1
|
Khaw SC, Martins da Silva S. From Bench to Babies - Drug Development for Male Subfertility. REPRODUCTION AND FERTILITY 2024; 5:RAF-24-0022. [PMID: 39226216 PMCID: PMC11466258 DOI: 10.1530/raf-24-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/03/2024] [Indexed: 09/05/2024] Open
Abstract
Infertility is estimated to affect more than 50 million couples around the world, with male factor accounting for half of these cases, yet there is a notable absence of effective treatment options for men, other than in-vitro fertilisation (IVF) or intracytoplasmic sperm injection (ICSI). This review considers unlicensed and empirical treatments used for male subfertility, including hormonal therapy, phosphodiesterase inhibitors, and antioxidants. Compounds generally demonstrate variable improvements in sperm function but benefits for fertility are less clear. There is a pressing need for effective treatment options for subfertile men, however, our knowledge of sperm function is limited, restricting the identification of precise treatment targets. The traditional drug discovery pathway is also notorious for its extensive resource and time requirements, often extending over decades and demanding significant financial investment. Unfortunately, a substantial number of potential therapies fail before reaching the marketplace. Furthermore, reliance on mammalian models is not possible in the drug development process for male subfertility, due to significant variability between animals and man. We review recent breakthroughs and highlight novel methods aimed at improving the effectiveness and efficiency of drug discovery for male subfertility. High-throughput screening, combinatorial chemistry, and the repurposing of established medications have great potential. These strategies offer the promise of accelerating the pace of drug development, curbing the extensive demand for resources, and, in the case of drug repurposing, diminish the demand for comprehensive pharmacokinetic and pharmacodynamic studies. As these innovative approaches are adopted, the feasibility of addressing male subfertility through scientific advancements appears to be increasingly attainable.
Collapse
Affiliation(s)
- Shen Chuen Khaw
- S Khaw, Reproductive Medicine Research Group, University of Dundee, Dundee, United Kingdom of Great Britain and Northern Ireland
| | - Sarah Martins da Silva
- S Martins da Silva, Reproductive Medicine Research Group, University of Dundee, Dundee, DD1 9SY, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
2
|
Lorenz J, Eisenhardt C, Mittermair T, Kulle AE, Holterhus PM, Fobker M, Boenigk W, Nordhoff V, Behre HM, Strünker T, Brenker C. The sperm-specific K + channel Slo3 is inhibited by albumin and steroids contained in reproductive fluids. Front Cell Dev Biol 2024; 12:1275116. [PMID: 39310227 PMCID: PMC11413451 DOI: 10.3389/fcell.2024.1275116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 07/19/2024] [Indexed: 09/25/2024] Open
Abstract
To locate and fertilize the egg, sperm probe the varying microenvironment prevailing at different stages during their journey across the female genital tract. To this end, they are equipped with a unique repertoire of mostly sperm-specific proteins. In particular, the flagellar Ca2+ channel CatSper has come into focus as a polymodal sensor used by human sperm to register ligands released into the female genital tract. Here, we provide the first comprehensive study on the pharmacology of the sperm-specific human Slo3 channel, shedding light on its modulation by reproductive fluids and their constituents. We show that seminal fluid and contained prostaglandins and Zn2+ do not affect the channel, whereas human Slo3 is inhibited in a non-genomic fashion by diverse steroids as well as by albumin, which are released into the oviduct along with the egg. This indicates that not only CatSper but also Slo3 harbours promiscuous ligand-binding sites that can accommodate structurally diverse molecules, suggesting that Slo3 is involved in chemosensory signalling in human sperm.
Collapse
Affiliation(s)
- Johannes Lorenz
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Clara Eisenhardt
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Teresa Mittermair
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Alexandra E. Kulle
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Christian-Albrechts-University, Kiel, Germany
| | - Paul Martin Holterhus
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Christian-Albrechts-University, Kiel, Germany
| | - Manfred Fobker
- Center for Laboratory Medicine, University Hospital, Münster, Germany
| | - Wolfgang Boenigk
- Max Planck Institute for Neurobiology of Behaviour—Caesar, Bonn, Germany
| | - Verena Nordhoff
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | | | - Timo Strünker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Christoph Brenker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| |
Collapse
|
3
|
He Y, Wang B, Huang J, Zhang D, Yuan Y. Environmental pollutants and male infertility: Effects on CatSper. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116341. [PMID: 38653022 DOI: 10.1016/j.ecoenv.2024.116341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Infertility is a growing health concern among many couples worldwide. Men account for half of infertility cases. CatSper, a sperm-specific Ca2+ channel, is expressed on the cell membrane of mammalian sperm. CatSper plays an important role in male fertility because it facilitates the entry of Ca2+ necessary for the rapid change in sperm motility, thereby allowing it to navigate the hurdles of the female reproductive tract and successfully locate the egg. Many pollutants present in the environment have been shown to affect the functions of CatSper and sperm, which is a matter of capital importance to understanding and solving male infertility issues. Environmental pollutants can act as partial agonists or inhibitors of CatSper or exhibit a synergistic effect. In this article, we briefly describe the structure, functions, and regulatory mechanisms of CatSper, and discuss the body of literature covering the effects of environmental pollutants on CatSper.
Collapse
Affiliation(s)
- Yuxin He
- Nanchang University Queen Mary School, Jiangxi Medical College of Nanchang University, Nanchang 330031, China
| | - Binhui Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Jian Huang
- Clinical Medical Experimental Center, Nanchang University, Nanchang 330031, China; Jiangxi Provincial Key Laboratory of Reproductive Physiology and Pathology, Nanchang University, Nanchang 330006, China
| | - Dalei Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China; Jiangxi Provincial Key Laboratory of Reproductive Physiology and Pathology, Nanchang University, Nanchang 330006, China
| | - Yangyang Yuan
- Clinical Medical Experimental Center, Nanchang University, Nanchang 330031, China; Jiangxi Provincial Key Laboratory of Reproductive Physiology and Pathology, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
4
|
Zhang Z, Xiong Y, Jiang H, Wang Q, Hu X, Wei X, Chen Q, Chen T. Vaginal extracellular vesicles impair fertility in endometriosis by favoring Th17/Treg imbalance and inhibiting sperm activity. J Cell Physiol 2024; 239:e31188. [PMID: 38192157 DOI: 10.1002/jcp.31188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/05/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024]
Abstract
Extracellular vesicles (EVs) play a key role in various diseases. However, their effect on endometriosis (EMs)-associated infertility is poorly understood. We co-cultured EVs from the female vaginal secretions with human sperm and also generated a mouse model of EMs by allogenic transplant to explore the effect of EVs on fertility. EVs from individuals with EMs-associated infertility (E-EVs) significantly inhibited the total motility (26.46% vs. 47.1%), progressive motility (18.78% vs. 41.06%), linear velocity (21.98 vs. 41.91 µm/s) and the acrosome reaction (AR) rate (5% vs. 22.3%) of human sperm in contrast to the control group (PBS). Furthermore, E-EVs dose-dependently decreased the intracellular Ca2+ ([Ca2+]i), a pivotal regulator of sperm function. Conversely, healthy women (H-EVs) increased human sperm motion parameters, the AR rate, and sperm [Ca2+]i. Importantly, the mouse model of EMs confirmed that E-EVs further decreased the conception rate and the mean number of embryo implantations (7.6 ± 3.06 vs. 4.5 ± 3.21) compared with the control mice by inducing the production of inflammatory cytokines leading to a Th17/Treg imbalance. H-EVs could restore impaired fertility by restoring the Th17/Treg balance. We determined the impact of EVs derived from the female genital tract on human sperm function and studied the possible mechanisms by which it affects fertility. Our findings provide a novel rationale to ameliorate EMs-associated infertility.
Collapse
Affiliation(s)
- Zuo Zhang
- Department of Obstetrics and Gynecology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yangbai Xiong
- International Tourism and Convention Management, Hong Kong Polytechnic University, Hong Kong, China
| | - Huifu Jiang
- Department of Obstetrics and Gynecology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qian Wang
- Department of Obstetrics and Gynecology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinyue Hu
- Department of Obstetrics and Gynecology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xin Wei
- Department of Obstetrics and Gynecology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qi Chen
- Department of Obstetrics and Gynecology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Tingtao Chen
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Gruber FS, Richardson A, Johnston ZC, Myles R, Norcross NR, Day DP, Georgiou I, Sesma-Sanz L, Wilson C, Read KD, Martins da Silva S, Barratt CLR, Gilbert IH, Swedlow JR. Sperm Toolbox-A selection of small molecules to study human spermatozoa. PLoS One 2024; 19:e0297666. [PMID: 38377053 PMCID: PMC10878532 DOI: 10.1371/journal.pone.0297666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Male contraceptive options and infertility treatments are limited, and almost all innovation has been limited to updates to medically assisted reproduction protocols and methods. To accelerate the development of drugs that can either improve or inhibit fertility, we established a small molecule library as a toolbox for assay development and screening campaigns using human spermatozoa. We have profiled all compounds in the Sperm Toolbox in several automated high-throughput assays that measure stimulation or inhibition of sperm motility or the acrosome reaction. We have assayed motility under non-capacitating and capacitating conditions to distinguish between pathways operating under these different physiological states. We also assayed cell viability to ensure any effects on sperm function are specific. A key advantage of our studies is that all compounds are assayed together in the same experimental conditions, which allows quantitative comparisons of their effects in complementary functional assays. We have combined the resulting datasets to generate fingerprints of the Sperm Toolbox compounds on sperm function. The data are included in an on-line R-based app for convenient querying.
Collapse
Affiliation(s)
- Franz S. Gruber
- Divisions of Computational Biology and Molecular, Cell and Developmental Biology, and National Phenotypic Screening Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Anthony Richardson
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Zoe C. Johnston
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Rachel Myles
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Neil R. Norcross
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - David P. Day
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Irene Georgiou
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Laura Sesma-Sanz
- Divisions of Computational Biology and Molecular, Cell and Developmental Biology, and National Phenotypic Screening Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Caroline Wilson
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Kevin D. Read
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sarah Martins da Silva
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Christopher L. R. Barratt
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Ian H. Gilbert
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Jason R. Swedlow
- Divisions of Computational Biology and Molecular, Cell and Developmental Biology, and National Phenotypic Screening Centre, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
6
|
Grahn E, Kaufmann SV, Askarova M, Ninov M, Welp LM, Berger TK, Urlaub H, Kaupp UB. Control of intracellular pH and bicarbonate by CO 2 diffusion into human sperm. Nat Commun 2023; 14:5395. [PMID: 37669933 PMCID: PMC10480191 DOI: 10.1038/s41467-023-40855-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 08/14/2023] [Indexed: 09/07/2023] Open
Abstract
The reaction of CO2 with H2O to form bicarbonate (HCO3-) and H+ controls sperm motility and fertilization via HCO3--stimulated cAMP synthesis. A complex network of signaling proteins participates in this reaction. Here, we identify key players that regulate intracellular pH (pHi) and HCO3- in human sperm by quantitative mass spectrometry (MS) and kinetic patch-clamp fluorometry. The resting pHi is set by amiloride-sensitive Na+/H+ exchange. The sperm-specific putative Na+/H+ exchanger SLC9C1, unlike its sea urchin homologue, is not gated by voltage or cAMP. Transporters and channels implied in HCO3- transport are not detected, and may be present at copy numbers < 10 molecules/sperm cell. Instead, HCO3- is produced by diffusion of CO2 into cells and readjustment of the CO2/HCO3-/H+ equilibrium. The proton channel Hv1 may serve as a unidirectional valve that blunts the acidification ensuing from HCO3- synthesis. This work provides a new framework for the study of male infertility.
Collapse
Affiliation(s)
- Elena Grahn
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Svenja V Kaufmann
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Malika Askarova
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Momchil Ninov
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Luisa M Welp
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Thomas K Berger
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Deutschhausstrasse 1-2, 35037, Marburg, Germany.
| | - Henning Urlaub
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany.
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
- Cluster of Excellence, Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Göttingen, Germany.
| | - U Benjamin Kaupp
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
- Life & Medical Sciences Institute (LIMES), University Bonn, Carl-Troll-Strasse 31, 53115, Bonn, Germany.
| |
Collapse
|
7
|
Ke S, Luo T. The Chemosensing Role of CatSper in Mammalian Sperm: An Updated Review. Curr Issues Mol Biol 2023; 45:6995-7010. [PMID: 37754226 PMCID: PMC10528052 DOI: 10.3390/cimb45090442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
After sperm enter the female reproductive tract, the physicochemical and biochemical microenvironment undergoes significant changes. In particular, the large changes in various ions encountered by sperm may alter the physiology of sperm, ultimately compromising capacitation and fertilization. Thus, the rapid response to environmental variations is vital for sperm functions. For example, Calcium, the most crucial ion for sperm functions, enters into sperm via Ca2+ permeable ion channels. The cation channel of sperm (CatSper) is a sperm-specific, pH-sensitive, and Ca2+-permeable ion channel. It is responsible for the predominant Ca2+ entry in mammalian sperm and is involved in nearly every event of sperm to acquire fertilizing capability. In addition, CatSper also serves as a pivotal polymodal chemosensor in mammalian sperm by responding to multiple chemical cues. Physiological chemicals (such as progesterone, prostaglandins, β-defensins, and odorants) provoke Ca2+ entry into sperm by activating CatSper and thus triggering sperm functions. Additionally, synthetic and natural chemicals (such as medicines, endocrine disrupting chemicals, drugs of abuse, and antioxidants) affect sperm functions by regulating CatSper-dependent Ca2+ signaling. Therefore, understanding the interactions between CatSper and extracellular ligands sheds light on the mechanisms underlying male infertility and offers innovative diagnostic and treatment approaches. This underscores the importance of CatSper as a crucial regulatory target in male reproduction, linking sperm function with the extracellular environment. In conclusion, this review comprehensively summarizes the relevant studies describing the environmental factors that affect CatSper in humans and rodents.
Collapse
Affiliation(s)
- Sulun Ke
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- Queen Mary School, Medical College, Nanchang University, Nanchang 330031, China
| | - Tao Luo
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- Key Laboratory of Reproductive Physiology and Pathology in Jiangxi Province, Nanchang University, Nanchang 330006, China
| |
Collapse
|
8
|
Wendler A, Wehling M. Many or too many progesterone membrane receptors? Clinical implications. Trends Endocrinol Metab 2022; 33:850-868. [PMID: 36384863 DOI: 10.1016/j.tem.2022.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/15/2022]
Abstract
Several receptors for nongenomically initiated actions of progesterone (P4) exist, namely membrane-associated P4 receptors (MAPRs), membrane progestin receptors (mPRs), receptors for neurosteroids [GABAA receptor (GABAAR), NMDA receptor, sigma-1 and -2 receptors (S1R/S2R)], the classical genomic P4 receptor (PGR), and α/β hydrolase domain-containing protein 2 (ABHD2). Two drugs related to this field have been approved: brexanolone (Zulresso™) for the treatment of postpartum depression, and ganaxolone (Ztalmy™) for the treatment of CDKL5 deficiency disorder. Both are derivatives of P4 and target the GABAAR. Several other indications are in clinical testing. CT1812 (Elayta™) is also being tested for the treatment of Alzheimer's disease (AD) in Phase 2 clinical trials, targeting the P4 receptor membrane component 1 (PGRMC1)/S2R complex. In this Review, we highlight emerging knowledge on the mechanisms of nongenomically initiated actions of P4 and its derivatives.
Collapse
Affiliation(s)
- Alexandra Wendler
- Clinical Pharmacology Mannheim, Faculty of Medicine Mannheim, Ruprecht-Karls-University of Heidelberg, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany
| | - Martin Wehling
- Clinical Pharmacology Mannheim, Faculty of Medicine Mannheim, Ruprecht-Karls-University of Heidelberg, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany.
| |
Collapse
|
9
|
Prajapati P, Kane S, McBrinn RC, Dean MS, Martins da Silva SJ, Brown SG. Elevated and Sustained Intracellular Calcium Signalling Is Necessary for Efficacious Induction of the Human Sperm Acrosome Reaction. Int J Mol Sci 2022; 23:ijms231911253. [PMID: 36232560 PMCID: PMC9570455 DOI: 10.3390/ijms231911253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Progesterone and prostaglandin E1 are postulated to trigger the human sperm acrosome reaction (AR). However, their reported efficacy is very variable which likely, in part, reflects the plethora of experimental conditions and methodologies used to detect this physiologically relevant event. The purpose of this study was to develop an assay for the robust induction and objective measurement of the complete AR. Sperm from healthy volunteers or patients undertaking IVF were treated with a variety of ligands (progesterone, prostaglandin E1 or NH4Cl, alone or in combinations). AR, motility and intracellular calcium measurements were measured using flow cytometry, computer-assisted sperm analysis (CASA) and fluorimetry, respectively. The AR was significantly increased by the simultaneous application of progesterone, prostaglandin E1 and NH4Cl, following an elevated and sustained intracellular calcium concentration. However, we observed notable inter- and intra-donor sample heterogeneity of the AR induction. When studying the patient samples, we found no relationship between the IVF fertilization rate and the AR. We conclude that progesterone and prostaglandin E1 alone do not significantly increase the percentage of live acrosome-reacted sperm. This assay has utility for drug discovery and sperm toxicology studies but is not predictive for IVF success.
Collapse
Affiliation(s)
- Priyanka Prajapati
- Reproductive Medicine Research Group, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Shruti Kane
- School of Applied Sciences, Abertay University, Dundee DD1 1HG, UK
| | | | - Morven S. Dean
- Reproductive Medicine Research Group, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
- Assisted Conception Unit, Ninewells Hospital, Dundee DD1 9SY, UK
| | - Sarah J. Martins da Silva
- Reproductive Medicine Research Group, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
- Assisted Conception Unit, Ninewells Hospital, Dundee DD1 9SY, UK
- Correspondence:
| | - Sean G. Brown
- School of Applied Sciences, Abertay University, Dundee DD1 1HG, UK
| |
Collapse
|
10
|
Cavarocchi E, Whitfield M, Saez F, Touré A. Sperm Ion Transporters and Channels in Human Asthenozoospermia: Genetic Etiology, Lessons from Animal Models, and Clinical Perspectives. Int J Mol Sci 2022; 23:ijms23073926. [PMID: 35409285 PMCID: PMC8999829 DOI: 10.3390/ijms23073926] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/18/2022] Open
Abstract
In mammals, sperm fertilization potential relies on efficient progression within the female genital tract to reach and fertilize the oocyte. This fundamental property is supported by the flagellum, an evolutionarily conserved organelle that provides the mechanical force for sperm propulsion and motility. Importantly several functional maturation events that occur during the journey of the sperm cells through the genital tracts are necessary for the activation of flagellar beating and the acquisition of fertilization potential. Ion transporters and channels located at the surface of the sperm cells have been demonstrated to be involved in these processes, in particular, through the activation of downstream signaling pathways and the promotion of novel biochemical and electrophysiological properties in the sperm cells. We performed a systematic literature review to describe the currently known genetic alterations in humans that affect sperm ion transporters and channels and result in asthenozoospermia, a pathophysiological condition defined by reduced or absent sperm motility and observed in nearly 80% of infertile men. We also present the physiological relevance and functional mechanisms of additional ion channels identified in the mouse. Finally, considering the state-of-the art, we discuss future perspectives in terms of therapeutics of asthenozoospermia and male contraception.
Collapse
Affiliation(s)
- Emma Cavarocchi
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France; (E.C.); (M.W.)
| | - Marjorie Whitfield
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France; (E.C.); (M.W.)
| | - Fabrice Saez
- UMR GReD Institute (Génétique Reproduction & Développement) CNRS 6293, INSERM U1103, Team «Mécanismes de L’Infertilité Mâle Post-Testiculaire», Université Clermont Auvergne, 63000 Clermont-Ferrand, France
- Correspondence: (F.S.); (A.T.)
| | - Aminata Touré
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France; (E.C.); (M.W.)
- Correspondence: (F.S.); (A.T.)
| |
Collapse
|
11
|
Gruber FS, Johnston ZC, Norcross NR, Georgiou I, Wilson C, Read KD, Gilbert IH, Swedlow JR, Martins da Silva S, Barratt CLR. Compounds enhancing human sperm motility identified using a high-throughput phenotypic screening platform. Hum Reprod 2022; 37:466-475. [PMID: 35048946 PMCID: PMC8888995 DOI: 10.1093/humrep/deac007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/04/2021] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION Can a high-throughput screening (HTS) platform facilitate male fertility drug discovery? SUMMARY ANSWER An HTS platform identified a large number of compounds that enhanced sperm motility. WHAT IS KNOWN ALREADY Several efforts to find small molecules modulating sperm function have been performed but none have used high-throughput technology. STUDY DESIGN, SIZE, DURATION Healthy donor semen samples were used and samples were pooled (3-5 donors per pool). Primary screening was performed singly; dose-response screening was performed in duplicate (using independent donor pools). PARTICIPANTS/MATERIALS, SETTING, METHODS Spermatozoa isolated from healthy donors were prepared by density gradient centrifugation and incubated in 384-well plates with compounds (6.25 μM) to identify those compounds with enhancing effects on motility. Approximately 17 000 compounds from the libraries, ReFRAME, Prestwick, Tocris, LOPAC, CLOUD and MMV Pathogen Box, were screened. Dose-response experiments of screening hits were performed to confirm the enhancing effect on sperm motility. Experiments were performed in a university setting. MAIN RESULTS AND THE ROLE OF CHANCE From our primary single concentration screening, 105 compounds elicited an enhancing effect on sperm motility compared to dimethylsulphoxide-treated wells. Confirmed enhancing compounds were grouped based on their annotated targets/target classes. A major target class, phosphodiesterase inhibitors, were identified, in particular PDE10A inhibitors as well as number of compounds not previously known to enhance human sperm motility, such as those related to GABA signalling. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Although this approach provides data about the activity of the compound, it is only a starting point. For example, further substantive experiments are necessary to provide a more comprehensive picture of each compound's activity, the effect on the kinetics of the cell populations and subpopulations, and their potential mechanisms of action. Compounds have been tested with prepared donor spermatozoa, incubated under non-capacitating conditions, and only incubated with compounds for a relatively short period of time. Therefore, the effect of compounds under different conditions, for example in whole semen, for longer incubation times, or using samples from patient groups, may be different and require further study. All experiments were performed in vitro. WIDER IMPLICATIONS OF THE FINDINGS This phenotypic screening assay identified a large number of compounds that increased sperm motility. In addition to furthering our understanding of human sperm function, for example identifying new avenues for discovery, we highlight potential compounds as promising start-point for a medicinal chemistry programme for potential enhancement of male fertility. Moreover, with disclosure of the results of screening, we present a substantial resource to inform further work in the field. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the Bill and Melinda Gates Foundation, Scottish Funding Council and Scottish Universities Life Science Alliance. C.L.R.B. is Editor for RBMO. C.L.R.B. receives funding from Chief Scientists Office (Scotland), ESHRE and Genus PLC, consulting fees from Exscientia and lecture fees from Cooper Surgical and Ferring. S.M.d.S. is an Associate Editor of Human Reproduction, and an Associate Editor of Reproduction and Fertility. S.M.d.S. receives funding from Cooper Surgical and British Dietetic Society. No other authors declared a COI.
Collapse
Affiliation(s)
- Franz S Gruber
- National Phenotypic Screening Centre, School of Life Sciences, University of Dundee, Dundee, UK
| | - Zoe C Johnston
- Reproductive Medicine Research Group, Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Neil R Norcross
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discover, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Irene Georgiou
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discover, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Caroline Wilson
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discover, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Kevin D Read
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discover, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Ian H Gilbert
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discover, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Jason R Swedlow
- National Phenotypic Screening Centre, School of Life Sciences, University of Dundee, Dundee, UK
- Division of Computational Biology and Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Sarah Martins da Silva
- Reproductive Medicine Research Group, Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Christopher L R Barratt
- Reproductive Medicine Research Group, Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
12
|
Li Y, Jin L, Li Y, Qian J, Wang Z, Zheng X, Xie C, Zhang X, Huang H, Zhou Y. Lysophosphatidic Acid Improves Human Sperm Motility by Enhancing Glycolysis and Activating L-Type Calcium Channels. Front Endocrinol (Lausanne) 2022; 13:896558. [PMID: 35903269 PMCID: PMC9317953 DOI: 10.3389/fendo.2022.896558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
Until now, the molecular mechanisms underlining sperm motility defect causing male infertility are still poorly understood. Safe and effective compounds or drugs that can improve sperm motility are also very limited. Lysophosphatidic acid (LPA) is a naturally occurring phospholipid and a bioactive intermediate with multiple biological activities. It has been detected in various body fluids such as serum, plasma, saliva, tears, blister fluids, hen egg white, and ascites from patients with ovarian cancer. LPA is also abundant in seminal plasma and follicular fluid. It enhances follicle stimulation, improves oocyte fertilization, and promotes early embryonic development and embryo implantation. However, the physiological role of LPA in the male reproductive system remains unknown. Here, our study showed that LPA significantly improved the motility parameters of human sperm hyperactivation in a dose-dependent manner. The LPA-induced elevation of sperm motility is dependent on bovine serum albumin (BSA) but independent of the classical BSA-induced sAC/cAMP/PKA signaling pathway. The enhancement of sperm motility by LPA could not be blocked by CCCP, a respiratory inhibitor suppressing mitochondrial ATP production. Moreover, LPA improved the activity of triosephosphate isomerase in glycolysis. Meanwhile, LPA treatment significantly increased ATP and phosphoenolpyruvate levels and decreased ADP content during sperm glycolysis. Notably, none of known or identified LPA receptors was detected in human sperm. Further investigations showed that LPA promoted sperm motility through L-type calcium channels. In summary, this study revealed the involvement of LPA in the regulation for human sperm motility by enhancing glycolysis and activating L-type calcium channels. The current findings may shed new light on the understanding of causes of asthenozoospermia, and indicate that LPA could be used as a novel therapeutic agent to improve sperm function and fertilizing capacity.
Collapse
Affiliation(s)
- Yinlam Li
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Li Jin
- Obstetrics & Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yanquan Li
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Jianing Qian
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Zhengquan Wang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoguo Zheng
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Chong Xie
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuelian Zhang
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
- *Correspondence: Yuchuan Zhou, ; Hefeng Huang, ; Xuelian Zhang,
| | - Hefeng Huang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- *Correspondence: Yuchuan Zhou, ; Hefeng Huang, ; Xuelian Zhang,
| | - Yuchuan Zhou
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- *Correspondence: Yuchuan Zhou, ; Hefeng Huang, ; Xuelian Zhang,
| |
Collapse
|
13
|
Rahban R, Rehfeld A, Schiffer C, Brenker C, Egeberg Palme DL, Wang T, Lorenz J, Almstrup K, Skakkebaek NE, Strünker T, Nef S. The antidepressant Sertraline inhibits CatSper Ca2+ channels in human sperm. Hum Reprod 2021; 36:2638-2648. [PMID: 34486673 PMCID: PMC8450872 DOI: 10.1093/humrep/deab190] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Do selective serotonin reuptake inhibitor (SSRI) antidepressants affect the function of human sperm? SUMMARY ANSWER The SSRI antidepressant Sertraline (e.g. Zoloft) inhibits the sperm-specific Ca2+ channel CatSper and affects human sperm function in vitro. WHAT IS KNOWN ALREADY In human sperm, CatSper translates changes of the chemical microenvironment into changes of the intracellular Ca2+ concentration ([Ca2+]i) and swimming behavior. CatSper is promiscuously activated by oviductal ligands, but also by synthetic chemicals that might disturb the fertilization process. It is well known that SSRIs have off-target actions on Ca2+, Na+ and K+ channels in somatic cells. Whether SSRIs affect the activity of CatSper is, however, unknown. STUDY DESIGN, SIZE, DURATION We studied the action of the seven drugs belonging to the most commonly prescribed class of antidepressants, SSRIs, on resting [Ca2+]i and Ca2+ influx via CatSper in human sperm. The SSRI Sertraline was selected for in-depth analysis of its action on steroid-, prostaglandin-, pH- and voltage-activation of human CatSper. Moreover, the action of Sertraline on sperm acrosomal exocytosis and penetration into viscous media was evaluated. PARTICIPANTS/MATERIALS, SETTING, METHODS The activity of CatSper was investigated in sperm of healthy volunteers, using kinetic Ca2+ fluorimetry and patch-clamp recordings. Acrosomal exocytosis was investigated using Pisum sativum agglutinin and image cytometry. Sperm penetration in viscous media was evaluated using the Kremer test. MAIN RESULTS AND THE ROLE OF CHANCE Several SSRIs affected [Ca2+]i and attenuated ligand-induced Ca2+ influx via CatSper. In particular, the SSRI Sertraline almost completely suppressed Ca2+ influx via CatSper. Remarkably, the drug was about four-fold more potent to suppress prostaglandin- versus steroid-induced Ca2+ influx. Sertraline also suppressed alkaline- and voltage-activation of CatSper, indicating that the drug directly inhibits the channel. Finally, Sertraline impaired ligand-induced acrosome reaction and sperm penetration into viscous media. LIMITATIONS, REASONS FOR CAUTION This is an in vitro study. Future studies have to assess the physiological relevance in vivo. WIDER IMPLICATIONS OF THE FINDINGS The off-target action of Sertraline on CatSper in human sperm might impair the fertilization process. In a research setting, Sertraline may be used to selectively inhibit prostaglandin-induced Ca2+ influx. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Swiss Centre for Applied Human Toxicology (SCAHT), the Département de l’Instruction Publique of the State of Geneva, the German Research Foundation (CRU326), the Interdisciplinary Center for Clinical Research, Münster (IZKF; Str/014/21), the Innovation Fund Denmark (grant numbers 14-2013-4) and the EDMaRC research grant from the Kirsten and Freddy Johansen’s Foundation. The authors declare that no conflict of interest could be perceived as prejudicing the impartiality of the research reported. TRIAL REGISTRATION NUMBER NA.
Collapse
Affiliation(s)
- Rita Rahban
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| | - Anders Rehfeld
- Department of Growth and Reproduction, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Christian Schiffer
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Christoph Brenker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | | | - Tao Wang
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany.,Institute of Life Science and School of Life Science, Nanchang University, Nanchang, Jiangxi, PR China
| | - Johannes Lorenz
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Kristian Almstrup
- Department of Growth and Reproduction, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Niels E Skakkebaek
- Department of Growth and Reproduction, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Timo Strünker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland.,Swiss Centre for Applied Human Toxicology, Basel, Switzerland
| |
Collapse
|
14
|
Campbell MJ, Sucquart IE, Whittaker A, Sanganee HJ, Barratt CLR, Martins da Silva SJ. Myeloperoxidase inhibitor AZD5904 enhances human sperm function in vitro. Hum Reprod 2021; 36:560-570. [PMID: 33393586 DOI: 10.1093/humrep/deaa328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/02/2020] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION Does AZD5904, a myeloperoxidase inhibitor (MPOi), have any effect on human sperm function in vitro? SUMMARY ANSWER AZD5904 improves sperm function in an in vitro model of oxidative stress (OS) and potentially offers a novel treatment approach for male infertility. WHAT IS KNOWN ALREADY Male infertility is an underlying or contributory cause in half of all couples experiencing difficulties conceiving, yet there is currently no effective treatment or cure. OS is a common pathology in a significant proportion of infertile men. It can negatively affect sperm motility and the ability to fertilize a mature oocyte, as well as DNA integrity, and therefore represents an attractive target for therapeutic intervention. STUDY DESIGN, SIZE, DURATION This study included population-based samples from men (23-50 years) attending Ninewells Assisted Conception Unit, Dundee for diagnostic semen analysis, July 2017-September 2018. Semen samples (n = 47) from 45 patients were used. PARTICIPANTS/MATERIALS, SETTING, METHODS Neutrophils activated using zymosan were incubated with prepared human spermatozoa for 2 h (T2) and 24 h (T24) to create an in vitro model of OS. Parallel samples were co-incubated with AZD5904, an MPOi, to examine its effects. Sperm motility was assessed by computer-assisted sperm analysis at T2 and T24. Functional motility was assessed by sperm penetration assay. Statistical analysis was performed using GraphPad Prism. MAIN RESULTS AND THE ROLE OF CHANCE There was no significant difference in total or progressive sperm motility between any treatment and control groups at T2 or T24. Nonetheless, significant positive effects on sperm function were observed with AZD5904, with 16/45 (35.6%) samples (with both normal and abnormal baseline semen analysis characteristics) displaying a ≥20% increase in sperm penetrated through viscous media (P < 0.003). LIMITATIONS, REASONS FOR CAUTION This was an in vitro study. WIDER IMPLICATIONS OF THE FINDINGS Treatment with AZD5904 resulted in significant increased sperm penetration in one of three samples treated, which is likely to represent improvement in sperm function required for fertilization. We are now planning a clinical trial to validate these results and hope that this could represent a new treatment for male infertility. STUDY FUNDING/COMPETING INTEREST(S) AZD5904 was shared through the AstraZeneca Open Innovation program. The study was funded by AstraZeneca and sponsored by the University of Dundee. Additional funding was provided by Chief Scientist Office/NHS Research Scotland (S.J.M.d.S.). A.W. and H.J.S. are both full time employees of AstraZeneca. A.W. and H.J.S. are inventors on a patent filed by AstraZeneca titled MPOi for use in medicine which includes MPOi for use in the treatment of male infertility (WO 2019/016074 Al). S.J.M.d.S. is Associate Editor of Human Reproduction and Editorial Board member of Reproduction & Fertility. C.L.R.B. is Editor of RBMO and has received lecturing fees from Merck and Ferring and is on the Scientific Advisory Panel for Ohana BioSciences. C.L.R.B. was chair of the World Health Organization Expert Synthesis Group on Diagnosis of Male infertility (2012-2016). C.L.R.B. has a patent WO2013054111 A1 issued. The other authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- M J Campbell
- Reproductive Medicine Research Group, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - I E Sucquart
- Reproductive Medicine Research Group, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - A Whittaker
- Emerging Innovations Unit, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - H J Sanganee
- Emerging Innovations Unit, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - C L R Barratt
- Reproductive Medicine Research Group, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - S J Martins da Silva
- Reproductive Medicine Research Group, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
15
|
Tricyclic Pyrazole-Based Compounds as Useful Scaffolds for Cannabinoid CB 1/CB 2 Receptor Interaction. Molecules 2021; 26:molecules26082126. [PMID: 33917187 PMCID: PMC8068016 DOI: 10.3390/molecules26082126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/29/2021] [Indexed: 11/17/2022] Open
Abstract
Cannabinoids comprise different classes of compounds, which aroused interest in recent years because of their several pharmacological properties. Such properties include analgesic activity, bodyweight reduction, the antiemetic effect, the reduction of intraocular pressure and many others, which appear correlated to the affinity of cannabinoids towards CB1 and/or CB2 receptors. Within the search aiming to identify novel chemical scaffolds for cannabinoid receptor interaction, the CB1 antagonist/inverse agonist pyrazole-based derivative rimonabant has been modified, giving rise to several tricyclic pyrazole-based compounds, most of which endowed of high affinity and selectivity for CB1 or CB2 receptors. The aim of this review is to present the synthesis and summarize the SAR study of such tricyclic pyrazole-based compounds, evidencing, for some derivatives, their potential in the treatment of neuropathic pain, obesity or in the management of glaucoma.
Collapse
|
16
|
Kumari S, Kotyan S, Sugunan S, Rajanikant GK, Kumar KSS, Adiga SK, Dasappa JP, Kalthur G. The synthesis of a novel pentoxifylline derivative with superior human sperm motility enhancement properties. NEW J CHEM 2021. [DOI: 10.1039/d0nj03522j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
mPTF1 demonstrated lower genotoxicity, higher affinity for PDEs, & reasonable pharmacokinetic, druglikeness & ADMET properties compared to Pentoxifylline.
Collapse
Affiliation(s)
- Sandhya Kumari
- Department of Clinical Embryology
- Kasturba Medical College of Manipal
- Manipal
- Academy of Higher Education
- Manipal
| | - Sukumar Kotyan
- Department of Chemistry
- Mangalore University
- Mangalagangothri
- Mangalore
- India
| | - Sinoy Sugunan
- School of Biotechnology
- National Institute of Technology Calicut
- Calicut
- India
| | - G. K. Rajanikant
- School of Biotechnology
- National Institute of Technology Calicut
- Calicut
- India
| | | | - Satish Kumar Adiga
- Department of Clinical Embryology
- Kasturba Medical College of Manipal
- Manipal
- Academy of Higher Education
- Manipal
| | | | - Guruprasad Kalthur
- Department of Clinical Embryology
- Kasturba Medical College of Manipal
- Manipal
- Academy of Higher Education
- Manipal
| |
Collapse
|
17
|
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
|
18
|
Rahban R, Nef S. CatSper: The complex main gate of calcium entry in mammalian spermatozoa. Mol Cell Endocrinol 2020; 518:110951. [PMID: 32712386 DOI: 10.1016/j.mce.2020.110951] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
Calcium ions (Ca2+) are involved in nearly every aspect of cellular life. They are one of the most abundant elements in mammals and play a vital role in physiological and biochemical processes acting mainly as intracellular messengers. In spermatozoa, several key functions are regulated by cytoplasmic Ca2+ concentration such as sperm capacitation, chemotaxis, hyperactive motility, and acrosome reaction. The sperm-specific ion channel CatSper is the principal calcium channel in sperm mediating the calcium influx into the sperm flagellum and acting as an essential modulator of downstream mechanisms involved in fertilization. This review aims to provide insights into the structure, localization, and function of the mammalian CatSper channel, primarily human and mice. The activation of CatSper by progesterone and prostaglandins, as well as the ligand-independent regulation of the channel by a change in the membrane voltage and intracellular pH are going to be addressed. Finally, major questions, challenges, and perspectives are discussed.
Collapse
Affiliation(s)
- Rita Rahban
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1206, Geneva, Switzerland.
| | - Serge Nef
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1206, Geneva, Switzerland.
| |
Collapse
|
19
|
Zhao X, Lorent K, Escobar-Zarate D, Rajagopalan R, Loomes KM, Gillespie K, Mesaros C, Estrada MA, Blair I, Winkler JD, Spinner NB, Devoto M, Pack M. Impaired Redox and Protein Homeostasis as Risk Factors and Therapeutic Targets in Toxin-Induced Biliary Atresia. Gastroenterology 2020; 159:1068-1084.e2. [PMID: 32505743 PMCID: PMC7856536 DOI: 10.1053/j.gastro.2020.05.080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/08/2020] [Accepted: 05/27/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Extrahepatic biliary atresia (BA) is a pediatric liver disease with no approved medical therapy. Recent studies using human samples and experimental modeling suggest that glutathione redox metabolism and heterogeneity play a role in disease pathogenesis. We sought to dissect the mechanistic basis of liver redox variation and explore how other stress responses affect cholangiocyte injury in BA. METHODS We performed quantitative in situ hepatic glutathione redox mapping in zebrafish larvae carrying targeted mutations in glutathione metabolism genes and correlated these findings with sensitivity to the plant-derived BA-linked toxin biliatresone. We also determined whether genetic disruption of HSP90 protein quality control pathway genes implicated in human BA altered biliatresone toxicity in zebrafish and human cholangiocytes. An in vivo screening of a known drug library was performed to identify novel modifiers of cholangiocyte injury in the zebrafish experimental BA model, with subsequent validation. RESULTS Glutathione metabolism gene mutations caused regionally distinct changes in the redox potential of cholangiocytes that differentially sensitized them to biliatresone. Disruption of human BA-implicated HSP90 pathway genes sensitized zebrafish and human cholangiocytes to biliatresone-induced injury independent of glutathione. Phosphodiesterase-5 inhibitors and other cyclic guanosine monophosphate signaling activators worked synergistically with the glutathione precursor N-acetylcysteine in preventing biliatresone-induced injury in zebrafish and human cholangiocytes. Phosphodiesterase-5 inhibitors enhanced proteasomal degradation and required intact HSP90 chaperone. CONCLUSION Regional variation in glutathione metabolism underlies sensitivity to the biliary toxin biliatresone and may account for the reported association between BA transplant-free survival and glutathione metabolism gene expression. Human BA can be causatively linked to genetic modulation of protein quality control. Combined treatment with N-acetylcysteine and cyclic guanosine monophosphate signaling enhancers warrants further investigation as therapy for BA.
Collapse
Affiliation(s)
- Xiao Zhao
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristin Lorent
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Diana Escobar-Zarate
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ramakrishnan Rajagopalan
- Division of Genomic Diagnostics, Department of Pathology, The Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kathleen M. Loomes
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kevin Gillespie
- Department of System Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Clementina Mesaros
- Department of System Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ian Blair
- Department of System Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey D. Winkler
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, USA
| | - Nancy B. Spinner
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marcella Devoto
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA.,Departments of Pediatrics and of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Translational and Precision Medicine, University La Sapienza, Rome, Italy
| | - Michael Pack
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
20
|
Wang T, Young S, Krenz H, Tüttelmann F, Röpke A, Krallmann C, Kliesch S, Zeng XH, Brenker C, Strünker T. The Ca 2+ channel CatSper is not activated by cAMP/PKA signaling but directly affected by chemicals used to probe the action of cAMP and PKA. J Biol Chem 2020; 295:13181-13193. [PMID: 32703901 DOI: 10.1074/jbc.ra120.013218] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
The sperm-specific Ca2+ channel CatSper (cation channel of sperm) controls the influx of Ca2+ into the flagellum and, thereby, the swimming behavior of sperm. A hallmark of human CatSper is its polymodal activation by membrane voltage, intracellular pH, and oviductal hormones. Whether CatSper is also activated by signaling pathways involving an increase of cAMP and ensuing activation of PKA is, however, a matter of controversy. To shed light on this question, we used kinetic ion-sensitive fluorometry, patch-clamp recordings, and optochemistry to study transmembrane Ca2+ flux and membrane currents in human sperm from healthy donors and from patients that lack functional CatSper channels. We found that human CatSper is neither activated by intracellular cAMP directly nor indirectly by the cAMP/PKA-signaling pathway. Instead, we show that nonphysiological concentrations of cAMP and membrane-permeable cAMP analogs used to mimic the action of intracellular cAMP activate human CatSper from the outside via a hitherto-unknown extracellular binding site. Finally, we demonstrate that the effects of common PKA inhibitors on human CatSper rest predominantly, if not exclusively, on off-target drug actions on CatSper itself rather than on inhibition of PKA. We conclude that the concept of an intracellular cAMP/PKA-activation of CatSper is primarily based on unspecific effects of chemical probes used to interfere with cAMP signaling. Altogether, our findings solve several controversial issues and reveal a novel ligand-binding site controlling the activity of CatSper, which has important bearings on future studies of cAMP and Ca2+ signaling in sperm.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, Jiangxi, China; Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Samuel Young
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Henrike Krenz
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Frank Tüttelmann
- Institute of Human Genetics, University of Münster, Münster, Germany
| | - Albrecht Röpke
- Institute of Human Genetics, University of Münster, Münster, Germany
| | - Claudia Krallmann
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany
| | - Xu-Hui Zeng
- Institute of Life Science and School of Life Science, Nanchang University, Nanchang, Jiangxi, China.
| | - Christoph Brenker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany.
| | - Timo Strünker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Münster, Germany; Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany.
| |
Collapse
|
21
|
McBrinn RC, Fraser J, Hope AG, Gray DW, Barratt CLR, Martins da Silva SJ, Brown SG. Novel pharmacological actions of trequinsin hydrochloride improve human sperm cell motility and function. Br J Pharmacol 2019; 176:4521-4536. [PMID: 31368510 PMCID: PMC6932944 DOI: 10.1111/bph.14814] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Asthenozoospermia is a leading cause of male infertility, but development of pharmacological agents to improve sperm motility is hindered by the lack of effective screening platforms and knowledge of suitable molecular targets. We have demonstrated that a high-throughput screening (HTS) strategy and established in vitro tests can identify and characterise compounds that improve sperm motility. Here, we applied HTS to identify new compounds from a novel small molecule library that increase intracellular calcium ([Ca2+ ]i ), promote human sperm cell motility, and systematically determine the mechanism of action. EXPERIMENTAL APPROACH A validated HTS fluorometric [Ca2+ ]i assay was used to screen an in-house library of compounds. Trequinsin hydrochloride (a PDE3 inhibitor) was selected for detailed molecular (plate reader assays, electrophysiology, and cyclic nucleotide measurement) and functional (motility and acrosome reaction) testing in sperm from healthy volunteer donors and, where possible, patients. KEY RESULTS Fluorometric assays identified trequinsin as an efficacious agonist of [Ca2+ ]i , although less potent than progesterone. Functionally, trequinsin significantly increased cell hyperactivation and penetration into viscous medium in all donor sperm samples and cell hyperactivation in 22/25 (88%) patient sperm samples. Trequinsin-induced [Ca2+ ]i responses were cross-desensitised consistently by PGE1 but not progesterone. Whole-cell patch clamp electrophysiology confirmed that trequinsin activated CatSper and partly inhibited potassium channel activity. Trequinsin also increased intracellular cGMP. CONCLUSION AND IMPLICATIONS Trequinsin exhibits a novel pharmacological profile in human sperm and may be a suitable lead compound for the development of new agents to improve patient sperm function and fertilisation potential.
Collapse
Affiliation(s)
- Rachel C McBrinn
- School of Science, Engineering and Technology, Abertay University, Dundee, UK
| | - Joanna Fraser
- School of Science, Engineering and Technology, Abertay University, Dundee, UK
| | - Anthony G Hope
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - David W Gray
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Christopher L R Barratt
- Reproductive and Developmental Biology, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Sarah J Martins da Silva
- Reproductive and Developmental Biology, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Sean G Brown
- School of Science, Engineering and Technology, Abertay University, Dundee, UK
| |
Collapse
|