Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
BMC Genom Data ; 24(1): 4, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36782118

RESUMO

Cytoskeleton-related proteins are essential for cell shape maintenance and cytoskeleton remodeling. The spermatozoa of Eriocheir sinensis (Chinese mitten crab) have a unique cellular structure, and the mechanism of spermatozoal metamorphosis during the acrosome reaction is not well understood. In this study, the E. sinensis spermatozoa were induced using calcium ionophore A23187 to undergo the acrosome reaction in vitro, and the acrosome-reacting and fresh (non-reacting) spermatozoa were collected separately. The differential expression of cytoskeleton-related protein genes in acrosome-reacting and fresh spermatozoa of E. sinensis was analyzed by whole transcriptome sequencing and bioinformatics analysis, and PPI network and miRNA-mRNA regulation network were constructed to analyze their possible function and regulation mechanism. The results showed that numerous differentially expressed cytoskeleton-related protein genes, miRNAs and lncRNAs were found in acrosome-reacting and fresh spermatozoa of E. sinensis; 27 cytoskeleton-related protein genes were down regulated and 687 miRNAs were up regulated in acrosome-reacting spermatozoa; 147 miRNAs target these 27 cytoskeleton-related protein genes. In the PPI networks, RAC1, BCAR1, RDX, NCKAP1, EPS8, CDC42BPA, LIMK1, ELMO2, GNAI1 and OCRL were identified as hub proteins. These proteins are mainly involved in the regulation of cytoskeleton organization, actin cytoskeleton organization, microtubule skeleton organization and small GTPase-mediated signal transduction and other biological processes, and play roles in pathways such as actin cytoskeletal regulation and axon guidance. miR-9, miR-31 and two novel miRNAs in the miRNA-mRNA regulatory network are the core miRNAs targeting cytoskeleton-related protein genes. miR-9 targets and regulates OBSCN, CDC42BPA, ELMO2, BCAS3, TPR and OCRL; while miR-31 targets and regulates CDC42BPA and TPR. This study provides a theoretical basis for revealing the mechanism of acrosome reaction under the special spermatozoa morphology of E. sinensis.


Assuntos
Reação Acrossômica , Braquiúros , Proteínas do Citoesqueleto , MicroRNAs , Espermatozoides , Masculino , Reação Acrossômica/genética , Reação Acrossômica/fisiologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/genética , MicroRNAs/genética , RNA Mensageiro/genética , Espermatozoides/metabolismo , Braquiúros/genética , Braquiúros/metabolismo
2.
Commun Biol ; 5(1): 984, 2022 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-36115925

RESUMO

SPACA6 is a sperm-expressed surface protein that is critical for gamete fusion during mammalian sexual reproduction. Despite this fundamental role, little is known about how SPACA6 specifically functions. We elucidated the crystal structure of the SPACA6 ectodomain at 2.2-Å resolution, revealing a two-domain protein containing a four-helix bundle and Ig-like ß-sandwich connected via a quasi-flexible linker. This structure is reminiscent of IZUMO1, another gamete fusion-associated protein, making SPACA6 and IZUMO1 founding members of a superfamily of fertilization-associated proteins, herein dubbed the IST superfamily. The IST superfamily is defined structurally by its distorted four-helix bundle and a pair of disulfide-bonded CXXC motifs. A structure-based search of the AlphaFold human proteome identified more protein members to this superfamily; remarkably, many of these proteins are linked to gamete fusion. The SPACA6 structure and its connection to other IST-superfamily members provide a missing link in our knowledge of mammalian gamete fusion.


Assuntos
Reação Acrossômica , Proteínas de Membrana , Espermatozoides , Reação Acrossômica/genética , Reação Acrossômica/fisiologia , Animais , Dissulfetos , Células Germinativas/metabolismo , Humanos , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Masculino , Mamíferos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteoma , Sêmen/metabolismo , Espermatozoides/metabolismo
3.
Hum Reprod ; 37(7): 1394-1405, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35551387

RESUMO

STUDY QUESTION: Are there new genetic factors responsible for male infertility with normal sperm quantity and morphology? SUMMARY ANSWER: We identified the bi-allelic variants in KCNU1 and confirmed it a novel pathogenetic gene for male infertility mainly due to impaired sperm acrosome reactions (ARs). WHAT IS KNOWN ALREADY: Until now, the underlying genetic determinants for male affected individuals exhibiting normal sperm quantity and morphology have been largely unknown. Potassium/calcium-activated channel subfamily U member 1 (KCNU1) is a sperm-specific potassium channel. The Kcnu1 null mutation in male mice causes infertility due to the impaired progressive motility and AR. STUDY DESIGN, SIZE, DURATION: We recruited a cohort of 126 male infertility individuals with typical asthenospermia or fertilization failure and focused on two infertile males from two consanguineous families from 2015 to 2020; whole-exome sequencing and homozygosity mapping were performed. We identified a homozygous missense variant (c.2144A>G, p.His715Arg) and a homozygous donor splice-site variant (c.1295 + 3A>C, p.Val405Glyfs*8) in KCNU1. Then, we generated a knock-in (KI) mouse model in September 2020 and have now carried out functional studies and possible treatment strategies. PARTICIPANTS/MATERIALS, SETTING, METHODS: The affected individuals with infertility were recruited from the Shanghai Ninth Hospital affiliated to Shanghai Jiao Tong University. Genomic DNA from the affected individual was extracted from peripheral blood. Whole-exome sequencing, homozygosity mapping and in silico analyses were used to screen and identify KCNU1 variants, and the variants were confirmed by Sanger sequencing. We used C57BL/6N mouse to construct KI mouse model to mimic the reproductive phenotype in vivo. We performed functional experiments by western blotting, AR assay and immunofluorescent Staining. Finally, we performed IVF and ICSI to explore the treatment strategies. MAIN RESULTS AND THE ROLE OF CHANCE: We identified a homozygous missense variant (c.2144A>G, p.His715Arg) and a homozygous donor splice-site variant (c.1295 + 3A>C, p.Val405Glyfs*8) in KCNU1 in two infertile males. We demonstrated that the splice-site variant affected normal alternative splicing of KCNU1, thus leading to the loss of function of KCNU1. Meanwhile, the missense pathogenic variant reduced the KCNU1 protein levels in sperm of both the affected individual and the KI mouse model, resulting in impaired ARs and male infertility. Intracytoplasmic sperm injection was able to rescue the deficiencies. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The exact molecular mechanism of KCNU1 and pathways need to be further explore in the future. WIDER IMPLICATIONS OF THE FINDINGS: This is the first report that establishes a causal relationship between KCNU1 deficiency and male infertility, confirming the critical role of KCNU1 in human reproduction. Our findings expand our knowledge of the genes that play critical roles in the human sperm AR and provide a new genetic marker for infertility. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the SHIPM-pi fund no. JY201801 from the Shanghai Institute of Precision Medicine, Ninth People's Hospital Shanghai Jiao Tong University School of Medicine, the National Natural Science Foundation of China (81725006, 81771649, 81822019, 81771581, 81971450, 81971382, 82001538 and 82071642). The authors declare no conflict of interest. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Reação Acrossômica , Infertilidade Masculina , Canais de Potássio Ativados por Cálcio de Condutância Alta , Reação Acrossômica/genética , Animais , China , Humanos , Infertilidade Masculina/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sêmen , Espermatozoides
4.
Reprod Biol Endocrinol ; 20(1): 5, 2022 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-34980136

RESUMO

BACKGROUND: Potassium channels are important for the structure and function of the spermatozoa. As a potassium transporter, the mSlo3 is essential for male fertility as Slo3 knockout male mice were infertile with the series of functional defects in sperm cells. However, no pathogenic variant has been detected in human SLO3 to date. Here we reported a human case with homozygous SLO3 mutation. The function of SLO3 in human sperm and the corresponding assisted reproductive strategy are also investigated. METHODS: We performed whole-exome sequencing analysis from a large cohort of 105 patients with asthenoteratozoospermia. The effects of the variant were investigated by quantitative RT-PCR, western blotting, and immunofluorescence assays using the patient spermatozoa. Sperm morphological and ultrastructural studies were conducted using haematoxylin and eosin staining, scanning and transmission electron microscopy. RESULTS: We identified a homozygous missense variant (c.1237A > T: p.Ile413Phe) in the sperm-specific SLO3 in one Chinese patient with male infertility. This SLO3 variant was rare in human control populations and predicted to be deleterious by multiple bioinformatic tools. Sperm from the individual harbouring the homozygous SLO3 variant exhibited severe morphological abnormalities, such as acrosome hypoplasia, disruption of the mitochondrial sheath, coiled tails, and motility defects. The levels of SLO3 mRNA and protein in spermatozoa from the affected individual were reduced. Furthermore, the acrosome reaction, mitochondrial membrane potential, and membrane potential during capacitation were also afflicted. The levels of acrosome marker glycoproteins and PLCζ1 as well as the mitochondrial sheath protein HSP60 and SLO3 auxiliary subunit LRRC52, were significantly reduced in the spermatozoa from the affected individual. The affected man was sterile due to acrosome and mitochondrial dysfunction; however, intra-cytoplasmic sperm injection successfully rescued this infertile condition. CONCLUSIONS: SLO3 deficiency seriously impact acrosome formation, mitochondrial sheath assembly, and the function of K+ channels. Our findings provided clinical implications for the genetic and reproductive counselling of affected families.


Assuntos
Acrossomo/patologia , Astenozoospermia/genética , Infertilidade Masculina/genética , Reação Acrossômica/genética , Adulto , Astenozoospermia/patologia , China , Estudos de Coortes , Consanguinidade , Características da Família , Feminino , Homozigoto , Humanos , Infertilidade Masculina/patologia , Infertilidade Masculina/terapia , Canais de Potássio Ativados por Cálcio de Condutância Alta , Masculino , Potencial da Membrana Mitocondrial/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Membranas Mitocondriais/patologia , Mutação de Sentido Incorreto , Linhagem , Gravidez , Injeções de Esperma Intracitoplásmicas , Espermatozoides/anormalidades , Espermatozoides/patologia
5.
Mol Reprod Dev ; 88(7): 479-481, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34114279

RESUMO

Many factors are involved in acrosome biogenesis in order for appropriate acrosome formation to occur. Here, we demonstrate that IZUMO family member 3, IZUMO3, plays an important role in acrosome biogenesis, as proven by gene disruption experiments. A loss of IZUMO3 in round spermatids affects acrosomal granule positioning due to lack of acrosomal granule contact with the inner acrosomal membrane, leading to the formation of grossly malformed spermatozoa associated with male subfertility. Thus, we suggest that mammalian spermiogenesis needs an elaborate acrosome biogenesis through IZUMO3 involvement.


Assuntos
Acrossomo/fisiologia , Fertilidade/genética , Proteínas de Membrana/fisiologia , Reação Acrossômica/genética , Animais , Infertilidade Masculina/genética , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Espermatogênese/genética , Espermatozoides/anormalidades , Espermatozoides/fisiologia
6.
Curr Mol Med ; 21(10): 850-859, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33475072

RESUMO

Male infertility is rising nowadays and accounts for a major part of infertility cases worldwide. Novel tests are being developed for better detection and management of male infertility. Though there are many tests available for diagnosing male infertility like acrosome reaction rate, hemizona assay, in vivo or in vitro sperm penetration assay, sperm DNA damage tests, however, a semen analysis is the most commonly used initial test for male infertility. It is usually associated with failure to detect the cause in many cases, as seminal composition gets affected by a number of factors and can give false reports. Furthermore, it does not give any information about defects in capacitation, sperm-zona pellucida interaction, and sperm's ability to fertilize oocytes. This results in failure of detection and delayed management of male infertility. Hence, the present review was conducted to identify various sperm proteins that play a significant role in spermatogenesis, sperm motility, sperm-zona pellucida interaction, and fertilization. These proteins can be used in the future as markers of male infertility and will aid in better detection and management of male infertility. Methodology: Search for literature was made from 1970 to 2020 from various databases like PUBMED, SCOPUS, Google Scholar on sperm proteins and their role in male fertility using keywords: "sperm protein as bio-markers", "novel sperm proteins as markers of infertility", "Sperm proteins essential for capacitation, sperm motility and oocyte fertilization". Inclusion criteria: All full-length research articles, systematic reviews, meta-analyses, or abstracts on sperm proteins and male infertility published in the English language in peer-reviewed journals were considered.


Assuntos
Infertilidade Masculina , Motilidade dos Espermatozoides/genética , Espermatogênese/genética , Reação Acrossômica/genética , Humanos , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Zona Pelúcida/metabolismo
7.
J Clin Endocrinol Metab ; 106(4): e1775-e1792, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33340048

RESUMO

CONTEXT: The calcium-sensing receptor (CaSR) is essential to maintain a stable calcium concentration in serum. Spermatozoa are exposed to immense changes in concentrations of CaSR ligands such as calcium, magnesium, and spermine during epididymal maturation, in the ejaculate, and in the female reproductive environment. However, the role of CaSR in human spermatozoa is unknown. OBJECTIVE: This work aimed to investigate the role of CaSR in human spermatozoa. METHODS: We identified CaSR in human spermatozoa and characterized the response to CaSR agonists on intracellular calcium, acrosome reaction, and 3',5'-cyclic adenosine 5'-monophosphate (cAMP) in spermatozoa from men with either loss-of-function or gain-of-function mutations in CASR and healthy donors. RESULTS: CaSR is expressed in human spermatozoa and is essential for sensing extracellular free ionized calcium (Ca2+) and Mg2+. Activators of CaSR augmented the effect of sperm-activating signals such as the response to HCO3- and the acrosome reaction, whereas spermatozoa from men with a loss-of-function mutation in CASR had a diminished response to HCO3-, lower progesterone-mediated calcium influx, and were less likely to undergo the acrosome reaction in response to progesterone or Ca2+. CaSR activation increased cAMP through soluble adenylyl cyclase (sAC) activity and increased calcium influx through CatSper. Moreover, external Ca2+ or Mg2+ was indispensable for HCO3- activation of sAC. Two male patients with a CASR loss-of-function mutation in exon 3 presented with normal sperm counts and motility, whereas a patient with a loss-of-function mutation in exon 7 had low sperm count, motility, and morphology. CONCLUSION: CaSR is important for the sensing of Ca2+, Mg2+, and HCO3- in spermatozoa, and loss-of-function may impair male sperm function.


Assuntos
Bicarbonatos/metabolismo , Cálcio/metabolismo , Receptores de Detecção de Cálcio/fisiologia , Espermatozoides/metabolismo , Reação Acrossômica/efeitos dos fármacos , Reação Acrossômica/genética , Adulto , Bicarbonatos/farmacologia , Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Estudos de Casos e Controles , Feminino , Humanos , Hipercalcemia/congênito , Hipercalcemia/genética , Hipercalcemia/metabolismo , Hipercalcemia/patologia , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipercalciúria/patologia , Hipocalcemia/genética , Hipocalcemia/metabolismo , Hipocalcemia/patologia , Hipoparatireoidismo/congênito , Hipoparatireoidismo/genética , Hipoparatireoidismo/metabolismo , Hipoparatireoidismo/patologia , Rim/metabolismo , Rim/patologia , Magnésio/metabolismo , Magnésio/farmacologia , Masculino , Mutação , Receptores de Detecção de Cálcio/genética , Motilidade dos Espermatozoides/efeitos dos fármacos , Motilidade dos Espermatozoides/genética , Espermatozoides/efeitos dos fármacos , Espermatozoides/fisiologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia
8.
Methods Mol Biol ; 2233: 139-168, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33222133

RESUMO

Acrosome reaction is an exocytic process that enables a sperm to penetrate the zona pellucida and fertilize an egg. The process involves the fenestration and vesiculation of the sperm plasma membrane and outer acrosomal membrane, releasing the acrosomal content. Given the importance of the acrosome secretion in fertilization, many different methods have been developed to detect the acrosome reaction of sperm. In this chapter, we describe detailed practical procedures to assess the acrosomal status of human spermatozoa. To do this, we resorted to light optical and epifluorescence microscopy, flow cytometry, and transmission electron microscopy. We also itemize the protocol for real-time measurements of the acrosome reaction by confocal microscopy. Further, we discuss the level of complexity, costs, and the reasons why a researcher should choose each technique.This chapter is designed to provide the user with sufficient background to measure acrosomal exocytosis in human sperm.


Assuntos
Reação Acrossômica/genética , Membrana Celular/ultraestrutura , Exocitose/genética , Espermatozoides/ultraestrutura , Acrossomo/metabolismo , Membrana Celular/genética , Humanos , Masculino , Espermatozoides/patologia , Zona Pelúcida/metabolismo
9.
FASEB J ; 34(12): 16224-16242, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33058343

RESUMO

Importin α proteins play a central role in the transport of cargo from the cytoplasm to the nucleus. In this study, we observed that male knock-out mice for importin α4, which is encoded by the Kpna4 gene (Kpna4-/- ), were subfertile and yielded smaller litter sizes than those of wild-type (WT) males. In contrast, mice lacking the closely related importin α3 (Kpna3-/- ) were fertile. In vitro fertilization and sperm motility assays demonstrated that sperm from Kpna4-/- mice had significantly reduced quality and motility. In addition, acrosome reaction was also impaired in Kpna4-/- mice. Transmission electron microscopy revealed striking defects, including abnormal head morphology and multiple axoneme structures in the flagella of Kpna4-/- mice. A five-fold increase in the frequency of abnormalities in Kpna4-/- mice compared to WT mice indicates the functional importance of importin α4 in normal sperm development. Moreover, Nesprin-2, which is a component of the linker of nucleus and cytoskeleton complex, was expressed at lower levels in sperm from Kpna4-/- mice and was localized with abnormal axonemes, suggesting incorrect formation of the nuclear membrane-cytoskeleton structure during spermiogenesis. Proteomics analysis of Kpna4-/- testis showed significantly altered expression of proteins related to sperm formation, which provided evidence that genetic loss of importin α4 perturbed chromatin status. Collectively, these findings indicate that importin α4 is critical for establishing normal sperm morphology in mice, providing new insights into male germ cell development by highlighting the requirement of importin α4 for normal fertility.


Assuntos
Fertilidade/genética , Infertilidade Masculina/genética , Carioferinas/genética , Motilidade dos Espermatozoides/genética , Espermatozoides/anormalidades , alfa Carioferinas/genética , Reação Acrossômica/genética , Animais , Flagelos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espermatogênese/genética , Testículo/anormalidades
10.
Proc Natl Acad Sci U S A ; 117(21): 11493-11502, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32393636

RESUMO

Sperm-oocyte membrane fusion is one of the most important events for fertilization. So far, IZUMO1 and Fertilization Influencing Membrane Protein (FIMP) on the sperm membrane and CD9 and JUNO (IZUMO1R/FOLR4) on the oocyte membrane have been identified as fusion-required proteins. However, the molecular mechanisms for sperm-oocyte fusion are still unclear. Here, we show that testis-enriched genes, sperm-oocyte fusion required 1 (Sof1/Llcfc1/1700034O15Rik), transmembrane protein 95 (Tmem95), and sperm acrosome associated 6 (Spaca6), encode sperm proteins required for sperm-oocyte fusion in mice. These knockout (KO) spermatozoa carry IZUMO1 but cannot fuse with the oocyte plasma membrane, leading to male sterility. Transgenic mice which expressed mouse Sof1, Tmem95, and Spaca6 rescued the sterility of Sof1, Tmem95, and Spaca6 KO males, respectively. SOF1 and SPACA6 remain in acrosome-reacted spermatozoa, and SPACA6 translocates to the equatorial segment of these spermatozoa. The coexpression of SOF1, TMEM95, and SPACA6 in IZUMO1-expressing cultured cells did not enhance their ability to adhere to the oocyte membrane or allow them to fuse with oocytes. SOF1, TMEM95, and SPACA6 may function cooperatively with IZUMO1 and/or unknown fusogens in sperm-oocyte fusion.


Assuntos
Reação Acrossômica , Proteínas de Membrana , Proteínas de Plasma Seminal , Espermatozoides/fisiologia , Reação Acrossômica/genética , Reação Acrossômica/fisiologia , Animais , Feminino , Infertilidade Masculina/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas de Plasma Seminal/genética , Proteínas de Plasma Seminal/metabolismo
11.
Mol Reprod Dev ; 87(5): 534-541, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32311190

RESUMO

Haprin (TRIM36) is a ubiquitin-protein ligase that mediates ubiquitination and subsequent proteasomal degradation of target proteins. It is expressed in the testes in both mice and humans and is thought to be involved in spermiogenesis, the acrosome reaction, and fertilization. However, the functional role of Haprin is poorly understood. The aim of this study was to investigate the physiological role of Haprin in fertility. Homozygous haprin-deficient mice were generated and these mice, and their spermatozoa, were analyzed to detect morphological and fertility-related abnormalities. In these models, normal spermatogenesis was observed but sperm quality was reduced with haprin-deficient mice having poorer sperm morphology and motility than wild-type mice. Interestingly, haprin-deficient mice showed normal in vivo fertility but could not fertilize oocytes under standard in vitro fertilization conditions. In conclusion, this study demonstrated that Haprin deficiency causes morphological abnormalities in spermatozoa, indicating that Haprin is involved in spermiogenesis.


Assuntos
Proteínas de Transporte/genética , Infertilidade Masculina/genética , Proteínas de Plasma Seminal/genética , Espermatozoides/fisiologia , Reação Acrossômica/genética , Animais , Proteínas de Transporte/metabolismo , Feminino , Fertilização/genética , Fertilização in vitro , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas de Plasma Seminal/metabolismo , Espermatogênese/genética , Espermatozoides/metabolismo
12.
Biochim Biophys Acta Mol Cell Res ; 1867(7): 118704, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32194132

RESUMO

Exocytosis of spermatozoon's secretory vesicle, named acrosome reaction (AR), is a regulated event that plays a central role in fertilization. It is coupled to a complex calcium signaling. Ceramide is a multitasking lipid involved in exocytosis. Nevertheless, its effect on secretion is controversial and the underlying cellular and molecular mechanisms remain unknown. Human spermatozoa are useful to dissect the role of ceramide in secretion given that the gamete is not capable to undergo any trafficking mechanisms other than exocytosis. We report for the first time, the presence of sphingolipid metabolism enzymes such as neutral-sphingomyelinase and ceramide synthase in sperm. Ceramidases are also present and active. Both the addition of cell-permeable ceramide and the rise of the endogenous one, increase intracellular calcium acting as potent inducers of exocytosis. Ceramide triggers AR in capacitated spermatozoa and enhances the gamete response to progesterone. The lipid induces physiological ultrastructural changes in the acrosome and triggers an exocytosis-signaling cascade involving protein tyrosine phosphatase 1B and VAMP2. Real-time imaging showed an increment of calcium in the cytosol upon ceramide treatment either in the absence or in the presence of extracellular calcium. Pharmacological experiments demonstrate that at early stages the process involves ryanodine receptors, CatSper (calcium channel of sperm), and store-operated calcium channels. We set out the signaling sequence of events that connect ceramide to internal calcium mobilization and external calcium signals during secretion. These results allow the coordination of lipids and proteins in a pathway that accomplishes secretion. Our findings contribute to the understanding of ceramide's role in regulated exocytosis and fertilization.


Assuntos
Reação Acrossômica/genética , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Espermatozoides/efeitos dos fármacos , Proteína 2 Associada à Membrana da Vesícula/genética , Acrossomo/efeitos dos fármacos , Acrossomo/metabolismo , Reação Acrossômica/efeitos dos fármacos , Adulto , Cálcio/química , Canais de Cálcio/genética , Sinalização do Cálcio/efeitos dos fármacos , Ceramidas/farmacologia , Citoplasma/efeitos dos fármacos , Citoplasma/genética , Exocitose/genética , Fertilização/genética , Humanos , Masculino , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/genética , Espermatozoides/patologia
13.
J Cell Physiol ; 235(6): 5340-5352, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31903553

RESUMO

Calmodulin is a small, highly conserved acidic protein present at high levels in spermatozoa that mediates numerous intracellular Ca2+ -dependent events. Sperm motility and fertilizing ability results from an array of biochemical pathways under Ca2+ control, in which the importance of calmodulin is not fully understood. The role of calmodulin in sperm function has been mostly assessed using antagonists. Nevertheless, few known calmodulin-regulated enzymes have been described in spermatozoa regarding their involvement in sperm function. To further understand the role of this important Ca2+ mediator in spermatozoa, different studies were also undertaken to investigate and to identify sperm calmodulin-binding proteins and determine their localization and subcellular distribution as an attempt to elucidate the role of this important Ca2+ mediator. In the present study, sperm calmodulin-binding proteins were identified by mass spectrometry after Ca2+ -dependent biotinylated-calmodulin binding on sperm head proteins subjected to 2D electrophoresis and transferred on a polyvinylidene difluoride membrane. Calmodulin binding protein identification was also done on detergent extracted whole sperm proteins pulled down in a Ca2+ -dependent manner by calmodulin-conjugated sepharose beads. In this latter group, 300 proteins were identified in at least two experiments out of three, and those identified in the three independent experiments were analyzed for overrepresented biological processes using the Bos taurus Gene Ontology database. Proteins with known function in reproductive processes, fertilization, sperm-egg recognition, sperm binding to the zona pellucida, regulation of sperm capacitation, and sperm motility were identified and further emphasize the importance of calmodulin in sperm function.


Assuntos
Cálcio/metabolismo , Proteínas de Ligação a Calmodulina/genética , Calmodulina/genética , Espermatozoides/crescimento & desenvolvimento , Reação Acrossômica/genética , Animais , Bovinos , Fertilização/genética , Humanos , Masculino , Ligação Proteica/genética , Capacitação Espermática , Motilidade dos Espermatozoides/genética , Espermatozoides/fisiologia , Zona Pelúcida/metabolismo
14.
Mol Hum Reprod ; 25(8): 458-470, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31194867

RESUMO

Human spermatozoa can fertilize an oocyte only after post-testicular maturation and capacitation. These processes involve dynamic modification and reorganization of the sperm plasma membrane, which allow them to bind to the zona pellucida (ZP) of the oocyte. Defective sperm-ZP binding is one of the major causes of male subfertility. Galectin-3 is a secretory lectin in human seminal plasma well known for its action on cell adhesion. The aim of this study was to determine the role of galectin-3 in spermatozoa-ZP interaction and its association with fertilization rate in clinical assisted reproduction. Our studies revealed that the acrosomal region of ejaculated and capacitated spermatozoa possess strong galectin-3 immunoreactivity, which is much stronger than that of epididymal spermatozoa. Expression of galectin-3 can also be detected on seminal plasma-derived extracellular vesicles (EVs) and can be transferred to the sperm surface. Blocking of sperm surface galectin-3 function by antibody or carbohydrate substrate reduced the ZP-binding capacity of spermatozoa. Purified galectin-3 is capable of binding to ZP, indicating that galectin-3 may serve as a cross-linking bridge between ZP glycans and sperm surface glycoproteins. Galectin-3 levels in seminal plasma-derived EVs were positively associated with fertilization rates. These results suggest that galectin-3 in EVs is transferred to the sperm surface during post-testicular maturation and plays a crucial role in spermatozoa-ZP binding after capacitation. Reduced galectin-3 expression in seminal plasma-derived EVs may be a cause behind a low fertilization rate. Further studies with more clinical samples are required to confirm the relationship between galectin-3 levels and IVF outcomes.


Assuntos
Fertilização/fisiologia , Galectina 3/metabolismo , Zona Pelúcida/metabolismo , Reação Acrossômica/genética , Reação Acrossômica/fisiologia , Adesão Celular/fisiologia , Fertilização/genética , Galectina 3/genética , Humanos , Masculino , Oócitos/metabolismo , Sêmen/metabolismo , Capacitação Espermática/fisiologia , Interações Espermatozoide-Óvulo/genética , Interações Espermatozoide-Óvulo/fisiologia , Espermatozoides/metabolismo
15.
J Cell Mol Med ; 23(4): 2583-2594, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30680919

RESUMO

Oligoasthenozoospermia is a major cause of male infertility; however, its etiology and pathogenesis are unclear and may be associated with specific gene abnormalities. This study focused on Tppp2 (tubulin polymerization promoting protein family member 2), whose encoded protein localizes in elongating spermatids at stages IV-VIII of the seminiferous epithelial cycle in testis and in mature sperm in the epididymis. In human and mouse sperm, in vitro inhibition of TPPP2 caused significantly decreased motility and ATP content. Studies on Tppp2 knockout (KO) mice demonstrated that deletion of TPPP2 resulted in male subfertility with a significantly decreased sperm count and motility. In Tppp2-/- mice, increased irregular mitochondria lacking lamellar cristae, abnormal expression of electron transfer chain molecules, lower ATP levels, decreased mitochondrial membrane potential and increased apoptotic index were observed in sperm, which could be the potential causes for its oligoasthenozoospermia phenotype. Moreover, we identified a potential TPPP2-interactive protein, eEf1b (eukaryotic translation elongation factor 1 beta), which plays an important role in protein translation extension. Thus, TPPP2 is probably a potential pathogenic factor in oligoasthenozoospermia. Deficiency of TPPP2 might affect the translation of specific proteins, altering the structure and function of sperm mitochondria, and resulting in decreased sperm count, motility and fertility.


Assuntos
Trifosfato de Adenosina/deficiência , Mitocôndrias/metabolismo , Proteínas do Tecido Nervoso/genética , Oligospermia/genética , Fatores de Alongamento de Peptídeos/genética , Espermatozoides/metabolismo , Reação Acrossômica/genética , Animais , Epididimo/metabolismo , Epididimo/patologia , Feminino , Expressão Gênica , Humanos , Tamanho da Ninhada de Vivíparos , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/patologia , Proteínas do Tecido Nervoso/deficiência , Oligospermia/metabolismo , Oligospermia/patologia , Fatores de Alongamento de Peptídeos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Capacitação Espermática/genética , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Espermatozoides/patologia , Testículo/metabolismo , Testículo/patologia
16.
J Reprod Dev ; 65(2): 97-102, 2019 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-30606959

RESUMO

Gene-knockout mice lacking ACRBP, a proacrosin-binding protein localized in the acrosome of sperm, have been shown to exhibit male subfertility, owing to abnormal formation of the acrosome. In this study, to elucidate the mechanism contributing to the subfertility phenotype, we examined the behavior of ACRBP-deficient mouse sperm in the female reproductive tract. When sperm that had migrated into the uterus and oviduct after mating were counted, the number of ACRBP-deficient sperm was noticeably smaller in the oviduct of mice post mating. However, ACRBP-deficient sperm recovered from the oviduct possessed morphologically normal head shape and retained normal motility. Importantly, ACRBP-deficient sperm displayed a marked reduction in the ability to successfully gain access to unfertilized oocytes. These data suggest that male subfertility of ACRBP-deficient mice may be attributed to incompleteness of the acrosome reaction rather than impairment in sperm migration from the uterus to the oviduct.


Assuntos
Reação Acrossômica/genética , Proteínas de Transporte/genética , Genitália Feminina/metabolismo , Interações Espermatozoide-Óvulo/genética , Espermatozoides/metabolismo , Acrossomo/metabolismo , Animais , Proteínas de Transporte/metabolismo , Tubas Uterinas/metabolismo , Feminino , Fertilização/genética , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Especificidade de Órgãos/genética , Análise do Sêmen , Motilidade dos Espermatozoides/genética
17.
J Reprod Dev ; 65(2): 147-153, 2019 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-30662011

RESUMO

Previously, we reported that neurotensin (NT), which is expressed in the uterus and oviduct, enhanced bovine sperm capacitation and acrosome reactions. As NT mRNA expression in bovine oviducts increases dramatically in the follicular phase, we hypothesized that NT modulates fertilization and subsequent conception in cattle. The objective of this study was to evaluate the effect of NT on embryo development and blastocyst quality. The rate of embryo cleavage was significantly increased by the addition of NT to the fertilization medium. Furthermore, the total number of cells and numbers of cells in the inner cell mass of blastocysts were significantly increased by NT during in vitro fertilization (IVF). These results suggested that NT enhanced the efficiency of early bovine embryo development and blastocyst quality. The expression of NT receptors (NTRs) in sperm, testes, oocytes, and cumulus cells was evaluated to determine whether NT acted via NTRs in sperm alone or in both male and female reproductive cells during IVF. Immunocytochemistry and reverse transcription polymerase chain reaction revealed that NTR1 and NTR2 were expressed in sperm and testes, but not in oocytes and cumulus cells. We propose that NT selectively acts upon sperm via NTR1 and NTR2 during IVF to improve the cleavage rate and quality of blastocysts, which are important determinants of sperm quality for successful conception. This research supports our hypothesis that NT acts as a key modulator of fertilization and conception in cattle. Further studies are necessary to apply our findings to the industrial framework of bovine reproduction.


Assuntos
Blastocisto/citologia , Blastocisto/efeitos dos fármacos , Fertilização in vitro , Neurotensina/farmacologia , Receptores de Neurotensina/fisiologia , Espermatozoides/efeitos dos fármacos , Reação Acrossômica/efeitos dos fármacos , Reação Acrossômica/genética , Animais , Blastocisto/fisiologia , Bovinos/embriologia , Células Cultivadas , Técnicas de Cultura Embrionária/métodos , Técnicas de Cultura Embrionária/veterinária , Embrião de Mamíferos , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Feminino , Fertilização/efeitos dos fármacos , Fertilização/genética , Fertilização in vitro/veterinária , Masculino , Neurotensina/metabolismo , Neurotensina/fisiologia , Receptores de Neurotensina/genética , Receptores de Neurotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Capacitação Espermática/efeitos dos fármacos , Capacitação Espermática/genética , Espermatozoides/fisiologia
18.
PLoS One ; 13(11): e0206224, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30412589

RESUMO

Calcium signalling is critical for successful fertilization. In spermatozoa, capacitation, hyperactivation of motility and acrosome reactions are all mediated by increases in intracellular Ca2+. Our previous reports have shown that deficiency of MTMR14, a novel phosphoinositide phosphatase, induces a muscle disorder by disrupting Ca2+ homeostasis. Recently, we found that MTMR14 is also expressed in the testes; however, whether deficiency of MTMR14 in the testes also alters the Ca2+ concentration and impairs male fertility remains entirely unknown. In the present study, we found that MTMR14 is also expressed in the testes and mature sperm cells, suggesting that deficiency of MTMR14 might have some effect on male fertility. Both in vivo fertility and in vitro fertilization tests were then performed, and we found that MTMR14-/- male mice showed decreased fertility. A series of experiments were then arranged to test the testis and sperm parameters; we found that MTMR14 deficiency caused small size of the testes, small numbers of both total and immotile sperm, expanded membrane of sperm tail, a decreased proportion of acrosome reaction, and in contrast, an increased proportion of abnormal sperm and augmented apoptosis, etc. Further study also found that the muscle force of the vas deferens decreased significantly in KO mice. Intracellular calcium homeostasis in the testes and epididymis was impaired by MTMR14 deletion; moreover, the relative mRNA expression levels of Itpr1, Itpr2, and Ryr3 were dramatically decreased in MTMR14 KO mice. Thus, MTMR14 deletion impairs male fertility by causing decreased muscle force of the vas deferens and intracellular calcium imbalance.


Assuntos
Sinalização do Cálcio/genética , Fertilidade/genética , Monoéster Fosfórico Hidrolases/genética , Espermatozoides/metabolismo , Reação Acrossômica/genética , Animais , Epididimo/crescimento & desenvolvimento , Epididimo/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Masculino , Camundongos , Camundongos Knockout , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Capacitação Espermática/genética , Espermatozoides/patologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
19.
Biol Reprod ; 99(4): 817-827, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29733332

RESUMO

Male infertility has become an increasingly common health concern in recent years. Apart from environmental factors, nutrition, lifestyle, and sexually transmitted diseases, genetic defects are important causes of male infertility. Many genes have been demonstrated to be associated with male infertility. However, the roles of some functional genes in infertility, especially those that are specifically expressed in the reproductive system, remain to be elucidated. Here, we demonstrated that the testis-specific gene coiled-coil domain-containing 87 (Ccdc87) is critical for male fertility. Reverse-transcriptase polymerase chain reaction and western blot analyses revealed that the Ccdc87 mRNA and protein were only expressed in mouse testis. Ccdc87 expression first appeared at postnatal day 14 and remained at a relatively high level until adulthood. Male mice lacking Ccdc87 gene (Ccdc87-/-) were found to be subfertile. Approximately 20% of Ccdc87-null sperm from the testis and epididymis displayed severe abnormity of acrosome and cell nucleus. Sperm isolated from the cauda epididymides of Ccdc87-/- mice exhibited decreased initial motility but did not show any change in capacitation. Additionally, Ccdc87 disruption led to the impotency of sperm spontaneous and progesterone-induced acrosome reaction. Moreover, in vitro fertilization assays indicated that the fertilizing capacity of Ccdc87-/- sperm was significantly reduced. Taken together, these findings provide a new clue to understand the genetic causes of male infertility.


Assuntos
Fertilidade/fisiologia , Proteínas/genética , Proteínas/fisiologia , Espermatozoides/fisiologia , Reação Acrossômica/genética , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Feminino , Fertilidade/genética , Expressão Gênica , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Infertilidade Masculina/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Motilidade dos Espermatozoides/genética , Interações Espermatozoide-Óvulo/genética , Espermatozoides/anormalidades , Espermatozoides/ultraestrutura , Testículo/metabolismo
20.
Biol Reprod ; 99(2): 373-383, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29481619

RESUMO

Epididymal sperm protein CRISP1 has the ability to both regulate murine CatSper, a key sperm calcium channel, and interact with egg-binding sites during fertilization. In spite of its relevance for sperm function, Crisp1-/-mice are fertile. Considering that phenotypes can be influenced by the genetic background, in the present work mice from the original mixed Crisp1-/- colony (129/SvEv*C57BL/6) were backcrossed onto the C57BL/6 strain for subsequent analysis of their reproductive phenotype. Whereas fertility and fertilization rates of C57BL/6 Crisp1-/- males did not differ from those reported for mice from the mixed background, several sperm functional parameters were clearly affected by the genetic background. Crisp1-/- sperm from the homogeneous background exhibited defects in both the progesterone-induced acrosome reaction and motility not observed in the mixed background, and normal rather than reduced protein tyrosine phosphorylation. Additional studies revealed a significant decrease in sperm hyperactivation as well as in cAMP and protein kinase A (PKA) substrate phosphorylation levels in sperm from both colonies. The finding that exposure of mutant sperm to a cAMP analog and phosphodiesterase inhibitor overcame the sperm functional defects observed in each colony indicated that a common cAMP-PKA signaling defect led to different phenotypes depending on the genetic background. Altogether, our observations indicate that the phenotype of CRISP1 null males is modulated by the genetic context and reveal new roles for the protein in both the functional events and signaling pathways associated to capacitation.


Assuntos
Fertilidade/genética , Fertilização/genética , Glicoproteínas de Membrana/genética , Reprodução/genética , Espermatozoides/metabolismo , Reação Acrossômica/efeitos dos fármacos , Reação Acrossômica/genética , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Patrimônio Genético , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Progesterona/farmacologia , Motilidade dos Espermatozoides/genética , Espermatozoides/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA