Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Ovarian Res ; 11(1): 19, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29490681

RESUMO

BACKGROUND: Retinoic acid (RA) signaling has been identified as a key driver in male and female gamete development. The presence of RA is a critical step in the initiation of meiosis and is required for the production of competent oocytes from primordial germ cells. Meiosis has been identified as a difficult biological process to recapitulate in vitro, when differentiating stem cells to germ cells. We have previously shown that primordial germ cell-like cells, and more advanced oocyte-like cells (OLCs), can be formed by differentiating mouse skin-derived stem cells. However, the OLCs remain unable to function due to what appears to be failure of meiotic initiation. The aim of this study was to determine the effect of RA treatment, during stem cell differentiation to germ cells, particularly on the initiation of meiosis. RESULTS: Using qPCR we found significant increases in the meiosis markers Stra8 and Sycp3 and a significant reduction in the meiosis inhibitor Nanos2, in the differentiating populations. Furthermore, OLCs from the RA treated group, expressed significantly more of the meiosis regulatory gene Marf1 and the oocyte marker Oct4. At the protein level RA treatment was found to increase the expression of the gap junction protein CX43 and the pluripotency marker OCT4. Moreover, the expression of SYCP3 was significantly upregulated and the localization pattern better matched that of control fetal ovarian cells. RA treatment also improved the structural integrity of the OLCs produced by initiating the expression of all three zona pellucida transcripts (Zp1-3) and improving ZP3 expression levels and localization. Finally, the addition of RA during differentiation led to an almost two-fold increase in the number of OLCs recovered and increased their in vitro growth. CONCLUSION: RA is a key driver in the formation of functioning gametes and its addition during stem cell to germ cell differentiation improves OLCs entry into meiosis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Germinativas/citologia , Células Germinativas/efeitos dos fármacos , Pele/citologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Biomarcadores , Conexina 43/genética , Conexina 43/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células Germinativas/metabolismo , Meiose/genética , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Transporte Proteico , Células-Tronco/metabolismo , Zona Pelúcida/efeitos dos fármacos , Zona Pelúcida/metabolismo
2.
Semin Cell Dev Biol ; 50: 22-30, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26780117

RESUMO

The development and differentiation of cells involved in spermatogenesis requires highly regulated and coordinated interactions between cells. Intercellular communication, particularly via connexin43 (Cx43) gap junctions, plays a critical role in the development of germ cells during fetal development and during spermatogenesis in the adult. Loss of Cx43 in the fetus results in a decreased number of germ cells, while the loss of Cx43 in the adult Sertoli cells results in complete inhibition of spermatogenesis. Connexins 26, 32, 33, 36, 45, 46 and 50 have also been localized to specific compartments of the testis in various mammals. Loss of Cx46 is associated with an increase in germ cell apoptosis and loss of the integrity of the blood-testis barrier, while loss of other connexins appears to have more subtle effects within the seminiferous tubule. Outside the seminiferous tubule, the interstitial Leydig cells express connexins 36 and 45 along with Cx43; deletion of the latter connexin did not reveal it to be crucial for steroidogenesis or for the development and differentiation of Leydig cells. In contrast, loss of Cx43 from Sertoli cells results in Leydig cell hyperplasia, suggesting important cross-talk between Sertoli and Leydig cells. In the epididymis connexins 26, 30.3, Cx31.1, 32, and 43 have been identified and differentiation of the epithelium is associated with dramatic changes in their expression. Decreased expression of Cx43 results in decreased sperm motility, a function acquired by spermatozoa during epididymal transit. Clearly, intercellular gap junctional communication within the testis and epididymis represents a critical aspect of male reproductive function and fertility. The implications of this mode of intercellular communication for male fertility remains a poorly understood but important facet of male reproduction.


Assuntos
Conexinas/metabolismo , Espermatogênese , Testículo/embriologia , Testículo/metabolismo , Animais , Epididimo/metabolismo , Fertilidade , Células Germinativas/metabolismo , Humanos , Masculino
3.
Hum Reprod Update ; 21(3): 340-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25667189

RESUMO

BACKGROUND: Connexins comprise a family of ~20 proteins that form intercellular membrane channels (gap junction channels) providing a direct route for metabolites and signalling molecules to pass between cells. This review provides a critical analysis of the evidence for essential roles of individual connexins in female reproductive function, highlighting implications for women's reproductive health. METHODS: No systematic review has been carried out. Published literature from the past 35 years was surveyed for research related to connexin involvement in development and function of the female reproductive system. Because of the demonstrated utility of genetic manipulation for elucidating connexin functions in various organs, much of the cited information comes from research with genetically modified mice. In some cases, a distinction is drawn between connexin functions clearly related to the formation of gap junction channels and those possibly linked to non-channel roles. RESULTS AND CONCLUSIONS: Based on work with mice, several connexins are known to be required for female reproductive functions. Loss of connexin43 (CX43) causes an oocyte deficiency, and follicles lacking or expressing less CX43 in granulosa cells exhibit reduced growth, impairing fertility. CX43 is also expressed in human cumulus cells and, in the context of IVF, has been correlated with pregnancy outcome, suggesting that this connexin may be a determinant of oocyte and embryo quality in women. Loss of CX37, which exclusively connects oocytes with granulosa cells in the mouse, caused oocytes to cease growing without acquiring meiotic competence. Blocking of CX26 channels in the uterine epithelium disrupted implantation whereas loss or reduction of CX43 expression in the uterine stroma impaired decidualization and vascularization in mouse and human. Several connexins are important in placentation and, in the human, CX43 is a key regulator of the fusogenic pathway from the cytotrophoblast to the syncytiotrophoblast, ensuring placental growth. CX40, which characterizes the extravillous trophoblast (EVT), supports proliferation of the proximal EVTs while preventing them from differentiating into the invasive pathway. Furthermore, women with recurrent early pregnancy loss as well as those with endometriosis exhibit reduced levels of CX43 in their decidua. The antimalaria drug mefloquine, which blocks gap junction function, is responsible for increased risk of early pregnancy loss and stillbirth, probably due to inhibition of intercellular communication in the decidua or between trophoblast layers followed by an impairment of placental growth. Gap junctions also play a critical role in regulating uterine blood flow, contributing to the adaptive response to pregnancy. Given that reproductive impairment can result from connexin mutations in mice, it is advised that women suffering from somatic disease symptoms associated with connexin gene mutations be additionally tested for impacts on reproductive function. Better knowledge of these essential connexin functions in human female reproductive organs is important for safeguarding women's reproductive health.


Assuntos
Comunicação Celular/efeitos dos fármacos , Conexina 43/antagonistas & inibidores , Células do Cúmulo/metabolismo , Junções Comunicantes/efeitos dos fármacos , Mefloquina/efeitos adversos , Animais , Conexina 26 , Conexina 43/genética , Conexinas , Implantação do Embrião/fisiologia , Perda do Embrião/induzido quimicamente , Feminino , Fertilidade/fisiologia , Junções Comunicantes/fisiologia , Humanos , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/metabolismo , Camundongos , Oócitos/metabolismo , Oogênese/fisiologia , Placenta/metabolismo , Gravidez , Resultado da Gravidez , RNA Mensageiro/genética , Saúde Reprodutiva , Trofoblastos/citologia , Trofoblastos/metabolismo
4.
Stem Cells Dev ; 23(14): 1636-46, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24694074

RESUMO

Expression of the gap junction protein, connexin43 (Cx43), begins early during embryogenesis and is maintained in many different cell types. Several stem cell populations have been shown to express Cx43 and to form functional gap junctions. While it is clear that Cx43 is critical to the function of many organs, whether the same is true for stem cells has not been clearly demonstrated. Recently, stem cells isolated from newborn mouse skin were shown to form oocyte-like cells (OLCs) in vitro, hence the present study focussed on the role Cx43 plays in the proliferation and differentiation of these cells. The stem cells express Cx43 and those from knockout mice (Cx43 KO) exhibited significantly reduced cell-cell coupling. Loss of Cx43 reduced the rate of cellular migration [Cx43 KO, 1.57±0.65 radial cell units (RCU); wildtype (WT), 5.57±0.37 RCU] but increased the proliferation rate of the stem cells (Cx43 KO, 29.40%±2.02%; WT, 12.76%±1.50%). The expression of the pluripotency markers OCT4 and Nanog were found to be reduced in the Cx43 KO population, suggesting an inhibition of differentiation potential. To test the differentiation ability, the stem cells were induced to form neuronal cell types in vitro. While both the WT and KO cells were able to form GFAP-positive astrocytic cells, only WT stem cells were able to form ßIII tubulin-positive neurons. Similarly, the ability of the stem cells to form OLCs was ablated by the loss of Cx43. These data reveal a role for Cx43 in maintaining multipotency within the skin-derived stem cell population.


Assuntos
Diferenciação Celular/genética , Conexina 43/metabolismo , Desenvolvimento Embrionário , Neurônios/citologia , Células-Tronco/citologia , Animais , Proliferação de Células/genética , Conexina 43/genética , Junções Comunicantes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/biossíntese , Oócitos/citologia , Oócitos/crescimento & desenvolvimento , Pele/citologia , Pele/embriologia , Células-Tronco/metabolismo , Tubulina (Proteína)/biossíntese
5.
Adv Med Educ Pract ; 4: 201-16, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24159264

RESUMO

BACKGROUND: A training program in Reproduction, Early Development, and the Impact on Health (REDIH) was initiated in 2009 by researchers specializing in biomedical, clinical, population health, and ethics research from seven collaborating universities in Quebec and Ontario, and Health Canada. This paper reports the findings from the first three years of the 6-year program. OBJECTIVES: The objective of the REDIH program is to provide increased opportunities for excellent training in reproduction and early development for graduate students and fellows, in order to build research, clinical, regulatory, decision-making, and industry capacity in Canada. METHODS: A mixed methods approach was used to evaluate the REDIH training program, so as to combine the strengths of both qualitative and quantitative studies. A total of four focus groups (two with mentors and two with trainees) were run during the June 2012 REDIH meeting. Surveys were administered directly after each training module. The W(e)Learn framework was used as a guide to design and evaluate the program and answer the research questions. RESULTS: The data from the analysis of the focus group interviews, in corroboration with the survey data, suggested trainees enjoyed and benefited from the REDIH experience. Trainees provided several examples of new knowledge and skills they had acquired from REDIH sessions, regarding reproductive and early developmental biology, and health. A few trainees who had been in the program for over a year provided examples of knowledge and skills acquired during the REDIH session that they were using in their place of work. Next steps will include following up on REDIH graduates to see if the program has had any impact on trainees' employment opportunities and career development. CONCLUSION: Trainees and mentors concluded that the curricular design, which focuses on modules in 2-day learning sessions over a 6-year period, with opportunities for application in the workplace, enabled the sessions to be tailored to the outcomes of the formative evaluation. By sharing our experiences with REDIH, we hope that others can benefit from this unique emerging design, which focuses on the flexibility and receptivity of the mentors, and results in a program that lends itself to curriculum modification and tailoring as learners' needs are solicited and addressed.

6.
Biol Reprod ; 89(5): 111, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24048574

RESUMO

Connexin43 (CX43), encoded by Gja1 in the mouse, is highly expressed in decidual cells and is known to be important for the transformation of stromal cells into the compact decidua and for neoangiogenesis. Here we investigated if the dominant Gja1(Jrt) mutation encoding CX43(G60S) in mice, which results in a phenotype resembling oculodentodigital dysplasia in humans, has an impact on decidualization, angiogenesis, and implantation. We found a reduced mean weight of fetuses at Gestational Day 17.5 in dams carrying this mutation, with the growth deficiency being independent of fetal genotype. Although the mutant implantation sites exhibited a reduction in CX43 protein, with most immunoreactivity being cytoplasmic, the decidua was morphologically intact at Embryonic Days 5.5 to 7.5. However, the mutation resulted in enhanced and irregular angiogenesis and an increased level of expression of the angiogenic factor-encoding genes Vegfa, Flt1, Kdr, and Fgf2 as well as the prolactin-related gene Prl6a. Moreover, immunolocalization of VEGFA, FLT1, and KDR revealed a homogeneous distribution pattern in the mesometrial as well as antimesometrial decidua of the mutants. Most obviously, uterine NK cells are drastically diminished in the mesometrial decidua of the mutant mice. Invasion of ectoplacental cone cells was disoriented, and placentation was established more laterally in the implantation chambers. It was concluded that the CX43(G60S) mutant impairs control of decidual angiogenesis, leading to dysmorphic placentation and fetal growth restriction. This phenomenon could contribute to the reduced fetal weights and viability of pups born of Gja1(Jrt)/+ dams.


Assuntos
Conexina 43/genética , Decídua/irrigação sanguínea , Neovascularização Fisiológica/genética , Placenta/citologia , Placenta/fisiologia , Animais , Animais Recém-Nascidos , Polaridade Celular/genética , Códon sem Sentido , Feminino , Retardo do Crescimento Fetal/genética , Genes Dominantes , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Circulação Placentária/genética , Placentação/genética , Gravidez
7.
Biol Reprod ; 89(2): 39, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23843235

RESUMO

WNTs are extracellular signaling molecules that exert their actions through receptors of the frizzled (FZD) family. Previous work indicated that WNT2 regulates cell proliferation in mouse granulosa cells acting through CTNNB1 (beta-catenin), a key component in canonical WNT signaling. In other cells, WNT signaling has been shown to regulate expression of connexin43 (CX43), a gap junction protein, as well as gap junction assembly. Since previous work demonstrated that CX43 is also essential in ovarian follicle development, the objective of this study was to determine if WNT2 regulates CX43 expression and/or gap-junctional intercellular communication (GJIC) in granulosa cells. WNT2 knockdown via siRNA markedly reduced CX43 expression and GJIC. CX43 expression, the extent of CX43-containing gap junction membrane, and GJIC were also reduced by CTNNB1 transient knockdown. CTNNB1 is mainly localized to the membranes between granulosa cells but disappeared from this location after WNT2 knockdown. Furthermore, CTNNB1 knockdown interfered with the ability of follicle-stimulating hormone (FSH) to promote the mobilization of CX43 into gap junctions. We propose that the WNT2/CTNNB1 pathway regulates CX43 expression and GJIC in granulosa cells by modulating CTNNB1 stability and localization in adherens junctions, and that this is essential for FSH stimulation of GJIC.


Assuntos
Conexina 43/metabolismo , Hormônio Foliculoestimulante/metabolismo , Junções Comunicantes/metabolismo , Células da Granulosa/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt2/metabolismo , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Feminino , Hormônio Foliculoestimulante/farmacologia , Junções Comunicantes/efeitos dos fármacos , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Camundongos , Fase S/efeitos dos fármacos , Fase S/fisiologia , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
8.
Can J Physiol Pharmacol ; 91(2): 157-64, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23458200

RESUMO

Connexin43 has been recognized as forming gap junctions in Leydig cells. However, previous work has shown that mouse Leydig cells lacking this connexin do not suffer any limitation of their ability to produce testosterone when stimulated with luteinizing hormone. The objective of this study was to identify additional connexins in mouse Leydig cells that could be required for steroidogenesis. A reverse transcription - polymerase chain reaction screen involving isolated adult Leydig cells identified connexin36 and connexin45 as expressed along with connexin43. Treatment of dissociated testes with carbenoxolone, a nonspecific blocker of gap junctional coupling, significantly reduced testosterone output as did treatment with quinine, which disrupts coupling provided by connexin36 and connexin45 gap junctions but not those composed of connexin43, indicating that either or both of connexins 36 and 45 could be involved in supporting Leydig cell steroidogenesis. Immunolabeling of adult mouse testis sections confirmed the localization of connexin36 along with connexin43 in Leydig cell gap junctions but not connexin45, which is distributed throughout the cells. It was concluded that connexin36, connexin43, and connexin45 are coexpressed in Leydig cells with connexins 36 and 43 contributing to gap junctions. The role of connexin45 remains to be elucidated.


Assuntos
Conexina 43/metabolismo , Conexinas/metabolismo , Células Intersticiais do Testículo/metabolismo , Testosterona/biossíntese , Animais , Carbenoxolona/farmacologia , Células Cultivadas , Conexina 43/genética , Conexinas/genética , Imunofluorescência , Junções Comunicantes/metabolismo , Células Intersticiais do Testículo/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Masculino , Camundongos , Quinina/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/metabolismo , Proteína delta-2 de Junções Comunicantes
9.
J Biol Chem ; 288(10): 7077-85, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23344951

RESUMO

Restoration of the functional potency of pancreatic islets either through enhanced proliferation (hyperplasia) or increase in size (hypertrophy) of beta cells is a major objective for intervention in diabetes. We have obtained experimental evidence that global knock-out of a small, single-span regulatory subunit of Na,K-ATPase, FXYD2, alters glucose control. Adult Fxyd2(-/-) mice showed significantly lower blood glucose levels, no signs of peripheral insulin resistance, and improved glucose tolerance compared with their littermate controls. Strikingly, there was a substantial hyperplasia in pancreatic beta cells from the Fxyd2(-/-) mice compared with the wild type littermates, compatible with an observed increase in the level of circulating insulin. No changes were seen in the exocrine compartment of the pancreas, and the mice had only a mild, well-adapted renal phenotype. Morphometric analysis revealed an increase in beta cell mass in KO compared with WT mice. This appears to explain a phenotype of hyperinsulinemia. By RT-PCR, Western blot, and immunocytochemistry we showed the FXYD2b splice variant in pancreatic beta cells from wild type mice. Phosphorylation of Akt kinase was significantly higher under basal conditions in freshly isolated islets from Fxyd2(-/-) mice compared with their WT littermates. Inducible expression of FXYD2 in INS 832/13 cells produced a reduction in the phosphorylation level of Akt, and phosphorylation was restored in parallel with degradation of FXYD2. Thus we suggest that in pancreatic beta cells FXYD2 plays a role in Akt signaling pathways associated with cell growth and proliferation.


Assuntos
Glicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/sangue , ATPase Trocadora de Sódio-Potássio/metabolismo , Processamento Alternativo , Animais , Western Blotting , Linhagem Celular Tumoral , Feminino , Regulação Enzimológica da Expressão Gênica , Teste de Tolerância a Glucose , Hiperplasia , Imuno-Histoquímica , Células Secretoras de Insulina/patologia , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , ATPase Trocadora de Sódio-Potássio/genética
10.
Front Genet ; 3: 129, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22798963

RESUMO

Growth and maturation of healthy oocytes within follicles requires bidirectional signaling and intercellular gap junctional communication. Aberrant endocrine signaling and loss of gap junctional communication between the oocyte and granulosa cells leads to compromised folliculogenesis, oocyte maturation, and oocyte competency, consequently impairing fertility. Given that oocyte-specific DNA methylation establishment at imprinted genes occurs during this growth phase, we determined whether compromised endocrine signaling and gap junctional communication would disrupt de novo methylation acquisition using ERß and connexin37 genetic models. To compare mutant oocytes to control oocytes, DNA methylation acquisition was first examined in individual, 20-80 µm control oocytes at three imprinted genes, Snrpn, Peg3, and Peg1. We observed that each gene has its own size-dependent acquisition kinetics, similar to previous studies. To determine whether compromised endocrine signaling and gap junctional communication disrupted de novo methylation acquisition,individual oocytes from Esr2- and Gja4-deficient mice were also assessed for DNA methylation establishment. We observed no aberrant or delayed acquisition of DNA methylation at Snrpn, Peg3, or Peg1 in oocytes from Esr2-deficient females, and no perturbation in Snrpn or Peg3de novo methylation in oocytes from Gja4-null females. However, Gja4 deficiency resulted in a loss or delay in methylation acquisition at Peg1. One explanation for this difference between the three loci analyzed is the late establishment of DNA methylation at the Peg1 gene. These results indicate that compromised fertility though impaired intercellular communication can lead to imprinting acquisition errors. Further studies are required to determine the effects of subfertility/infertility originating from impaired signaling and intercellular communication during oogenesis on imprint maintenance during preimplantation development.

11.
J Membr Biol ; 245(5-6): 291-301, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22729691

RESUMO

Connexin43 (Cx43) forms gap junctions that couple the granulosa cells of ovarian follicles. In Cx43 knockout mice, follicle growth is restricted as a result of impaired granulosa cell proliferation. We have used these mice to examine the importance of specific Cx43 phosphorylation sites in follicle growth. Serines at residues 255, 262, 279, and 282 are MAP kinase substrates that, when phosphorylated, reduce junctional conductance. Mutant forms of Cx43 were constructed with these serines replaced with amino acids that cannot be phosphorylated. These mutants were transduced into Cx43 knockout ovarian somatic cells that were combined with wild-type oocytes and grafted into immunocompromised female mice permitting follicle growth in vivo. Despite residues 255 or 262 being mutated to prevent their being phosphorylated, recombinant ovaries constructed with these mutants were able to rescue the null phenotype, restoring complete folliculogenesis. In contrast, Cx43 with serine to alanine mutations at both residues 279 and 282 or at all four residues failed to rescue folliculogenesis; the mutant molecules were largely confined to intracellular sites, with few gap junctions. Using an in vitro proliferation assay, we confirmed a decrease in proliferation of granulosa cells expressing the double mutant construct. These results indicate that Cx43 phosphorylation by MAP kinase at serines 279 and 282 occurs in granulosa cells of early follicles and that this is involved in regulating follicle development.


Assuntos
Conexina 43/metabolismo , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Ovário/citologia , Ovário/metabolismo , Serina/metabolismo , Animais , Proliferação de Células , Feminino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ovário/crescimento & desenvolvimento , Fosforilação
12.
PLoS One ; 6(5): e20339, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21629667

RESUMO

We previously reported that fetal porcine skin-derived stem cells were capable of differentiation into oocyte-like cells (OLCs). Here we report that newborn mice skin-derived stem cells are also capable of differentiating into early OLCs. Using stem cells from mice that are transgenic for Oct4 germline distal enhancer-GFP, germ cells resulting from their differentiation are expected to be GFP(+). After differentiation, some GFP(+) OLCs reached 40-45 µM and expressed oocyte markers. Flow cytometric analysis revealed that ∼ 0.3% of the freshly isolated skin cells were GFP(+). The GFP-positive cells increased to ∼ 7% after differentiation, suggesting that the GFP(+) cells could be of in vivo origin, but are more likely induced upon being cultured in vitro. To study the in vivo germ cell potential of skin-derived cells, they were aggregated with newborn ovarian cells, and transplanted under the kidney capsule of ovariectomized mice. GFP(+) oocytes were identified within a subpopulation of follicles in the resulting growth. Our finding that early oocytes can be differentiated from mice skin-derived cells in defined medium may offer a new in vitro model to study germ cell formation and oogenesis.


Assuntos
Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Feminino , Citometria de Fluxo , Imunofluorescência , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Oogênese/genética , Oogênese/fisiologia , Ovário/citologia , Ovário/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/citologia , Pele/metabolismo , Células-Tronco/metabolismo
13.
Invest Ophthalmol Vis Sci ; 52(6): 3539-47, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21273537

RESUMO

PURPOSE: Oculodentodigital dysplasia (ODDD) is a human disorder caused by mutations in the gap junction alpha 1 (GJA1) gene encoding the connexin43 (Cx43) gap junction protein. Causal links between GJA1 mutations and glaucoma are not understood. The purpose in this study was to examine the ocular phenotype for Gja1(Jrt/+) mice harboring a Cx43 G60S mutation. METHODS; In young Gja1(Jrt/+) mice, Cx43 abundance was assessed with a Western blot, and Cx43 localization was visualized using immunohistochemistry and confocal microscopy. Intraocular pressure (IOP) was measured by rebound tonometry, and eye anatomy was imaged using ocular coherence tomography (OCT). Hematoxylin and eosin (H&E)-stained eye sections were examined for ocular histopathology related to the development of glaucoma. RESULTS: Decreased Cx43 protein levels were evident in whole eyes from Gja1(Jrt/+) mice compared with those of wild-type mice at postnatal day 1 (P = 0.005). Cx43 immunofluorescence in ciliary bodies of Gja1(Jrt/+) mice was diffuse and intracellular, unlike the gap junction plaques prevalent in wild-type mice. IOP in Gja1(Jrt/+) mice changed during postnatal development, with significantly lower IOP at 21 weeks of age in comparison to the IOP of wild-type eyes. Microphthalmia, enophthalmia, anterior angle closure, and reduced pupil diameter were observed in Gja1(Jrt/+) mice at all ages examined. Ocular histology showed prominent separations between the pigmented and nonpigmented ciliary epithelium of Gja1(Jrt/+) mice, split irides, and alterations in the number and distribution of nuclei in the retina. CONCLUSIONS: Detailed phenotyping of Gja1(Jrt/+) eyes offers a framework for elucidating human ODDD ocular disease mechanisms and evaluating new treatments designed to protect ocular synaptic network integrity.


Assuntos
Conexina 43/genética , Esmalte Dentário/anormalidades , Modelos Animais de Doenças , Anormalidades do Olho/genética , Glaucoma/diagnóstico , Sindactilia/genética , Animais , Segmento Anterior do Olho/patologia , Western Blotting , Corpo Ciliar/metabolismo , Esmalte Dentário/patologia , Anormalidades do Olho/diagnóstico , Glaucoma/genética , Imuno-Histoquímica , Pressão Intraocular , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Microscopia Confocal , Mutação , Fenótipo , Sindactilia/diagnóstico , Tomografia de Coerência Óptica
14.
Stem Cells Dev ; 20(5): 809-19, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21054136

RESUMO

We previously reported the differentiation of cells derived from porcine female fetal skin into cells resembling germ cells and oocytes. A subpopulation of these cells expressed germ cell markers and formed aggregates resembling cumulus-oocyte complexes. Some of these aggregates extruded large oocyte-like cells (OLCs) that expressed markers consistent with those of oocytes. The objective of the current study was to further characterize OLCs differentiated from porcine skin-derived stem cells. Reverse transcriptase (RT)-polymerase chain reaction and Western blot revealed the expression of connexin37 and connexin43, both of which are characteristic of ovarian follicles. The expression of meiosis markers DMC1 and synaptonemal complex protein, but not STRA8 and REC8, was detected in the OLC cultures. Immunofluorescence with an antibody against synaptonemal complex protein on chromosome spreads revealed a very small subpopulation of stained OLCs that had a similar pattern to leptotene, zytotene, or pachytene nuclei during prophase I of meiosis. Sodium bisulfite sequencing of the differentially methylated region of H19 indicated that this region is almost completely demethylated in OLCs, similar to in vivo-derived oocytes. We also investigated the differentiation potential of male skin-derived stem cells in the same differentiation medium. Large cells with oocyte morphology were generated in the male stem cell differentiation cultures. These OLCs expressed oocyte genes such as octamer-binding transcription factor 4 (OCT4), growth differentiation factor-9b (GDF9B), deleted in azoospermia-like (DAZL), VASA, zona pellucida B (ZPB), and zona pellucida C (ZPC). It was concluded that skin-derived stem cells from both male and female porcine fetuses are capable of entering an oocyte differentiation pathway, but the culture system currently in place is inadequate to support the complete development of competent oocytes.


Assuntos
Biomarcadores/análise , Conexinas/metabolismo , Células-Tronco Fetais/metabolismo , Oócitos/metabolismo , Pele , Animais , Western Blotting , Diferenciação Celular , Células Cultivadas , Conexinas/genética , Metilação de DNA , Feminino , Células-Tronco Fetais/citologia , Feto , Imunofluorescência , Expressão Gênica , Humanos , Masculino , Oócitos/citologia , Oogênese/genética , Pele/citologia , Pele/metabolismo , Sus scrofa
15.
Can J Physiol Pharmacol ; 88(4): 399-413, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20555408

RESUMO

Female fertility is determined to a large extent by the quality (developmental competence) of the oocyte as reflected in its ability to undergo meiosis, be fertilized, and give rise to a healthy embryo. Growth of the mammalian oocyte is coordinated with that of the follicle that encloses it by the actions of signals that pass in both directions between the germline and somatic components. This review summarizes what is known about the roles played by 2 different modes of intrafollicular signalling in oogenesis: paracrine factors activating receptors on the opposite cell type, and direct sharing of small molecules throughout the follicle via gap junction channels. Recent evidence indicates that these 2 modes of signalling interact to regulate oocyte growth and granulosa cell proliferation and that defects in either can contribute to female infertility.


Assuntos
Comunicação Celular/fisiologia , Células da Granulosa/fisiologia , Oócitos/fisiologia , Oogênese/fisiologia , Folículo Ovariano/fisiologia , Animais , Proliferação de Células , Feminino , Fertilidade/fisiologia , Junções Comunicantes/fisiologia , Humanos , Masculino , Comunicação Parácrina , Transdução de Sinais
16.
Biol Reprod ; 82(5): 865-75, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20107203

RESUMO

WNTs are secreted extracellular signaling molecules that transduce their signals by binding to G protein-coupled receptors of the frizzled (FZD) family. They control diverse developmental processes, such as cell fate specification, cell proliferation, cell differentiation, and apoptosis. Although WNT signaling has been shown to be essential for development of the ovary, its mechanistic role in folliculogenesis within the adult ovary has not been studied extensively. Therefore, the objective of this study was to investigate the regulation and function of WNT2 signaling in mouse granulosa cells. Immunostaining identified WNT2 as being expressed in granulosa cells throughout folliculogenesis, but with varying signal strength: in sequential sections, WNT2 immunoreactivity was strongest in healthy antral follicles but weak in atretic follicles. Knockdown of WNT2 expression using transfected short interfering RNA decreased DNA synthesis in granulosa cells, whereas WNT2 overexpression using a recombinant viral vector enhanced it. WNT2 knockdown led to accumulation of glycogen synthase kinase-3beta (GSK3B) in the cytoplasm but reduced the expression of beta-catenin. Conversely, WNT2 overexpression reduced the expression of GSK3B in the cytoplasm and induced beta-catenin translocation from the membrane into the nucleus. Beta-catenin knockdown also inhibited DNA synthesis in granulosa cells and neutralized the effect of WNT2 overexpression. WNT2/beta-catenin signaling had a slight effect on the apoptosis of granulosa cells. Taken together, the data indicate that WNT2 regulates beta-catenin localization in granulosa cells, and WNT2/beta-catenin signaling contributes to regulating their proliferation.


Assuntos
DNA/biossíntese , Células da Granulosa/metabolismo , Transdução de Sinais/fisiologia , Proteína Wnt2/fisiologia , beta Catenina/metabolismo , Animais , Proliferação de Células , Feminino , Técnicas de Silenciamento de Genes , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Camundongos , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/metabolismo , Sistemas do Segundo Mensageiro/fisiologia
17.
Am J Physiol Heart Circ Physiol ; 297(1): H93-H101, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19429814

RESUMO

We have previously shown that increased nitric oxide (NO) production in sepsis impairs arteriolar-conducted vasoconstriction cGMP independently and that the gap junction protein connexin (Cx) 37 is required for this conducted response. In the present study, we hypothesized that NO impairs interendothelial electrical coupling in sepsis by targeting Cx37. We examined the effect of exogenous NO on coupling in monolayers of cultured microvascular endothelial cells derived from the hindlimb skeletal muscle of wild-type (WT), Cx37 null, Cx40 null, and Cx43(G60S) (nonfunctional mutant) mice. To assess coupling, we measured the spread of electrical current injected in the monolayer and calculated the monolayer intercellular resistance (inverse measure of coupling). The NO donor 2,2'-(hydroxynitrosohydrazino)bis-ethanamine (DETA) rapidly and reversibly reduced coupling in cells from WT mice, cGMP independently. NO scavenger HbO(2) did not affect baseline coupling, but it eliminated DETA-induced reduction in coupling. Reduced coupling in response to DETA was also seen in cells from Cx40 null and Cx43(G60S) mice, but not in cells from Cx37 null mice. DETA did not alter the expression of Cx37, Cx40, and Cx43 in WT cells analyzed by immunoblotting and immunofluorescence. Furthermore, neither the peroxynitrite scavenger 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron (III), superoxide scavenger Mn(III)tetrakis(4-benzoic acid)porphyrin chloride, nor preloading of WT cells with the antioxidant ascorbate affected this reduction. We conclude that NO-induced reduction of electrical coupling between microvascular endothelial cells depends on Cx37 and propose that NO in sepsis impairs arteriolar-conducted vasoconstriction by targeting Cx37 within the arteriolar wall.


Assuntos
Conexinas/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Óxido Nítrico/farmacologia , Animais , Western Blotting , Capilares/citologia , Capilares/fisiologia , Separação Celular , Conexina 43/genética , Conexina 43/fisiologia , Conexinas/genética , Eletrofisiologia , Endotélio Vascular/citologia , Imunofluorescência , Indicadores e Reagentes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doadores de Óxido Nítrico/farmacologia , Nitritos/farmacologia , Ácido Peroxinitroso/metabolismo , Proteína alfa-5 de Junções Comunicantes , Proteína alfa-4 de Junções Comunicantes
18.
Dis Model Mech ; 2(3-4): 157-67, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19259389

RESUMO

The essential role of connexin43 (Cx43) during oogenesis has been demonstrated by the severe germ cell deficiency and arrested folliculogenesis observed in Cx43 knockout mice. Recently, another mutant mouse strain became available (Gja1(Jrt)/+) that carries the dominant loss-of-function Cx43 mutation, Cx43(G60S). Gja1(Jrt)/+ mice display features of the human disease oculodentodigital dysplasia (ODDD), which is caused by mutations in the GJA1 gene. We used this new mutant strain to study how a disease-linked Cx43 mutant affects oogenesis. We found that female mutant mice are subfertile with significantly reduced mating success and small litters. The phosphorylated species of the Cx43 protein are reduced in the mutant ovaries in association with impaired trafficking and assembly of gap junctions in the membranes of granulosa cells, confirming that the mutant protein acts dominantly on its wild-type counterpart. Correspondingly, although starting with a normal abundance of germ cells, ovaries of the mutant mice contain significantly fewer pre-ovulatory follicles and do not respond to superovulation by gonadotropins, which is at least partially the result of reduced proliferation and increased apoptosis of granulosa cells. We conclude that the Gja1(Jrt) mutation has a dominant negative effect on Cx43 function in the ovary, rendering the females subfertile. Given these findings, closer examination of reproductive function in ODDD human females is warranted.


Assuntos
Anormalidades Múltiplas/genética , Conexina 43/genética , Modelos Animais de Doenças , Anormalidades do Olho/genética , Deformidades Congênitas dos Membros/genética , Oogênese/genética , Anormalidades Dentárias/genética , Animais , Apoptose , Conexina 43/fisiologia , Feminino , Junções Comunicantes/metabolismo , Células da Granulosa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação
19.
Biol Reprod ; 80(6): 1099-106, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19176884

RESUMO

Expression of GJA1 (commonly known as connexin43 or Cx43), a major myometrial gap junction protein, is upregulated before the onset of delivery, suggesting an essential role for Cx43-mediated gap junctional intercellular communication (GJIC) in normal uterine contraction during parturition. To determine how a disease-linked Cx43 mutation affects myometrial function, we studied a mutant mouse model carrying an autosomal dominant mutation (Gja1(Jrt)) in the gene encoding Cx43 that displays features of the human genetic disease oculodentodigital dysplasia. We found that Cx43 level, specifically the phosphorylated species of the protein, is significantly reduced in the myometrium of the mutant mice (Gja1(Jrt)/+), as revealed by Western blotting and immunostaining. Patch-clamp electrophysiological measurements demonstrated that coupling between myometrial smooth muscle cells is reduced to <15% of wild-type, indicating that the mutant protein acts dominantly on its wild-type counterpart. The phosphorylated species of Cx43 in the mutant myometrium failed to increase prior to parturition as well as in response to exogenous estrogen. Correspondingly, in vitro experiments with uterine strips revealed weaker contraction of the mutant myometrium and reduced responsiveness to oxytocin, providing an explanation for the prolonged gestation and presence of suffocated fetuses in the uteri that were observed in some of the mutant mice. We conclude that the Gja1(Jrt) mutation has a dominant-negative effect on Cx43 function in the myometrium, severely reducing GJIC, leading to impaired parturition.


Assuntos
Conexina 43/genética , Junções Comunicantes/fisiologia , Miócitos de Músculo Liso/fisiologia , Parto , Contração Uterina , Animais , Conexina 43/metabolismo , Estrogênios/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Miométrio/metabolismo , Gravidez , Prenhez
20.
Mol Hum Reprod ; 15(1): 11-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19038973

RESUMO

Signaling via the conserved WNT/beta-CATENIN pathway controls diverse developmental processes. To explore its potential role in the ovary, we investigated the expression of WNTs, frizzled (FZD) receptors and other pathway components in human cumulus cells obtained from oocytes collected for in vitro fertilization. Proteins were detected in cultured cells using immunofluorescence microscopy. Protein-protein interactions were analyzed by means of immunoprecipitation. WNT2, FZD2, FZD3 and FZD9 were identified but WNT1, WNT4 and FZD4 were not detected. WNT2 is co-expressed with FZD2, FZD3 and FZD9. Co-immunoprecipitation using WNT2 antibody demonstrated that WNT2 interacts with both FZD3 and FZD9, but only FZD9 antibody precipitated WNT2. We also identified DVL (disheveled), AXIN, GSK-3beta (glycogen synthase kinase-3beta) and beta-CATENIN. beta-CATENIN is concentrated in the plasma membranes. DVL co-localizes with FZD9 and AXIN in the membranes, but GSK-3beta has little co-localization with AXIN and beta-CATENIN. Interestingly, beta-CATENIN is highly co-localized with FZD9 and AXIN. CDH1 (E-cadherin) was also detected in the plasma membranes and cytoplasm, co-localized with beta-CATENIN, and CDH1 antibody precipitated beta-CATENIN. The results suggest that WNT2 could act through its receptor FZD9 to regulate the beta-CATENIN pathway in cumulus cells, recruiting beta-CATENIN into plasma membranes and promoting the formation of adherens junctions involving CDH1.


Assuntos
Células do Cúmulo/metabolismo , Transdução de Sinais/fisiologia , Proteína Wnt2/metabolismo , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Axina , Membrana Celular/metabolismo , Células Cultivadas , Proteínas Desgrenhadas , Feminino , Receptores Frizzled/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Immunoblotting , Imunoprecipitação , Microscopia de Fluorescência , Fosfoproteínas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...