Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Metallomics ; 15(10)2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37740571

RESUMO

Currently, clinical analysis of male infertility mainly relies on parameters of semen and sperm cells. However, the high diagnostic failure rates indicate that the current assessment methods are not sufficient and a new approach to evaluating sperm function still needs to be developed. Here we explored the feasibility of single-cell inductively coupled plasma mass spectrometry (sc-ICP-MS)-derived profiles to determine the elemental characteristics in viable capacitated sperm under normal and deficient conditions. To validate the measurements, we used male sterile Pmca4-knockout (KO) mice with impaired calcium clearance, known to be dysregulated due to loss of calcium efflux capacity during sperm capacitation. Consistently, we observed significantly increased calcium intensities in Pmca4-KO sperm upon capacitation stimulation compared with control sperm from the caudaepididymides of wild-type control (WT) mice. More importantly, we explored that the characteristic signatures of calcium intensities in individual spikes derived from sc-ICP-MS was consistent with the dynamics of relative calcium levels in single sperm reported in the literature. Prominent alterations were also observed in the dynamic signatures of sc-ICP-MS-derived profiles of essential elements, particularly the redox-labile elements including copper, iron, manganese, selenium, and zinc in Pmca4-KO sperm compared to WT controls. Therefore, our study demonstrates that elementomics of sc-ICP-MS-derived signals can reveal ionic dysregulation in plasma membrane Ca2+-ATPase isoform 4 protein deficient sperm, and that sc-ICP-MS assay can be applied for functional analysis of viable sperm in functional activities, such as capacitation stimulation. We propose that cell elementomics can be used as an alternative approach to assessing sperm quality and male fertility at the single-cell level.

2.
Asian J Androl ; 22(6): 583-589, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32859869

RESUMO

Prohibitin (PHB), an evolutionarily conserved mitochondrial inner membrane protein, is highly expressed in cells that require strong mitochondrial function. Recently, we demonstrated that the deletion of Phb in spermatocytes results in impaired mitochondrial function. In addition, PHB expression in the mitochondrial sheath of human sperm has a significantly negative correlation with mitochondrial reactive oxygen species levels, but a positive one with mitochondrial membrane potential and sperm motility. These results suggest that mitochondrial PHB expression plays a role in sperm motility. However, the mechanism of PHB-mediated regulation of sperm motility remains unknown. Here, we demonstrate for the first time that PHB interacts with protein kinase B (AKT) and exists in a complex with phospho-PHB (pT258) and phospho-AKT in the mitochondrial sheath of murine sperm, as determined using colocalization and coimmunoprecipitation assays. After blocking AKT activity using wortmannin (a phosphatidylinositol 3-kinase [PI3K] inhibitor), murine sperm have significantly ( P < 0.05) decreased levels of phospho-PHB (pT258) and the total and progressive motility. Furthermore, significantly ( P < 0.05) lower levels of phospho-PI3K P85 subunit α+γ (pY199 and pY467) and phospho-AKT (pS473; pT308) are found in sperm from infertile asthenospermic and oligoasthenospermic men compared with normospermic subjects, which suggest a reduced activity of the PI3K/AKT pathway in these infertile subjects. Importantly, these sperm from infertile subjects also have a significantly ( P < 0.05) lower level of phospho-PHB (pT258). Collectively, our findings suggest that the interaction of PHB with AKT in the mitochondrial sheath is critical for sperm motility, where PHB phosphorylation (pT258) level and PI3K/AKT activity are key regulatory factors.


Assuntos
Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Repressoras/metabolismo , Motilidade dos Espermatozoides , Adulto , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Imunofluorescência , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/fisiologia , Proibitinas , Proteínas Repressoras/fisiologia , Espermatozoides/metabolismo , Espermatozoides/fisiologia
3.
Nucleic Acids Res ; 48(9): 4780-4796, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32232334

RESUMO

Previously, we have shown that human sperm Prohibitin (PHB) expression is significantly negatively correlated with mitochondrial ROS levels but positively correlated with mitochondrial membrane potential and motility. However, the possible role of PHB in mammalian spermatogenesis has not been investigated. Here we document the presence of PHB in spermatocytes and its functional roles in meiosis by generating the first male germ cell-specific Phb-cKO mouse. Loss of PHB in spermatocytes resulted in complete male infertility, associated with not only meiotic pachytene arrest with accompanying apoptosis, but also apoptosis resulting from mitochondrial morphology and function impairment. Our mechanistic studies show that PHB in spermatocytes regulates the expression of STAG3, a key component of the meiotic cohesin complex, via a non-canonical JAK/STAT pathway, and consequently promotes meiotic DSB repair and homologous recombination. Furthermore, the PHB/JAK2 axis was found as a novel mechanism in the maintenance of stabilization of meiotic STAG3 cohesin complex and the modulation of heterochromatin formation in spermatocytes during meiosis. The observed JAK2-mediated epigenetic changes in histone modifications, reflected in a reduction of histone 3 tyrosine 41 phosphorylation (H3Y41ph) and a retention of H3K9me3 at the Stag3 locus, could be responsible for Stag3 dysregulation in spermatocytes with the loss of PHB.


Assuntos
Código das Histonas , Meiose/genética , Proteínas Repressoras/fisiologia , Espermatócitos/metabolismo , Espermatogênese/genética , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Pareamento Cromossômico , Epigenoma , Histonas/metabolismo , Recombinação Homóloga , Infertilidade/genética , Janus Quinase 2/metabolismo , Janus Quinases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/fisiologia , Mitocôndrias/ultraestrutura , Estágio Paquíteno , Fosforilação , Proibitinas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Espermatócitos/enzimologia , Espermatócitos/ultraestrutura , Testículo/metabolismo
4.
J Cell Physiol ; 234(8): 12745-12756, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30536802

RESUMO

Extracellular vesicles (EVs) were isolated by ultracentrifugation of vaginal luminal fluid (VLF) from superovulated mice and identified for the first time using transmission electron microscopy. Characterized by size and biochemical markers (CD9 and HSC70), EVs were shown to be both microvesicular and exosomal and were dubbed as "Vaginosomes" (VGS). Vaginal cross-sections were analyzed to visualize EVs in situ: EVs were present in the lumen and also embedded between squamous epithelial and keratinized cells, consistent with their endogenous origin. Western blots detected Plasma membrane Ca2+ -ATPase 1 (PMCA1) and tyrosine-phosphorylated proteins in the VGS cargo and also in uterosomes. Flow cytometry revealed that following coincubation of caudal sperm and VLF for 30 min, the frequencies of cells with the highest Sperm adhesion molecule 1 (SPAM1), PMCA1/4, and PMCA1 levels increased 16.4-, 8.2-, and 27-fold, respectively; compared with control coincubated in phosphate buffered saline (PBS). Under identical conditions, sperm tyrosine-phosphorylated proteins were elevated ~3.3-fold, after VLF coincubation. Progesterone-induced acrosome reaction (AR) rates were significantly (p < 0.001) elevated in sperm coincubated with VGS for 10-30 min, compared with PBS. Sperm artificially deposited in the vaginas of superovulated females for these periods also showed significant (p < 0.01) increases in AR rates, compared with PBS. Thus in vitro and in vivo, sperm acquire from the vaginal environment factors that induce capacitation, explaining recent findings for their acrosomal status in the isthmus. Overall, VGS appear to deliver higher levels of proteins involved in preventing premature capacitation and AR than those promoting them. Our findings which have implications for humans open the possibility of new approaches to infertility treatment with exosome therapeutics.


Assuntos
Membrana Celular/fisiologia , Vesículas Extracelulares/fisiologia , Fertilidade/fisiologia , Capacitação Espermática/fisiologia , Espermatozoides/fisiologia , Vagina/fisiologia , Acrossomo/metabolismo , Acrossomo/fisiologia , Animais , Membrana Celular/metabolismo , Exossomos/metabolismo , Exossomos/fisiologia , Vesículas Extracelulares/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Progesterona/metabolismo , Espermatozoides/metabolismo , Vagina/metabolismo
5.
Sci Rep ; 8(1): 16094, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30382141

RESUMO

Oviductosomes (OVS) are nano-sized extracellular vesicles secreted in the oviductal luminal fluid by oviductal epithelial cells and known to be involved in sperm capacitation and fertility. Although they have been shown to transfer encapsulated proteins to sperm, cargo constituents other than proteins have not been identified. Using next-generation sequencing, we demonstrate that OVS are carriers of microRNAs (miRNAs), with 272 detected throughout the estrous cycle. Of the 50 most abundant, 6 (12%) and 2 (4%) were expressed at significantly higher levels (P < 0.05) at metestrus/diestrus and proestrus/estrus. RT-qPCR showed that selected miRNAs are present in oviductal epithelial cells in significantly (P < 0.05) lower abundance than in OVS, indicating selective miRNA packaging. The majority (64%) of the top 25 OVS miRNAs are present in sperm. These miRNAs' potential target list is enriched with transcription factors, transcription regulators, and protein kinases and there are several embryonic developmentally-related genes. Importantly, OVS can deliver to sperm miRNAs, including miR-34c-5p which is essential for the first cleavage and is solely sperm-derived in the zygote. Z-stack of confocal images of sperm co-incubated with OVS loaded with labeled miRNAs showed the intracellular location of the delivered miRNAs. Interestingly, individual miRNAs were predominantly localized in specific head compartments, with miR-34c-5p being highly concentrated at the centrosome where it is known to function. These results, for the first time, demonstrate OVS' ability to contribute to the sperm's miRNA repertoire (an important role for solely sperm-derived zygotic miRNAs) and the physiological relevance of an OVS-borne miRNA that is delivered to sperm.


Assuntos
Centrossomo/metabolismo , Ciclo Estral/genética , Vesículas Extracelulares/metabolismo , Perfilação da Expressão Gênica , MicroRNAs/metabolismo , Oviductos/metabolismo , Espermatozoides/metabolismo , Animais , Proliferação de Células , Centrossomo/ultraestrutura , Desenvolvimento Embrionário , Endocitose , Vesículas Extracelulares/ultraestrutura , Feminino , Regulação da Expressão Gênica , Ontologia Genética , Masculino , Camundongos , MicroRNAs/genética , Oviductos/embriologia , Oviductos/ultraestrutura , Reprodutibilidade dos Testes
6.
Mol Hum Reprod ; 24(3): 143-157, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29370405

RESUMO

STUDY QUESTIONS: Are extracellular vesicles (EVs) in the murine oviduct (oviductosomes, OVS) conserved in humans and do they play a role in the fertility of Pmca4-/- females? SUMMARY ANSWER: OVS and their fertility-modulating proteins are conserved in humans, arise via the apocrine pathway, and mediate a compensatory upregulation of PMCA1 (plasma membrane Ca2+-ATPase 1) in Pmca4-/- female mice during proestrus/estrus, to account for their fertility. WHAT IS KNOWN ALREADY: Recently murine OVS were identified and shown during proestrus/estrus to express elevated levels of PMCA4 which they can deliver to sperm. PMCA4 is the major Ca2+ efflux pump in murine sperm and Pmca4 deletion leads to loss of sperm motility and male infertility as there is no compensatory upregulation of the remaining Ca2+ pump, PMCA1. Of the four family members of PMCAs (PMCA1-4), PMCA1 and PMCA4 are ubiquitous, and to date there have been no reports of one isoform being upregulated to compensate for another in any organ/tissue. Since Pmca4-/- females are fertile, despite the abundant expression of PMCA4 in wild-type (WT) OVS, we propose that OVS serve a role of packaging and delivering to sperm elevated levels of PMCA1 in Pmca4-/- during proestrus/estrus to compensate for PMCA4's absence. STUDY DESIGN, SIZE, DURATION: Fallopian tubes from pre-menopausal women undergoing hysterectomy were used to study EVs in the luminal fluid. Oviducts from sexually mature WT mice were sectioned after perfusion fixation to detect EVs in situ. Oviducts were recovered from WT and Pmca4-/- after hormonally induced estrus and sectioned for PMCA1 immunofluorescence (IF) (detected with confocal microscopy) and hematoxylin and eosin staining. Reproductive tissues, luminal fluids and EVs were recovered after induced estrus and after natural cycling for western blot analysis of PMCA1 and qRT-PCR of Pmca1 to compare expression levels in WT and Pmca4-/-. OVS, uterosomes, and epididymal luminal fluid were included in the comparisons. WT and Pmca4-/- OVS were analyzed for the presence of known PMCA4 partners in sperm and their ability to interact with PMCA1, via co-immunoprecipitation. In vitro uptake of PMCA1 from OVS was analyzed in capacitated and uncapacitated sperm via quantitative western blot analysis, IF localization and flow cytometry. Caudal sperm were also assayed for uptake of tyrosine-phosphorylated proteins which were shown to be present in OVS. Finally, PMCA1 and PMCA4 in OVS and that delivered to sperm were assayed for enzymatic activity. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human fallopian tubes were flushed to recover luminal fluid which was processed for OVS via ultracentrifugation. Human OVS were negatively stained for transmission electron microscopy (TEM) and subjected to immunogold labeling, to detect PMCA4. Western analysis was used to detect HSC70 (an EV biomarker), PMCA1 and endothelial nitric oxide synthase (eNOS) which is a fertility-modulating protein delivered to human sperm by prostasomes. Oviducts of sexually mature female mice were sectioned after perfusion fixation for TEM tomography to obtain 3D information and to distinguish cross-sections of EVs from those of microvilli and cilia. Murine tissues, luminal fluids and EVs were assayed for PMCA1 (IF and western blot) or qRT-PCR. PMCA1 levels from western blots were quantified, using band densities and compared in WT and Pmca4-/- after induced estrus and in proestrus/estrus and metestrus/diestrus in cycling females. In vitro uptake of PMCA1 and tyrosine-phosphorylated proteins was quantified with flow cytometry and/or quantitative western blot. Ca2+-ATPase activity in OVS and sperm before and after PMCA1 and PMCA4 uptake was assayed, via the enzymatic hydrolysis rate of ATP. MAIN RESULTS AND THE ROLE OF CHANCE: TEM revealed that human oviducts contain EVs (exosomal and microvesicular). These EVs contain PMCA4 (immunolabeling), eNOS and PMCA1 (western blot) in their cargo. TEM tomography showed the murine oviduct with EV-containing blebs which typify the apocrine pathway for EV biogenesis. Western blots revealed that during proestrus/estrus PMCA1 was significantly elevated in the oviductal luminal fluid (OLF) (P = 0.02) and in OVS (P = 0.03) of Pmca4-/-, compared to WT. Further, while PMCA1 levels did not fluctuate in OLF during the cycle in WT, they were significantly (P = 0.02) higher in proestrus/estrus than at metestrus/diestrus in Pmca4-/-. The elevated levels of PMCA1 in proestrus/estrus, which mimics PMCA4 in WT, is OLF/OVS-specific, and is not seen in oviductal tissues, uterosomes or epididymal luminal fluid of Pmca4-/-. However, qRT-PCR revealed significantly elevated levels of Pmca1 transcript in Pmca4-/- oviductal tissues, compared to WT. PMCA1 could be transferred from OVS to sperm and the levels were significantly higher for capacitated vs uncapacitated sperm, as assessed by flow cytometry (P = 0.001) after 3 h co-incubation, quantitative western blot (P < 0.05) and the frequency of immuno-labeled sperm (P < 0.001) after 30 min co-incubation. Tyrosine phosphorylated proteins were discovered in murine OVS and could be delivered to sperm after their co-incubation with OVS, as detected by western, immunofluorescence localization, and flow cytometry. PMCA1 and PMCA4 in OVS were shown to be enzymatically active and this activity increased in sperm after OVS interaction. LARGE SCALE DATA: None. LIMITATIONS REASONS FOR CAUTION: Although oviductal tissues of WT and Pmca4-/- showed no significant difference in PMCA1 levels, Pmca4-/- levels of OVS/OLF during proestrus/estrus were significantly higher than in WT. We have attributed this enrichment or upregulation of PMCA1 in Pmca4-/- partly to selective packaging in OVS to compensate for the lack of PMCA4. However, in the absence of a difference between WT and Pmca4-/- in the PMCA1 levels in oviductal tissues as a whole, we cannot rule out significantly higher PMCA1 expression in the oviductal epithelium that gives rise to the OVS as significantly higher Pmca1 transcripts were detected in Pmca4-/-. WIDER IMPLICATIONS OF THE FINDINGS: Since OVS and fertility-modulating cargo components are conserved in humans, it suggests that murine OVS role in regulating the expression of proteins required for capacitation and fertility is also conserved. Secondly, OVS may explain some of the differences in in vivo and in vitro fertilization for mouse mutants, as seen in mice lacking the gene for FER which is the enzyme required for sperm protein tyrosine phosphorylation. Our observation that murine OVS carry and can modulate sperm protein tyrosine phosphorylation by delivering them to sperm provides an explanation for the in vivo fertility of Fer mutants, not seen in vitro. Finally, our findings have implications for infertility treatment and exosome therapeutics. STUDY FUNDING AND COMPETING INTEREST(S): The work was supported by National Institute of Health (RO3HD073523 and 5P20RR015588) grants to P.A.M.-D. There are no conflicts of interests.


Assuntos
Capacitação Espermática/fisiologia , Animais , ATPases Transportadoras de Cálcio/genética , ATPases Transportadoras de Cálcio/metabolismo , Tubas Uterinas/citologia , Tubas Uterinas/metabolismo , Tubas Uterinas/ultraestrutura , Feminino , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Oviductos/citologia , Oviductos/metabolismo , Oviductos/ultraestrutura , ATPases Transportadoras de Cálcio da Membrana Plasmática , Pré-Menopausa , Capacitação Espermática/genética , Motilidade dos Espermatozoides/genética , Motilidade dos Espermatozoides/fisiologia
7.
J Cell Physiol ; 233(1): 11-22, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28247940

RESUMO

Reduced sperm motility (asthenospermia) and resulting infertility arise from deletion of the Plasma Membrane Ca2+ -ATPase 4 (Pmca4) gene which encodes the highly conserved Ca2+ efflux pump, PMCA4. This is the major Ca2+ clearance protein in murine sperm. Since the mechanism underlying asthenospermia in PMCA4's absence or reduced activity is unknown, we investigated if sperm PMCA4 negatively regulates nitric oxide synthases (NOSs) and when absent NO, peroxynitrite, and oxidative stress levels are increased. Using co-immunoprecipitation (Co-IP) and Fluorescence Resonance Energy Transfer (FRET), we show an association of PMCA4 with the NOSs in elevated cytosolic [Ca2+ ] in capacitated and Ca2+ ionophore-treated sperm and with neuronal (nNOS) at basal [Ca2+ ] (ucapacitated sperm). FRET efficiencies for PMCA4-eNOS were 35% and 23% in capacitated and uncapacitated sperm, significantly (p < 0.01) different, with the molecules being <10 nm apart. For PMCA4-nNOS, this interaction was seen only for capacitated sperm where FRET efficiency was 24%, significantly (p < 0.05) higher than in uncapacitated sperm (6%). PMCA4 and the NOSs were identified as interacting partners in a quaternary complex that includes Caveolin1, which co-immunoprecipitated with eNOS in a Ca2+ -dependent manner. In Pmca4-/- sperm NOS activity was elevated twofold in capacitated/uncapacitated sperm (vs. wild-type), accompanied by a twofold increase in peroxynitrite levels and significantly (p < 0.001) increased numbers of apoptotic germ cells. The data support a quaternary complex model in which PMCA4 co-ordinates Ca2+ and NO signaling to maintain motility, with increased NO levels resulting in asthenospermia in Pmca4-/- males. They suggest the involvement of PMCA4 mutations in human asthenospermia, with diagnostic relevance.


Assuntos
Astenozoospermia/enzimologia , Sinalização do Cálcio , ATPases Transportadoras de Cálcio/metabolismo , Membrana Celular/enzimologia , Óxido Nítrico/metabolismo , Motilidade dos Espermatozoides , Espermatozoides/enzimologia , Animais , Apoptose , Astenozoospermia/genética , Astenozoospermia/patologia , Astenozoospermia/fisiopatologia , ATPases Transportadoras de Cálcio/deficiência , ATPases Transportadoras de Cálcio/genética , Caveolina 1/metabolismo , Fertilidade , Transferência Ressonante de Energia de Fluorescência , Predisposição Genética para Doença , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Complexos Multienzimáticos , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Ácido Peroxinitroso/metabolismo , Fenótipo , Espermatozoides/patologia
8.
Heliyon ; 3(2): e00250, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28239673

RESUMO

A walnut supplement for a Western-style diet in men was shown to improve sperm motility, vitality, and morphology. To gain further insights into factors underlying this improvement, we administered a parallel walnut-enriched diet to mice [including those with a defect in sperm motility due to deletion of Plasma Membrane Ca2+-ATPase 4 (Pmca4-/- )] to determine if there is a similar improvement that is accompanied by reduced sperm membrane peroxidative damage. Although sperm vitality and acrosome reaction rate were unaffected, the diet led to a significant improvement in motility (P < 0.05) and morphology (P < 0.04) in wild-type sperm and in morphology (P < 0.01) in Pmca4-/- , confirming the diet's efficacy, which appeared to be more modest in mice than in humans. In both strains of mice, the diet resulted in a significant decrease in sperm lipid peroxidation (oxidative stress) levels, but did not rescue the significantly increased apoptotic levels seen in the testis and epididymis of Pmca4 nulls. Our findings support the effectiveness of walnuts on sperm quality, associated with reduced peroxidative damage; and suggest that oxidative stress is involved in the mechanism(s) underlying male reproductive defects in Pmca4-/- .

9.
Mol Hum Reprod ; 23(2): 132-140, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28062807

RESUMO

STUDY QUESTION: Is junctional adhesion molecule A (JAM-A), a sperm protein essential for normal motility, expressed in the murine post-testicular pathway and involved in sperm maturation? SUMMARY ANSWER: JAM-A is present in the prostate and seminal vesicles and in all three regions of the epididymis where it is secreted in epididymosomes in the luminal fluid and can be delivered to sperm in vitro. WHAT IS KNOWN ALREADY: JAM-A shares with the plasma membrane Ca2+ATPase 4 (PMCA4, the major Ca2+ efflux pump in murine sperm) a common interacting partner, CASK (Ca2+/CaM-dependent serine kinase). JAM-A, like PMCA4, plays a role in Ca2+ regulation, since deletion of Jam-A results in significantly elevated intracellular Ca2+ levels and reduced sperm motility. Recently, PMCA4 was reported to be expressed in the epididymis and along with CASK was shown to be in a complex on epididymosomes where it was transferred to sperm. Because of the association of JAM-A with CASK in sperm and because of the presence of PMCA4 and CASK in the epididymis, the present study was performed to determine whether JAM-A is expressed in the epididymis and delivered to sperm during their maturation. STUDY DESIGN, SIZE, DURATION: The epididymides, prostate and seminal vesicles were collected from sexually mature C57BL/6J and Institute for Cancer Research mice and antibodies specific for JAM-A and Ser285 -phosphorylated JAM-A (pJAM-A) were used for the analysis. Tissues, sperm and epididymal luminal fluid (ELF) were studied. Epididymosomes were also isolated for study. Caput and caudal sperm were co-incubated with ELF individually to determine their abilities to acquire JAM-A in vitro. PARTICIPANTS/MATERIALS, SETTING, METHODS: Sections of all three regions of the epididymis were subjected to indirect immunofluorescence analysis. Epididymal tissues, fluid, sperm, prostate and seminal vesicle tissues were analyzed for JAM-A and/or pJAM-A via western blotting analysis. The relative amounts of JAM-A and pJAM-A among epididymal tissues, ELF and sperm were detected by western blot via quantification of band intensities. Epididymosomes were isolated by ultracentrifugation of the ELF after it was clarified to remove cells and tissue fragments, and the proteins western blotted for JAM-A and pJAM-A, and exosomal biochemical markers. FACS analysis was used to quantify the amount of JAM-A present on caput and caudal sperm, as well as the amount of JAM-A acquired in vitro after their co-incubation with ELF. MAIN RESULTS AND THE ROLE OF CHANCE: Western blots revealed that JAM-A is expressed in all three regions of the epididymis, the prostate and seminal vesicles. As confirmed by indirect immunofluorescence, a western blot showed that JAM-A has a higher expression in the corpus and caudal regions, where it is significantly (P < 0.01) more abundant than in the caput. Both JAM-A and Ser285-phosphorylated JAM-A (pJAM-A) are secreted into the ELF where it is highest in the distal regions. In the ELF, both JAM-A and pJAM-A were detected in epididymosomes. Western blotting of sperm proteins showed a significant (P < 0.01) increase of JAM-A and pJAM-A in caudal, compared with caput, sperm. Flow-cytometric analysis confirmed the increase in JAM-A in caudal sperm where it was 1.4-fold higher than in caput ones. Co-incubation of caput and caudal sperm with ELF demonstrated ~2.3- and ~1.3-fold increases, respectively, in JAM-A levels indicating that epididymosomes transfer more JAM-A to caput sperm that are less saturated with the protein than caudal ones. LARGE SCALE DATA: Not applicable. LIMITATIONS, REASONS FOR CAUTION: First, although the ELF was clarified prior to ultracentrifugation for epididymosome isolation, we cannot rule out contamination of the epididymosomal proteins by those from epididymal epithelial cells. Second, the JAM-A detected in the prostate and seminal vesicles might not necessarily be secreted from those organs and may only be present within the tissues, where it would be unable to impact sperm in the ejaculate. WIDER IMPLICATIONS OF THE FINDINGS: Although performed in the mouse the study has implications for humans, as the highly conserved JAM-A is a signaling protein in human sperm. There is physiological significance to the finding that JAM-A, which regulates sperm motility and intracellular Ca2+, exists in elevated levels in the cauda where sperm gain motility and fertilizing ability. The study suggests that the acquisition of JAM-A in the epididymal tract is involved in the mechanism by which sperm gain their motility during epididymal maturation. This increased understanding of sperm physiology is important for aspects of ART. STUDY FUNDING AND COMPETING INTEREST(S): The work was supported by NIH-RO3HD073523 and NIH-5P20RR015588 grants to P.A.M.-D. The authors declare there are no conflicts of interests.


Assuntos
Cálcio/metabolismo , Epididimo/metabolismo , Molécula A de Adesão Juncional/genética , Maturação do Esperma/genética , Motilidade dos Espermatozoides/genética , Espermatozoides/metabolismo , Animais , Sinalização do Cálcio , ATPases Transportadoras de Cálcio/genética , ATPases Transportadoras de Cálcio/metabolismo , Epididimo/citologia , Epididimo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Guanilato Quinases/genética , Guanilato Quinases/metabolismo , Humanos , Molécula A de Adesão Juncional/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Próstata/citologia , Próstata/crescimento & desenvolvimento , Próstata/metabolismo , Transporte Proteico , Glândulas Seminais/citologia , Glândulas Seminais/crescimento & desenvolvimento , Glândulas Seminais/metabolismo , Espermatozoides/citologia , Espermatozoides/crescimento & desenvolvimento
10.
J Cell Mol Med ; 21(1): 121-129, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27558591

RESUMO

Prohibitin (PHB), a major mitochondrial membrane protein, has been shown earlier in our laboratoryto regulate sperm motility via an alteration in mitochondrial membrane potential (MMP) in infertile men with poor sperm quality. To test if PHB expression is associated with sperm mitochondrial superoxide (mROS) levels, here we examined sperm mROS levels, high MMP and lipid peroxidation in infertile men with poor sperm motility (asthenospermia, A) and/or low sperm concentrations (oligoasthenospermia, OA). The diaphorase-type activity of sperm mitochondrial complex I (MCI) and PHB expression were also determined. We demonstrate that mROS and lipid peroxidation levels are significantly higher in sperm from A and OA subjects than in normospermic subjects, whereas high MMP and PHB expression are significantly lower. A positive correlation between mROS and lipid peroxidation and a negative correlation of mROS with PHB expression, high MMP, and sperm motility were found in these subjects. The finding of similar diaphorase-type activity levels of sperm MCI in the three groups studied suggests that the catalytic subunits of MCI in the matrix arm may produce mROS on its own. There may be a dysfunction of electron transport at MCI associated with decreased expression of PHB in sperm with poor quality. We conclude that mROS level is increased and associated with decreased PHB expression, and it may regulate sperm motility via increases in low MMP and lipid peroxidation. This is the first report on the involvement of PHB in human sperm motility loss associated with increased generation of mROS at MCI.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias/efeitos dos fármacos , Proteínas Repressoras/farmacologia , Espermatozoides/efeitos dos fármacos , Superóxidos/metabolismo , Adulto , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Proibitinas , Espécies Reativas de Oxigênio/metabolismo , Contagem de Espermatozoides/métodos , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo
11.
Front Biosci (Landmark Ed) ; 21(6): 1278-85, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27100506

RESUMO

The oviductal epithelial membrane releases into the luminal environment extracellular vesicles (EVs) which are pleomorphic in nature and fall into two categories: exosomes and microvesicles. Both of these membrane vesicles are referred to as Oviductosomes (OVS), and to date have been identified in the murine and bovine species. Bovine EVs derived in vivo and from in vitro culture show differences in their protein cargo which includes CD9 and HSC70 biochemical markers and fertility-modulating proteins such as oviduct-specific glycoprotein (OVGP) and Plasma Membrane Ca(2+) ATPase 4 (PMCA4). PMCA4, an essential multifunctional sperm protein, is hormonally-regulated with elevated levels seen in proestrus/estrus. OVS deliver PMCA4 to sperm via a fusogenic mechanism involving the interaction between CD9 and integrins which are present on their surfaces. Studies of OVS are needed to determine the components of their cargoes and their interaction with oocytes and the very early embryo. Based on our present knowledge of their interaction with sperm, they are expected to play pivotal roles in regulating fertility and promise to inform the current IVF practice.


Assuntos
Exossomos/fisiologia , Tubas Uterinas/fisiologia , Animais , Blastocisto/fisiologia , Micropartículas Derivadas de Células/fisiologia , Micropartículas Derivadas de Células/ultraestrutura , Exossomos/ultraestrutura , Feminino , Humanos , Masculino , ATPases Transportadoras de Cálcio da Membrana Plasmática/fisiologia , Gravidez , Espermatozoides/fisiologia
12.
Front Biosci (Schol Ed) ; 8(1): 115-22, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26709901

RESUMO

The term "uterosomes" was first used to classify extracellular membrane vesicles released into the uterine luminal fluid. These extracellular vesicles (EVs), varying in sizes, fit the classification of exosomes and microvesicles on the basis of size, the presence of the CD9 biochemical marker, and lateral orientation of the membrane. Uterosomes appear to be formed by the apocrine pathway, similar to other reproductive EVs. In the murine system, the protein cargo carried by uterosomes includes glycosyl phosphatidylinositol (GPI)-linked and transmembrane proteins and these are hormonally regulated, appearing at high levels during proestrus/estrus and only marginally present at diestrus /metestrus. Uterosomes have been shown to deliver proteins in their cargo to sperm, with a functional impact, and are thought to participate in promoting sperm capacitation. Further studies are warranted, particularly those aimed at identifying the contents of their cargo during the estrus and menstrual cycle and the role they play n sperm maturation.


Assuntos
Ciclo Estral , Exossomos/fisiologia , Útero/fisiologia , Animais , Estro , Feminino , Fertilização , Humanos , Masculino , Metestro , Biogênese de Organelas , Proteínas/metabolismo , Reprodução , Maturação do Esperma , Espermatozoides
13.
Mol Hum Reprod ; 21(11): 832-43, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26345709

RESUMO

Deletion of the gene encoding the widely conserved plasma membrane calcium ATPase 4 (PMCA4), a major Ca(2+) efflux pump, leads to loss of sperm motility and male infertility in mice. PMCA4's partners in sperm and how its absence exerts its effect on fertility are unknown. We hypothesize that in sperm PMCA4 interacts with endothelial nitric oxide synthase (eNOS) and neuronal nitric oxide synthase (nNOS) which are rapidly activated by Ca(2+), and that these fertility-modulating proteins are present in prostasomes, which deliver them to sperm. We show that in human sperm PMCA4 is present on the acrosome, inner acrosomal membrane, posterior head, neck, midpiece and the proximal principal piece. PMCA4 localization showed inter- and intra-individual variation and was most abundant at the posterior head/neck junction, co-localizing with NOSs. Co-immunoprecipitations (Co-IP) revealed a close association of PMCA4 and the NOSs in Ca(2+) ionophore-treated sperm but much less so in uncapacitated untreated sperm. Fluorescence resonance energy transfer (FRET) showed a similar Ca(2+)-related association: PMCA4 and the NOSs are within 10 nm apart, and preferentially so in capacitated, compared with uncapacitated, sperm. FRET efficiencies varied, being significantly (P < 0.001) higher at high cytosolic Ca(2+) concentration ([Ca(2+)]c) in capacitated sperm than at low [Ca(2+)]c in uncapacitated sperm for the PMCA4-eNOS complex. These dynamic interactions were not seen for PMCA4-nNOS complexes, which had the highest FRET efficiencies. Further, along with Ca(2+)/CaM-dependent serine kinase (CASK), PMCA4 and the NOSs are present in the seminal plasma, specifically in prostasomes where Co-IP showed complexes similar to those in sperm. Finally, flow cytometry demonstrated that following co-incubation of sperm and seminal plasma, PMCA4 and the NOSs can be delivered in vitro to sperm via prostasomes. Our findings indicate that PMCA4 interacts simultaneously with the NOSs preferentially at high [Ca(2+)]c in sperm to down-regulate them, and thus prevent elevated levels of NO, known to induce asthenozoospermia via oxidative stress. Our studies point to the potential underlying cause of infertility in PMCA4's absence, and suggest that inactivating mutations of PMCA4 could lead to asthenozoospermia and human infertility. Screening for these mutations may serve both diagnostic and therapeutic purposes.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Óxido Nítrico Sintase/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Cálcio/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Transferência Ressonante de Energia de Fluorescência , Humanos , Técnicas In Vitro , Masculino , Óxido Nítrico Sintase/genética , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Ligação Proteica , Motilidade dos Espermatozoides/genética , Motilidade dos Espermatozoides/fisiologia , Espermatozoides/metabolismo
14.
Asian J Androl ; 17(5): 720-5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26112481

RESUMO

A variety of glycosylphosphatidylinositol (GPI)-linked proteins are acquired on spermatozoa from epididymal luminal fluids (ELF) during sperm maturation. These proteins serve roles in immunoprotection and in key steps of fertilization such as capacitation, acrosomal exocytosis and sperm-egg interactions. Their acquisition on sperm cells is mediated both by membrane vesicles (epididymosomes, EP) which were first reported to dock on the sperm surface, and by lipid carriers which facilitate the transfer of proteins associated with the membrane-free fraction of ELF. While the nonvesicular fraction is more efficient, both pathways are dependent on hydrophobic interactions between the GPI-anchor and the external lipid layer of the sperm surface. More recently proteomic and hypothesis-driven studies have shown that EP from several mammals carry transmembrane (TM) proteins, including plasma membrane Ca 2 + -ATPase 4 (PMCA4). Synthesized in the testis, PMCA4 is an essential protein and the major Ca 2 + efflux pump in murine spermatozoa. Delivery of PMCA4 to spermatozoa from bovine and mouse EP during epididymal maturation and in vitro suggests that the docking of EP on the sperm surface precedes fusion, and experimental evidence supports a fusogenic mechanism for TM proteins. Fusion is facilitated by CD9, which generates fusion-competent sites on membranes. On the basis of knowledge of PMCA4's interacting partners a number of TM and membrane-associated proteins have been identified or are predicted to be present, in the epididymosomal cargo deliverable to spermatozoa. These Ca 2 + -dependent proteins, undetected in proteomic studies, play essential roles in sperm motility and fertility, and their detection highlights the usefulness of the hypothesis-driven approach.


Assuntos
Epididimo/metabolismo , Proteínas de Membrana/metabolismo , Maturação do Esperma/fisiologia , Espermatozoides/metabolismo , Animais , Humanos , Masculino , Proteômica , Interações Espermatozoide-Óvulo/fisiologia
15.
J Biol Chem ; 290(29): 17710-17723, 2015 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-26023236

RESUMO

Oviductosomes ((OVS), exosomes/microvesicles), which deliver the Ca(2+) efflux pump, plasma membrane Ca(2+)ATPase 4 (PMCA4), to sperm are likely to play an important role in sperm fertilizing ability (Al-Dossary, A. A., Strehler, E. E., and Martin-DeLeon, P. A. (2013) PloS one 8, e80181). It is unknown how exosomes/microvesicles deliver transmembrane proteins such as PMCA4 to sperm. Here we define a novel experimental approach for the assessment of the interaction of OVS with sperm at a nanoscale level, using a lipophilic dye (FM4-64FX) and three-dimensional SR/SIM, which has an 8-fold increase in volumetric resolution, compared with conventional confocal microscopy. Coincubation assays detected fusion of prelabeled OVS with sperm, primarily over the head and midpiece. Immunofluorescence revealed oviductosomal delivery of PMCA4a to WT and Pmca4 KO sperm, and also endogenous PMCA4a on the inner acrosomal membrane. Fusion was confirmed by transmission immunoelectron microscopy, showing immunogold particles in OVS, and fusion stalks on sperm membrane. Immunofluorescence colocalized OVS with the αv integrin subunit which, along with CD9, resides primarily on the sperm head and midpiece. In capacitated and acrosome reacted sperm, fusion was significantly (p < 0.001) inhibited by blocking integrin/ligand interactions via antibodies, exogenous ligands (vitronectin and fibronectin), and their RGD recognition motif. Our results provide evidence that receptor/ligand interactions, involving αvß3 and α5ß1integrins on sperm and OVS, facilitate fusion of OVS in the delivery of transmembrane proteins to sperm. The mechanism uncovered is likely to be also involved in cargo delivery of prostasomes, epididymosomes, and uterosomes.


Assuntos
Exossomos/metabolismo , Tubas Uterinas/metabolismo , Integrinas/metabolismo , Fusão de Membrana , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Espermatozoides/metabolismo , Animais , Células Cultivadas , Tubas Uterinas/citologia , Tubas Uterinas/ultraestrutura , Feminino , Fertilização , Imunofluorescência , Corantes Fluorescentes/análise , Integrinas/análise , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , ATPases Transportadoras de Cálcio da Membrana Plasmática/análise , Transporte Proteico , Espermatozoides/ultraestrutura
16.
Asian J Androl ; 17(2): 261-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25370207

RESUMO

Titanium dioxide (TiO 2 ) nanoparticles (TNPs) are widely used commercially and exist in a variety of products. To determine if anatase TNPs (ATNPs) in doses smaller than previously used reach the scrotum after entry in the body at a distant location and induce sperm defects, 100% ATNP (2.5 or 5 mg kg-1 body weight) was administered intraperitoneally to adult males for three consecutive days, followed by sacrifice 1, 2, 3, or 5 weeks later (long-) or 24, 48 or 120 h (short-term exposure). Transmission electron microscopy revealed the presence of ANTP in scrotal adipose tissues collected 120 h postinjection when cytokine evaluation showed an inflammatory response in epididymal tissues and fluid. At 120 h and up to 3 weeks postinjection, testicular histology revealed enlarged interstitial spaces. Significantly increased numbers of terminal deoxyribonucleotidyl transferase-mediated dUTP nick-end labeling-positive (apoptotic) germ (P = 0.002) and interstitial space cells (P = 0.04) were detected in treated males. Caudal epididymal sperm from the short-term, but not a long-term, arm showed significantly (P < 0.001) increased frequencies of flagellar abnormalities, excess residual cytoplasm (ERC), and unreacted acrosomes in treated versus controls (dose-response relationship). A novel correlation between ERC and unreacted acrosomes was uncovered. At 120 h, there were significant decreases in hyperactivated motility (P < 0.001) and mitochondrial membrane potential (P < 0.05), and increased reactive oxygen species levels (P < 0.00001) in treated versus control sperm. These results indicate that at 4-8 days postinjection, ANTP induce structural and functional sperm defects associated with infertility, and DNA damage via oxidative stress. Sperm defects were transient as they were not detected 10 days to 5 weeks postinjection.


Assuntos
Nanopartículas/efeitos adversos , Fármacos Fotossensibilizantes/efeitos adversos , Fármacos Fotossensibilizantes/farmacologia , Espermatozoides/efeitos dos fármacos , Titânio/efeitos adversos , Titânio/farmacologia , Acrossomo/efeitos dos fármacos , Acrossomo/patologia , Acrossomo/fisiologia , Animais , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Relação Dose-Resposta a Droga , Flagelos/efeitos dos fármacos , Flagelos/patologia , Flagelos/fisiologia , Injeções Intraperitoneais , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Modelos Animais , Nanopartículas/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Fármacos Fotossensibilizantes/administração & dosagem , Espermatozoides/patologia , Espermatozoides/fisiologia , Fatores de Tempo , Titânio/administração & dosagem
17.
Biol Reprod ; 91(5): 109, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25232017

RESUMO

To initiate the crucial cell adhesion events necessary for fertilization, sperm must penetrate extracellular matrix barriers containing hyaluronic acid (HA), a task thought to be accomplished by neutral-active hyaluronidases. Here we report that the ~57 kDa hyaluronidase 2 (HYAL2) that in somatic tissues has been highly characterized to be acid-active is present in mouse and human sperm, as detected by Western blot, flow cytometric, and immunoprecipitation assays. Immunofluorescence revealed its presence on the plasma membrane over the acrosome, the midpiece, and proximal principal piece in mice where protein fractionation demonstrated a differential distribution in subcellular compartments. It is significantly more abundant in the acrosome-reacted (P = 0.04) and soluble acrosomal fractions (P = 0.006) (microenvironments where acid-active hyaluronidases function) compared to that of the plasma membrane where neutral hyaluronidases mediate cumulus penetration. Using HA substrate gel electrophoresis, immunoprecipitated HYAL 2 was shown to have catalytic activity at pH 4.0. Colocalization and coimmunoprecipitation assays reveal that HYAL2 is associated with its cofactor, CD44, consistent with CD44-dependent HYAL2 activity. HYAL2 is also present throughout the epididymis, where Hyal2 transcripts were detected, and in the epididymal luminal fluids. In vitro assays demonstrated that HYAL2 can be acquired on the sperm membrane from epididymal luminal fluids, suggesting that it plays a role in epididymal maturation. Because similar biphasic kinetics are seen for HYAL2 and SPAM1 (Sperm adhesion molecule 1), it is likely that HYAL2 plays a redundant role in the catalysis of megadalton HA to its 20 kDa intermediate during fertilization.


Assuntos
Moléculas de Adesão Celular/fisiologia , Epididimo/metabolismo , Células Germinativas/metabolismo , Hialuronoglucosaminidase/fisiologia , Espermatozoides/metabolismo , Animais , Moléculas de Adesão Celular/genética , Epididimo/enzimologia , Feminino , Fertilização , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/fisiologia , Expressão Gênica , Células Germinativas/enzimologia , Humanos , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/genética , Masculino , Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Interações Espermatozoide-Óvulo , Espermatozoides/enzimologia
19.
PLoS One ; 8(11): e80181, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24244642

RESUMO

PMCA4, a membrane protein, is the major Ca(2+) efflux pump in murine sperm where its deletion leads to a severe loss of hyperactivated motility and to male infertility. We have previously shown that the PMCA4b splice variant interacts with CASK (Ca(2+/)CaM-dependent serine kinase) in regulating sperm Ca(2+). More recently we detected that PMCA4a isoform, in addition to its presence in testis, is secreted in the epididymal luminal fluid and transferred to sperm. Here we show that Pmca4 mRNA is expressed in both the 4a and 4b variants in the vagina, uterus, and oviduct. Immunofluorescence reveals that PMCA4a is similarly expressed and is elevated during estrus, appearing in the glandular and luminal epithelia. Western analysis detected PMCA4a in all tissues and in the luminal fluids (LF) of the vagina (VLF), uterus (ULF), and the oviduct (OLF) collected during estrus. It was ~9- and 4-fold higher in OLF than in VLF and ULF, and only marginally present in LF collected at metestrus/diestrus. Fractionation of the LF collected at estrus, via ultracentrifugation, revealed that 100% of the PMCA4a resides in the vesicular fraction of the ULF and OLF. Transmission electron microscopy (TEM) revealed that OLF vesicles have an exosomal orientation (with the cytoplasmic-side inward), a size range of 25-100 nm, with the characteristic CD9 biomarker. Thus, we dubbed these vesicles "oviductosomes", to which PMCA4a was immunolocalized. Incubation of caudal sperm in the combined LF or exosomes resulted in up to a ~3-fold increase of sperm PMCA4a, as detected by flow cytometry, indicating in vitro uptake. Our results are consistent with the increased requirement of Ca(2+) efflux in the oviduct. They show for the first time the presence of oviductal exosomes and highlight their role, along with uterosomes and vaginal exosomes, in post-testicular sperm acquisition of PMCA4a which is essential for hyperactivated motility and fertility.


Assuntos
ATPases Transportadoras de Cálcio/genética , Estro/fisiologia , Exossomos/metabolismo , Fertilidade/fisiologia , Reprodução/fisiologia , Espermatozoides/metabolismo , Animais , Biomarcadores/metabolismo , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Vesículas Citoplasmáticas/metabolismo , Vesículas Citoplasmáticas/ultraestrutura , Exossomos/ultraestrutura , Feminino , Expressão Gênica , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Oviductos/metabolismo , Oviductos/ultraestrutura , Transporte Proteico , Tetraspanina 29/genética , Tetraspanina 29/metabolismo , Útero/metabolismo , Útero/ultraestrutura , Vagina/metabolismo , Vagina/ultraestrutura
20.
Proc (Bayl Univ Med Cent) ; 26(3): 295-7, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23814397

RESUMO

Mucinous carcinoma of the breast is one of the rarer forms of intramammary cancer, often presenting as a lobulated, fairly well circumscribed mass on mammography, sonography, and gadolinium-enhanced magnetic resonance imaging. It accounts for 1% to 7% of all breast cancers and generally carries a better prognosis than other types of malignant breast cancers. Metastatic disease occurs at a lower frequency than in other types of invasive carcinoma. We present an atypical case of mucinous carcinoma in a woman who presented with a palpable intramammary lymph node metastasis from an unknown breast primary. Subsequent magnetic resonance imaging and percutaneous biopsy demonstrated histologic findings consistent with a mixed mucinous neoplasm with a micropapillary pattern.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...