Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 104(2): 331-340, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30686508

RESUMEN

Male infertility is a major health concern. Among its different causes, multiple morphological abnormalities of the flagella (MMAF) induces asthenozoospermia and is one of the most severe forms of qualitative sperm defects. Sperm of affected men display short, coiled, absent, and/or irregular flagella. To date, six genes (DNAH1, CFAP43, CFAP44, CFAP69, FSIP2, and WDR66) have been found to be recurrently associated with MMAF, but more than half of the cases analyzed remain unresolved, suggesting that many yet-uncharacterized gene defects account for this phenotype. Here, whole-exome sequencing (WES) was performed on 168 infertile men who had a typical MMAF phenotype. Five unrelated affected individuals carried a homozygous deleterious mutation in ARMC2, a gene not previously linked to the MMAF phenotype. Using the CRISPR-Cas9 technique, we generated homozygous Armc2 mutant mice, which also presented an MMAF phenotype, thus confirming the involvement of ARMC2 in human MMAF. Immunostaining experiments in AMRC2-mutated individuals and mutant mice evidenced the absence of the axonemal central pair complex (CPC) proteins SPAG6 and SPEF2, whereas the other tested axonemal and peri-axonemal components were present, suggesting that ARMC2 is involved in CPC assembly and/or stability. Overall, we showed that bi-allelic mutations in ARMC2 cause male infertility in humans and mice by inducing a typical MMAF phenotype, indicating that this gene is necessary for sperm flagellum structure and assembly.


Asunto(s)
Alelos , Astenozoospermia/genética , Astenozoospermia/patología , Proteínas del Citoesqueleto/genética , Flagelos/genética , Mutación , Espermatozoides/anomalías , Espermatozoides/patología , Animales , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/deficiencia , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino , Ratones , Proteínas de Microtúbulos/deficiencia , Proteínas
2.
Biol Reprod ; 106(3): 463-476, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-34875016

RESUMEN

Infertility represents a growing burden worldwide, with one in seven couples presenting difficulties conceiving. Among these, 10-15% of the men have idiopathic infertility that does not correlate with any defect in the classical sperm parameters measured. In the present study, we used a mouse model to investigate the effects of maternal undernutrition on fertility in male progeny. Our results indicate that mothers fed on a low-protein diet during gestation and lactation produce male offspring with normal sperm morphology, concentration, and motility but exhibiting an overall decrease of fertility when they reach adulthood. Particularly, in contrast to control, sperm from these offspring show a remarkable lower capacity to fertilize oocytes when copulation occurs early in the estrus cycle relative to ovulation, due to an altered sperm capacitation. Our data demonstrate for the first time that maternal nutritional stress can have long-term consequences on the reproductive health of male progeny by affecting sperm physiology, especially capacitation, with no observable impact on spermatogenesis and classical quantitative and qualitative sperm parameters. Moreover, our experimental model could be of major interest to study, explain, and ultimately treat certain categories of infertilities.


Asunto(s)
Infertilidad Masculina , Desnutrición , Adulto , Animales , Femenino , Fertilidad , Humanos , Infertilidad Masculina/etiología , Lactancia , Masculino , Desnutrición/complicaciones , Ratones , Embarazo , Capacitación Espermática , Motilidad Espermática , Espermatozoides/fisiología
3.
Int J Mol Sci ; 23(14)2022 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-35887380

RESUMEN

The group X secreted phospholipase A2 (PLA2G10) is present at high levels in mouse sperm acrosome. The enzyme is secreted during capacitation and amplifies the acrosome reaction and its own secretion via an autocrine loop. PLA2G10 also improves the rate of fertilization. In in vitro fertilization (IVF) experiments, sperm from Pla2g10-deficient mice produces fewer two-cell embryos, and the absence of PLA2G10 is rescued by adding recombinant enzymes. Moreover, wild-type (WT) sperm treated with recombinant PLA2G10 produces more two-cell embryos. The effects of PLA2G10 on mouse fertility are inhibited by sPLA2 inhibitors and rescued by products of the enzymatic reaction such as free fatty acids, suggesting a role of catalytic activity. However, PLA2G10 also binds to mouse PLA2R1, which may play a role in fertility. To determine the relative contribution of enzymatic activity and PLA2R1 binding in the profertility effect of PLA2G10, we tested H48Q-PLA2G10, a catalytically-inactive mutant of PLA2G10 with low enzymatic activity but high binding properties to PLA2R1. Its effect was tested in various mouse strains, including Pla2r1-deficient mice. H48Q-PLA2G10 did not trigger the acrosome reaction but was as potent as WT-PLA2G10 to improve IVF in inbred C57Bl/6 mice; however, this was not the case in OF1 outbred mice. Using gametes from these mouse strains, the effect of H48Q-PLA2G10 appeared dependent on both spermatozoa and oocytes. Moreover, sperm from C57Bl/6 Pla2r1-deficient mice were less fertile and lowered the profertility effects of H48Q-PLA2G10, which were completely suppressed when sperm and oocytes were collected from Pla2r1-deficient mice. Conversely, the effect of WT-PLA2G10 was not or less sensitive to the absence of PLA2R1, suggesting that the effect of PLA2G10 is polymodal and complex, acting both as an enzyme and a ligand of PLA2R1. This study shows that the action of PLA2G10 on gametes is complex and can simultaneously activate the catalytic pathway and the PLA2R1-dependent receptor pathway. This work also shows for the first time that PLA2G10 binding to gametes' PLA2R1 participates in fertilization optimization.


Asunto(s)
Semen , Espermatozoides , Animales , Fertilización , Fertilización In Vitro , Fosfolipasas A2 Grupo X/metabolismo , Fosfolipasas A2 Grupo X/farmacología , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Semen/metabolismo , Espermatozoides/metabolismo
4.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096770

RESUMEN

Animal venoms are small natural mixtures highly enriched in bioactive components. They are known to target at least two important pharmacological classes of cell surface receptors: ion channels and G protein coupled receptors. Since sperm cells express a wide variety of ion channels and membrane receptors, required for the control of cell motility and acrosome reaction, two functions that are defective in infertility issues, animal venoms should contain interesting compounds capable of modulating these two essential physiological functions. Herein, we screened for bioactive compounds from the venom of the Egyptian black snake Walterinnesia aegyptia (Wa) that possess the property to activate sperm motility in vitro from male mice OF1. Using RP-HPLC and cation exchange chromatography, we identified a new toxin of 6389.89 Da (termed walterospermin) that activates sperm motility. Walterospermin was de novo sequenced using a combination of matrix assisted laser desorption ionization time of flight mass spectrometry (MALDI-TOF/TOF MS/MS) and liquid chromatography electrospray ionization quadrupole time-of-flight mass spectrometry (LC-ESI-QTOF MS/MS) following reduction, alkylation, and enzymatic proteolytic digestion with trypsin, chymotrypsin or V8 protease. The peptide is 57 amino acid residues long and contains three disulfide bridges and was found to be identical to the previously cloned Wa Kunitz-type protease inhibitor II (Wa Kln-II) sequence. Moreover, it has strong homology with several other hitherto cloned Elapidae and Viperidae snake toxins suggesting that it belongs to a family of compounds able to regulate sperm function. The synthetic peptide shows promising activation of sperm motility from a variety of species, including humans. Its fluorescently-labelled analog predominantly marks the flagellum, a localization in agreement with a receptor that controls motility function.


Asunto(s)
Venenos Elapídicos/química , Péptidos/química , Péptidos/farmacología , Motilidad Espermática/efectos de los fármacos , Animales , Cromatografía por Intercambio Iónico , Disulfuros/química , Egipto , Venenos Elapídicos/farmacología , Elapidae , Humanos , Macaca fascicularis , Masculino , Ratones Endogámicos , Péptidos/síntesis química , Péptidos/aislamiento & purificación , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Cola del Espermatozoide/química , Cola del Espermatozoide/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/fisiología , Espectrometría de Masas en Tándem
5.
Hum Mol Genet ; 25(5): 878-91, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26721930

RESUMEN

In mammals, sperm-oocyte fusion initiates Ca(2+) oscillations leading to a series of events called oocyte activation, which is the first stage of embryo development. Ca(2+) signaling is elicited by the delivery of an oocyte-activating factor by the sperm. A sperm-specific phospholipase C (PLCZ1) has emerged as the likely candidate to induce oocyte activation. Recently, PAWP, a sperm-born tryptophan domain-binding protein coded by WBP2NL, was proposed to serve the same purpose. Here, we studied two infertile brothers exhibiting normal sperm morphology but complete fertilization failure after intracytoplasmic sperm injection. Whole exomic sequencing evidenced a missense homozygous mutation in PLCZ1, c.1465A>T; p.Ile489Phe, converting Ile 489 into Phe. We showed the mutation is deleterious, leading to the absence of the protein in sperm, mislocalization of the protein when injected in mouse GV and MII oocytes, highly abnormal Ca(2+) transients and early embryonic arrest. Altogether these alterations are consistent with our patients' sperm inability to induce oocyte activation and initiate embryo development. In contrast, no deleterious variants were identified in WBP2NL and PAWP presented normal expression and localization. Overall we demonstrate in humans, the absence of PLCZ1 alone is sufficient to prevent oocyte activation irrespective of the presence of PAWP. Additionally, it is the first mutation located in the C2 domain of PLCZ1, a domain involved in targeting proteins to cell membranes. This opens the door to structure-function studies to identify the conserved amino acids of the C2 domain that regulate the targeting of PLCZ1 and its selectivity for its lipid substrate(s).


Asunto(s)
Proteínas Portadoras/genética , Infertilidad Masculina/genética , Mutación , Fosfoinositido Fosfolipasa C/genética , Proteínas de Plasma Seminal/genética , Interacciones Espermatozoide-Óvulo/genética , Espermatozoides/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Señalización del Calcio , Proteínas Portadoras/metabolismo , Pérdida del Embrión , Femenino , Regulación de la Expresión Génica , Homocigoto , Humanos , Técnicas de Maduración In Vitro de los Oocitos , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Oocitos/citología , Oocitos/metabolismo , Fosfoinositido Fosfolipasa C/deficiencia , Transporte de Proteínas , Proteínas de Plasma Seminal/metabolismo , Alineación de Secuencia , Hermanos , Motilidad Espermática , Espermatozoides/patología
6.
Reproduction ; 156(6): 463-476, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30422808

RESUMEN

For artificial insemination (AI) to be successful, it is essential that sperm delivery be perfectly timed relative to ovulation, as sperm lifespan is limited due to oxidative metabolism induced by capacitation. Extending the window of sperm capacitation could therefore increase sperm lifespan, prolong sperm-fertilizing competence and increase AI efficiency. Hyperpolarization of sperm is a crucial step in capacitation and is induced by activation of the potassium calcium-activated channel subfamily U member 1 (KCNU1, also named Slo3 or KSper). Given the essential role played by KCNU1 in capacitation, this study assessed the impact of its pharmacological inhibition on sperm lifespan. We showed that treatment of murine sperm with sub-micromolar concentrations of clofilium, a specific inhibitor of KCNU1, slowed down capacitation, decreased the rate of acrosome reaction and extended the fertilizing competence of capacitated sperm for 12 h. Clofilium also extended fertilizing competence and motility of bovine-capacitated sperm, and increased the rate of fertilization with sperm capacitated for 24 h by 100%, and the rate of blastocyst formation by 150%. Finally, toxicity experiments showed clofilium to have no impact on sperm DNA and no embryotoxicity at the concentration used to extend sperm lifespan. Our results demonstrate that clofilium prolongs fertilizing competence of aging capacitated sperm in vitro in both rodent and bovine species. To our knowledge, this is the first time the duration of sperm-fertilizing competence is shown to be extended by potassium channels blockers.


Asunto(s)
Fertilización/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Compuestos de Amonio Cuaternario/farmacología , Capacitación Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Animales , Bovinos , Masculino , Ratones , Motilidad Espermática/efectos de los fármacos , Espermatozoides/fisiología
7.
J Biol Chem ; 291(32): 16448-61, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27317664

RESUMEN

Selective transport of pyruvate across the inner mitochondrial membrane by the mitochondrial pyruvate carrier (MPC) is a fundamental step that couples cytosolic and mitochondrial metabolism. The recent molecular identification of the MPC complex has revealed two interacting subunits, MPC1 and MPC2. Although in yeast, an additional subunit, MPC3, can functionally replace MPC2, no alternative MPC subunits have been described in higher eukaryotes. Here, we report for the first time the existence of a novel MPC subunit termed MPC1-like (MPC1L), which is present uniquely in placental mammals. MPC1L shares high sequence, structural, and topological homology with MPC1. In addition, we provide several lines of evidence to show that MPC1L is functionally equivalent to MPC1: 1) when co-expressed with MPC2, it rescues pyruvate import in a MPC-deleted yeast strain; 2) in mammalian cells, it can associate with MPC2 to form a functional carrier as assessed by bioluminescence resonance energy transfer; 3) in MPC1 depleted mouse embryonic fibroblasts, MPC1L rescues the loss of pyruvate-driven respiration and stabilizes MPC2 expression; and 4) MPC1- and MPC1L-mediated pyruvate imports show similar efficiency. However, we show that MPC1L has a highly specific expression pattern and is localized almost exclusively in testis and more specifically in postmeiotic spermatids and sperm cells. This is in marked contrast to MPC1/MPC2, which are ubiquitously expressed throughout the organism. To date, the biological importance of this alternative MPC complex during spermatogenesis in placental mammals remains unknown. Nevertheless, these findings open up new avenues for investigating the structure-function relationship within the MPC complex.


Asunto(s)
Proteínas de Transporte de Anión/biosíntesis , Regulación de la Expresión Génica/fisiología , Proteínas de Transporte de Membrana Mitocondrial/biosíntesis , Espermátides/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Proteínas de Transporte de Anión/genética , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Transportadores de Ácidos Monocarboxílicos , Espermátides/citología , Testículo/citología
8.
J Biol Chem ; 291(6): 3076-89, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26655718

RESUMEN

Phospholipase A2 (PLA2) activity has been shown to be involved in the sperm acrosome reaction (AR), but the molecular identity of PLA2 isoforms has remained elusive. Here, we have tested the role of two intracellular (iPLA2ß and cytosolic PLA2α) and one secreted (group X) PLA2s in spontaneous and progesterone (P4)-induced AR by using a set of specific inhibitors and knock-out mice. iPLA2ß is critical for spontaneous AR, whereas both iPLA2ß and group X secreted PLA2 are involved in P4-induced AR. Cytosolic PLA2α is dispensable in both types of AR. P4-induced AR spreads over 30 min in the mouse, and kinetic analyses suggest the presence of different sperm subpopulations, using distinct PLA2 pathways to achieve AR. At low P4 concentration (2 µm), sperm undergoing early AR (0-5 min post-P4) rely on iPLA2ß, whereas sperm undergoing late AR (20-30 min post-P4) rely on group X secreted PLA2. Moreover, the role of PLA2s in AR depends on P4 concentration, with the PLA2s being key actors at low physiological P4 concentrations (≤2 µm) but not at higher P4 concentrations (~10 µm).


Asunto(s)
Reacción Acrosómica/efectos de los fármacos , Acrosoma/enzimología , Exocitosis/efectos de los fármacos , Fosfolipasas A2 Grupo VI/metabolismo , Fosfolipasas A2 Grupo X/metabolismo , Progesterona/farmacología , Animales , Fosfolipasas A2 Grupo VI/genética , Fosfolipasas A2 Grupo X/genética , Masculino , Ratones , Ratones Noqueados , Progesterona/metabolismo
9.
Mol Hum Reprod ; 23(2): 116-131, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-27932550

RESUMEN

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.


Asunto(s)
Embrión de Mamíferos/efectos de los fármacos , Fármacos para la Fertilidad/farmacología , Péptidos/farmacología , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Venenos de Araña/química , Adulto , Secuencia de Aminoácidos , Animales , Bovinos , Criopreservación , Embrión de Mamíferos/citología , Epidídimo/citología , Epidídimo/efectos de los fármacos , Epidídimo/fisiopatología , Femenino , Fármacos para la Fertilidad/síntesis química , Fármacos para la Fertilidad/aislamiento & purificación , Fertilización In Vitro , Humanos , Infertilidad Masculina/tratamiento farmacológico , Infertilidad Masculina/fisiopatología , Macaca fascicularis , Masculino , Ratones , Biblioteca de Péptidos , Péptidos/síntesis química , Péptidos/aislamiento & purificación , Escorpiones , Análisis de Semen , Motilidad Espermática/fisiología , Espermatozoides/citología , Espermatozoides/patología , Venenos de Araña/síntesis química , Venenos de Araña/aislamiento & purificación , Venenos de Araña/farmacología , Testículo/citología , Testículo/efectos de los fármacos , Testículo/fisiopatología
10.
J Cell Physiol ; 230(8): 1758-1769, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25597298

RESUMEN

Mammalian sperm acquire fertilizing ability in the female tract in a process known as capacitation. At the molecular level, capacitation is associated with up-regulation of a cAMP-dependent pathway, changes in intracellular pH, intracellular Ca(2+), and an increase in tyrosine phosphorylation. How these signaling systems interact during capacitation is not well understood. Results presented in this study indicate that Ca(2+) ions have a biphasic role in the regulation of cAMP-dependent signaling. Media without added Ca(2+) salts (nominal zero Ca(2+)) still contain micromolar concentrations of this ion. Sperm incubated in this medium did not undergo PKA activation or the increase in tyrosine phosphorylation suggesting that these phosphorylation pathways require Ca(2+). However, chelation of the extracellular Ca(2+) traces by EGTA induced both cAMP-dependent phosphorylation and the increase in tyrosine phosphorylation. The EGTA effect in nominal zero Ca(2+) media was mimicked by two calmodulin antagonists, W7 and calmidazolium, and by the calcineurin inhibitor cyclosporine A. These results suggest that Ca(2+) ions regulate sperm cAMP and tyrosine phosphorylation pathways in a biphasic manner and that some of its effects are mediated by calmodulin. Interestingly, contrary to wild-type mouse sperm, sperm from CatSper1 KO mice underwent PKA activation and an increase in tyrosine phosphorylation upon incubation in nominal zero Ca(2+) media. Therefore, sperm lacking Catsper Ca(2+) channels behave as wild-type sperm incubated in the presence of EGTA. This latter result suggests that Catsper transports the Ca(2+) involved in the regulation of cAMP-dependent and tyrosine phosphorylation pathways required for sperm capacitation.


Asunto(s)
Calcio/metabolismo , Transducción de Señal/fisiología , Capacitación Espermática/fisiología , Animales , Western Blotting , Canales de Calcio/metabolismo , AMP Cíclico/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación , Motilidad Espermática/fisiología , Tirosina/metabolismo
11.
Biol Reprod ; 92(5): 121, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25855261

RESUMEN

To gain fertilizing capacity, mammalian sperm should reside in the female tract for a period of time. The physiological changes that render the sperm able to fertilize are known as capacitation. Capacitation is associated with an increase in intracellular pH, an increase in intracellular calcium, and phosphorylation of different proteins. This process is also accompanied by the hyperpolarization of the sperm plasma membrane potential (Em). In the present work, we used flow cytometry to analyze changes in sperm Em during capacitation in individual cells. Our results indicate that a subpopulation of hyperpolarized mouse sperm can be clearly distinguished by sperm flow cytometry analysis. Using sperm bearing green fluorescent protein in their acrosomes, we found that this hyperpolarized subpopulation is composed of sperm with intact acrosomes. In addition, we show that the capacitation-associated hyperpolarization is blocked by high extracellular K(+), by PKA inhibitors, and by SLO3 inhibitors in CD1 mouse sperm, and undetectable in Slo3 knockout mouse sperm. On the other hand, in sperm incubated in conditions that do not support capacitation, sperm membrane hyperpolarization can be induced by amiloride, high extracellular NaHCO3, and cAMP agonists. Altogether, our observations are consistent with a model in which sperm Em hyperpolarization is downstream of a cAMP-dependent pathway and is mediated by the activation of SLO3 K(+) channels.


Asunto(s)
Potenciales de la Membrana/fisiología , Capacitación Espermática/fisiología , Espermatozoides/fisiología , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/fisiología , Activación del Canal Iónico , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Masculino , Ratones , Ratones Noqueados , Permeabilidad , Potasio , Transducción de Señal/fisiología , Sodio , Coloración y Etiquetado
12.
Mol Hum Reprod ; 21(2): 169-85, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25354700

RESUMEN

We recently identified the DPY19L2 gene as the main genetic cause of human globozoospermia. Non-genetically characterized cases of globozoospermia were associated with DNA alterations, suggesting that DPY19L2-dependent globozoospermia may be associated with poor DNA quality. However the origins of such defects have not yet been characterized and the consequences on the quality of embryos generated with globozoospermic sperm remain to be determined. Using the mouse model lacking Dpy19l2, we compared several key steps of nuclear compaction. We show that the kinetics of appearance and disappearance of the histone H4 acetylation waves and of transition proteins are defective. More importantly, the nuclear invasion by protamines does not occur. As a consequence, we showed that globozoospermic sperm presented with poor sperm chromatin compaction and sperm DNA integrity breakdown. We next assessed the developmental consequences of using such faulty sperm by performing ICSI. We showed in the companion article that oocyte activation (OA) with globozoospermic sperm is very poor and due to the absence of phospholipase Cζ; therefore artificial OA (AOA) was used to bypass defective OA. Herein, we evaluated the developmental potential of embryos generated by ICSI + AOA in mice. We demonstrate that although OA was fully rescued, preimplantation development was impaired when using globozoospermic sperm. In human, a small number of embryos could be generated with sperm from DPY19L2-deleted patients in the absence of AOA and these embryos also showed a poor developmental potential. In conclusion, we show that chromatin compaction during spermiogenesis in Dpy19l2 KO mouse is defective and leads to sperm DNA damage. Most of the DNA breaks were already present when the sperm reached the epididymis, indicating that they occurred inside the testis. This result thus suggests that testicular sperm extraction in Dpy19l2-dependent globozoospermia is not recommended. These defects may largely explain the poor embryonic development of most mouse and human embryos obtained with globozoospermic sperm.


Asunto(s)
Proteínas de la Membrana/deficiencia , Espermatozoides/metabolismo , Animales , Daño del ADN/genética , Daño del ADN/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Oocitos/metabolismo , Protaminas/metabolismo , Espermátides/metabolismo , Espermatogénesis/genética , Espermatogénesis/fisiología , Espermatozoides/fisiología
13.
Mol Hum Reprod ; 21(2): 157-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25354701

RESUMEN

We recently identified the DPY19L2 gene as the main genetic cause of human globozoospermia (70%) and described that Dpy19l2 knockout (KO) mice faithfully reproduce the human phenotype of globozoospermia making it an excellent model to characterize the molecular physiopathology of globozoospermia. Recent case studies on non-genetically characterized men with globozoospermia showed that phospholipase C, zeta (PLCζ), the sperm factor thought to induce the Ca(2+) oscillations at fertilization, was absent from their sperm, explaining the poor fertilization potential of these spermatozoa. Since 30% of globozoospermic men remain genetically uncharacterized, the absence of PLCζ in DPY19L2 globozoospermic men remains to be formally established. Moreover, the precise localization of PLCζ and the reasons underlying its loss during spermatogenesis in globozoospermic patients are still not understood. Herein, we show that PLCζ is absent, or its presence highly reduced, in human and mouse sperm with DPY19L2-associated globozoospermia. As a consequence, fertilization with sperm from Dpy19l2 KO mice failed to initiate Ca(2+) oscillations and injected oocytes remained arrested at the metaphase II stage, although a few human oocytes injected with DPY19L2-defective sperm showed formation of 2-pronuclei embryos. We report for the first time the subcellular localization of PLCζ in control human sperm, which is along the inner acrosomal membrane and in the perinuclear theca, in the area corresponding to the equatorial region. Because these cellular components are absent in globozoospermic sperm, the loss of PLCζ in globozoospermic sperm is thus consistent and reinforces the role of PLCζ as an oocyte activation factor necessary for oocyte activation. In our companion article, we showed that chromatin compaction during spermiogenesis in Dpy19l2 KO mouse is defective and leads to sperm DNA damage. Together, these defects explain the poor fertilization potential of DPY19L2-globozoospermic sperm and the compromised developmental potential of embryos obtained using sperm from patients with a deletion of the DPY19L2 gene.


Asunto(s)
Proteínas de la Membrana/deficiencia , Oocitos/metabolismo , Espermatozoides/enzimología , Espermatozoides/fisiología , Fosfolipasas de Tipo C/metabolismo , Acrosoma/metabolismo , Animales , Femenino , Humanos , Infertilidad Masculina/enzimología , Infertilidad Masculina/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados
14.
J Biol Chem ; 288(49): 35307-20, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24129574

RESUMEN

Fertilization competence is acquired in the female tract in a process known as capacitation. Capacitation is needed for the activation of motility (e.g. hyperactivation) and to prepare the sperm for an exocytotic process known as acrosome reaction. Although the HCO3(-)-dependent soluble adenylyl cyclase Adcy10 plays a role in motility, less is known about the source of cAMP in the sperm head. Transmembrane adenylyl cyclases (tmACs) are another possible source of cAMP. These enzymes are regulated by stimulatory heterotrimeric Gs proteins; however, the presence of Gs or tmACs in mammalian sperm has been controversial. In this study, we used Western blotting and cholera toxin-dependent ADP-ribosylation to show the Gs presence in the sperm head. Also, we showed that forskolin, a tmAC-specific activator, induces cAMP accumulation in sperm from both WT and Adcy10-null mice. This increase is blocked by the tmAC inhibitor SQ22536 but not by the Adcy10 inhibitor KH7. Although Gs immunoreactivity and tmAC activity are detected in the sperm head, PKA is only found in the tail, where Adcy10 was previously shown to reside. Consistent with an acrosomal localization, Gs reactivity is lost in acrosome-reacted sperm, and forskolin is able to increase intracellular Ca(2+) and induce the acrosome reaction. Altogether, these data suggest that cAMP pathways are compartmentalized in sperm, with Gs and tmAC in the head and Adcy10 and PKA in the flagellum.


Asunto(s)
Adenilil Ciclasas/metabolismo , AMP Cíclico/metabolismo , Espermatozoides/metabolismo , Acrosoma/metabolismo , Reacción Acrosómica/efectos de los fármacos , Adenina/análogos & derivados , Adenina/farmacología , Adenilil Ciclasas/deficiencia , Adenilil Ciclasas/genética , Animales , Calcio/metabolismo , Compartimento Celular , Colforsina/farmacología , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Capacitación Espermática/efectos de los fármacos , Cabeza del Espermatozoide/metabolismo , Cola del Espermatozoide/metabolismo , Espermatozoides/efectos de los fármacos
15.
Am J Hum Genet ; 88(3): 351-61, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21397064

RESUMEN

An increasing number of couples require medical assistance to achieve a pregnancy, and more than 2% of the births in Western countries now result from assisted reproductive technologies. To identify genetic variants responsible for male infertility, we performed a whole-genome SNP scan on patients presenting with total globozoospermia, a primary infertility phenotype characterized by the presence of 100% round acrosomeless spermatozoa in the ejaculate. This strategy allowed us to identify in most patients (15/20) a 200 kb homozygous deletion encompassing only DPY19L2, which is highly expressed in the testis. Although there was no known function for DPY19L2 in humans, previous work indicated that its ortholog in C. elegans is involved in cell polarity. In man, the DPY19L2 region has been described as a copy-number variant (CNV) found to be duplicated and heterozygously deleted in healthy individuals. We show here that the breakpoints of the deletions are located on a highly homologous 28 kb low copy repeat (LCR) sequence present on each side of DPY19L2, indicating that the identified deletions were probably produced by nonallelic homologous recombination (NAHR) between these two regions. We demonstrate that patients with globozoospermia have a homozygous deletion of DPY19L2, thus indicating that DPY19L2 is necessary in men for sperm head elongation and acrosome formation. A molecular diagnosis can now be proposed to affected men; the presence of the deletion confirms the diagnosis of globozoospermia and assigns a poor prognosis for the success of in vitro fertilization.


Asunto(s)
Acrosoma/patología , Eliminación de Gen , Infertilidad Masculina/genética , Proteínas de la Membrana/genética , Cabeza del Espermatozoide/patología , Acrosoma/metabolismo , Variaciones en el Número de Copia de ADN/genética , Familia , Femenino , Ligamiento Genético , Sitios Genéticos/genética , Homocigoto , Humanos , Jordania , Masculino , Linaje , Cabeza del Espermatozoide/metabolismo
16.
J Cell Sci ; 125(Pt 2): 473-85, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22302997

RESUMEN

Mammalian sperm require time in the female tract in order to be able to fertilize an egg. The physiological changes that render the sperm able to fertilize are known as capacitation. Capacitation is associated with an increase in intracellular pH, an increase in intracellular calcium and phosphorylation of different proteins. This process is also accompanied by the hyperpolarization of the sperm plasma membrane potential. Recently, we presented evidence showing that epithelial Na+ channels (ENaC) are present in mature sperm and that ENaCs are blocked during capacitation. In the present work, we used flow cytometry to analyze changes in intracellular Na+ concentration ([Na+](i)) during capacitation in individual cells. Our results indicate that capacitated sperm have lower Na+ concentrations. Using sperm with green fluorescent protein in their acrosomes, it was shown that the lower [Na+](i) concentration only occurs in sperm having intact acrosomes. ENaC inhibition has been shown in other cell types to depend on the activation of cystic fibrosis transmembrane conductance regulator (CFTR). In non-capacitated sperm, amiloride, an ENaC inhibitor, and genistein, a CFTR activator, caused a decrease in [Na+](i), suggesting that also in these cells [Na+](i) is dependent on the crosstalk between ENaC and CFTR. In addition, PKA inhibition blocked [Na+](i) decrease in capacitated sperm. Altogether, these data are consistent with the hypothesis that the capacitation-associated hyperpolarization involves a decrease in [Na+](i) mediated by inhibition of ENaC and regulated by PKA through activation of CFTR channels.


Asunto(s)
Sodio/metabolismo , Capacitación Espermática , Espermatozoides/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Bloqueadores del Canal de Sodio Epitelial/farmacología , Citometría de Flujo , Genisteína/farmacología , Masculino , Ratones , Espermatozoides/efectos de los fármacos
17.
Elife ; 122024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38441556

RESUMEN

From a cohort of 167 infertile patients suffering from multiple morphological abnormalities of the flagellum (MMAF), pathogenic bi-allelic mutations were identified in the CCDC146 gene. In somatic cells, CCDC146 is located at the centrosome and at multiple microtubule-related organelles during mitotic division, suggesting that it is a microtubule-associated protein (MAP). To decipher the molecular pathogenesis of infertility associated with CCDC146 mutations, a Ccdc146 knock-out (KO) mouse line was created. KO male mice were infertile, and sperm exhibited a phenotype identical to CCDC146 mutated patients. CCDC146 expression starts during late spermiogenesis. In the spermatozoon, the protein is conserved but is not localized to centrioles, unlike in somatic cells, rather it is present in the axoneme at the level of microtubule doublets. Expansion microscopy associated with the use of the detergent sarkosyl to solubilize microtubule doublets suggests that the protein may be a microtubule inner protein (MIP). At the subcellular level, the absence of CCDC146 impacted all microtubule-based organelles such as the manchette, the head-tail coupling apparatus (HTCA), and the axoneme. Through this study, a new genetic cause of infertility and a new factor in the formation and/or structure of the sperm axoneme were characterized.


Asunto(s)
Anomalías Múltiples , Infertilidad Masculina , Animales , Humanos , Masculino , Ratones , Centriolos , Infertilidad Masculina/genética , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Semen
18.
J Cell Physiol ; 228(3): 590-601, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22833409

RESUMEN

Mammalian sperm must undergo a maturational process, named capacitation, in the female reproductive tract to fertilize the egg. Sperm capacitation is regulated by a cAMP/protein kinase A (PKA) pathway and involves increases in intracellular Ca(2+), pH, Cl(-), protein tyrosine phosphorylation, and in mouse and some other mammals a membrane potential hyperpolarization. The cystic fibrosis transmembrane conductance regulator (CFTR), a Cl(-) channel modulated by cAMP/PKA and ATP, was detected in mammalian sperm and proposed to modulate capacitation. Our whole-cell patch-clamp recordings from testicular mouse sperm now reveal a Cl(-) selective component to membrane current that is ATP-dependent, stimulated by cAMP, cGMP, and genistein (a CFTR agonist, at low concentrations), and inhibited by DPC and CFTR(inh) -172, two well-known CFTR antagonists. Furthermore, the Cl(-) current component activated by cAMP and inhibited by CFTR(inh) -172 is absent in recordings on testicular sperm from mice possessing the CFTR ΔF508 loss-of-function mutation, indicating that CFTR is responsible for this component. A Cl(-) selective like current component displaying CFTR characteristics was also found in wild type epididymal sperm bearing the cytoplasmatic droplet. Capacitated sperm treated with CFTR(inh) -172 undergo a shape change, suggesting that CFTR is involved in cell volume regulation. These findings indicate that functional CFTR channels are present in mouse sperm and their biophysical properties are consistent with their proposed participation in capacitation.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Espermatozoides/metabolismo , Animales , Benzoatos/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fenómenos Electrofisiológicos , Femenino , Genisteína/farmacología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Técnicas de Placa-Clamp , Capacitación Espermática/fisiología , Tiazolidinas/farmacología , ortoaminobenzoatos/farmacología
19.
Elife ; 122023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37934199

RESUMEN

Male infertility is common and complex, presenting a wide range of heterogeneous phenotypes. Although about 50% of cases are estimated to have a genetic component, the underlying cause often remains undetermined. Here, from whole-exome sequencing on samples from 168 infertile men with asthenoteratozoospermia due to severe sperm flagellum, we identified homozygous ZMYND12 variants in four unrelated patients. In sperm cells from these individuals, immunofluorescence revealed altered localization of DNAH1, DNALI1, WDR66, and TTC29. Axonemal localization of ZMYND12 ortholog TbTAX-1 was confirmed using the Trypanosoma brucei model. RNAi knock-down of TbTAX-1 dramatically affected flagellar motility, with a phenotype similar to the sperm from men bearing homozygous ZMYND12 variants. Co-immunoprecipitation and ultrastructure expansion microscopy in T. brucei revealed TbTAX-1 to form a complex with TTC29. Comparative proteomics with samples from Trypanosoma and Ttc29 KO mice identified a third member of this complex: DNAH1. The data presented revealed that ZMYND12 is part of the same axonemal complex as TTC29 and DNAH1, which is critical for flagellum function and assembly in humans, and Trypanosoma. ZMYND12 is thus a new asthenoteratozoospermia-associated gene, bi-allelic variants of which cause severe flagellum malformations and primary male infertility.


Asunto(s)
Astenozoospermia , Infertilidad Masculina , Humanos , Masculino , Animales , Ratones , Semen , Flagelos , Fertilidad , Proteínas de Unión al Calcio , Dineínas
20.
Med Sci (Paris) ; 28(5): 512-8, 2012 May.
Artículo en Francés | MEDLINE | ID: mdl-22643005

RESUMEN

The spermatozoon is one of the most differentiated cells in mammals and its production requires an extremely complex machinery. Subtle but critical molecular changes take place during capacitation, which comprises the last series of maturation steps that naturally occur between the cauda epididymidis where spermatozoa are stored and their ultimate destination inside the oocyte. Phospholipases, by hydrolyzing various phospholipids, have been found to be critical in sperm processes such as 1) the control of flagellum beats, 2) capacitation - the molecular transformations preparing the sperm for fertilization, 3) acrosome reaction and 4) oocyte activation by eliciting calcium oscillations. The emerging important role of phospholipases is also emphasized by the fact that alterations of sperm lipids can lead to infertility. Phospholipases may represent valuable targets to develop anti- and pro-fertility drugs. Results obtained in mice are encouraging, since treatment of sperm with recombinant sPLA(2) of group X, known to be involved in capacitation, improves fertilization in vitro, while co-injection of PLCζ RNA with infertile sperm restores oocyte activation.


Asunto(s)
Infertilidad Masculina/terapia , Terapia Molecular Dirigida/métodos , Fosfolipasas/fisiología , Espermatozoides/enzimología , Espermatozoides/fisiología , Reacción Acrosómica/genética , Reacción Acrosómica/fisiología , Animales , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/metabolismo , Humanos , Infertilidad Masculina/etiología , Infertilidad Masculina/metabolismo , Masculino , Ratones , Modelos Biológicos , Fosfolipasas/antagonistas & inhibidores , Fosfolipasas/genética , Fosfolipasas/metabolismo , Motilidad Espermática/genética , Motilidad Espermática/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA