Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 134(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34313315

RESUMEN

Ca2+ influx during oocyte maturation and after sperm entry is necessary to fill the internal Ca2+ stores and for complete egg activation. We knocked out the transient receptor potential vanilloid member 3 (TRPV3) and the T-type channel, CaV3.2, to determine their necessity for maintaining these functions in mammalian oocytes/eggs. Double-knockout (dKO) females were subfertile, their oocytes and eggs showed reduced internal Ca2+ stores, and, following sperm entry or Plcz (also known as Plcz1) cRNA injection, fewer dKO eggs displayed Ca2+ responses compared to wild-type eggs, which were also of lower frequency. These parameters were rescued and/or enhanced by removing extracellular Mg2+, suggesting that the residual Ca2+ influx could be mediated by the TRPM7 channel, consistent with the termination of divalent-cation oscillations in dKO eggs by a TRPM7 inhibitor. In total, we demonstrated that TRPV3 and CaV3.2 mediate the complete filling of the Ca2+ stores in mouse oocytes and eggs. We also showed that they are required for initiating and maintaining regularly spaced-out oscillations, suggesting that Ca2+ influx through PM ion channels dictates the periodicity and persistence of Ca2+ oscillations during mammalian fertilization.


Asunto(s)
Canales de Calcio Tipo T , Calcio , Oocitos , Canales Catiónicos TRPV , Animales , Calcio/metabolismo , Canales de Calcio Tipo T/genética , Femenino , Fertilidad , Fertilización , Eliminación de Gen , Homeostasis , Ratones , Ratones Noqueados , Oocitos/metabolismo , Canales Catiónicos TRPM , Canales Catiónicos TRPV/genética
2.
Reproduction ; 165(1): 79-91, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36215093

RESUMEN

In brief: Oocyte quality remains the most important and unsolved issue in reproduction. Our data show that multidrug resistance transporters and oocyte mitochondria are involved in determining oocyte quality in a mouse model. Abstract: Multidrug resistance transporter-1 (MDR-1) is a transmembrane ATP-dependent effluxer present in organs that transport a variety of xenobiotics and by-products. Previous findings by our group demonstrated that this transporter is also present in the oocyte mitochondrial membrane and that its mutation led to abnormal mitochondrial homeostasis. Considering the importance of these organelles in the female gamete, we assessed the impact of MDR-1 dysfunction on mouse oocyte quality, with a particular focus on the meiotic spindle organization, aneuploidies, Ca2+ homeostasis, ATP production and mtDNA mutations. Our results demonstrate that young Mdr1a mutant mice produce oocytes characterized by lower quality, with a significant delay in the germinal vesicle to germinal vesicle breakdown transition, an increased percentage of symmetric divisions, chromosome misalignments and a severely altered meiotic spindle shape compared to the wild types. Mutant oocytes exhibit 7000 more SNPs in the exomic DNA and twice the amount of mitochondrial DNA (mtDNA) SNPs compared to the wild-type ones. Ca2+ analysis revealed the inability of MDR-1 mutant oocytes to manage Ca2+ storage content and oscillations in response to several stimuli, and ATP quantification shows that mutant oocytes trend toward lower ATP levels compared to wild types. Finally, 1-year-old mutant ovaries express a lower amount of SIRT1, SIRT3, SIRT5, SIRT6 and SIRT7 compared to wild-type levels. These results together emphasize the importance of MDR-1 in mitochondrial physiology and highlight the influence of MDR-1 on oocyte quality and ovarian aging.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Calcio , Meiosis , Oocitos , Sirtuinas , Animales , Femenino , Ratones , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , ADN Mitocondrial/genética , Resistencia a Múltiples Medicamentos , Homeostasis , Oocitos/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo , Huso Acromático/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo
3.
Reproduction ; 164(1): F9-F20, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35521902

RESUMEN

The discovery of PLCZ1 nearly 20 years ago as the primary Ca2+ oscillation-inducing factor in the sperm of mammals represented a significant breakthrough in our quest to elucidate the molecules and pathways that promote egg activation during fertilization. The advent of the intracytoplasmic sperm injection (ICSI) technique, which made fertilization possible without sperm capacitation, acrosome reaction, and gamete fusion, strengthened the research that led to the discovery of PLCZ1 and became an essential clinical tool for humans. The use of ICSI combined with the detection of PLCZ1 expression and mutations in infertile patients established the fundamental role of PLCZ1 in human fertility while leading to the discovery of novel components of the perinuclear theca, the site of the residence of PLCZ1 in sperm before fertilization. Remarkably, the more extensive use of ICSI in species other than humans and mice revealed poor success and exposed gaps in our understanding of PLCZ1 release and/or activation. Similarly, fertilization using sperm from mouse models lacking Plcz1 has produced striking results whose true implications are yet to be determined. Nevertheless, answers to these unresolved questions will produce a complete picture of the adaptations and molecular players that mammalian species employ to ensure the success of the triggering event of embryo development that has linked generations since the beginning of times.


Asunto(s)
Oocitos , Inyecciones de Esperma Intracitoplasmáticas , Animales , Fertilización , Humanos , Masculino , Mamíferos/metabolismo , Ratones , Fosfoinositido Fosfolipasa C/genética , Fosfoinositido Fosfolipasa C/metabolismo , Espermatozoides/metabolismo
4.
J Cell Sci ; 132(3)2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30659110

RESUMEN

In mammals, fertilization initiates Ca2+ oscillations in metaphase II oocytes, which are required for the activation of embryo development. Germinal vesicle (GV) oocytes also display Ca2+ oscillations, although these unfold spontaneously in the absence of any known agonist(s) and their function remains unclear. We found that the main intracellular store of Ca2+ in GV oocytes, the endoplasmic reticulum ([Ca2+]ER), constitutively 'leaks' Ca2+ through the type 1 inositol 1,4,5-trisphosphate receptor. The [Ca2+]ER leak ceases around the resumption of meiosis, the GV breakdown (GVBD) stage, which coincides with the first noticeable accumulation of Ca2+ in the stores. It also concurs with downregulation of the Ca2+ influx and termination of the oscillations, which seemed underpinned by the inactivation of the putative plasma membrane Ca2+ channels. Lastly, we demonstrate that mitochondria take up Ca2+ during the Ca2+ oscillations, mounting their own oscillations that stimulate the mitochondrial redox state and increase the ATP levels of GV oocytes. These distinct features of Ca2+ homeostasis in GV oocytes are likely to underpin the acquisition of both maturation and developmental competence, as well as fulfill stage-specific cellular functions during oocyte maturation.


Asunto(s)
Señalización del Calcio/genética , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Mitocondrias/metabolismo , Oocitos/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Femenino , Regulación de la Expresión Génica , Homeostasis/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Metafase , Ratones , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Oocitos/citología , Oogénesis/genética , Cultivo Primario de Células , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Proteína 25 Asociada a Sinaptosomas/deficiencia , Proteína 25 Asociada a Sinaptosomas/genética
5.
Mol Cell ; 52(3): 325-39, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24095281

RESUMEN

Active metabolism regulates oocyte cell death via calcium/calmodulin-dependent protein kinase II (CaMKII)-mediated phosphorylation of caspase-2, but the link between metabolic activity and CaMKII is poorly understood. Here we identify coenzyme A (CoA) as the key metabolic signal that inhibits Xenopus laevis oocyte apoptosis by directly activating CaMKII. We found that CoA directly binds to the CaMKII regulatory domain in the absence of Ca(2+) to activate CaMKII in a calmodulin-dependent manner. Furthermore, we show that CoA inhibits apoptosis not only in X. laevis oocytes but also in Murine oocytes. These findings uncover a direct mechanism of CaMKII regulation by metabolism and further highlight the importance of metabolism in preserving oocyte viability.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Coenzima A/metabolismo , Oocitos/metabolismo , Xenopus laevis/metabolismo , Animales , Apoptosis/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Caspasa 2/metabolismo , Supervivencia Celular/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Oocitos/crecimiento & desarrollo , Fosforilación/genética , Unión Proteica , Transducción de Señal , Activación Transcripcional , Xenopus laevis/crecimiento & desarrollo
6.
Proc Natl Acad Sci U S A ; 115(44): E10370-E10378, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30322909

RESUMEN

The success of mammalian development following fertilization depends on a series of transient increases in egg cytoplasmic Ca2+, referred to as Ca2+ oscillations. Maintenance of these oscillations requires Ca2+ influx across the plasma membrane, which is mediated in part by T-type, CaV3.2 channels. Here we show using genetic mouse models that TRPM7 channels are required to support this Ca2+ influx. Eggs lacking both TRPM7 and CaV3.2 stop oscillating prematurely, indicating that together they are responsible for the majority of Ca2+ influx immediately following fertilization. Fertilized eggs lacking both channels also frequently display delayed resumption of Ca2+ oscillations, which appears to require sperm-egg fusion. TRPM7 and CaV3.2 channels almost completely account for Ca2+ influx observed following store depletion, a process previously attributed to canonical store-operated Ca2+ entry mediated by STIM/ORAI interactions. TRPM7 serves as a membrane sensor of extracellular Mg2+ and Ca2+ concentrations and mediates the effects of these ions on Ca2+ oscillation frequency. When bred to wild-type males, female mice carrying eggs lacking TRPM7 and CaV3.2 are subfertile, and their offspring have increased variance in postnatal weight. These in vivo findings confirm previous observations linking in vitro experimental alterations in Ca2+ oscillatory patterns with developmental potential and offspring growth. The identification of TRPM7 and CaV3.2 as key mediators of Ca2+ influx following fertilization provides a mechanistic basis for the rational design of culture media that optimize developmental potential in research animals, domestic animals, and humans.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Fertilización/fisiología , Canales Catiónicos TRPM/metabolismo , Cigoto/metabolismo , Animales , Membrana Celular/metabolismo , Citoplasma/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Oocitos/metabolismo , Espermatozoides/metabolismo , Molécula de Interacción Estromal 1/metabolismo
7.
J Biol Chem ; 294(31): 11876-11891, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31201271

RESUMEN

Ca2+ oscillations and consequent Ca2+/calmodulin-dependent protein kinase II (CaMKII) activation are required for embryogenesis, as well as neuronal, immunological, and cardiac signaling. Fertilization directly results in Ca2+ oscillations, but the resultant pattern of CaMKII activity remains largely unclear. To address this gap, we first employed the one existing biosensor for CaMKII activation. This sensor, Camui, comprises CaMKIIα and therefore solely reports on the activation of this CaMKII variant. Additionally, to detect the activity of all endogenous CaMKII variants simultaneously, we constructed a substrate-based sensor for CaMKII activity, FRESCA (FRET-based sensor for CaMKII activity). To examine the differential responses of the Camui and FRESCA sensors, we used several approaches to stimulate Ca2+ release in mouse eggs, including addition of phospholipase Cζ cRNA, which mimics natural fertilization. We found that the Camui response is delayed or terminates earlier than the FRESCA response. FRESCA enables assessment of endogenous CaMKII activity in real-time by both fertilization and artificial reagents, such as Sr2+, which also leads to CaMKII activation. FRESCA's broad utility will be important for optimizing artificial CaMKII activation for clinical use to manage infertility. Moreover, FRESCA provides a new view on CaMKII activity, and its application in additional biological systems may reveal new signaling paradigms in eggs, as well as in neurons, cardiomyocytes, immune cells, and other CaMKII-expressing cells.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Animales , Técnicas Biosensibles/métodos , Fertilización , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Ionomicina/farmacología , Ratones , Óvulo/efectos de los fármacos , Óvulo/metabolismo , Fosfoinositido Fosfolipasa C/metabolismo
8.
J Cell Physiol ; 235(5): 4351-4360, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31612508

RESUMEN

Compensatory endocytosis (CE) is one of the primary mechanisms through which cells maintain their surface area after exocytosis. Considering that in eggs massive exocytosis of cortical granules (CG) takes place after fertilization, the aim of this study was to evaluate the occurrence of CE following cortical exocytosis in mouse eggs. For this purpose, we developed a pulse-chase assay to detect CG membrane internalization. Results showed internalized labeling in SrCl2 -activated and fertilized eggs when chasing at 37°C, but not at a nonpermissive temperature (4°C). The use of kinase and calcineurin inhibitors led us to conclude that this internal labeling corresponded to CE. Further experiments showed that CE in mouse eggs is dependent on actin dynamics and dynamin activity, and could be associated with a transient exposure of phosphatidylserine. Finally, CE was impaired in A23187 ionophore-activated eggs, highlighting once again the mechanistic differences between the activation methods. Altogether, these results demonstrate for the first time that egg activation triggers CE in mouse eggs after exocytosis of CG, probably as a plasma membrane homeostasis mechanism.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Endocitosis/fisiología , Exocitosis/fisiología , Óvulo/fisiología , Animales , Calcio/metabolismo , Femenino , Fertilización/fisiología , Masculino , Ratones
9.
Development ; 144(16): 2914-2924, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28694258

RESUMEN

Activation of the egg by the sperm is the first, vital stage of embryogenesis. The sperm protein PLCζ has been proposed as the physiological agent that triggers the Ca2+ oscillations that normally initiate embryogenesis. Consistent with this, recombinant PLCζ induces Ca2+ oscillations in eggs and debilitating mutations in the PLCZ1 gene are associated with infertility in men. However, there has been no evidence that knockout of the gene encoding PLCζ abolishes the ability of sperm to induce Ca2+ oscillations in eggs. Here, we show that sperm derived from Plcz1-/- male mice fail to trigger Ca2+ oscillations in eggs, cause polyspermy and thus demonstrate that PLCζ is the physiological trigger of these Ca2+ oscillations. Remarkably, some eggs fertilized by PLCζ-null sperm can develop, albeit at greatly reduced efficiency, and after a significant time-delay. In addition, Plcz1-/- males are subfertile but not sterile, suggesting that in the absence of PLCζ, spontaneous egg activation can eventually occur via an alternative route. This is the first demonstration that in vivo fertilization without the normal physiological trigger of egg activation can result in offspring. PLCζ-null sperm now make it possible to resolve long-standing questions in fertilization biology, and to test the efficacy and safety of procedures used to treat human infertility.


Asunto(s)
Calcio/metabolismo , Desarrollo Embrionario/fisiología , Fosfoinositido Fosfolipasa C/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiología , Desarrollo Embrionario/genética , Edición Génica , Masculino , Mamíferos , Ratones , Ratones Mutantes , Fosfoinositido Fosfolipasa C/genética , Espermatogénesis/genética , Espermatogénesis/fisiología
10.
Mol Reprod Dev ; 86(1): 4-19, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30411426

RESUMEN

More than 15 years have elapsed since the identification of phospholipase C ζ1 (PLCζ) from a genomic search for mouse testis/sperm-specific PLCs. This molecule was proposed to represent the sperm factor responsible for the initiation of calcium (Ca2+ ) oscillations required for egg activation and embryo development in mammals. Supporting evidence for this role emerged from studies documenting its expression in all mammals and other vertebrate species, the physiological Ca2+ rises induced by injection of its messenger RNA into mammalian and nonmammalian eggs, and the lack of expression in infertile males that fail intracytoplasmic sperm injection. In the last year, genetic animal models have added support to its role as the long sought-after sperm factor. In this review, we highlight the findings that demonstrated the role of Ca2+ as the universal signal of egg activation and the experimental buildup that culminated with the identification of PLCζ as the soluble sperm factor. We also discuss the structural-functional properties that make PLCζ especially suited to evoke oscillations in eggs. Lastly, we examine unresolved aspects of the function and regulation of PLCζ and whether or not it is the only sperm factor in mammalian sperm.


Asunto(s)
Señalización del Calcio , Embrión de Mamíferos/enzimología , Desarrollo Embrionario , Fosfoinositido Fosfolipasa C/metabolismo , Interacciones Espermatozoide-Óvulo , Espermatozoides/enzimología , Animales , Femenino , Humanos , Infertilidad Masculina/enzimología , Masculino , Ratones , Inyecciones de Esperma Intracitoplasmáticas
11.
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
12.
Anim Biotechnol ; 29(3): 190-198, 2018 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28799828

RESUMEN

Phospholipase C zeta, a novel sperm-specific protein which is widely known to induce oocyte activation following fertilization, had already been characterized in various mammalian species, but not in water buffaloes thus far. The present study was conducted to initially characterize and compare the sequences of PLCZ1 gene of swamp and riverine buffaloes. Semen samples were collected; total RNA was extracted and reverse-transcribed. PLCZ1 cDNA was then amplified, and submitted for sequencing. Buffalo PLCZ1 gene yielded a sequence of 1905 base pair nucleotides translated into 634 bp amino acids. In general, the buffalo PLCZ1 gene was found to have high sequence identity with cattle and other domestic species. Similarly, significant residues and motifs in PLCZ1 gene sequence are found conserved in water buffaloes. However, there are variations in sequences identified between types of water buffaloes that may play a role in species-specific differences in terms of gene and protein expression, physiological mechanisms, and biological functions. The molecular information on buffalo PLCZ1 gene is highly valuable in subsequent works such as correlation studies on the identified gene variations with semen quality and fertility, and the development of biomarkers for bull fertility.


Asunto(s)
Búfalos/genética , Fosfoinositido Fosfolipasa C/genética , Animales , Fertilidad/genética , Marcadores Genéticos/genética , Masculino , Tipificación Molecular , Filogenia , ARN/genética , ARN/aislamiento & purificación , Análisis de Secuencia de ADN , Espermatozoides/química
13.
Reproduction ; 154(3): 307-318, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28751536

RESUMEN

The efficiency of intracytoplasmic sperm injection (ICSI) in the bovine is low compared to other species. It is unknown whether defective oocyte activation and/or sperm head decondensation limit the success of this technique in this species. To elucidate where the main obstacle lies, we used homologous and heterologous ICSI and parthenogenetic activation procedures. We also evaluated whether in vitro maturation negatively impacted the early stages of activation after ICSI. Here we showed that injected bovine sperm are resistant to nuclear decondensation by bovine oocytes and this is only partly overcome by exogenous activation. Remarkably, when we used heterologous ICSI, in vivo-matured mouse eggs were capable of mounting calcium oscillations and displaying normal PN formation following injection of bovine sperm, although in vitro-matured mouse oocytes were unable to do so. Together, our data demonstrate that bovine sperm are especially resistant to nuclear decondensation by in vitro-matured oocytes and this deficiency cannot be simply overcome by exogenous activation protocols, even by inducing physiological calcium oscillations. Therefore, the inability of a suboptimal ooplasmic environment to induce sperm head decondensation limits the success of ICSI in the bovine. Studies aimed to improve the cytoplasmic milieu of in vitro-matured oocytes and to replicate the molecular changes associated with in vivo capacitation and acrosome reaction will deepen our understanding of the mechanism of fertilization and improve the success of ICSI in this species.


Asunto(s)
Enfermedades de los Bovinos/terapia , Núcleo Celular/patología , Ensamble y Desensamble de Cromatina , Infertilidad Masculina/veterinaria , Cabeza del Espermatozoide/patología , Inyecciones de Esperma Intracitoplasmáticas/veterinaria , Interacciones Espermatozoide-Óvulo , Animales , Señalización del Calcio , Bovinos , Enfermedades de los Bovinos/metabolismo , Enfermedades de los Bovinos/patología , Núcleo Celular/metabolismo , Células Cultivadas , Técnicas de Cultivo de Embriones/veterinaria , Femenino , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Infertilidad Masculina/terapia , Masculino , Ratones , Partenogénesis , Especificidad de la Especie , Capacitación Espermática , Cabeza del Espermatozoide/metabolismo
14.
Exp Cell Res ; 347(1): 42-51, 2016 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-27423421

RESUMEN

Fusion of cortical granules with the oocyte plasma membrane is the most significant event to prevent polyspermy. This particular exocytosis, also known as cortical reaction, is regulated by calcium and its molecular mechanism is still not known. Rab3A, a member of the small GTP-binding protein superfamily, has been implicated in calcium-dependent exocytosis and is not yet clear whether Rab3A participates in cortical granules exocytosis. Here, we examine the involvement of Rab3A in the physiology of cortical granules, particularly, in their distribution during oocyte maturation and activation, and their participation in membrane fusion during cortical granule exocytosis. Immunofluorescence and Western blot analysis showed that Rab3A and cortical granules have a similar migration pattern during oocyte maturation, and that Rab3A is no longer detected after cortical granule exocytosis. These results suggested that Rab3A might be a marker of cortical granules. Overexpression of EGFP-Rab3A colocalized with cortical granules with a Pearson correlation coefficient of +0.967, indicating that Rab3A and cortical granules have almost a perfect colocalization in the egg cortical region. Using a functional assay, we demonstrated that microinjection of recombinant, prenylated and active GST-Rab3A triggered cortical granule exocytosis, indicating that Rab3A has an active role in this secretory pathway. To confirm this active role, we inhibited the function of endogenous Rab3A by microinjecting a polyclonal antibody raised against Rab3A prior to parthenogenetic activation. Our results showed that Rab3A antibody microinjection abolished cortical granule exocytosis in parthenogenetically activated oocytes. Altogether, our findings confirm that Rab3A might function as a marker of cortical granules and participates in cortical granule exocytosis in mouse eggs.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Exocitosis , Oocitos/citología , Oocitos/metabolismo , Proteína de Unión al GTP rab3A/metabolismo , Animales , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Caballos , Humanos , Metafase , Ratones , Microinyecciones , Proteínas Recombinantes de Fusión/metabolismo
15.
Mar Drugs ; 15(10)2017 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-29027927

RESUMEN

Voltage-gated calcium (CaV) channels are widely expressed and are essential for the completion of multiple physiological processes. Close regulation of their activity by specific inhibitors and agonists become fundamental to understand their role in cellular homeostasis as well as in human tissues and organs. CaV channels are divided into two groups depending on the membrane potential required to activate them: High-voltage activated (HVA, CaV1.1-1.4; CaV2.1-2.3) and Low-voltage activated (LVA, CaV3.1-3.3). HVA channels are highly expressed in brain (neurons), heart, and adrenal medulla (chromaffin cells), among others, and are also classified into subtypes which can be distinguished using pharmacological approaches. Cone snails are marine gastropods that capture their prey by injecting venom, "conopeptides", which cause paralysis in a few seconds. A subset of conopeptides called conotoxins are relatively small polypeptides, rich in disulfide bonds, that target ion channels, transporters and receptors localized at the neuromuscular system of the animal target. In this review, we describe the structure and properties of conotoxins that selectively block HVA calcium channels. We compare their potency on several HVA channel subtypes, emphasizing neuronal calcium channels. Lastly, we analyze recent advances in the therapeutic use of conotoxins for medical treatments.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Conotoxinas/farmacología , Animales , Bloqueadores de los Canales de Calcio/química , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio/metabolismo , Conotoxinas/química , Humanos , Potenciales de la Membrana/efectos de los fármacos , Caracoles
16.
J Biol Chem ; 289(23): 16085-99, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24764305

RESUMEN

The endoplasmic reticulum (ER) is organized in part by adapter proteins that nucleate the formation of large protein complexes. Tetratricopeptide repeats (TPR) are well studied protein structural motifs that support intermolecular protein-protein interactions. TMTC1 and TMTC2 were identified by an in silico search as TPR-containing proteins possessing N-terminal ER targeting signal sequences and multiple hydrophobic segments, suggestive of polytopic membrane proteins that are targeted to the secretory pathway. A variety of cell biological and biochemical assays was employed to demonstrate that TMTC1 and TMTC2 are both ER resident integral membrane proteins with multiple clusters of TPR domains oriented within the ER lumen. Proteomic analysis followed by co-immunoprecipitation verification found that both proteins associated with the ER calcium uptake pump SERCA2B, and TMTC2 also bound to the carbohydrate-binding chaperone calnexin. Live cell calcium measurements revealed that overexpression of either TMTC1 or TMTC2 caused a reduction of calcium released from the ER following stimulation, whereas the knockdown of TMTC1 or TMTC2 increased the stimulated calcium released. Together, these results implicate TMTC1 and TMTC2 as ER proteins involved in ER calcium homeostasis.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Calcio/metabolismo , Proteínas Portadoras/fisiología , Retículo Endoplásmico/metabolismo , Homeostasis , Proteínas de la Membrana/fisiología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Secuencia de Bases , Células COS , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Cartilla de ADN , ADN Complementario , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
J Cell Sci ; 126(Pt 24): 5714-24, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24101727

RESUMEN

Changes in the intracellular concentration of free calcium ([Ca(2+)]i) regulate diverse cellular processes including fertilization. In mammalian eggs, the [Ca(2+)]i changes induced by the sperm unfold in a pattern of periodical rises, also known as [Ca(2+)]i oscillations. The source of Ca(2+) during oscillations is the endoplasmic reticulum ([Ca(2+)]ER), but it is presently unknown how [Ca(2+)]ER is regulated. Here, we show using mouse eggs that [Ca(2+)]i oscillations induced by a variety of agonists, including PLCζ, SrCl2 and thimerosal, provoke simultaneous but opposite changes in [Ca(2+)]ER and cause differential effects on the refilling and overall load of [Ca(2+)]ER. We also found that Ca(2+) influx is required to refill [Ca(2+)]ER, because the loss of [Ca(2+)]ER was accelerated in medium devoid of Ca(2+). Pharmacological inactivation of the function of the mitochondria and of the Ca(2+)-ATPase pumps PMCA and SERCA altered the pattern of oscillations and abruptly reduced [Ca(2+)]ER, especially after inactivation of mitochondria and SERCA functions. We also examined the expression of SERCA2b protein and found that it was expressed throughout oocyte maturation and attained a conspicuous cortical cluster organization in mature eggs. We show that its overexpression reduces the duration of inositol-1,4,5-trisphosphate-induced [Ca(2+)]i rises, promotes initiation of oscillations and enhances refilling of [Ca(2+)]ER. Collectively, our results provide novel insights on the regulation of [Ca(2+)]ER oscillations, which underlie the unique Ca(2+)-signalling system that activates the developmental program in mammalian eggs.


Asunto(s)
Señalización del Calcio , Retículo Endoplásmico/metabolismo , Oocitos/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Ionóforos de Calcio/farmacología , Células Cultivadas , Femenino , Expresión Génica , Ionomicina/farmacología , Ratones , Fosfoinositido Fosfolipasa C/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Timerosal/farmacología
18.
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
19.
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
20.
J Assist Reprod Genet ; 32(9): 1415-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26174123

RESUMEN

PURPOSE: The purpose of this study is to describe impaired oocyte fertilization from phospholipase C-zeta (PLC-ζ) deficiency in normal-appearing sperm that was successfully treated using calcium (Ca(2+)) ionophore with intracytoplasmic sperm injection (ICSI) of oocytes matured in vitro. METHODS: An infertile couple undergoing in vitro fertilization (IVF) experienced failed oocyte fertilization following ICSI with normal-appearing sperm. A semen sample collected from the patient was used to assess the expression of sperm PLC- ζ protein by Western blot analysis and immunofluorescence and PLC-ζ bioactivity by an in vitro model of Ca(2+) release. A second IVF cycle was performed using Ca(2+) ionophore with ICSI to enhance Ca(2+)-induced oocyte activation of oocytes matured in vitro. RESULTS: Sperm PLC-ζ protein deficiency was demonstrated by Western blot analysis and immunofluorescence and confirmed by reduced PLC-ζ bioactivity using an in vitro model of Ca(2+) release. Nevertheless, with this sperm and supplementation of Ca(2+) ionophore following ICSI, fertilization of four of six oocytes matured in vitro was obtained. In addition, four embryos underwent cleavage and two of them reached the blastocyst stage. Transfer of these blastocysts into the uterus led to a single pregnancy and live birth. CONCLUSIONS: Deficiency of PLC-ζ in normal-appearing human sperm is associated with impaired Ca(2+)-dependent oocyte activation during ICSI. Under this condition, use of Ca(2+) ionophore following ICSI of oocytes matured in vitro improves embryo developmental competence, possibly through the activation of Ca(2+)-dependent mechanisms governing fertilization and preimplantation embryogenesis.


Asunto(s)
Fertilización/fisiología , Infertilidad/etiología , Oocitos/citología , Inducción de la Ovulación , Fosfoinositido Fosfolipasa C/deficiencia , Inyecciones de Esperma Intracitoplasmáticas/métodos , Espermatozoides/fisiología , Adulto , Western Blotting , Transferencia de Embrión , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Recién Nacido , Infertilidad/enzimología , Masculino , Oocitos/fisiología , Embarazo , Insuficiencia del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA