5 are expressed in bovine testis as well as in primary spermatocytes and spermatids. We also reveal using a combination of immunological techniques and proteomics analytical tools that the PDE10A isoform X4 is present in the area of the developing acrosome of spermatids and of the acrosome of mature spermatozoa.
Collapse
Affiliation(s)
- Serge Goupil
- Département d’obstétrique, gynécologie et reproduction, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - Loïze Maréchal
- Département d’obstétrique, gynécologie et reproduction, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - Hassan El Hajj
- Département de médecine moléculaire, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Département de médecine moléculaire, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - François J. Richard
- Département des sciences animales, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
| | - Pierre Leclerc
- Département d’obstétrique, gynécologie et reproduction, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
- * E-mail:
| |
Collapse
|
39
|
Abstract
Izumo1 is a testis-specific gene product, whose function is essential for sperm-egg fusion. Throughout its lifespan, Izumo1 is posttranslationally modified, being both N-linked glycosylated on its extracellular domain and phosphorylated on the intracellular C-terminal tail. Within the caput regions of the rat epididymis, two phosphorylation events have been documented. However, as sperm pass through the epididymis, this cytoplasmic portion of Izumo1 has been shown to contain up to seven phosphorylation sites. Remarkably, in the rat, in correlation with these events, Izumo1 undergoes sub-cellular re-location, moving from the head/tail regions of the spermatozoa, to a predominantly equatorial segment location once they have reached the caudal end of the epididymis.
Collapse
Affiliation(s)
| | | | - Mark A Baker
- Priority Research Centre in Reproductive Science, Discipline of Biological Sciences, Faculty of Science and IT, University of Newcastle, Callaghan, NSW, Australia,
| |
Collapse
|
40
|
Whitfield M, Pollet-Villard X, Levy R, Drevet JR, Saez F. Posttesticular sperm maturation, infertility, and hypercholesterolemia. Asian J Androl 2016; 17:742-8. [PMID: 26067871 PMCID: PMC4577583 DOI: 10.4103/1008-682x.155536] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cholesterol is a key molecule in the mammalian physiology of especial particular importance for the reproductive system as it is the common precursor for steroid hormone synthesis. Cholesterol is also a recognized modulator of sperm functions, not only at the level of gametogenesis. Cholesterol homeostasis regulation is crucial for posttesticular sperm maturation, and imbalanced cholesterol levels may particularly affect these posttesticular events. Metabolic lipid disorders (dyslipidemia) affect male fertility but are most of the time studied from the angle of endocrine/testicular consequences. This review will focus on the deleterious effects of a particular dyslipidemia, i.e., hypercholesterolemia, on posttesticular maturation of mammalian spermatozoa.
Collapse
Affiliation(s)
| | | | | | - Joël R Drevet
- Team "Mechanisms of post testicular infertility", Génétique Reproduction et Développement, UMR CNRS 6293, INSERM U1103, Clermont Université, 24 Avenue des Landais, BP80026, 63171 Aubière Cedex, France
| | | |
Collapse
|
41
|
Alvau A, Battistone MA, Gervasi MG, Navarrete FA, Xu X, Sánchez-Cárdenas C, De la Vega-Beltran JL, Da Ros VG, Greer PA, Darszon A, Krapf D, Salicioni AM, Cuasnicu PS, Visconti PE. The tyrosine kinase FER is responsible for the capacitation-associated increase in tyrosine phosphorylation in murine sperm. Development 2016; 143:2325-33. [PMID: 27226326 DOI: 10.1242/dev.136499] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/12/2016] [Indexed: 11/20/2022]
Abstract
Sperm capacitation is required for fertilization. At the molecular level, this process is associated with fast activation of protein kinase A. Downstream of this event, capacitating conditions lead to an increase in tyrosine phosphorylation. The identity of the tyrosine kinase(s) mediating this process has not been conclusively demonstrated. Recent experiments using stallion and human sperm have suggested a role for PYK2 based on the use of small molecule inhibitors directed against this kinase. However, crucially, loss-of-function experiments have not been reported. Here, we used both pharmacological inhibitors and genetically modified mice models to investigate the identity of the tyrosine kinase(s) mediating the increase in tyrosine phosphorylation in mouse sperm. Similar to stallion and human, PF431396 blocks the capacitation-associated increase in tyrosine phosphorylation. Yet, sperm from Pyk2(-/-) mice displayed a normal increase in tyrosine phosphorylation, implying that PYK2 is not responsible for this phosphorylation process. Here, we show that PF431396 can also inhibit FER, a tyrosine kinase known to be present in sperm. Sperm from mice targeted with a kinase-inactivating mutation in Fer failed to undergo capacitation-associated increases in tyrosine phosphorylation. Although these mice are fertile, their sperm displayed a reduced ability to fertilize metaphase II-arrested eggs in vitro.
Collapse
Affiliation(s)
- Antonio Alvau
- Department of Veterinary and Animal Science, Integrated Sciences Building, University of Massachusetts, Amherst, MA 01003, USA
| | | | - Maria Gracia Gervasi
- Department of Veterinary and Animal Science, Integrated Sciences Building, University of Massachusetts, Amherst, MA 01003, USA
| | - Felipe A Navarrete
- Department of Veterinary and Animal Science, Integrated Sciences Building, University of Massachusetts, Amherst, MA 01003, USA
| | - Xinran Xu
- Department of Electrical and Computer Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80521, USA
| | - Claudia Sánchez-Cárdenas
- Departamento de Genética del Desarrollo y Fisiología Molecular, IBT-UNAM, Cuernavaca 62210, México
| | | | - Vanina G Da Ros
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires C1428ADN, Argentina
| | - Peter A Greer
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada, K7L 3N6
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, IBT-UNAM, Cuernavaca 62210, México
| | - Diego Krapf
- Department of Electrical and Computer Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80521, USA
| | - Ana Maria Salicioni
- Department of Veterinary and Animal Science, Integrated Sciences Building, University of Massachusetts, Amherst, MA 01003, USA
| | - Patricia S Cuasnicu
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires C1428ADN, Argentina
| | - Pablo E Visconti
- Department of Veterinary and Animal Science, Integrated Sciences Building, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
42
|
Stanger SJ, Law EA, Jamsai D, O'Bryan MK, Nixon B, McLaughlin EA, Aitken RJ, Roman SD. A novel germ cell protein, SPIF (sperm PKA interacting factor), is essential for the formation of a PKA/TCP11 complex that undergoes conformational and phosphorylation changes upon capacitation. FASEB J 2016; 30:2777-91. [DOI: 10.1096/fj.201500136r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/12/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Simone J. Stanger
- Centre for Chemical Biology, University of NewcastleCallaghanNew South WalesAustralia
- Priority Research Centre for Reproductive ScienceUniversity of NewcastleCallaghanNew South WalesAustralia
- School of Environmental and Life SciencesUniversity of NewcastleCallaghanNew South WalesAustralia
- Australian Research Council Centre of Excellence in Biotechnology and DevelopmentUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Estelle A. Law
- School of Environmental and Life SciencesUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Duangporn Jamsai
- Australian Research Council Centre of Excellence in Biotechnology and DevelopmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Department of Anatomy and Developmental BiologyMonash UniversityMelbourneVictoriaAustralia
| | - Moira K. O'Bryan
- Australian Research Council Centre of Excellence in Biotechnology and DevelopmentUniversity of NewcastleCallaghanNew South WalesAustralia
- Department of Anatomy and Developmental BiologyMonash UniversityMelbourneVictoriaAustralia
| | - Brett Nixon
- Centre for Chemical Biology, University of NewcastleCallaghanNew South WalesAustralia
- Priority Research Centre for Reproductive ScienceUniversity of NewcastleCallaghanNew South WalesAustralia
- School of Environmental and Life SciencesUniversity of NewcastleCallaghanNew South WalesAustralia
- Australian Research Council Centre of Excellence in Biotechnology and DevelopmentUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Eileen A. McLaughlin
- Centre for Chemical Biology, University of NewcastleCallaghanNew South WalesAustralia
- Priority Research Centre for Reproductive ScienceUniversity of NewcastleCallaghanNew South WalesAustralia
- School of Environmental and Life SciencesUniversity of NewcastleCallaghanNew South WalesAustralia
- Australian Research Council Centre of Excellence in Biotechnology and DevelopmentUniversity of NewcastleCallaghanNew South WalesAustralia
| | - R. John Aitken
- Priority Research Centre for Reproductive ScienceUniversity of NewcastleCallaghanNew South WalesAustralia
- Australian Research Council Centre of Excellence in Biotechnology and DevelopmentUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Shaun D. Roman
- Centre for Chemical Biology, University of NewcastleCallaghanNew South WalesAustralia
- Priority Research Centre for Reproductive ScienceUniversity of NewcastleCallaghanNew South WalesAustralia
- School of Environmental and Life SciencesUniversity of NewcastleCallaghanNew South WalesAustralia
- Australian Research Council Centre of Excellence in Biotechnology and DevelopmentUniversity of NewcastleCallaghanNew South WalesAustralia
| |
Collapse
|
43
|
Kerns K, Morales P, Sutovsky P. Regulation of Sperm Capacitation by the 26S Proteasome: An Emerging New Paradigm in Spermatology. Biol Reprod 2016; 94:117. [PMID: 27053366 DOI: 10.1095/biolreprod.115.136622] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/24/2016] [Indexed: 12/12/2022] Open
Abstract
The ubiquitin proteasome system (UPS) participates in many biological processes ranging from cell cycle and antigen processing to cellular defense and signaling. Work of the last decade has made it evident that the UPS is involved in many sperm-related processes leading up to and as part of fertilization. The current knowledge of UPS involvement and changes during sperm capacitation are reviewed together with a list of known proteasome-associated sperm proteins and a discussion of the relationships between these proteins and the proteasome. Proteasomal inhibitors such as MG-132 and epoxomicin significantly alter capacitation and prevent acrosome reaction. The 26S proteasome degrades AKAP3, an A-kinase anchoring protein, partially regulating the release of protein-kinase A (PKA), a vital component necessary for the steps leading up to capacitation. Further, changes occur in 20S core subunit localization and abundance throughout capacitation. Proteasome-interacting valosine-containing protein (VCP) undergoes tyrosine phosphorylation; however, its physiological roles in capacitation and fertilization remain unknown. The E1-type ubiquitin-activating enzyme (UBA1) inhibitor PYR-41 also alters acrosomal membrane remodeling during capacitation. Furthermore, after capacitation, the acrosomal proteasomes facilitate the degradation of zona pellucida glycoproteins leading up to fertilization. Methods to modulate the sperm proteasome activity during sperm storage and capacitation may translate to increased reproductive efficiency in livestock animals. Human male infertility diagnostics may benefit from incorporation of research outcomes built upon relationships between UPS and capacitation. Altogether, the studies reviewed here support the involvement of UPS in sperm capacitation and present opportunities for new discoveries.
Collapse
Affiliation(s)
- Karl Kerns
- Division of Animal Sciences, University of Missouri, Columbia, Missouri
| | - Patricio Morales
- Department of Biomedicine, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile Instituto Antofagasta, Antofagasta, Chile
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, Missouri Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri
| |
Collapse
|
44
|
Breitbart H, Finkelstein M. Regulation of Sperm Capacitation and the Acrosome Reaction by PIP 2 and Actin Modulation. Asian J Androl 2016; 17:597-600. [PMID: 25966627 PMCID: PMC4492050 DOI: 10.4103/1008-682x.154305] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Actin polymerization and development of hyperactivated (HA) motility are two processes that take place during sperm capacitation. Actin polymerization occurs during capacitation and prior to the acrosome reaction, fast F-actin breakdown takes place. The increase in F-actin during capacitation depends upon inactivation of the actin severing protein, gelsolin, by its binding to phosphatydilinositol-4, 5-bisphosphate (PIP 2 ) and its phosphorylation on tyrosine-438 by Src. Activation of gelsolin following its release from PIP 2 is known to cause F-actin breakdown and inhibition of sperm motility, which can be restored by adding PIP 2 to the cells. Reduction of PIP 2 synthesis inhibits actin polymerization and motility, while increasing PIP 2 synthesis enhances these activities. Furthermore, sperm demonstrating low motility contained low levels of PIP 2 and F-actin. During capacitation there was an increase in PIP 2 and F-actin levels in the sperm head and a decrease in the tail. In spermatozoa with high motility, gelsolin was mainly localized to the sperm head before capacitation, whereas in low motility sperm, most of the gelsolin was localized to the tail before capacitation and translocated to the head during capacitation. We also showed that phosphorylation of gelsolin on tyrosine-438 depends upon its binding to PIP 2 . Stimulation of phospholipase C, by Ca 2 + -ionophore or by activating the epidermal-growth-factor-receptor, inhibits tyrosine phosphorylation of gelsolin and enhances enzyme activity. In conclusion, these data indicate that the increase of PIP 2 and/or F-actin in the head during capacitation enhances gelsolin translocation to the head. As a result, the decrease of gelsolin in the tail allows the maintenance of high levels of F-actin in this structure, which is essential for the development of HA motility.
Collapse
Affiliation(s)
- Haim Breitbart
- The Mina and Everard Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | |
Collapse
|
45
|
Tyrosine kinase-mediated axial motility of basal cells revealed by intravital imaging. Nat Commun 2016; 7:10666. [PMID: 26868824 PMCID: PMC4754344 DOI: 10.1038/ncomms10666] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/08/2016] [Indexed: 12/28/2022] Open
Abstract
Epithelial cells are generally considered to be static relative to their neighbours. Basal cells in pseudostratified epithelia display a single long cytoplasmic process that can cross the tight junction barrier to reach the lumen. Using in vivo microscopy to visualize the epididymis, a model system for the study of pseudostratified epithelia, we report here the surprising discovery that these basal cell projections--which we call axiopodia--periodically extend and retract over time. We found that axiopodia extensions and retractions follow an oscillatory pattern. This movement, which we refer to as periodic axial motility (PAM), is controlled by c-Src and MEK1/2-ERK1/2. Therapeutic inhibition of tyrosine kinase activity induces a retraction of these projections. Such unexpected cell motility may reflect a novel mechanism by which specialized epithelial cells sample the luminal environment.
Collapse
|
46
|
Hurtado de Llera A, Martin-Hidalgo D, Gil M, Garcia-Marin L, Bragado M. New insights into transduction pathways that regulate boar sperm function. Theriogenology 2016; 85:12-20. [DOI: 10.1016/j.theriogenology.2015.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 10/23/2022]
|
47
|
Agarwal A, Bertolla RP, Samanta L. Sperm proteomics: potential impact on male infertility treatment. Expert Rev Proteomics 2016; 13:285-296. [PMID: 26853600 DOI: 10.1586/14789450.2016.1151357] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Spermatozoa are unique cells that have highly compact DNA, motility (and hypermotility) patterns, a specific morphology, localized mitochondria and an apical acrosome. They are the end product of a dynamic process termed spermatogenesis. Sperm are therefore produced with specific proteins in order to effect different traits, such as the presence of cysteine-rich protamines in DNA, which effectively compacts DNA. Moreover, specific proteins are transferred during epididymal maturation and after ejaculation in order to render sperm capable of undergoing post-ejaculatory alterations, generally termed capacitation, which confers capacity to fertilize a mature oocyte. In addition, sperm exhibit several post-translational modifications, which are fundamental to their function, such as SUMOylation and ubiquitination. Discussed in this review is the current knowledge of the sperm proteome in terms of its composition and the function that these proteins determine, as well as their post-translational modifications and how these alter sperm functional integrity. Studies are emphasized that focus on shotgun proteomics--untargeted determination of the protein constituent of a cell in a given biological condition--and technologies currently applied toward that end are reviewed.
Collapse
Affiliation(s)
- Ashok Agarwal
- a American Center for Reproductive Medicine, Department of Urology , Cleveland Clinic , Cleveland , OH , USA
| | - Ricardo Pimenta Bertolla
- b Department of Surgery, Division of Urology, Human Reproduction Section , Federal University of São Paulo , São Paulo , Brazil
| | - Luna Samanta
- c Department of Zoology, School of Life Sciences , Ravenshaw University , Cuttack , India
| |
Collapse
|
48
|
Sperm Capacitation and Acrosome Reaction in Mammalian Sperm. SPERM ACROSOME BIOGENESIS AND FUNCTION DURING FERTILIZATION 2016; 220:93-106. [DOI: 10.1007/978-3-319-30567-7_5] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
49
|
Lackey BR, Gray SL. Second messengers, steroids and signaling cascades: Crosstalk in sperm development and function. Gen Comp Endocrinol 2015; 224:294-302. [PMID: 26188217 DOI: 10.1016/j.ygcen.2015.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/08/2015] [Accepted: 07/14/2015] [Indexed: 10/23/2022]
Abstract
Signaling cascades control numerous aspects of sperm physiology, ranging from creation to fertilization. Novel aspects of several kinases and their influence on sperm development will be discussed in the first section and cover proliferation, chromatin remodeling and morphology. In the second section, protein kinases (A, B and C) that affect sperm function and their regulation by second messengers, cyclic-AMP and phosphoinositides, as well as steroids will be featured. Key areas of integration will be presented on the topics of sperm motility, capacitation, acrosome reaction and fertilization.
Collapse
Affiliation(s)
- B R Lackey
- Endocrine Physiology Laboratory, AVS Department, Clemson University, Clemson, SC, USA
| | - S L Gray
- Endocrine Physiology Laboratory, AVS Department, Clemson University, Clemson, SC, USA.
| |
Collapse
|
50
|
Wang W, Guo C, Zhu P, Lu J, Li W, Liu C, Xie H, Myatt L, Chen ZJ, Sun K. Phosphorylation of STAT3 mediates the induction of cyclooxygenase-2 by cortisol in the human amnion at parturition. Sci Signal 2015; 8:ra106. [PMID: 26508788 DOI: 10.1126/scisignal.aac6151] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The induction of cyclooxygenase-2 (COX-2) and subsequent production of prostaglandin E2 (PGE2) by cortisol in the amnion contrast with the effect of cortisol on most other tissues, but this proinflammatory effect of cortisol may be a key event in human parturition (labor). We evaluated the underlying mechanism activated by cortisol in primary human amnion fibroblasts. Exposure of the amnion fibroblasts to cortisol led to the activation of the cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) pathway, which induced the phosphorylation of the kinase SRC and STAT3 (signal transducer and activator of transcription 3). STAT3 interacted with the glucocorticoid receptor (GR) and the transcription factor CREB-1 (cAMP response element-binding protein 1) at the promoter of the gene encoding COX-2, which promoted the production of the secreted prostaglandin PGE2. PGE2 activates the prostaglandin receptors EP2 and EP4, which stimulate cAMP-PKA signaling. Thus, cortisol reinforced the activation of cAMP-PKA signaling through an SRC-STAT3-COX-2-PGE2-mediated feedback loop. Inhibiting STAT3, SRC, or the cAMP-PKA pathway attenuated the cortisol-stimulated induction of COX-2 and PGE2 production in amnion fibroblasts. In human amnion tissue, the amount of phosphorylated STAT3 correlated positively with that of cortisol, COX-2, and PGE2, and all were more abundant in tissue obtained after active labor than in tissue obtained from cesarean surgeries in the absence of labor. These results indicated that the coordinated recruitment of STAT3, CREB-1, and GR to the promoter of the gene encoding COX-2 contributes to the feed-forward induction of COX-2 activity and prostaglandin synthesis in the amnion during parturition.
Collapse
Affiliation(s)
- Wangsheng Wang
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P. R. China. Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P. R. China
| | - Chunming Guo
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ping Zhu
- Department of Obstetrics and Gynecology, No. 401 Hospital, Qingdao 266100, P. R. China
| | - Jiangwen Lu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P. R. China. Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P. R. China
| | - Wenjiao Li
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P. R. China. Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P. R. China
| | - Chao Liu
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P. R. China. Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P. R. China
| | - Huiliang Xie
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P. R. China
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P. R. China. Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P. R. China
| | - Kang Sun
- Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, P. R. China. Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, P. R. China.
| |
Collapse
|