1
|
Ogneva IV, Zhdankina YS, Gogichaeva KK, Malkov AA, Biryukov NS. The Motility of Mouse Spermatozoa Changes Differentially After 30-Minute Exposure Under Simulating Weightlessness and Hypergravity. Int J Mol Sci 2024; 25:13561. [PMID: 39769324 PMCID: PMC11678010 DOI: 10.3390/ijms252413561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
Research into the mechanisms by which gravity influences spermatozoa has implications for maintaining the species in deep space exploration and may provide new approaches to reproductive technologies on Earth. Changes in the speed of mouse spermatozoa after 30 min exposure to simulated weightlessness (by 3D-clinostat) and 2 g hypergravity (by centrifugation) were studied using inhibitory analysis. Simulated microgravity after 30 min led to an increase in the speed of spermatozoa and against the background of an increase in the relative calcium content in the cytoplasm. This effect was prevented by the introduction of 6-(dimethylamino) purine, wortmannin, and calyculin A. Hypergravity led to a decrease in the speed of spermatozoa movement, which was prevented by sodium orthovanadate and calyculin A. At the same time, under microgravity conditions, there was a redistribution of proteins forming microfilament bundles between the membrane and cytoplasmic compartments and under hypergravity conditions-proteins forming networks. The obtained results indicate that even a short exposure of spermatozoa to altered gravity leads to the launch of mechanotransduction pathways in them and a change in motility.
Collapse
Affiliation(s)
- Irina V. Ogneva
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia; (Y.S.Z.); (K.K.G.); (A.A.M.); (N.S.B.)
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, 119991 Moscow, Russia
- Yu.A. Gagarin Research and Test Cosmonaut Training Center, 141160 Star City, Moscow Region, Russia
| | - Yulia S. Zhdankina
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia; (Y.S.Z.); (K.K.G.); (A.A.M.); (N.S.B.)
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, 119991 Moscow, Russia
| | - Ksenia K. Gogichaeva
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia; (Y.S.Z.); (K.K.G.); (A.A.M.); (N.S.B.)
| | - Artyom A. Malkov
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia; (Y.S.Z.); (K.K.G.); (A.A.M.); (N.S.B.)
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, 119991 Moscow, Russia
| | - Nikolay S. Biryukov
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia; (Y.S.Z.); (K.K.G.); (A.A.M.); (N.S.B.)
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, 119991 Moscow, Russia
| |
Collapse
|
2
|
Wang W, Guo L, Jiang B, Yan B, Li Y, Ye X, Yang Y, Liu S, Shao Z, Diao H. Role of the Glycogen Synthase Kinase 3-Cyclic AMP/Protein Kinase A in the Immobilization of Human Sperm by Tideglusib. Reprod Sci 2023; 30:1281-1290. [PMID: 36207578 DOI: 10.1007/s43032-022-01086-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/15/2022] [Indexed: 10/10/2022]
Abstract
Tideglusib is considered to be a promising alternative to nonyl alcohol-9 contraceptives. Previous studies have demonstrated that the rapid spermicidal effect of tideglusib at a high concentration (≥ 10 μM) may occur through detergent-like activity; however, the effect of low concentrations of tideglusib (< 5 μM) on sperm is unknown. We explored the intracellular mechanism of tideglusib (< 5 μM) on the immobilization of human sperm by exploring related signaling pathways in human sperm. After treatment with tideglusib (1.25 μM) for 2 h, sperm motility rate decreased to 0, while sperm membrane integrity rate was 70%. Protein tyrosine phosphorylation level and intracellular cyclic adenosine 3,5-monophosphate (cAMP) concentration decreased significantly compared to those in the control group. Isobutylmethylxanthine and 8-Bromo-cAMP relieved the inhibition of spermatozoa tyrosine phosphorylation, while tyrosine phosphorylation of sperm protein in the H89 and CALP1 treatment groups was significantly inhibited, and there was no difference in the tideglusib treatment group. H-89 and CALP1 reduced the level of serine phosphorylation of GSK-3α/β (Ser21/9), while its level was enhanced by IBMX and 8-Bromo-cAMP. Our results show the existence of the GSK3-cAMP/PKA regulatory loop in human sperm, which may mediate the immobilization effect of tideglusib at low of concentrations (e.g., 1.25 μM) on sperm motility.
Collapse
Affiliation(s)
- Weiwei Wang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lina Guo
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Bingbing Jiang
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Bin Yan
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuhua Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xin Ye
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Yiting Yang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhiyu Shao
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China.
| | - Hua Diao
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Karanwal S, Pal A, Chera JS, Batra V, Kumaresan A, Datta TK, Kumar R. Identification of protein candidates in spermatozoa of water buffalo ( Bubalus bubalis) bulls helps in predicting their fertility status. Front Cell Dev Biol 2023; 11:1119220. [PMID: 36891514 PMCID: PMC9986327 DOI: 10.3389/fcell.2023.1119220] [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: 12/08/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
The water buffalo (Bubalus bubalis) is an indispensable part of the Indian dairy sector and in several instances, the farmers incur economic losses due to failed pregnancy after artificial insemination (AI). One of the key factors for the failure of conception is the use of semen from the bulls of low fertilizing potential and hence, it becomes important to predict the fertility status before performing AI. In this study, the global proteomic profile of high fertile (HF) and low fertile (LF) buffalo bull spermatozoa was established using a high-throughput LC-MS/MS technique. A total of 1,385 proteins (≥1 high-quality PSM/s, ≥1 unique peptides, p < 0.05, FDR < 0.01) were identified out of which, 1,002 were common between both the HF and LF groups while 288 and 95 proteins were unique to HF and LF groups respectively. We observed 211 and 342 proteins were significantly high (log Fc ≥ 2) and low abundant (log Fc ≤ 0.5) in HF spermatozoa (p < 0.05). Gene ontology analysis revealed that the fertility associated high abundant proteins in HF were involved in spermatogenesis, sperm motility, acrosome integrity, zona pellucida binding and other associated sperm functions. Besides this, the low abundant proteins in HF were involved in glycolysis, fatty acid degradation and inflammation. Furthermore, fertility related differentially abundant proteins (DAPs) on sperm viz., AKAP3, Sp17, and DLD were validated through Western blotting and immunocytochemistry which was in coherence with the LC-MS/MS data. The DAPs identified in this study may be used as potential protein candidates for predicting fertility in buffaloes. Our findings provide an opportunity in mitigating the economic losses that farmers incur due to male infertility.
Collapse
Affiliation(s)
- Seema Karanwal
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Ankit Pal
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Jatinder Singh Chera
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Vipul Batra
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Arumugam Kumaresan
- Theriogenelogy Laboratory, SRS of National Dairy Research Institute, Bengaluru, India
| | - Tirtha K Datta
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Rakesh Kumar
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| |
Collapse
|
4
|
Cordero-Martínez J, Jimenez-Gutierrez GE, Aguirre-Alvarado C, Alacántara-Farfán V, Chamorro-Cevallos G, Roa-Espitia AL, Hernández-González EO, Rodríguez-Páez L. Participation of signaling proteins in sperm hyperactivation. Syst Biol Reprod Med 2022; 68:315-330. [DOI: 10.1080/19396368.2022.2122761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Joaquín Cordero-Martínez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | | | - Charmina Aguirre-Alvarado
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Unidad de Investigación Médica en Inmunología e Infectología Centro Médico Nacional La Raza, IMSS, Ciudad de México, Mexico
| | - Verónica Alacántara-Farfán
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Germán Chamorro-Cevallos
- Laboratorio de Toxicología Preclínica Departamento de Farmacia Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ana L. Roa-Espitia
- Departamento de Biología Celular Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional, México City, Mexico
| | - Enrique O. Hernández-González
- Departamento de Biología Celular Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional, México City, Mexico
| | - Lorena Rodríguez-Páez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
5
|
Swain DK, Sharma P, Shah N, Sethi M, Mahajan A, Gupta S, Mishra AK, Yadav S. Introduction to the pathways involved in the activation and regulation of sperm motility: A review of the relevance of ion channels. Anim Reprod Sci 2022; 246:107052. [PMID: 35987804 DOI: 10.1016/j.anireprosci.2022.107052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022]
Abstract
To participate in sperm-oocyte fusion, spermatozoa need to be motile. In the testes, spermatozoa are immotile, although these gametes acquire the capacity for motility during the transit through the epididymis. During the period of epididymal transport from the male genital tract to the female genital tract, spermatozoa exhibit various types of motility that are regulated by complex signalling and communication mechanisms. Because motility is very dynamic, it can be affected by small changes in the external or internal environment of spermatozoa within a very short time. This indicates that regulatory membrane proteins, known as sperm ion channels, are involved in the regulation of sperm motility. Research results from studies, where there was use of electrophysiological, pharmacological, molecular and knock-out approaches, indicate ion channels are possibly involved in the regulation of sperm membrane polarisation, intracellular pH, motility, energy homeostasis, membrane integrity, capacitation, hyperactivity, acrosome reaction and fertilisation processes. In this review, there is summarisation of the key functions that ion channels have in the regulation, initiation, maintenance, and modulation of sperm motility. In addition, in this review there is highlighting of novel insights about the pathways of ion channels that are activated in spermatozoa while these gametes are located in the oviduct leading to the fertilisation capacity of these cells.
Collapse
Affiliation(s)
- Dilip Kumar Swain
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India.
| | - Pratishtha Sharma
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India
| | - Nadeem Shah
- Department of Veterinary Gynaecology and Obstetrics, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Manisha Sethi
- Department of Veterinary Gynaecology and Obstetrics, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Abhishek Mahajan
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India
| | - Shashikant Gupta
- Animal Reproduction Division, ICAR-Indian Veterinary Research Institute, Izzatnagar, Bareilly 243122, Uttar Pradesh, India
| | | | - Sarvajeet Yadav
- Sperm Signaling Laboratory, Department of Veterinary Physiology, College of Veterinary Science & Animal Husbandry, U.P. Pandit Deendayal Upadhayaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan, Mathura 281001, Uttar Pradesh, India
| |
Collapse
|
6
|
Qu X, Han Y, Chen X, Lv Y, Zhang Y, Cao L, Zhang J, Jin Y. Inhibition of 26 S Proteasome Enhances AKAP3-mediated cAMP-PKA Signaling During Boar Sperm Capacitation. Anim Reprod Sci 2022; 247:107079. [DOI: 10.1016/j.anireprosci.2022.107079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/20/2022] [Accepted: 09/21/2022] [Indexed: 11/01/2022]
|
7
|
Vanderkooi SC, Zhao Y, Lima PDA, Kan FWK. Recombinant human OVGP1 increases intracellular calcium and further potentiates the effects of progesterone on human sperm. J Assist Reprod Genet 2022; 39:2287-2301. [PMID: 35972586 PMCID: PMC9596666 DOI: 10.1007/s10815-022-02591-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose To investigate the effects of recombinant human oviduct–specific glycoprotein (rHuOVGP1) alone and in combination with progesterone (P4) on intracellular Ca2+ concentration [Ca2+]i and to investigate if rHuOVGP1 in combination with P4 can further enhance tyrosine phosphorylation (pY) of sperm proteins during human sperm capacitation. Methods Fluorometric flow cytometry was performed to examine the effects of rHuOVGP1 on [Ca2+]i in human sperm during capacitation. Confocal microscopy was used in conjunction with live cell imaging to analyze the influence of rHuOVGP1 and P4 on [Ca2+]i in the sperm tail and to examine the involvement of CatSper channels in their effect on [Ca2+]i. Western blot analysis was performed to assess the protein levels of p105, a major tyrosine-phosphorylated sperm protein. Results rHuOVGP1 increases [Ca2+]i in human sperm at the beginning of capacitation and further increases and sustains the level of [Ca2+]i in the sperm tail following the addition of P4. Inhibition of CatSper channels impedes the effects of rHuOVGP1 on [Ca2+]i in the sperm tail. P4 alone can increase pY of a major human sperm protein, p105, yet yields a further increase when used in combination with rHuOVGP1. Conclusion The present study revealed that rHuOVGP1 may work with P4 to upregulate [Ca2+]i at the beginning of capacitation in part through CatSper channels which, in turn, leads to the downstream event of pY of sperm proteins and enhancement of sperm capacitation. Supplementary information The online version contains supplementary material available at 10.1007/s10815-022-02591-0.
Collapse
Affiliation(s)
- Sydney C Vanderkooi
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Yuewen Zhao
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
- Yale Fertility Center, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University, Orange, Connecticut, 06477, USA
| | - Patricia D A Lima
- Queen's CardioPulmonary Unit, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Frederick W K Kan
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada.
| |
Collapse
|
8
|
Sperm of Fruit Fly Drosophila melanogaster under Space Flight. Int J Mol Sci 2022; 23:ijms23147498. [PMID: 35886847 PMCID: PMC9319090 DOI: 10.3390/ijms23147498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
Studies of reproductive function under long-term space flight conditions are of interest in planning the exploration of deep space. Motility, including the use of various inhibitors, cellular respiration, and the content of cytoskeletal proteins were studied, assessing the level of expression of the corresponding genes in spermatozoa of Drosophila melanogaster, which were in space flight conditions for 12 days. The experiment was carried out twice on board the Russian Segment of the International Space Station. Sperm motility speed after space flight, and subsequently 16 h after landing, is reduced relative to the control by 20% (p < 0.05). In comparison with the simulation experiment, we showed that this occurs as a result of the action of overloads and readaptation to the Earth’s gravity. At the same time, cellular respiration, the content of proteins of the respiratory chain, and the expression of their genes do not change. We used kinase inhibitor 6-(dimethylamino)purine (6-DMAP) and phosphatase inhibitors; 6-DMAP restored the reduced the speed of spermatozoa in the flight group to that of the control. These results can be useful in developing a strategy for protecting reproductive health during the development of other bodies in the solar system.
Collapse
|
9
|
Rafaee A, Kashani-Amin E, Meybodi AM, Ebrahim-Habibi A, Sabbaghian M. Structural modeling of human AKAP3 protein and in silico analysis of single nucleotide polymorphisms associated with sperm motility. Sci Rep 2022; 12:3656. [PMID: 35256641 PMCID: PMC8901789 DOI: 10.1038/s41598-022-07513-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/28/2022] [Indexed: 11/20/2022] Open
Abstract
AKAP3 is a member of the A-kinase anchoring proteins and it is a constituent of the sperm fibrous sheath. AKAP3 is needed for the formation of sperm flagellum structure, sperm motility, and male fertility. This study aims to model the AKAP3 tertiary structure and identify the probable impact of four mutations characterized in infertile men on the AKAP3 structure. The T464S, I500T, E525K, and I661T substitutions were analyzed using in silico methods. The secondary structure and three-dimensional model of AKAP3 were determined using PSI-BLAST based secondary structure prediction and Robetta servers. The TM-score was used to quantitatively measure the structural similarities between native and mutated models. All of the desired substitutions were classified as benign. I-Mutant results showed all of the substitutions decreased AKAP3 stability; however, the I500T and I661T were more effective. Superposition and secondary structure comparisons between native and mutants showed no dramatic deviations. Our study provided an appropriate model for AKAP3. Destabilization of AKAP3 caused by these substitutions did not appear to induce structural disturbances. As AKAP3 is involved in male infertility, providing more structural insights and the impact of mutations that cause protein functional diversity could elucidate the etiology of male fertility problems at molecular level.
Collapse
Affiliation(s)
- Alemeh Rafaee
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elaheh Kashani-Amin
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahita Mohseni Meybodi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Azadeh Ebrahim-Habibi
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marjan Sabbaghian
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
10
|
Chakraborty S, Saha S. Understanding sperm motility mechanisms and the implication of sperm surface molecules in promoting motility. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2022. [DOI: 10.1186/s43043-022-00094-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Abstract
Background
It is estimated that approximately 8–12% of couples globally face problems associated with infertility. A large number of men exhibit suboptimal sperm parameters. Sperm motility is one of the factors that is measured when analysing sperm parameters. The indication of several crucial sperm surface molecules, having the ability to modulate motility, has opened new avenues in understanding the complex processes involved in motility.
Main body of the abstract
There are various mechanisms that regulate and enhance sperm motility. Several surface molecules on sperm cells can also regulate motility, thus showing their possible application as a treatment for infertility caused by impaired motility. Sperm motility is regulated by intracellular and extracellular pH, along with calcium ions (Ca2+) and carbonate ion (HCO3−) concentrations. Moreover, sperm cells have an array of surface proteins which play a critical role in their function and motility. The indication of surface molecules presented new opportunities for understanding sperm motility and the possibility of treating infertility caused by impaired sperm function. Infertility and problems associated with conception can cause underlying stress and mental trauma. Although there are several methods for treating infertility, most are complex, invasive, and expensive.
Conclusion
It is important to understand how surface molecules and proteins on the sperm cell regulate motility. This will enable us to treat anomalies associated with proper sperm function. This review highlights the general mechanisms that regulate sperm motility, and it stresses the importance and relevance of sperm surface molecules in regulating sperm motility.
Collapse
|
11
|
Wang Y, Yuan X, Ali MA, Qin Z, Zhang Y, Zeng C. piR-121380 Is Involved in Cryo-Capacitation and Regulates Post-Thawed Boar Sperm Quality Through Phosphorylation of ERK2 via Targeting PTPN7. Front Cell Dev Biol 2022; 9:792994. [PMID: 35155446 PMCID: PMC8826432 DOI: 10.3389/fcell.2021.792994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/24/2021] [Indexed: 01/06/2023] Open
Abstract
Cryopreservation induces capacitation-like (cryo-capacitation) changes, similar to natural capacitation, and affects the fertility potential of post-thawed sperm. The molecular mechanism of sperm cryo-capacitation during cryopreservation remains unknown. PIWI-interacting RNAs (piRNAs) have been reported to be involved in cryo-capacitation of post-thawed sperm and regulation of sperm motility, capacitation, and chemotaxis. In this study, protein tyrosine phosphatase nonreceptor type 7 (PTPN7) was positively targeted by piR-121380 after a dual luciferase assay. The mRNA expression of PTPN7 and piR-121380 was significantly decreased (p < 0.01); however, PTPN7 protein was significantly increased (p < 0.01) in post-thawed boar sperm. Furthermore, E1RK1/2 phosphorylation was reduced during cryopreservation. Six hours after transfection with piR-121380 mimic and inhibitor, the phosphorylation of ERK2 was significantly increased and decreased (p < 0.01), respectively. Furthermore, the highest and lowest total sperm motility, forward motility, and capacitation rate were observed after piR-121380 mimic and inhibitor treatments, respectively. The concentration of intracellular calcium ([Ca2+]i) showed no significant difference after transfection with either piR-121380 mimic or inhibitor at 1, 3, and 6 h. In conclusion, we demonstrated that piR-121380 modulates ERK2 phosphorylation by targeting PTPN7, which induces sperm cryo-capacitation, and eventually affects the motility and fertility potential of post-thawed sperm.
Collapse
Affiliation(s)
- Yihan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xiang Yuan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Malik Ahsan Ali
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- Department of Theriogenology, Riphah College of Veterinary Sciences, Lahore, Pakistan
| | - Ziyue Qin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yan Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Changjun Zeng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Changjun Zeng,
| |
Collapse
|
12
|
Zhao Y, Vanderkooi S, Kan FWK. The role of oviduct-specific glycoprotein (OVGP1) in modulating biological functions of gametes and embryos. Histochem Cell Biol 2022; 157:371-388. [PMID: 34993641 PMCID: PMC8979936 DOI: 10.1007/s00418-021-02065-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 01/13/2023]
Abstract
Diverse lines of evidence indicate that the mammalian oviduct makes important contributions to the complex process of reproduction other than being simply a conduit for the transport of gametes and embryos. The cumulative synthesis and transport of proteins secreted by oviductal secretory cells into the oviductal lumen create a microenvironment supporting important reproductive events, including sperm capacitation, fertilization, and early embryo development. Among the components that have been identified in the oviductal fluid is a family of glycosylated proteins known collectively as oviduct-specific glycoprotein (OVGP1) or oviductin. OVGP1 has been identified in several mammalian species, including humans. The present review summarizes the work carried out, in various mammalian species, by many research groups revealing the synthesis and secretion of OVGP1, its fate in the female reproductive tract upon secretion by the oviductal epithelium, and its role in modulating biological functions of gametes and embryos. The production and functions of recombinant human OVGP1 and recombinant OVGP1 of other mammalian species are also discussed. Some of the findings obtained with immunocytochemistry will be highlighted in the present review. It is hoped that the findings obtained from recent studies carried out with recombinant OVGP1 from various species will rekindle researchers’ interest in pursuing further the role of the oviductal microenvironment, of which OVGP1 is a major component, in contributing to the successful occurrence of early reproductive events, and the potential use of OVGP1 in improving the current assisted reproductive technology in alleviating infertility.
Collapse
Affiliation(s)
- Yuewen Zhao
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, K7L 3N, Canada
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale Fertility Center, Yale University, Orange, CT, 06477, USA
| | - Sydney Vanderkooi
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, K7L 3N, Canada
| | - Frederick W K Kan
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, K7L 3N, Canada.
| |
Collapse
|
13
|
Impact of Cryopreservation on Motile Subpopulations and Tyrosine-Phosphorylated Regions of Ram Spermatozoa during Capacitating Conditions. BIOLOGY 2021; 10:biology10111213. [PMID: 34827206 PMCID: PMC8614982 DOI: 10.3390/biology10111213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/09/2021] [Accepted: 11/17/2021] [Indexed: 11/24/2022]
Abstract
Simple Summary Spermatozoa go through diverse changes to achieve their fertilizing potential (capacitation) and develop a specific motility pattern (hyperactivation). However, to ensure a greater reproductive success, not all the spermatozoa present in an ejaculate react equally or at the same time. Therefore, a comparative analysis was performed in the present study to improve our current understanding about how cryopreservation may affect the heterogeneous nature of fresh ejaculates during these two events. Among the four motile sperm subpopulations identified in fresh and frozen-thawed ram semen, one of them developed a hyperactivated-like movement and was the main group involve in those changes associated with sperm capacitation based on the marked increase and the positive correlation with mitochondrial activity and tyrosine phosphorylation, two relevant parameters that usually increase during capacitation. In addition, cryopreservation altered the distribution of the motile sperm subpopulations. Although the subpopulation with hyperactivated-like movement increased at the beginning of incubation in frozen-thawed samples, this subpopulation together with the subpopulation of rapid and progressive spermatozoa were replaced after a prolonged incubation by the subpopulation of slow spermatozoa with the lowest mitochondrial activity, which clearly indicate the reduction in sperm quality. These findings will aid to optimize the current cryopreservation and in vitro fertilization protocols. Abstract The heterogeneous nature of ejaculates highlights the relevance of studying the behavior of different sperm subpopulations. Changes in sperm motility and the increase in tyrosine phosphorylation are key events that usually occur during capacitation and can be modified by the cryopreservation process. However, the relationship between both events remains poorly defined throughout capacitation in the different sperm subpopulations. Fresh and frozen-thawed spermatozoa were incubated in capacitating (CAP) and non-capacitating (NC) media up to 240 min. Sperm kinematics, tyrosine phosphorylation and mitochondrial activity were measured by the CASA system and imaging flow cytometry. Four motile sperm subpopulations (SP) were identified in fresh and frozen-thawed ram semen after the cluster analysis. Incubation under CAP conditions over time led to greater changes in the percentage of spermatozoa included in each subpopulation compared to NC conditions, being different between fresh and frozen-thawed spermatozoa. The SP1, characterized by slow spermatozoa, progressively increased after 15 min in frozen-thawed samples incubated in both media but not in fresh ones. The SP4, characterized by fast and non-linear spermatozoa, showed a marked increase during CAP, but not under NC conditions, occurring more rapidly in frozen-thawed spermatozoa. This subpopulation (SP4) was also the only one positively and strongly correlated with mitochondrial activity and all phosphorylated sperm regions during capacitation, either in fresh or frozen-thawed samples. Our results indicated that in vitro capacitation induced significant changes in the distribution of motile sperm subpopulations, being affected by cryopreservation. Notwithstanding, the subpopulation which probably represents hyperactivated-like spermatozoa (SP4) also increased in frozen-thawed samples, occurring faster and simultaneously to the increment of mitochondrial activity and tyrosine phosphorylation of different sperm regions.
Collapse
|
14
|
|
15
|
Balbach M, Ghanem L, Rossetti T, Kaur N, Ritagliati C, Ferreira J, Krapf D, Puga Molina LC, Santi CM, Hansen JN, Wachten D, Fushimi M, Meinke PT, Buck J, Levin LR. Soluble adenylyl cyclase inhibition prevents human sperm functions essential for fertilization. Mol Hum Reprod 2021; 27:6360468. [PMID: 34463764 DOI: 10.1093/molehr/gaab054] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/16/2021] [Indexed: 01/07/2023] Open
Abstract
Soluble adenylyl cyclase (sAC: ADCY10) has been genetically confirmed to be essential for male fertility in mice and humans. In mice, ex vivo studies of dormant, caudal epididymal sperm demonstrated that sAC is required for initiating capacitation and activating motility. We now use an improved sAC inhibitor, TDI-10229, for a comprehensive analysis of sAC function in mouse and human sperm. In contrast to caudal epididymal mouse sperm, human sperm are collected post-ejaculation, after sAC activity has already been stimulated. In addition to preventing the capacitation-induced stimulation of sAC and protein kinase A activities, tyrosine phosphorylation, alkalinization, beat frequency and acrosome reaction in dormant mouse sperm, sAC inhibitors interrupt each of these capacitation-induced changes in ejaculated human sperm. Furthermore, we show for the first time that sAC is required during acrosomal exocytosis in mouse and human sperm. These data define sAC inhibitors as candidates for non-hormonal, on-demand contraceptives suitable for delivery via intravaginal devices in women.
Collapse
Affiliation(s)
- Melanie Balbach
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA
| | - Lubna Ghanem
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA
| | - Thomas Rossetti
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA
| | - Navpreet Kaur
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA
| | - Carla Ritagliati
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA.,Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario, Rosario, Argentina
| | - Jacob Ferreira
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA
| | - Dario Krapf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario, Rosario, Argentina
| | - Lis C Puga Molina
- Department of OB/GYN, Washington University School of Medicine, Saint Louis, MO, USA
| | - Celia Maria Santi
- Department of OB/GYN, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jan Niklas Hansen
- Biophysical Imaging, Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Dagmar Wachten
- Biophysical Imaging, Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Makoto Fushimi
- Tri-Institutional Therapeutics Discovery Institute, New York City, NY, USA
| | - Peter T Meinke
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA.,Tri-Institutional Therapeutics Discovery Institute, New York City, NY, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medicine, New York City, NY, USA
| |
Collapse
|
16
|
Reynoso S, Castillo V, Katkar GD, Lopez-Sanchez I, Taheri S, Espinoza C, Rohena C, Sahoo D, Gagneux P, Ghosh P. GIV/Girdin, a non-receptor modulator for Gαi/s, regulates spatiotemporal signaling during sperm capacitation and is required for male fertility. eLife 2021; 10:69160. [PMID: 34409938 PMCID: PMC8376251 DOI: 10.7554/elife.69160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
For a sperm to successfully fertilize an egg, it must first undergo capacitation in the female reproductive tract and later undergo acrosomal reaction (AR) upon encountering an egg surrounded by its vestment. How premature AR is avoided despite rapid surges in signaling cascades during capacitation remains unknown. Using a combination of conditional knockout (cKO) mice and cell-penetrating peptides, we show that GIV (CCDC88A), a guanine nucleotide-exchange modulator (GEM) for trimeric GTPases, is highly expressed in spermatocytes and is required for male fertility. GIV is rapidly phosphoregulated on key tyrosine and serine residues in human and murine spermatozoa. These phosphomodifications enable GIV-GEM to orchestrate two distinct compartmentalized signaling programs in the sperm tail and head; in the tail, GIV enhances PI3K→Akt signals, sperm motility and survival, whereas in the head it inhibits cAMP surge and premature AR. Furthermore, GIV transcripts are downregulated in the testis and semen of infertile men. These findings exemplify the spatiotemporally segregated signaling programs that support sperm capacitation and shed light on a hitherto unforeseen cause of infertility in men.
Collapse
Affiliation(s)
- Sequoyah Reynoso
- Department of Pathology, School of Medicine, University of California San Diego, San Diego, United States
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Gajanan Dattatray Katkar
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Inmaculada Lopez-Sanchez
- Department of Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Sahar Taheri
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, San Diego, United States
| | - Celia Espinoza
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Cristina Rohena
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States
| | - Debashis Sahoo
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, San Diego, United States.,Moore's Comprehensive Cancer Center, University of California San Diego, San Diego, United States.,Department of Pediatrics, School of Medicine, University of California San Diego, San Diego, United States
| | - Pascal Gagneux
- Department of Pathology, School of Medicine, University of California San Diego, San Diego, United States
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, San Diego, United States.,Department of Medicine, School of Medicine, University of California San Diego, San Diego, United States.,Moore's Comprehensive Cancer Center, University of California San Diego, San Diego, United States.,Veterans Affairs Medical Center, Washington DC, United States
| |
Collapse
|
17
|
Selection of Boar Sperm by Reproductive Biofluids as Chemoattractants. Animals (Basel) 2020; 11:ani11010053. [PMID: 33396764 PMCID: PMC7824399 DOI: 10.3390/ani11010053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/21/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Both in natural breeding and some assisted reproduction technologies, spermatozoa are deposited into the uterus. The journey the spermatozoa must take from the place of semen deposition to the fertilization site is long, hostile, and selective of the best spermatozoa. For the fertilization to succeed, spermatozoa are guided by chemical stimuli (chemoattractants) to the fertilization site, mainly secreted by the oocyte, cumulus cells, and other substances poured into the oviduct in the periovulatory period. This work studied some sources of chemotactic factors and their action on spermatozoa functionality in vitro, including the fertility. A special chemotactic chamber for spermatozoa selection was designed which consists of two wells communicated by a tube. The spermatozoa are deposited in well A, and the chemoattractants in well B. This study focuses on the use of follicular fluid (FF), periovulatory oviductal fluid (pOF), conditioned medium from the in vitro maturation of oocytes (CM), and progesterone (P4) as chemoattractants to sperm. The chemotactic potential of these substances is also investigated as related to their action on CatSper which is a calcium channel in the spermatozoa known to be sensitive to chemoattractants and essential for motility. Abstract Chemotaxis is a spermatozoa guidance mechanism demonstrated in vitro in several mammalian species including porcine. This work focused on follicular fluid (FF), periovulatory oviductal fluid (pOF), the medium surrounding oocytes during in vitro maturation (conditioned medium; CM), progesterone (P4), and the combination of those biofluids (Σ) as chemotactic agents and modulators of spermatozoa fertility in vitro. A chemotaxis chamber was designed consisting of two independent wells, A and B, connected by a tube. The spermatozoa are deposited in well A, and the chemoattractants in well B. The concentrations of biofluids that attracted a higher proportion of spermatozoa to well B were 0.25% FF, 0.25% OF, 0.06% CM, 10 pM P4 and 0.25% of a combination of biofluids (Σ2), which attracted between 3.3 and 12.3% of spermatozoa (p < 0.05). The motility of spermatozoa recovered in well B was determined and the chemotactic potential when the sperm calcium channel CatSper was inhibited, which significantly reduced the % of spermatozoa attracted (p < 0.05). Regarding the in vitro fertility, the spermatozoa attracted by FF produced higher rates of penetration of oocytes and development of expanded blastocysts. In conclusion, porcine reproductive biofluids show an in vitro chemotactic effect on spermatozoa and modulate their fertilizing potential.
Collapse
|
18
|
Dcunha R, Hussein RS, Ananda H, Kumari S, Adiga SK, Kannan N, Zhao Y, Kalthur G. Current Insights and Latest Updates in Sperm Motility and Associated Applications in Assisted Reproduction. Reprod Sci 2020; 29:7-25. [PMID: 33289064 PMCID: PMC7721202 DOI: 10.1007/s43032-020-00408-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/08/2020] [Accepted: 11/01/2020] [Indexed: 02/07/2023]
Abstract
Spermatozoon is a motile cell with a special ability to travel through the woman’s reproductive tract and fertilize an oocyte. To reach and penetrate the oocyte, spermatozoa should possess progressive motility. Therefore, motility is an important parameter during both natural and assisted conception. The global trend of progressive reduction in the number and motility of healthy spermatozoa in the ejaculate is associated with increased risk of infertility. Therefore, developing approaches for maintaining or enhancing human sperm motility has been an important area of investigation. In this review we discuss the physiology of sperm, molecular pathways regulating sperm motility, risk factors affecting sperm motility, and the role of sperm motility in fertility outcomes. In addition, we discuss various pharmacological agents and biomolecules that can enhance sperm motility in vitro and in vivo conditions to improve assisted reproductive technology (ART) outcomes. This article opens dialogs to help toxicologists, clinicians, andrologists, and embryologists in understanding the mechanism of factors influencing sperm motility and various management strategies to improve treatment outcomes.
Collapse
Affiliation(s)
- Reyon Dcunha
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Reda S Hussein
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Obstetrics and Gynecology, Assiut University, Assiut City, Egypt
| | - Hanumappa Ananda
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Sandhya Kumari
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Satish Kumar Adiga
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nagarajan Kannan
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, 55905, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yulian Zhao
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Guruprasad Kalthur
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India. .,Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
19
|
Baro Graf C, Ritagliati C, Stival C, Luque GM, Gentile I, Buffone MG, Krapf D. Everything you ever wanted to know about PKA regulation and its involvement in mammalian sperm capacitation. Mol Cell Endocrinol 2020; 518:110992. [PMID: 32853743 DOI: 10.1016/j.mce.2020.110992] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/29/2022]
Abstract
The 3', 5'-cyclic adenosine monophosphate (cAMP) dependent protein kinase (PKA) is a tetrameric holoenzyme comprising a set of two regulatory subunits (PKA-R) and two catalytic (PKA-C) subunits. The PKA-R subunits act as sensors of cAMP and allow PKA-C activity. One of the first signaling events observed during mammalian sperm capacitation is PKA activation. Thus, understanding how PKA activity is restricted in space and time is crucial to decipher the critical steps of sperm capacitation. It is widely accepted that PKA specificity depends on several levels of regulation. Anchoring proteins play a pivotal role in achieving proper localization signaling, subcellular targeting and cAMP microdomains. These multi-factorial regulation steps are necessary for a precise spatio-temporal activation of PKA. Here we discuss recent understanding of regulatory mechanisms of PKA in mammalian sperm, such as post-translational modifications, in the context of its role as the master orchestrator of molecular events conducive to capacitation.
Collapse
Affiliation(s)
- Carolina Baro Graf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina; Laboratorio de Medicina Reproductiva (LMR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Carla Ritagliati
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina
| | - Cintia Stival
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina
| | - Guillermina M Luque
- Laboratory of Cellular and Molecular Reproductive Biology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Iñaki Gentile
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina
| | - Mariano G Buffone
- Laboratory of Cellular and Molecular Reproductive Biology, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Dario Krapf
- Laboratory of Cell Signal Transduction Networks, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario, Argentina; Laboratorio de Medicina Reproductiva (LMR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina.
| |
Collapse
|
20
|
Zhao Y, Kan FWK. Human OVGP1 enhances tyrosine phosphorylation of proteins in the fibrous sheath involving AKAP3 and increases sperm-zona binding. J Assist Reprod Genet 2019; 36:1363-1377. [PMID: 31254143 PMCID: PMC6642236 DOI: 10.1007/s10815-019-01502-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/07/2019] [Indexed: 12/26/2022] Open
Abstract
Purpose To investigate if the recombinant human oviduct-specific glycoprotein (rHuOVGP1)–enhanced tyrosine-phosphorylated (pY) proteins are components of specific structure(s) of the sperm tail and if rHuOVGP1 binds to the oocyte and enhances sperm-egg binding. Methods Immunofluorescent staining and confocal microscopy were performed to examine the localization of pY proteins, outer dense fiber (ODF), and A-Kinase Associated Protein 3 (AKAP3) in human sperm during capacitation. Western blot and immunoprecipitation were employed to analyze protein levels of pY proteins and AKAP3. Immunofluorescent staining was performed to examine the binding of rHuOVGP1 to human oocytes. The effect of rHuOVGP1 on enhancing sperm-zona binding was examined using hemizona assay. Results pY proteins were detected mainly in the fibrous sheath (FS) surrounding the ODF with a relatively weak immunoreaction in the neck and mid-piece. Western blot analysis revealed co-migration of the pY 105 kDa protein with AKAP3, which was further confirmed by immunoprecipitation correlating immunofluorescent results of co-localization of pY proteins with AKAP3 in the sperm tail. rHuOVGP1 binds specifically to the zona pellucida (ZP) of human oocytes. Prior incubation of sperm and/or ZP with rHuOVGP1 increased sperm-egg binding. Conclusions The present study revealed that one of the major rHuOVGP1-enhanced pY proteins could be AKAP3 of the FS and that rHuOVGP1 is capable of binding to human ZP and its presence in the medium results in an increase in sperm-zona binding. Supplement of rHuOVGP1 in in vitro fertilization media could be beneficial for enhancement of the fertilizing ability of human sperm.
Collapse
Affiliation(s)
- Yuewen Zhao
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Frederick W K Kan
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, K7L 3N6, Canada.
| |
Collapse
|
21
|
Puga Molina LC, Pinto NA, Torres NI, González-Cota AL, Luque GM, Balestrini PA, Romarowski A, Krapf D, Santi CM, Treviño CL, Darszon A, Buffone MG. CFTR/ENaC-dependent regulation of membrane potential during human sperm capacitation is initiated by bicarbonate uptake through NBC. J Biol Chem 2018; 293:9924-9936. [PMID: 29743243 DOI: 10.1074/jbc.ra118.003166] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/29/2018] [Indexed: 12/16/2022] Open
Abstract
To fertilize an egg, sperm must reside in the female reproductive tract to undergo several maturational changes that are collectively referred to as capacitation. From a molecular point of view, the HCO3--dependent activation of the atypical soluble adenylyl cyclase (ADCY10) is one of the first events that occurs during capacitation and leads to the subsequent cAMP-dependent activation of protein kinase A (PKA). Capacitation is also accompanied by hyperpolarization of the sperm plasma membrane. We previously reported that PKA activation is necessary for CFTR (cystic fibrosis transmembrane conductance regulator channel) activity and for the modulation of membrane potential (Em). However, the main HCO3- transporters involved in the initial transport and the PKA-dependent Em changes are not well known nor characterized. Here, we analyzed how the activity of CFTR regulates Em during capacitation and examined its relationship with an electrogenic Na+/HCO3- cotransporter (NBC) and epithelial Na+ channels (ENaCs). We observed that inhibition of both CFTR and NBC decreased HCO3- influx, resulting in lower PKA activity, and that events downstream of the cAMP activation of PKA are essential for the regulation of Em. Addition of a permeable cAMP analog partially rescued the inhibitory effects caused by these inhibitors. HCO3- also produced a rapid membrane hyperpolarization mediated by ENaC channels, which contribute to the regulation of Em during capacitation. Altogether, we demonstrate for the first time, that NBC cotransporters and ENaC channels are essential in the CFTR-dependent activation of the cAMP/PKA signaling pathway and Em regulation during human sperm capacitation.
Collapse
Affiliation(s)
- Lis C Puga Molina
- From the Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), C1425FQB Buenos Aires, Argentina
| | - Nicolás A Pinto
- From the Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), C1425FQB Buenos Aires, Argentina
| | - Nicolás I Torres
- From the Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), C1425FQB Buenos Aires, Argentina
| | - Ana L González-Cota
- the Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Guillermina M Luque
- From the Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), C1425FQB Buenos Aires, Argentina
| | - Paula A Balestrini
- From the Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), C1425FQB Buenos Aires, Argentina
| | - Ana Romarowski
- From the Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), C1425FQB Buenos Aires, Argentina
| | - Dario Krapf
- the Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET-UNR, Rosario 2000, Argentina, and
| | - Celia M Santi
- the Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Claudia L Treviño
- the Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, 62210 Morelos, México
| | - Alberto Darszon
- the Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, 62210 Morelos, México
| | - Mariano G Buffone
- From the Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), C1425FQB Buenos Aires, Argentina,
| |
Collapse
|
22
|
Selvaraju S, Parthipan S, Somashekar L, Binsila BK, Kolte AP, Arangasamy A, Ravindra JP, Krawetz SA. Current status of sperm functional genomics and its diagnostic potential of fertility in bovine (Bos taurus). Syst Biol Reprod Med 2018. [DOI: 10.1080/19396368.2018.1444816] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Sellappan Selvaraju
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, India
| | - Sivashanmugam Parthipan
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, India
| | - Lakshminarayana Somashekar
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, India
| | - B. Krishnan Binsila
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, India
| | - Atul P. Kolte
- Omics Laboratory, Animal Nutrition Division, ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, India
| | - Arunachalam Arangasamy
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, India
| | - Janivara Parameshwaraiah Ravindra
- Reproductive Physiology Laboratory, Animal Physiology Division, ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, India
| | - Stephen A. Krawetz
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| |
Collapse
|
23
|
Martinez G, Hograindleur JP, Voisin S, Abi Nahed R, Abd El Aziz TM, Escoffier J, Bessonnat J, Fovet CM, De Waard M, Hennebicq S, Aucagne V, Ray PF, Schmitt E, Bulet P, Arnoult C. Spermaurin, an La1-like peptide from the venom of the scorpion Scorpio maurus palmatus, improves sperm motility and fertilization in different mammalian species. Mol Hum Reprod 2018; 23:116-131. [PMID: 27932550 DOI: 10.1093/molehr/gaw075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/18/2016] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Is it possible to identify original compounds that are able to enhance sperm motility from the venom of the scorpion Scorpio maurus palmatus? SUMMARY ANSWER We identified a potent disulfide-rich peptide (DRP) of 73 amino acids that significantly improved the motility of fresh and frozen-thawed sperm in different mammalian species, including human, and improved fertilization outcome in mouse IVF experiments. WHAT IS KNOWN ALREADY Any disturbance of sperm motility has a strong impact on fertilization and can lead to subfertility or infertility. Significant efforts have, therefore, been made to identify pharmacological drugs that might improve sperm motility. Such compounds are particularly useful in azoospermia to improve testicular sperm extraction and in the domain of cryopreservation because the motility of frozen-thawed sperm is reduced. STUDY DESIGN, SIZE, DURATION This was a basic science/medical research study aimed at identifying original compounds from a library of venoms able to enhance mammalian sperm motility, including human. We first identified in the venom of a scorpion S. m. palmatus a fraction able to potently activate sperm motility. We next purified and characterized the compound by liquid chromatography, mass spectrometry and peptide synthesis. Finally, the potency and toxicity of both purified and synthetic versions of the identified compound on sperm motility were assessed using different in vitro tests in different mammalian species. PARTICIPANTS/MATERIALS, SETTING, METHODS For human sperm, biological samples were collected from normozoospermic donors and subfertile patients attending a reproduction department for diagnostic semen analysis. Testicular sperm was collected from cynomolgus monkeys (Macaca fascicularis) euthanized for the needs of specific authorized research projects. The peptide was also tested on bovine and mouse epidydimal sperm. We measured different sperm motility parameters with a computer-assisted sperm analysis system in the presence or absence of the peptide. MAIN RESULTS AND THE ROLE OF CHANCE Size exclusion chromatography enabled us to isolate a fraction of the venom of S. m. palmatus able to increase sperm motility. By liquid chromatography and mass spectrometry, a peptide comprising 73 amino acids with 4 disulfide bridges was identified as responsible for the biological activity and called 'spermaurin'. The identity of spermaurin was confirmed by chemical synthesis. We showed that the peptide increased the motility of fresh and frozen-thawed human sperm. We observed that the potency of the peptide was higher on fresh ejaculated spermatozoa with a low motility, achieving a 100% increase of curvilinear velocity in poorly performing sperm. We also demonstrated that peptide is effective on bovine and mouse fresh epididymal, bovine frozen-thawed ejaculated and fresh non-human primate testicular sperm. Finally, in mouse IVF, the production of 2-cell embryos was increased by 24% when sperm were treated with the peptide. LIMITATIONS, REASONS FOR CAUTION This work is an in vitro evaluation of the ability of spermaurin to improve sperm motility parameters. Another limitation of this study is the small number of human sperm samples tested with the natural (n = 36) and synthetic (n = 12) peptides. Moreover, the effect of the peptide on IVF outcome was only tested in mouse and further tests with human and bovine gametes are required to confirm and extend this result in other mammalian species. WIDER IMPLICATIONS OF THE FINDINGS This work confirms our initial study showing that venoms represent an interesting source of molecules that are able to modify sperm physiology. Moreover, this work presents the first demonstrated biological action of a venom peptide from the scorpion S. m. palmatus with sequence similarities to La1 peptide from Liocheles australasiae (Wood scorpion), a widespread family of DRPs. LARGE SCALE DATA Not applicable. STUDY FUNDING/COMPETING INTEREST(S) This work is part of the project 'LAB COM-14 LAB7 0004 01-LIPAV', funded by the program LabCom 2014 from the French Research Agency (ANR). Dr Arnoult reports grants from IMV Technologies during the conduct of the study. In addition, Drs Arnoult, Martinez, Ray and Schmitt have a patent EP16305642.7 pending containing some of the information presented in this manuscript.
Collapse
Affiliation(s)
- Guillaume Martinez
- IMV Technologies, ZI N° 1 Est, F-61300 L'Aigle, France.,Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France
| | - Jean-Pascal Hograindleur
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France
| | - Sébastien Voisin
- Plateforme BioPark d'Archamps, Archamps Technopole, Saint Julien en Genevois F-74160, France
| | - Roland Abi Nahed
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France
| | - Tarek M Abd El Aziz
- L'institut du thorax, Inserm UMR 1087/CNRS UMR 6291, Université de Nantes, NantesF44007, France
| | - Jessica Escoffier
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France
| | - Julien Bessonnat
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France.,CHU Grenoble Alpes, UF de Biologie de la procréation, Grenoble F-38000, France
| | - Claire-Maëlle Fovet
- Molecular Imaging Research Center, MIRCen CEA/INSERM UMR1169, Fontenay-aux-Roses F-92265, France
| | - Michel De Waard
- L'institut du thorax, Inserm UMR 1087/CNRS UMR 6291, Université de Nantes, NantesF44007, France
| | - Sylviane Hennebicq
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France.,CHU Grenoble Alpes, UF de Biologie de la procréation, Grenoble F-38000, France
| | - Vincent Aucagne
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Orléans F-45071, France
| | - Pierre F Ray
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France.,CHU Grenoble Alpes, UF de Biochimie Génétique et Moléculaire, Grenoble F-38000, France
| | - Eric Schmitt
- IMV Technologies, ZI N° 1 Est, F-61300 L'Aigle, France
| | - Philippe Bulet
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France.,Plateforme BioPark d'Archamps, Archamps Technopole, Saint Julien en Genevois F-74160, France
| | - Christophe Arnoult
- Université Grenoble Alpes, Grenoble F-38000, France.,Institute for Advanced Biosciences INSERM U1209, CNRS UMR5309, Grenoble F-38000, France
| |
Collapse
|
24
|
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]
|
25
|
Abstract
The genetic bases and molecular mechanisms involved in the assembly and function of the flagellum components as well as in the regulation of the flagellar movement are not fully understood, especially in humans. There are several causes for sperm immotility, of which some can be avoided and corrected, whereas other are related to genetic defects and deserve full investigation to give a diagnosis to patients. This review was performed after an extensive literature search on the online databases PubMed, ScienceDirect, and Web of Science. Here, we review the involvement of regulatory pathways responsible for sperm motility, indicating possible causes for sperm immotility. These included the calcium pathway, the cAMP-dependent protein kinase pathway, the importance of kinases and phosphatases, the function of reactive oxygen species, and how the regulation of cell volume and osmolarity are also fundamental components. We then discuss main gene defects associated with specific morphological abnormalities. Finally, we slightly discuss some preventive and treatments approaches to avoid development of conditions that are associated with unspecified sperm immotility. We believe that in the near future, with the development of more powerful techniques, the genetic causes of sperm immotility and the regulatory mechanisms of sperm motility will be better understand, thus enabling to perform a full diagnosis and uncover new therapies.
Collapse
Affiliation(s)
- Rute Pereira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal and Multidisciplinary Unit for Biomedical Research-UMIB, ICBAS-UP, Portugal
| | - Rosália Sá
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal and Multidisciplinary Unit for Biomedical Research-UMIB, ICBAS-UP, Portugal
| | - Alberto Barros
- Centre for Reproductive Genetics Alberto Barros, Av. do Bessa, 240, 1° Dto. Frente, 4100-012 Porto, Portugal.,Department of Genetics, Faculty of Medicine, University of Porto. Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal and Institute of Health Research an Innovation (I3S), University of Porto, Portugal
| | - Mário Sousa
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal and Multidisciplinary Unit for Biomedical Research-UMIB, ICBAS-UP, Portugal
| |
Collapse
|
26
|
Guasti PN, Monteiro GA, Maziero RRD, Carmo MT, Dell'Aqua JA, Crespilho AM, Rifai EA, Papa FO. Pentoxifylline effects on capacitation and fertility of stallion epididymal sperm. Anim Reprod Sci 2017; 179:27-34. [PMID: 28238445 DOI: 10.1016/j.anireprosci.2017.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 01/15/2017] [Accepted: 01/29/2017] [Indexed: 10/20/2022]
Abstract
The aims of this study were to determinate whether pentoxifylline (PTX) increases the motion parameters of fresh and frozen-thawed equine epididymal spermatozoa, to evaluate the tyrosine phosphorylation of frozen-thawed epididymal sperm in the presence of PTX and to determine whether the PTX-treatment of stallion epididymal sperm prior to freezing improves the fertility response of mares to a reduced number of spermatozoa per insemination dose. Fifty epididymis were flushed with a skim milk based extender with or without PTX. The pre-treatment with PTX enhanced the sperm motility after being harvested (P<0.05); however the freeze-thaw process did not alter the sperm kinematics between control and treated samples (P>0.05). Plasma membrane integrity did not differ between control and PTX group after recovery and after thawing (P>0.05), as observed in tyrosine phosphorylation, which the PTX treatment did not alter the percentage of tail-associated immunofluorescence of cryopreserved epididymal sperm (P>0.05). For the fertility trial, different insemination groups were tested: 800×106 epididymal sperm (C800); 100×106 epididymal sperm (C100); 100×106 epididymal sperm recovered in an extender containing PTX (PTX100). The conception rates for C800; C100 and PTX100 were 68.7% (11/16); 31.5% (5/16) and 50% (8/16), respectively. The conception rate did not differ among groups (P>0.05), however, a low number of animals was used in this study. A trend toward significance (P=0.07) was observed between C800 and C100 groups. In conclusion, PTX has no deleterious effect on sperm motility, viability and capacitation of cryopreserved stallion epididymal sperm. The conventional artificial insemination with 100×106 sperm recovered with PTX ensures acceptable conception rates and maximize the limited number of doses of cryopreserved stallion epididymal sperm.
Collapse
Affiliation(s)
- P N Guasti
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, Brazil.
| | - G A Monteiro
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, Brazil
| | - R R D Maziero
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, Brazil
| | - M T Carmo
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, Brazil
| | - J A Dell'Aqua
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, Brazil
| | | | - E A Rifai
- Division of Molecular & Computational Toxicology, Vrije Universiteit Amsterdam, The Netherlands
| | - F O Papa
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, Brazil
| |
Collapse
|
27
|
Puga Molina LC, Pinto NA, Torres Rodríguez P, Romarowski A, Vicens Sanchez A, Visconti PE, Darszon A, Treviño CL, Buffone MG. Essential Role of CFTR in PKA-Dependent Phosphorylation, Alkalinization, and Hyperpolarization During Human Sperm Capacitation. J Cell Physiol 2016; 232:1404-1414. [PMID: 27714810 DOI: 10.1002/jcp.25634] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/05/2016] [Indexed: 12/17/2022]
Abstract
Mammalian sperm require to spend a limited period of time in the female reproductive tract to become competent to fertilize in a process called capacitation. It is well established that HCO3- is essential for capacitation because it activates the atypical soluble adenylate cyclase ADCY10 leading to cAMP production, and promotes alkalinization of cytoplasm, and membrane hyperpolarization. However, how HCO3- is transported into the sperm is not well understood. There is evidence that CFTR activity is involved in the human sperm capacitation but how this channel is integrated in the complex signaling cascades associated with this process remains largely unknown. In the present work, we have analyzed the extent to which CFTR regulates different events in human sperm capacitation. We observed that inhibition of CFTR affects HCO3- -entrance dependent events resulting in lower PKA activity. CFTR inhibition also affected cAMP/PKA-downstream events such as the increase in tyrosine phosphorylation, hyperactivated motility, and acrosome reaction. In addition, we demonstrated for the first time, that CFTR and PKA activity are essential for the regulation of intracellular pH, and membrane potential in human sperm. Addition of permeable cAMP partially recovered all the PKA-dependent events altered in the presence of inh-172 which is consistent with a role of CFTR upstream of PKA activation. J. Cell. Physiol. 232: 1404-1414, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lis C Puga Molina
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Nicolás A Pinto
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Paulina Torres Rodríguez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, México
| | - Ana Romarowski
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| | - Alberto Vicens Sanchez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, México
| | - Pablo E Visconti
- Department of Veterinary and Animal Science, Paige Labs, University of Massachusetts, Amherst, Massachusetts
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, México
| | - Claudia L Treviño
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, México
| | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
28
|
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
|
29
|
Mukherjee S, Jansen V, Jikeli JF, Hamzeh H, Alvarez L, Dombrowski M, Balbach M, Strünker T, Seifert R, Kaupp UB, Wachten D. A novel biosensor to study cAMP dynamics in cilia and flagella. eLife 2016; 5. [PMID: 27003291 PMCID: PMC4811770 DOI: 10.7554/elife.14052] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/05/2016] [Indexed: 01/09/2023] Open
Abstract
The cellular messenger cAMP regulates multiple cellular functions, including signaling in cilia and flagella. The cAMP dynamics in these subcellular compartments are ill-defined. We introduce a novel FRET-based cAMP biosensor with nanomolar sensitivity that is out of reach for other sensors. To measure cAMP dynamics in the sperm flagellum, we generated transgenic mice and reveal that the hitherto methods determining total cAMP levels do not reflect changes in free cAMP levels. Moreover, cAMP dynamics in the midpiece and principal piece of the flagellum are distinctively different. The sole cAMP source in the flagellum is the soluble adenylate cyclase (SACY). Although bicarbonate-dependent SACY activity requires Ca2+, basal SACY activity is suppressed by Ca2+. Finally, we also applied the sensor to primary cilia. Our new cAMP biosensor features unique characteristics that allow gaining new insights into cAMP signaling and unravel the molecular mechanisms underlying ciliary function in vitro and in vivo. DOI:http://dx.doi.org/10.7554/eLife.14052.001 Cells can change the way they grow, move or develop in response to information from their environment. This information is first detected at the surface of the cell and then the information is relayed around the interior of the cell by signaling molecules known as “second messengers”. A molecule called cAMP is a well-known second messenger that is involved in many different signaling pathways. Therefore, the levels of cAMP in specific areas of the cell need to be precisely regulated to enable different signaling pathways to be activated at specific times and locations. Some cells have hair-like structures called cilia or flagella on their surface. Cilia and flagella are able to move the fluid that surrounds the cells or even move the cells themselves. The second messenger cAMP plays an essential role in making cilia move, but it is challenging to analyze the dynamics of cAMP – that this, how the levels of this molecule change over time – in these structures. The levels of cAMP in live cells can only be measured using fluorescent biosensors. Introducing these biosensors into specific cell structures is difficult and they are not sensitive enough to respond to low levels of cAMP. Furthermore, it is difficult to measure cAMP activity inside such tiny structures using these biosensors. Mukherjee, Jansen, Jikeli et al. now address some of these challenges by creating a new cAMP biosensor that has several unique features. Most importantly, it can respond to very low levels of cAMP, making it more sensitive than previous biosensors. Mukherjee et al. test this new biosensor in the flagella of sperm cells from mice, which reveals how the production of cAMP is regulated in the flagellum. The new biosensor also shows that different parts of the flagellum can have different cAMP dynamics. In the future, this new biosensor could be used to study cAMP in other structures and compartments within cells. DOI:http://dx.doi.org/10.7554/eLife.14052.002
Collapse
Affiliation(s)
- Shatanik Mukherjee
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, Bonn, Germany
| | - Vera Jansen
- Minerva Max Planck Research Group, Molecular Physiology, Center of Advanced European Studies and Research, Bonn, Germany
| | - Jan F Jikeli
- Minerva Max Planck Research Group, Molecular Physiology, Center of Advanced European Studies and Research, Bonn, Germany
| | - Hussein Hamzeh
- Minerva Max Planck Research Group, Molecular Physiology, Center of Advanced European Studies and Research, Bonn, Germany
| | - Luis Alvarez
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, Bonn, Germany
| | - Marco Dombrowski
- Minerva Max Planck Research Group, Molecular Physiology, Center of Advanced European Studies and Research, Bonn, Germany
| | - Melanie Balbach
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, Bonn, Germany
| | - Timo Strünker
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, Bonn, Germany.,Centrum für Reproduktionsmedizin und Andrologie, Universitätsklinikum Münster, Münster, Germany
| | - Reinhard Seifert
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, Bonn, Germany
| | - U Benjamin Kaupp
- Department of Molecular Sensory Systems, Center of Advanced European Studies and Research, Bonn, Germany
| | - Dagmar Wachten
- Minerva Max Planck Research Group, Molecular Physiology, Center of Advanced European Studies and Research, Bonn, Germany
| |
Collapse
|
30
|
Baker MA. Proteomics of post-translational modifications of mammalian spermatozoa. Cell Tissue Res 2015; 363:279-287. [DOI: 10.1007/s00441-015-2249-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 06/18/2015] [Indexed: 12/25/2022]
|
31
|
Tamburrino L, Marchiani S, Vicini E, Muciaccia B, Cambi M, Pellegrini S, Forti G, Muratori M, Baldi E. Quantification of CatSper1 expression in human spermatozoa and relation to functional parameters. Hum Reprod 2015; 30:1532-44. [PMID: 25983333 DOI: 10.1093/humrep/dev103] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/20/2015] [Indexed: 01/03/2023] Open
Abstract
STUDY QUESTION Is CatSper1 expression in human spermatozoa related to semen parameter values and sperm functions? SUMMARY ANSWER CatSper1 expression is positively related to progressive and hyperactivated (HA) motility, [Ca(2+)]i responsiveness to progesterone but not the acrosome reaction (AR). WHAT IS KNOWN ALREADY The role of cationic channel of sperm (CatSper) in sperm functions is clear in animal models but less defined in human sperm cells. Current knowledge is mostly based on low specificity CatSper inhibitors showing agonistic and toxic effects on human spermatozoa and is thus of little help in clarifying the role of the CatSper channel in human sperm functions. STUDY DESIGN, SIZE, DURATION CatSper1 protein expression was evaluated in 115 men undergoing semen analysis for couple infertility. CatSper1 expression was related to routine semen parameters, motility kinematic parameters and basal and progesterone-stimulated [Ca(2+)]i and the AR. PARTICIPANTS/MATERIALS, SETTING, METHODS CatSper1 expression was evaluated (n = 85 normozoospermic, n = 30 asthenozoospermic patients) by immunofluorescence coupled to flow cytometry leading to quantitative measurement of the percentage of ejaculated sperm cells expressing the protein. Semen analysis was evaluated according to World Health Organization guidelines. Kinematic parameters were evaluated by a computer-aided sperm analysis system. [Ca(2+)]i was measured by a spectrofluorimetric method in fura-2-loaded spermatozoa. The AR was evaluated in live sperm cells by fluorescent-labeled lectin. MAIN RESULTS AND THE ROLE OF CHANCE CatSper1 protein expression in spermatozoa was reduced in asthenozoospermic men (mean ± SD: 53.0 ± 15.5%, n = 30 versus 67.9 ± 17.1% in normozoospermic, n = 85, P < 0.01) and was significantly correlated with progressive (r = 0.36, P < 0.001), total (r = 0.35, P < 0.001) and HA (r = 0.41, P < 0.005) motility. In addition to a higher percentage of spermatozoa not expressing CatSper1, asthenozoospermic men showed a large number of spermatozoa with immunofluorescent signal localized outside the principal piece compared with those in normozoospermia. A significant positive correlation was found between CatSper1 protein expression and the increase of [Ca(2+)]i in response to progesterone (r = 0.36, P < 0.05, n = 40) but not with basal [Ca(2+)]i. No correlation was found with the AR, either basal or in response to progesterone. LIMITATIONS, REASONS FOR CAUTION The study is partly descriptive. Furthermore, we cannot rule out the possibility that some round cells remain after a single round of 40% density gradient centrifugation or that this step may have removed some defective or slow swimming sperm, and therefore this preparation may not be representative of the entire sperm sample. Although our data suggest that CatSper1 may be a useful marker for infertility, and a possible contraceptive target, any clinical application is limited without further research. WIDER IMPLICATIONS OF THE FINDINGS Our results demonstrate an association of CatSper1 expression with human sperm progressive and HA motility and provide preliminary evidence that lack of expression or mislocalization of CatSper1 in spermatozoa may be involved in the pathogenesis of asthenozoospermia. However, mechanistic studies are needed to confirm that the correlations between CatSper1 expression and sperm functions are causative. STUDY FUNDING/COMPETING INTERESTS Supported by grants from Ministry of University and Scientific Research (PRIN project to E.B. and FIRB project to S.M.) and by Regione Toscana (to G.F.). L.T. was recipient of a grant from Accademia dei Lincei (Rome, Italy). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- L Tamburrino
- Department of Biomedical Experimental and Clinical Sciences, Center of Excellence DENOthe, University of Florence, Viale Pieraccini 6, Florence I-50139, Italy
| | - S Marchiani
- Department of Biomedical Experimental and Clinical Sciences, Center of Excellence DENOthe, University of Florence, Viale Pieraccini 6, Florence I-50139, Italy
| | - E Vicini
- Fondazione Pasteur Cenci Bolognetti, Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Italy
| | - B Muciaccia
- Fondazione Pasteur Cenci Bolognetti, Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Italy
| | - M Cambi
- Department of Biomedical Experimental and Clinical Sciences, Center of Excellence DENOthe, University of Florence, Viale Pieraccini 6, Florence I-50139, Italy
| | - S Pellegrini
- Centro Procreazione Assistita 'Demetra' of Florence, Via Caccini 18, Florence I-50139, Italy
| | - G Forti
- Department of Biomedical Experimental and Clinical Sciences, Center of Excellence DENOthe, University of Florence, Viale Pieraccini 6, Florence I-50139, Italy
| | - M Muratori
- Department of Biomedical Experimental and Clinical Sciences, Center of Excellence DENOthe, University of Florence, Viale Pieraccini 6, Florence I-50139, Italy
| | - E Baldi
- Department of Biomedical Experimental and Clinical Sciences, Center of Excellence DENOthe, University of Florence, Viale Pieraccini 6, Florence I-50139, Italy
| |
Collapse
|
32
|
Naresh S, Atreja SK. The protein tyrosine phosphorylation during in vitro capacitation and cryopreservation of mammalian spermatozoa. Cryobiology 2015; 70:211-6. [PMID: 25828199 DOI: 10.1016/j.cryobiol.2015.03.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 03/07/2015] [Accepted: 03/21/2015] [Indexed: 11/26/2022]
Abstract
Before the process of fertilization, spermatozoa necessitate a period of residence in the female reproductive environment, and undergo a sequence of physiological and biochemical changes collectively referred to as capacitation. Accumulated evidences from several laboratories indicated that the protein tyrosine phosphorylation (PTP) is one of the most important intracellular signaling events regulating sperm function, and is a meaningful indicator of capacitation. Different factors that affect PTP are cholesterol efflux, influx of HCO3(-), increased intracellular Ca(2+), cAMP and reactive oxygen species (ROS). cAMP/PKA and extracellular signal regulated kinases (ERKs) are the known important signaling pathways primarily involved in PTP. Advanced proteomics approaches have revealed several proteins that undergo tyrosine phosphorylation during capacitation. Semen cryopreservation subjects spermatozoa to frequent stressors, which result in capacitation like changes (cryo-capacitation). The cryo-capacitated spermatozoa usually show different patterns of PTP than the normal in vitro capacitated spermatozoa. In the current manuscript, we have summarized some information about the proteins undergoing tyrosine phosphorylation during capacitation and the effect of cryopreservation on PTP as well as the possibilities to reduce the changes associated with cryopreservation process.
Collapse
Affiliation(s)
- Sai Naresh
- Reproductive Biochemistry Laboratory, Animal Biochemistry Division, National Dairy Research Institute, Karnal, Haryana 132001, India.
| | - Suresh Kumar Atreja
- Reproductive Biochemistry Laboratory, Animal Biochemistry Division, National Dairy Research Institute, Karnal, Haryana 132001, India.
| |
Collapse
|
33
|
Mizuno Y, Isono A, Kojima A, Arai MM, Noda T, Sakase M, Fukushima M, Harayama H. Distinct segment-specific functions of calyculin A-sensitive protein phosphatases in the regulation of cAMP-triggered events in ejaculated bull spermatozoa. Mol Reprod Dev 2015; 82:232-50. [PMID: 25735235 DOI: 10.1002/mrd.22465] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/02/2015] [Indexed: 12/19/2022]
Abstract
Livestock spermatozoa possess more tenacious suppressors of cAMP-triggered events-including capacitation-associated changes-than laboratory animal spermatozoa, leading to flagellar hyperactivation. In order to identify the suppressors, we examined effects of an inhibitor of serine/threonine protein phosphatases (calyculin A) on cAMP-triggered changes in the protein phosphorylation state, and subsequent occurrence of hyperactivation and acrosome reaction in ejaculated bull spermatozoa. Ejaculated spermatozoa were incubated in cAMP-supplemented medium, then assessed for motility, acrosome morphology, and phosphorylated protein localization. The addition of calyculin A greatly enhanced cAMP-triggered protein phosphorylation at serine/threonine and tyrosine residues in the connecting piece and induction of flagellar hyperactivation. Most hyperactivated spermatozoa exhibited extremely asymmetrical bends at the middle piece, which produced intensive twisting or figure-eight movements. In the sperm head, however, cAMP-triggered dephosphorylation of serine/threonine-phosphorylated proteins and subsequent acrosome reaction were abolished by the addition of calyculin A. Based on these results, we suggest that calyculin A-sensitive protein phosphatases in the connecting piece are suppressors of cAMP-triggered events leading to hyperactivation. By contrast, similar protein phosphatases in the sperm head accelerate cAMP-triggered events leading to the acrosome reaction. These findings are consistent with the indication that calyculin A-sensitive protein phosphatases have distinct functions in the regulation of cAMP-triggered events in different regions of ejaculated bull spermatozoa.
Collapse
Affiliation(s)
- Yohei Mizuno
- Laboratory of Reproductive Biology, Division of Animal Science, Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe, 657-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Marchiani S, Tamburrino L, Ricci B, Nosi D, Cambi M, Piomboni P, Belmonte G, Forti G, Muratori M, Baldi E. SUMO1 in human sperm: new targets, role in motility and morphology and relationship with DNA damage. Reproduction 2014; 148:453-67. [PMID: 25118297 DOI: 10.1530/rep-14-0173] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In studies carried out previously, we demonstrated that small ubiquitin-like modifier 1 (SUMO1) is associated with poor sperm motility when evaluated with a protocol that reveals mostly SUMO1-ylated live sperm. Recently, with another protocol, it has been demonstrated that SUMO is expressed in most sperm and is related to poor morphology and motility, suggesting that sumoylation may have multiple roles depending on its localisation and targets. We show herein, by confocal microscopy and co-immunoprecipitation, that dynamin-related protein 1 (DRP1), Ran GTPase-activating protein 1 (RanGAP1) and Topoisomerase IIα, SUMO1 targets in somatic and/or germ cells, are SUMO1-ylated in mature human spermatozoa. DRP1 co-localises with SUMO1 in the mid-piece, whereas RanGAP1 and Topoisomerase IIα in the post-acrosomal region of the head. Both SUMO1 expression and co-localisation with the three proteins were significantly higher in morphologically abnormal sperm, suggesting that sumoylation represents a marker of defective sperm. DRP1 sumoylation at the mid-piece level was higher in the sperm of asthenospermic men. As in somatic cells, DRP1 sumoylation is associated with mitochondrial alterations, this protein may represent the link between SUMO and poor motility. As SUMO pathways are involved in responses to DNA damage, another aim of our study was to investigate the relationship between sumoylation and sperm DNA fragmentation (SDF). By flow cytometry, we demonstrated that SUMO1-ylation and SDF are correlated (r=0.4, P<0.02, n=37) and most sumoylated sperm shows DNA damage in co-localisation analysis. When SDF was induced by stressful conditions (freezing and thawing and oxidative stress), SUMO1-ylation increased. Following freezing and thawing, SUMO1-Topoisomerase IIα co-localisation and co-immunoprecipitation increased, suggesting an involvement in the formation/repair of DNA breakage.
Collapse
Affiliation(s)
- S Marchiani
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - L Tamburrino
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - B Ricci
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - D Nosi
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - M Cambi
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - P Piomboni
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - G Belmonte
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - G Forti
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - M Muratori
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| | - E Baldi
- Department of BiomedicalExperimental and Clinical Sciences, Center of Excellence DeNotheDepartment of Experimental and Clinical MedicineUniversity of Florence, Viale Pieraccini 6, I-50139 Florence, ItalyDepartments of Molecular and Developmental MedicineMedicalSurgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
35
|
Xu K, Yang L, Zhao D, Wu Y, Qi H. AKAP3 synthesis is mediated by RNA binding proteins and PKA signaling during mouse spermiogenesis. Biol Reprod 2014; 90:119. [PMID: 24648398 DOI: 10.1095/biolreprod.113.116111] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mammalian spermatogenesis is regulated by coordinated gene expression in a spatiotemporal manner. The spatiotemporal regulation of major sperm proteins plays important roles during normal development of the male gamete, of which the underlying molecular mechanisms are poorly understood. A-kinase anchoring protein 3 (AKAP3) is one of the major components of the fibrous sheath of the sperm tail that is formed during spermiogenesis. In the present study, we analyzed the expression of sperm-specific Akap3 and the potential regulatory factors of its protein synthesis during mouse spermiogenesis. Results showed that the transcription of Akap3 precedes its protein synthesis by about 2 wk. Nascent AKAP3 was found to form protein complex with PKA and RNA binding proteins (RBPs), including PIWIL1, PABPC1, and NONO, as revealed by coimmunoprecipitation and protein mass spectrometry. RNA electrophoretic gel mobility shift assay showed that these RBPs bind sperm-specific mRNAs, of which proteins are synthesized during the elongating stage of spermiogenesis. Biochemical and cell biological experiments demonstrated that PIWIL1, PABPC1, and NONO interact with each other and colocalize in spermatids' RNA granule, the chromatoid body. In addition, NONO was found in extracytoplasmic granules in round spermatids, whereas PIWIL1 and PABPC1 were diffusely localized in cytoplasm of elongating spermatids, indicating their participation at different steps of mRNA metabolism during spermatogenesis. Interestingly, type I PKA subunits colocalize with PIWIL1 and PABPC1 in the cytoplasm of elongating spermatids and cosediment with the RBPs in polysomal fractions on sucrose gradients. Further biochemical analyses revealed that activation of PKA positively regulates AKAP3 protein synthesis without changing its mRNA level in elongating spermatids. Taken together, these results indicate that PKA signaling directly participates in the regulation of protein translation in postmeiotic male germ cells, underscoring molecular mechanisms that regulate protein synthesis during mouse spermiogenesis.
Collapse
Affiliation(s)
- Kaibiao Xu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Lele Yang
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Danyun Zhao
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yaoyao Wu
- Department of Biology, University of Science and Technology of China, Hefei, China
| | - Huayu Qi
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
36
|
Tamburrino L, Marchiani S, Minetti F, Forti G, Muratori M, Baldi E. The CatSper calcium channel in human sperm: relation with motility and involvement in progesterone-induced acrosome reaction. Hum Reprod 2014; 29:418-28. [PMID: 24430778 DOI: 10.1093/humrep/det454] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
STUDY QUESTION Does CatSper have a role in the achievement of human sperm motility and in the Progesterone (P)-induced acrosome reaction (AR)? SUMMARY ANSWER CatSper1 expression is associated with human sperm progressive motility and the P-induced AR; it may have a role in the pathogenesis of asthenozoospermia. WHAT IS KNOWN ALREADY Knockout mice for any of the Catsper family genes fail to acquire hyperactivated motility and are infertile. CatSper channels mediate P-induced Ca(2+) influx in human sperm. The role of CatSper in human sperm hyperactivated/activated motility and in asthenospermia is less clear. A few men with CatSper mutations have been described but the phenotype regarding sperm motility has not been well established. STUDY DESIGN, SIZE, DURATION The effects of two Catsper inhibitors, NNC55-0396 (NNC, 10 and 20 µM) and Mibefradil (Mib, 30 and 40 µM), were tested on human sperm motility parameters and the P-induced AR. Catsper1 protein expression was evaluated in unselected and swim-up selected sperm samples and in sperm from normo- and astheno-zoospermic subjects. PARTICIPANTS/MATERIALS, SETTING, METHODS Semen sample kinematic parameters were analysed by a CASA system. A fluorescent-labelled lectin was used to evaluate P-induced AR in live sperm by fluorescence microscopy. CatSper1 protein expression was determined by western blot analysis and by flow cytometry. Intracellular calcium concentrations ([Ca(2+)]i) were evaluated by a spectrofluorimetric method following sperm loading with the calcium-sensitive probe fura 2/AM. MAIN RESULTS AND THE ROLE OF CHANCE CatSper1 protein was localized in the tail of human sperm. CatSperI expression was higher in swim up selected than unselected sperm both when measured by western blot or by flow cytometry (52.7 ± 15.8% versus 27.2 ± 9.01%, n = 7, P < 0.01). Basal and P-stimulated [Ca(2+)]i were significantly higher in swim-up selected compared with unselected sperm. CatSper1 expression (western blot analysis) was found to be decreased in sperm from asthenozoospermic (n = 10) compared with those from normozoospermic (n = 9) men (intensity values relative to β-actin: 244.4 ± 69.3 versus 385.8 ± 139.5, P < 0.01). A positive correlation was found between CatSper1 protein expression and the percentage of sperm with progressive motility (n = 19, r = 0.59, P = 0.007). NNC (10 µM) and Mib (30 µM) significantly reduced the percentage of sperm with progressive motility and several kinematic parameters but did not affect the percentage of hyperactivated sperm. Their effects were the same whether they were added to swim-up selected and capacitated sperm or were added to the swim-up medium. Mib was found to have a slight but significant effect on sperm viability at both concentrations tested. P-stimulated AR was significantly reduced by both inhibitors (P < 0.05). Overall, our results indicate that, in human sperm, CatSper channel expression and function are associated with progressive motility and may be involved in the pathogenesis of asthenozoospermia. LIMITATIONS, REASONS FOR CAUTION In general, studies evaluating the effect of inhibitors have the limitation of the specificity of the molecules. We show here that Mib may have toxic effect on human sperm. Although most of the parameters have been evaluated in live sperm, the toxic effect could have contributed to the observed decreases. More studies are necessary to evaluate further the role of CatSper1 in asthenozoospermia. WIDER IMPLICATIONS OF THE FINDINGS In view of the involvement in P-induced AR and of the evidence of a role in the pathogenesis of astenozoospermia, CatSper channels may represent a promising target for the development of new drugs for the treatment of male infertility and for non-hormonal contraception. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from Ministry of University and Scientific Research (Prin project to E.B. and FIRB project to S.M) and Regione Toscana (to G.F.). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Lara Tamburrino
- Department of Biomedical, Experimental and Clinical Sciences, Centre of Excellence DeNothe, University of Florence, Florence, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Yu J, Zhou S, Jiang X, Bai J, Wang G. Knockdown of sAC affects sperm hyperactivation by cAMP-signaling pathway in male rat (Rattus norvegicus). ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-5928-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
38
|
Lachance C, Goupil S, Leclerc P. Stattic V, a STAT3 inhibitor, affects human spermatozoa through regulation of mitochondrial activity. J Cell Physiol 2013; 228:704-13. [PMID: 22911368 DOI: 10.1002/jcp.24215] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 08/08/2012] [Indexed: 11/10/2022]
Abstract
We have recently shown that many mediators of the JAK/STAT signaling pathway are present in ejaculated human spermatozoa. Among them, STAT3 is detected mainly in membranes and flagellar cytoskeletal fractions. In order to determine the importance of STAT3-mediated signaling, sperm were incubated with Stattic V, a specific inhibitor. Effects on motility were evaluated by CASA, sperm acrosomal integrity was evaluated by FITC conjugated lectin (PSA or PNA) staining, and protein phosphotyrosine content was assessed by Western blot using a monoclonal anti-phosphotyrosine antibody. INDO1-AM and JC-1 were used to measure sperm intracellular calcium and mitochondrial membrane potential, respectively, by flow cytometry, and reactive oxygen species (ROS) production was investigated by luminol-based assay. Percentages of motility and motility parameters were significantly affected by Stattic V. This later also significantly increased intracellular Ca(2+) levels, progesterone- and calcium ionophore (A23187)-induced acrosome reaction. On the other hand, a significant decrease in ATP content was measured when sperm were treated with Stattic V, associated with depolarization of mitochondrial membrane and elevated ROS production. These results suggest that STAT3 is involved in sperm functions, at least through regulation of mitochondrial activity. This further emphasizes that STAT3 mediates cellular activities in a manner different than strictly the activation of gene transcription.
Collapse
Affiliation(s)
- Catherine Lachance
- Département d'Obstétrique et de Gynécologie, Centre de recherche en biologie de la reproduction, Université Laval, Axe reproduction, santé périnatale et santé de l'enfant, Centre de recherche du CHUQ-CHUL, Québec, Canada
| | | | | |
Collapse
|
39
|
De Amicis F, Perrotta I, Santoro M, Guido C, Morelli C, Cesario MG, Bruno R, Aquila S. Human Sperm Anatomy: Different Expression and Localization of Phosphatidylinositol 3-Kinase in Normal and Varicocele Human Spermatozoa. Ultrastruct Pathol 2013; 37:176-82. [DOI: 10.3109/01913123.2013.763881] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Redgrove KA, Nixon B, Baker MA, Hetherington L, Baker G, Liu DY, Aitken RJ. The molecular chaperone HSPA2 plays a key role in regulating the expression of sperm surface receptors that mediate sperm-egg recognition. PLoS One 2012; 7:e50851. [PMID: 23209833 PMCID: PMC3510172 DOI: 10.1371/journal.pone.0050851] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 10/25/2012] [Indexed: 01/18/2023] Open
Abstract
A common defect encountered in the spermatozoa of male infertility patients is an idiopathic failure of sperm–egg recognition. In order to resolve the molecular basis of this condition we have compared the proteomic profiles of spermatozoa exhibiting an impaired capacity for sperm-egg recognition with normal cells using label free mass spectrometry (MS)-based quantification. This analysis indicated that impaired sperm–zona binding was associated with reduced expression of the molecular chaperone, heat shock 70 kDa protein 2 (HSPA2), from the sperm proteome. Western blot analysis confirmed this observation in independent patients and demonstrated that the defect did not extend to other members of the HSP70 family. HSPA2 was present in the acrosomal domain of human spermatozoa as a major component of 5 large molecular mass complexes, the most dominant of which was found to contain HSPA2 in close association with just two other proteins, sperm adhesion molecule 1 (SPAM1) and arylsulfatase A (ARSA), both of which that have previously been implicated in sperm-egg interaction. The interaction between SPAM1, ARSA and HSPA2 in a multimeric complex mediating sperm-egg interaction, coupled with the complete failure of this process when HSPA2 is depleted in infertile patients, provides new insights into the mechanisms by which sperm function is impaired in cases of male infertility.
Collapse
Affiliation(s)
- Kate A. Redgrove
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Brett Nixon
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Mark A. Baker
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Louise Hetherington
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Gordon Baker
- Melbourne In Vitro Fertilisation, East Melbourne, Victoria, Australia, and Department of Obstetrics and Gynaecology, The University of Melbourne, Royal Women’s Hospital, Carlton, Victoria, Australia
| | - De-Yi Liu
- Melbourne In Vitro Fertilisation, East Melbourne, Victoria, Australia, and Department of Obstetrics and Gynaecology, The University of Melbourne, Royal Women’s Hospital, Carlton, Victoria, Australia
| | - R. John Aitken
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, New South Wales, Australia
- * E-mail:
| |
Collapse
|
41
|
Ickowicz D, Finkelstein M, Breitbart H. Mechanism of sperm capacitation and the acrosome reaction: role of protein kinases. Asian J Androl 2012; 14:816-21. [PMID: 23001443 PMCID: PMC3720105 DOI: 10.1038/aja.2012.81] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/30/2012] [Accepted: 07/08/2012] [Indexed: 12/19/2022] Open
Abstract
Mammalian sperm must undergo a series of biochemical and physiological modifications, collectively called capacitation, in the female reproductive tract prior to the acrosome reaction (AR). The mechanisms of these modifications are not well characterized though protein kinases were shown to be involved in the regulation of intracellular Ca(2+) during both capacitation and the AR. In the present review, we summarize some of the signaling events that are involved in capacitation. During the capacitation process, phosphatidyl-inositol-3-kinase (PI3K) is phosphorylated/activated via a protein kinase A (PKA)-dependent cascade, and downregulated by protein kinase C α (PKCα). PKCα is active at the beginning of capacitation, resulting in PI3K inactivation. During capacitation, PKCα as well as PP1γ2 is degraded by a PKA-dependent mechanism, allowing the activation of PI3K. The activation of PKA during capacitation depends mainly on cyclic adenosine monophosphate (cAMP) produced by the bicarbonate-dependent soluble adenylyl cyclase. This activation of PKA leads to an increase in actin polymerization, an essential process for the development of hyperactivated motility, which is necessary for successful fertilization. Actin polymerization is mediated by PIP(2) in two ways: first, PIP(2) acts as a cofactor for phospholipase D (PLD) activation, and second, as a molecule that binds and inhibits actin-severing proteins such as gelsolin. Tyrosine phosphorylation of gelsolin during capacitation by Src family kinase (SFK) is also important for its inactivation. Prior to the AR, gelsolin is released from PIP(2) and undergoes dephosphorylation/activation, resulting in fast F-actin depolymerization, leading to the AR.
Collapse
Affiliation(s)
- Debby Ickowicz
- The Mina & Everard Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52100, Israel
| | | | | |
Collapse
|
42
|
Sperm DNA fragmentation induced by cryopreservation: new insights and effect of a natural extract from Opuntia ficus-indica. Fertil Steril 2012; 98:326-33. [DOI: 10.1016/j.fertnstert.2012.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 05/02/2012] [Accepted: 05/02/2012] [Indexed: 11/23/2022]
|
43
|
Li YF, He W, Mandal A, Kim YH, Digilio L, Klotz K, Flickinger CJ, Herr JC, Herr JC. CABYR binds to AKAP3 and Ropporin in the human sperm fibrous sheath. Asian J Androl 2011; 13:266-74. [PMID: 21240291 DOI: 10.1038/aja.2010.149] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Calcium-binding tyrosine phosphorylation-regulated protein (CABYR) is a highly polymorphic calcium-binding tyrosine- and serine-/threonine-phosphorylated fibrous sheath (FS) protein involved in capacitation. A putative domain (amino acids 12-48) homologous to the regulatory subunit of type II cAMP-dependent protein kinase A (RII) dimerisation and A kinase-anchoring protein (AKAP)-binding domains of protein kinase A at the N-terminus suggests that CABYR may self-assemble and bind to AKAPs. Moreover, there is evidence that CABYR has limited interaction with AKAPs. However, further evidence and new relationships between CABYR and other FS proteins, including AKAPs, will be helpful in understanding the basic physiology of FS. In this study, a new strategy for co-immunoprecipitation of insoluble proteins, as well as the standard co-immunoprecipitation method in combination with mass spectrometry and western blot, was employed to explore the relationship between CABYR, AKAP3 and Ropporin. The results showed that AKAP3 was co-immunoprecipitated with CABYR by the anti-CABYR-A polyclonal antibody, and, conversely, CABYR was also co-immunoprecipitated with AKAP3 by the anti-AKAP3 polyclonal antibody. Another RII-like domain containing protein, Ropporin, was also co-immunoprecipitated with CABYR, indicating that Ropporin is one of CABYR's binding partners. The interactions between CABYR, AKAP3 and Ropporin were confirmed by yeast two-hybrid assays. Further analysis showed that CABYR not only binds to AKAP3 by its RII domain but binds to Ropporin through other regions besides the RII-like domain. This is the first demonstration that CABYR variants form a complex not only with the scaffolding protein AKAP3 but also with another RII-like domain-containing protein in the human sperm FS.
Collapse
Affiliation(s)
- Yan-Feng Li
- Department of Urology, Daping Hospital, Institute of Surgery Research, Third Military Medical University, Chongqing 400042, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Identification of capacitation associated tyrosine phosphoproteins in buffalo (Bubalus bubalis) and cattle spermatozoa. Anim Reprod Sci 2011; 123:40-7. [DOI: 10.1016/j.anireprosci.2010.11.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 10/28/2010] [Accepted: 11/25/2010] [Indexed: 11/22/2022]
|
45
|
De Amicis F, Guido C, Perrotta I, Avena P, Panza S, Andò S, Aquila S. Conventional progesterone receptors (PR) B and PRA are expressed in human spermatozoa and may be involved in the pathophysiology of varicocoele: a role for progesterone in metabolism. ACTA ACUST UNITED AC 2010; 34:430-45. [PMID: 20946440 DOI: 10.1111/j.1365-2605.2010.01111.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The physiological roles of intracellular progesterone (PRG) receptors (PRs) have been studied intensively in female mammals, while their functions in male are scarce. Conventional PRs were evidenced in our study by Western blotting, concomitantly in healthy spermatozoa and in oligoasthenoteratozoospermic samples without and with varicocoele. Transmission electron microscopy revealed the presence of the PRs on the membrane as well as in the nucleus, mitochondria and flagellum. A reduced expression of the PRs was observed only in varicocoele spermatozoa. Responses to PRG treatment on cholesterol efflux, tyrosine phosphorylation, src and Akt activities, acrosin activity and acrosome reaction in varicocoele spermatozoa were reduced or absent. To further investigate PRG significance in human male gamete, we focused its action on lipid and glucose metabolism. The evaluation of the triglycerides content, lipase and acyl-CoA dehydrogenase activities suggests that PRG through the PRs exerts a lipolytic effect on human spermatozoa. An increase in glucose-6-phosphate dehydrogenase activity was also obtained, evidencing a role for PRG on glucose metabolism. In 'varicocoele' spermatozoa, the PRG did not induce energy consumption. The action of PRs on sperm metabolism is a novel finding that renews the importance of PRG in male fertility. Our results showed that varicocoele may lead to male factor infertility by a mechanism involving a decreased PR expression in human spermatozoa that evidences a detrimental effect on spermatozoa at the molecular level, going beyond the abnormal sperm morphology described to date.
Collapse
Affiliation(s)
- F De Amicis
- Centro Sanitario Department of Pharmaco-Biology, University of Calabria, Arcavacata di Rende (CS), Italy
| | | | | | | | | | | | | |
Collapse
|
46
|
Marchiani S, Tamburrino L, Giuliano L, Nosi D, Sarli V, Gandini L, Piomboni P, Belmonte G, Forti G, Baldi E, Muratori M. Sumo1-ylation of human spermatozoa and its relationship with semen quality. ACTA ACUST UNITED AC 2010; 34:581-93. [DOI: 10.1111/j.1365-2605.2010.01118.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
47
|
Liu B, Wang P, Wang Z, Jia Y, Niu X, Wang W, Zhang W. Analysis and difference of voltage-dependent anion channel mRNA in ejaculated spermatozoa from normozoospermic fertile donors and infertile patients with idiopathic asthenozoospermia. J Assist Reprod Genet 2010; 27:719-24. [PMID: 20809416 DOI: 10.1007/s10815-010-9466-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 08/12/2010] [Indexed: 11/25/2022] Open
Abstract
PURPOSE to analyze the abundance and difference of voltage-dependent anion channel (VDAC) mRNA in ejaculated spermatozoa from normozoospermic fertile donors and infertile patients with idiopathic asthenozoospermia. METHODS high motile and low motile spermatozoa were separated respectively from ejaculates of 36 donors and 40 patients using a discontinuous Percoll gradient centrifugation. Real-Time PCR was performed to detect mRNA abundance and difference of three VDAC subtypes between two groups with different sperm motility. RESULTS real-Time PCR demonstrated that three VDAC mRNAs were present in mature spermatozoa. The VDAC2 mRNA level in ejaculated spermatozoa of patients was significantly higher than that of donors. No significant differences of VDAC1 and VDAC3 mRNA levels were found between two groups. CONCLUSION the high abundance of VDAC2 mRNA seemed to have a positive correlation with low sperm motility. The abnormal expression of VDAC might be related to male infertility with idiopathic asthenozoospermia.
Collapse
Affiliation(s)
- Bianjiang Liu
- Laboratory of Reproductive Medicine, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | |
Collapse
|
48
|
Purification and characterization of a sperm motility inhibiting factor from caprine epididymal plasma. PLoS One 2010; 5:e12039. [PMID: 20706623 PMCID: PMC2919373 DOI: 10.1371/journal.pone.0012039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 07/16/2010] [Indexed: 11/20/2022] Open
Abstract
Several studies have been reported on the occurrence of sperm motility inhibiting factors in the male reproductive fluids of different mammalian species, but these proteins have not been adequately purified and characterized. A novel sperm motility inhibiting factor (MIF-II) has been purified from caprine epididymal plasma (EP) by Hydroxylapatite gel adsorption chromatography, DEAE-Cellulose ion-exchange chromatography and chromatofocusing. The MIF-II has been purified to apparent homogeneity and the molecular weight estimated by Sephacryl S-300 gel filtration is 160 kDa. MIF-II is a dimeric protein, made up of two subunits each having a molecular mass of 80 kDa as shown by SDS-PAGE. The isoelectric point of MIF-II is 5.1 as determined by chromatofocusing and isoelectric focusing. It is a heat labile protein and maximal active at the pH 6.9 to 7.5. The sperm motility inhibiting protein factor at 2 µg/ml (12.5 nM) level showed maximal motility-inhibiting activity. The observation that the epididymal plasma factor lowered the intracellular cAMP level of spermatozoa in a concentration-dependent manner suggests that it may block the motility of caprine cauda spermatozoa by interfering the cAMP dependent motility function. The results revealed that the purified protein factor has the potential of sperm motility inhibition and may serve as a vaginal contraceptive. The antibody raised against the MIF-II has the potential for enhancement of forward motility of cauda-spermatozoa. This antibody may thus be useful for solving some of the problems of male infertility due to low sperm motility.
Collapse
|
49
|
Barbonetti A, Vassallo MRC, Cordeschi G, Venetis D, Carboni A, Sperandio A, Felzani G, Francavilla S, Francavilla F. Protein tyrosine phosphorylation of the human sperm head during capacitation: immunolocalization and relationship with acquisition of sperm-fertilizing ability. Asian J Androl 2010; 12:853-61. [PMID: 20694019 DOI: 10.1038/aja.2010.52] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The occurrence of tyrosine phosphorylation (TP) in the sperm head during capacitation has been poorly investigated, and no data exist on the relationship of its dynamics with the acquisition of sperm fertilizing ability. This study localized TP of head proteins in human spermatozoa during capacitation and explored its relationship with acquisition of the ability to display progesterone (P)-stimulated acrosome reactions (ARs) and to penetrate zona-free hamster oocytes. By immunofluorescence, TP immunoreactivity was revealed in the acrosomal region of formaldehyde-fixed/unpermeabilized samples, whereas it was abolished in fixed/permeabilized samples, in which TP immunoreactivity was high in the principal piece. No TP immunoreactivity was detectable in unfixed spermatozoa. Head TP immunoreactivity was localized externally to the acrosome, close to the cytoplasmic membrane, as assessed by transmission electron microscopy. The increase in head TP was an early event during capacitation, occurring within 1 h in capacitating conditions. At this time, the P-stimulated ARs were also increased, whereas egg penetration was as poor as in uncapacitated spermatozoa. At 5 h of capacitation, the extent of neither head TP nor the P-induced ARs were greater than that at 1 h, whereas egg penetration had significantly increased. Seminal plasma inhibited head TP, P-induced ARs and egg penetration. None of these inhibitory effects, unlike those on tail TP, were prevented by the cAMP analogue dbcAMP (N,2-O-dibutyryladenosine 3',5'-cyclic monophosphate). In conclusion, head TP is a subsurface event occurring early during capacitation and is closely related to acquisition of the ability to display P-stimulated ARs, whereas the ability to fuse with oolemma and to decondense is a later capacitation-related event.
Collapse
Affiliation(s)
- Arcangelo Barbonetti
- Andrology Unit, Department of Internal Medicine, University of L'Aquila, Coppito 67100, L'Aquila, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nixon B, Bielanowicz A, Anderson AL, Walsh A, Hall T, Mccloghry A, Aitken RJ. Elucidation of the signaling pathways that underpin capacitation-associated surface phosphotyrosine expression in mouse spermatozoa. J Cell Physiol 2010; 224:71-83. [PMID: 20232304 DOI: 10.1002/jcp.22090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent studies from within our laboratory have demonstrated a causal relationship between capacitation-associated surface phosphotyrosine expression and the ability of mouse spermatozoa to recognize the oocyte and engage in sperm-zona pellucida interaction. In the studies described herein we have sought to investigate the signaling pathways that underpin the tyrosine phosphorylation of sperm surface protein targets and validate the physiological significance of these pathways in relation to sperm-zona pellucida adhesion. Through selective pharmacological inhibition we have demonstrated that surface phosphotyrosine expression is unlikely to be mediated by the canonical cAMP-dependent protein kinase A (PKA) signaling cascade that has been most widely studied in relation to sperm capacitation. Rather, it appears to be primarily driven by the extracellular signal-regulated kinase (ERK) module of the mitogen-activated protein kinase (MAPK) pathway. Consistent with this notion, the main components of the ERK module (RAS, RAF1, MEK, and ERK1/2) were localized to the periacrosomal region of the head of mature mouse spermatozoa and their phosphorylation status within this region of the cell was positively modulated by capacitation. Furthermore, inhibition of several elements of this pathway suppressed sperm surface phosphotyrosine expression and induced a concomitant reduction sperm-zona pellucida interaction. Collectively, these data highlight a previously unappreciated role of the ERK module in the modification of the sperm surface during capacitation to render these cells functionally competent to engage in the process of fertilization.
Collapse
Affiliation(s)
- Brett Nixon
- Reproductive Science Group, School of Environmental and Life Sciences, University of Newcastle, Callaghan, Australia.
| | | | | | | | | | | | | |
Collapse
|