Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Biol Reprod ; 103(4): 750-759, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32667624

RESUMO

Uterine smooth muscle cells differentiate from mesenchymal cells, and gap junctions connect the muscle cells in the myometrium. At the neonatal stage, a uterine smooth muscle layer is situated away from the epithelium when smooth muscle cells are grafted near the epithelium, suggesting that the epithelium plays an important role in differentiation, proliferation, and/or migration of smooth muscle cells. In this study, developmental mechanisms regulating the formation of the smooth muscle layers in the mouse uterus were analyzed using an in vitro culture model. Differentiation of smooth muscle cells occurs at a neonatal stage because ACTA2 gene expression was increased at the outer layer, and GJA1 was not expressed in cellular membranes of uterine smooth muscle cells by postnatal day 15. To analyze the effects of the epithelium on the differentiation of smooth muscle cells, a bulk uterine mesenchymal cell line was established from p53-/- mice at postnatal day 3 (P3US cells). Co-culture with Müllerian ductal epithelial cells (E1 cells) induced repulsive migration of ACTA2-positive cells among bulk P3US cells from E1 cells, but it had no effects on the migration of any of 100% ACTA2-positive or negative smooth muscle cell lines cloned from P3US cells. Thus, uterine epithelial cells indirectly affected the repulsive migration of smooth muscle cells via mesenchymal cells. Conditioned medium by E1 cells inhibited differentiation into smooth muscle cells of clonal cells established from P3US cells. Therefore, the uterine epithelium inhibits the differentiation of stem-like progenitor mesenchymal cells adjacent to the epithelium into smooth muscle cells.


Assuntos
Células Epiteliais/fisiologia , Células-Tronco Mesenquimais/fisiologia , Ductos Paramesonéfricos/citologia , Músculo Liso/crescimento & desenvolvimento , Útero/crescimento & desenvolvimento , Actinas/genética , Actinas/metabolismo , Animais , Anticorpos , Diferenciação Celular , Movimento Celular , Técnicas de Cocultura , Feminino , Regulação da Expressão Gênica , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Knockout , Proteína Supressora de Tumor p53/genética , Vimentina/genética , Vimentina/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
2.
Nat Commun ; 10(1): 5378, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31772170

RESUMO

Wnt signaling is critical for directing epithelial gland development within the uterine lining to ensure successful gestation in adults. Wnt-dependent, Lgr5-expressing stem/progenitor cells are essential for the development of glandular epithelia in the intestine and stomach, but their existence in the developing reproductive tract has not been investigated. Here, we employ Lgr5-2A-EGFP/CreERT2/DTR mouse models to identify Lgr5-expressing cells in the developing uterus and to evaluate their stem cell identity and function. Lgr5 is broadly expressed in the uterine epithelium during embryogenesis, but becomes largely restricted to the tips of developing glands after birth. In-vivo lineage tracing/ablation/organoid culture assays identify these gland-resident Lgr5high cells as Wnt-dependent stem cells responsible for uterine gland development. Adjacent Lgr5neg epithelial cells within the neonatal glands function as essential niche components to support the function of Lgr5high stem cells ex-vivo. These findings constitute a major advance in our understanding of uterine development and lay the foundations for investigating potential contributions of Lgr5+ stem/progenitor cells to uterine disorders.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco , Útero/crescimento & desenvolvimento , Proteínas Wnt/metabolismo , Animais , Linhagem da Célula , Endométrio/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Transgênicos , Ductos Paramesonéfricos/citologia , Organoides , Gravidez , Receptores Acoplados a Proteínas G/genética , Células-Tronco/citologia , Células-Tronco/fisiologia , Proteínas Wnt/genética
3.
Development ; 146(20)2019 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-31575647

RESUMO

WNT signaling plays essential roles in the development and function of the female reproductive tract. Although crosstalk with the Hippo pathway is a key regulator of WNT signaling, whether Hippo itself plays a role in female reproductive biology remains largely unknown. Here, we show that conditional deletion of the key Hippo kinases Lats1 and Lats2 in mouse Müllerian duct mesenchyme cells caused them to adopt the myofibroblast cell fate, resulting in profound reproductive tract developmental defects and sterility. Myofibroblast differentiation was attributed to increased YAP and TAZ expression (but not to altered WNT signaling), leading to the direct transcriptional upregulation of Ctgf and the activation of the myofibroblast genetic program. Müllerian duct mesenchyme cells also became myofibroblasts in male mutant embryos, which impeded the development of the male reproductive tract and resulted in cryptorchidism. The inactivation of Lats1/2 in differentiated uterine stromal cells in vitro did not compromise their ability to decidualize, suggesting that Hippo is dispensable during implantation. We conclude that Hippo signaling is required to suppress the myofibroblast genetic program and maintain multipotency in Müllerian mesenchyme cells.


Assuntos
Ductos Paramesonéfricos/citologia , Ductos Paramesonéfricos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Imunoprecipitação da Cromatina , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Endométrio/citologia , Endométrio/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Supressoras de Tumor/genética
4.
Stem Cell Reports ; 11(5): 1136-1155, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30392973

RESUMO

Defective endometrial stromal fibroblasts (EMSFs) contribute to uterine factor infertility, endometriosis, and endometrial cancer. Induced pluripotent stem cells (iPSCs) derived from skin or bone marrow biopsies provide a patient-specific source that can be differentiated to various cells types. Replacement of abnormal EMSFs is a potential novel therapeutic approach for endometrial disease; however, the methodology or mechanism for differentiating iPSCs to EMSFs is unknown. The uterus differentiates from the intermediate mesoderm (IM) to form coelomic epithelium (CE) followed by the Müllerian duct (MD). Here, we successfully directed the differentiation of human iPSCs (hiPSCs) through IM, CE, and MD to EMSFs under molecularly defined embryoid body culture conditions using specific hormonal treatments. Activation of CTNNB1 was essential for expression of progesterone receptor that mediated the final differentiation step of EMSFs before implantation. These hiPSC-derived tissues illustrate the potential for iPSC-based endometrial regeneration for future cell-based treatments.


Assuntos
Endométrio/citologia , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Progesterona/farmacologia , Via de Sinalização Wnt , beta Catenina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Decídua/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mesoderma/citologia , Ductos Paramesonéfricos/citologia , Linha Primitiva/citologia , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Transcriptoma/genética , Via de Sinalização Wnt/efeitos dos fármacos
5.
Wiley Interdiscip Rev Dev Biol ; 7(3): e310, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29350886

RESUMO

The Müllerian ducts are part of the embryonic urogenital system. They give rise to mature structures that serve a critical function in the transport and development of the oocyte and/or embryo. In most vertebrates, both sexes initially develop Müllerian ducts during embryogenesis, but they regress in males under the influence of testis-derived Anti-Müllerian Hormone (AMH). A number of regulatory factors have been shown to be essential for proper duct development, including Bmp and Wnt signaling molecules, together with homeodomain transcription factors such as PAX2 and LIM1. Later in development, the fate of the ducts diverges between males and females and is regulated by AMH and Wnt signaling molecules (duct regression in males) and Hox genes (duct patterning in females). Most of the genes and molecular pathways known to be involved in Müllerian duct development have been elucidated through animal models, namely, the mouse and chicken. In addition, genetic analysis of humans with reproductive tract disorders has further defined molecular mechanisms of duct formation and differentiation. However, despite our current understanding of Müllerian duct development, some questions remain to be answered at the molecular genetic level. This article is categorized under: Early Embryonic Development > Development to the Basic Body Plan.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Ductos Paramesonéfricos/embriologia , Diferenciação Sexual , Animais , Linhagem da Célula , Feminino , Humanos , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Ductos Paramesonéfricos/citologia , Ductos Paramesonéfricos/metabolismo , Via de Sinalização Wnt
6.
Reproduction ; 153(4): 481-492, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28123059

RESUMO

The influence of the hedgehog signaling pathway on reproduction was studied in transgenic mice in which a dominant active allele of the hedgehog signal transducer, smoothened (Smo), was conditionally expressed in the developing Müllerian duct and gonads through recombination mediated by anti-Müllerian hormone receptor 2-cre (Amhr2cre ). Previous studies showed that development of the oviduct and uterus are abnormal in female Amhr2cre/+SmoM2 mice. In the current study, focusing on mutant males, litter size was reduced 53% in crosses with wild-type females. An extra band of undifferentiated tissue extended along each epididymis and vas deferens, a position suggesting derivation from Müllerian ducts that failed to regress fully. Hedgehog signaling was elevated in this tissue, based on mRNA levels of target genes. Amhr2 mRNA was dramatically reduced in the uterus of mutant females and in the extra tissue in the tract of mutant males, suggesting that AMHR2 signaling was inadequate for complete Müllerian duct regression. Spermatogenesis and sperm motility were normal, but testis weight was reduced 37% and epididymal sperm number was reduced 36%. The number of sperm recovered from the uteri of wild-type females after mating with mutant males was reduced 78%. This suggested that sperm transport through the male tract was reduced, resulting in fewer sperm in the ejaculate. Consistent with this, mutant males had unusually tortuous vas deferentia with constrictions within the lumen. We concluded that persistence of a relatively undifferentiated remnant of Müllerian tissue is sufficient to cause subtle changes in the male reproductive tract that reduce fertility.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Infertilidade/patologia , Ductos Paramesonéfricos/metabolismo , Receptores de Peptídeos/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Receptor Smoothened/fisiologia , Animais , Epididimo/citologia , Epididimo/metabolismo , Feminino , Infertilidade/etiologia , Infertilidade/metabolismo , Integrases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ductos Paramesonéfricos/citologia , Reprodução/fisiologia , Túbulos Seminíferos/citologia , Túbulos Seminíferos/metabolismo , Transdução de Sinais , Espermatogênese
7.
Proc Natl Acad Sci U S A ; 113(50): 14354-14359, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911779

RESUMO

The Müllerian duct develops into the oviduct, uterus, and vagina, all of which are quite distinct in their morphology and function. The epithelial fate of these female reproductive organs in developing mice is determined by factors secreted from the stroma; however, how stromal differentiation occurs in the female reproductive organs derived from the Müllerian duct is still unclear. In the present study, roles of retinoic acid (RA) signaling in developing female reproductive tracts were investigated. Retinol dehydrogenase 10 (RDH10) and aldehyde dehydrogenase family 1 subfamily A2 (ALDH1A2) mRNAs and proteins and transactivation activity of endogenous RA were found in the stroma of proximal Müllerian ducts and gradually decreased from the proximal to caudal regions in fetal mice. In organ-cultured Müllerian ducts, retinaldehyde or RA treatment induced uterine epithelial differentiation, defined as a layer of columnar epithelial cells negative for oviductal and vaginal epithelial markers. In contrast, inhibition of RA receptor (RAR) signaling induced vaginal epithelial differentiation, characterized as vaginal epithelial marker genes-positive stratified epithelium. Grafting experiments of the organ-cultured Müllerian duct revealed irreversible epithelial fate determination. Although RAR did not directly bind to the homeobox A10 (Hoxa10) promoter region, RA-RAR signaling stimulated Hoxa10 expression. Thus, RA-RAR signaling in the Müllerian duct determines the fate of stroma to form the future uterus and vagina.


Assuntos
Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/metabolismo , Tretinoína/metabolismo , Útero/embriologia , Útero/metabolismo , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ductos Paramesonéfricos/citologia , Técnicas de Cultura de Órgãos , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Ácido Retinoico/antagonistas & inibidores , Retinal Desidrogenase , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional , Útero/citologia , Vagina/citologia , Vagina/embriologia , Vagina/metabolismo
8.
Stem Cells Dev ; 25(22): 1733-1741, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27595922

RESUMO

A fundamental understanding of prostate development and tissue homeostasis has the high potential to reveal mechanisms for prostate disease initiation and identify novel therapeutic approaches for disease prevention and treatment. Our current understanding of prostate lineage specification stems from the use of developmental model systems that rely upon the embryonic preprostatic urogenital sinus mesenchyme to induce the formation of mature prostate epithelial cells. It is unclear, however, how the urogenital sinus epithelium can derive both adult urethral glands and prostate epithelia. Furthermore, the vast disparity in disease initiation between these two glands highlights key developmental factors that predispose prostate epithelia to hyperplasia and cancer. In this study we demonstrate that the caudal Müllerian duct mesenchyme (CMDM) drives prostate epithelial differentiation and is a key determinant in cell lineage specification between urethral glands and prostate epithelia. Utilizing both human embryonic stem cells and mouse embryonic tissues, we document that the CMDM is capable of inducing the specification of androgen receptor, prostate-specific antigen, NKX3.1, and Hoxb13-positive prostate epithelial cells. These results help to explain key developmental differences between prostate and urethral gland differentiation, and implicate factors secreted by the caudal Müllerian duct as novel targets for prostate disease prevention and treatment.


Assuntos
Mesoderma/embriologia , Ductos Paramesonéfricos/embriologia , Organogênese , Próstata/embriologia , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Epitélio , Células-Tronco Embrionárias Humanas/citologia , Humanos , Masculino , Mesoderma/citologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Ductos Paramesonéfricos/citologia , Próstata/citologia , Fatores de Transcrição/metabolismo , Uretra/citologia
9.
Development ; 143(19): 3549-3559, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27578782

RESUMO

The Müllerian duct (MD) and Wolffian duct (WD) are embryonic tubular tissues giving rise to female and male reproductive tracts, respectively. In amniote embryos, both MD and WD emerge in both sexes, but subsequently degenerate in the males and females, respectively. Here, by using MD-specific gene manipulations in chicken embryos, we identify the molecular and cellular mechanisms that link early MD specification to tubular invagination. Early (pre-)specification of MD precursors in the coelomic epithelium requires BMP signaling and its downstream target Pax2 in a WD-independent process. Subsequently, the BMP/Pax2 axis induces Lim1 expression, a hallmark of MD specification, for which FGF/ERK and WD-derived signals are also required. Finally, the sequential actions of the BMP/Pax2 and FGF/Lim1 axes culminate in epithelial invagination to form a tubular structure driven by an apical constriction, where apical accumulation of phospho-myosin light chain is positively regulated by FGF/ERK signaling. Our study delineates mechanisms governing the early formation of the MD, and also serves as a model of how an epithelial cell sheet is transformed to a tubular structure, a process seen in a variety of developmental contexts.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Ductos Paramesonéfricos/metabolismo , Fator de Transcrição PAX2/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Embrião de Galinha , Eletroporação , Feminino , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Masculino , Ductos Paramesonéfricos/citologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Ductos Mesonéfricos/citologia , Ductos Mesonéfricos/metabolismo
10.
Biotech Histochem ; 91(6): 428-37, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27437606

RESUMO

The sex of neonatal sea turtles is difficult to determine, because neonates lack heteromorphic sex chromosomes and dimorphic external characteristics; internal dimorphic morphology is defined at hatching. We used histochemical staining and made measurements in the gonads and paramesonephric ducts (PD) of both sexes to determine structural differences in female and male loggerhead sea turtle (Caretta caretta) hatchlings. We detected differences in the gonads and PD between the sexes including the amounts of mucopolysaccharides, collagen and elastic fibers. We determined that the thickness of the gonadal cortex and the diameter of the PD lumen are reliable sex-specific characteristics. We also assessed immunolocalization of aromatase, an enzyme complex that converts androgens to estrogens, and found differences in the localization and intensity of aromatase immunostaining in the gonads and PD of female and male hatchlings. Comprehensive studies of the sexual differences of sea turtles are important for conservation programs.


Assuntos
Gônadas/citologia , Imuno-Histoquímica , Mesonefro/citologia , Ductos Paramesonéfricos/citologia , Análise para Determinação do Sexo/métodos , Tartarugas/fisiologia , Animais , Aromatase/química , Feminino , Gônadas/enzimologia , Masculino , Caracteres Sexuais
11.
Mol Endocrinol ; 30(7): 783-95, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27164167

RESUMO

Cell fate of lower Müllerian duct epithelium (MDE), to become uterine or vaginal epithelium, is determined by the absence or presence of ΔNp63 expression, respectively. Previously, we showed that SMAD4 and runt-related transcription factor 1 (RUNX1) were independently required for MDE to express ΔNp63. Here, we report that vaginal mesenchyme directs vaginal epithelial cell fate in MDE through paracrine activation of fibroblast growth factor (FGF) receptor-MAPK pathway. In the developing reproductive tract, FGF7 and FGF10 were enriched in vaginal mesenchyme, whereas FGF receptor 2IIIb was expressed in epithelia of both the uterus and vagina. When Fgfr2 was inactivated, vaginal MDE underwent uterine cell fate, and this differentiation defect was corrected by activation of MEK-ERK pathway. In vitro, FGF10 in combination with bone morphogenetic protein 4 and activin A (ActA) was sufficient to induce ΔNp63 in MDE, and ActA was essential for induction of RUNX1 through SMAD-independent pathways. Accordingly, inhibition of type 1 receptors for activin in neonatal mice induced uterine differentiation in vaginal epithelium by down-regulating RUNX1, whereas conditional deletion of Smad2 and Smad3 had no effect on vaginal epithelial differentiation. In conclusion, vaginal epithelial cell fate in MDE is induced by FGF7/10-MAPK, bone morphogenetic protein 4-SMAD, and ActA-RUNX1 pathway activities, and the disruption in any one of these pathways results in conversion from vaginal to uterine epithelial cell fate.


Assuntos
Células Epiteliais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ductos Paramesonéfricos/citologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Animais Recém-Nascidos , Benzodioxóis/farmacologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células Epiteliais/citologia , Feminino , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 7 de Crescimento de Fibroblastos/genética , Fator 7 de Crescimento de Fibroblastos/metabolismo , Imunofluorescência , Imidazóis/farmacologia , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/genética , Piridinas/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Útero/citologia , Vagina/citologia
12.
Hum Mol Genet ; 25(6): 1059-73, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26721931

RESUMO

The Müllerian duct (MD) is the anlage of the oviduct, uterus and upper part of the vagina, the main parts of the female reproductive tract. Several wingless-type mouse mammary tumor virus (MMTV) integration site family member (Wnt) genes, including Wnt4, Wnt5a and Wnt7a, are involved in the development of MD and its derivatives, with Wnt4 particularly critical, since the MD fails to develop in its absence. We use, here, Wnt4(EGFPCre)-based fate mapping to demonstrate that the MD tip cells and the subsequent MD cells are derived from Wnt4+ lineage cells. Moreover, Wnt4 is required for the initiation of MD-forming cell migration. Application of anti-Wnt4 function-blocking antibodies after the initiation of MD elongation indicated that Wnt4 is necessary for the elongation as well, and consistent with this, cell culture wound-healing assays with NIH3T3 cells overexpressing Wnt4 promoted cell migration by comparison with controls. In contrast to the Wnt4 null embryos, some Wnt4(monomeric cherry/monomeric cherry) (Wnt4(mCh/mCh)) hypomorphic mice survived to adulthood and formed MD in ∼45% of cases. Nevertheless, the MD of the Wnt4(mCh/mCh) females had altered cell polarization and basement membrane deposition relative to the controls. Examination of the reproductive tract of the Wnt4(mCh/mCh) females indicated a poorly coiled oviduct, absence of the endometrial glands and an undifferentiated myometrium, and these mice were prone to develop a hydro-uterus. In conclusion, the results suggest that the Wnt4 gene encodes signals that are important for various aspects of female reproductive tract development.


Assuntos
Ductos Paramesonéfricos/metabolismo , Proteína Wnt4/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Movimento Celular/genética , Feminino , Humanos , Camundongos , Camundongos Knockout , Ductos Paramesonéfricos/citologia , Células NIH 3T3 , Reprodução , Útero/metabolismo , Proteína Wnt4/genética
13.
Rev. iberoam. fertil. reprod. hum ; 32(2): 42-51, abr.-jun. 2015. tab, graf, ilus
Artigo em Espanhol | IBECS | ID: ibc-144119

RESUMO

OBJETIVO: Revisión bibliográfica sobre la implicación de las células madre en reproducción. Células madre y endometrio: La regeneración que en cada ciclo experimenta el endometrio se lleva a cabo a partir de las células madre acantonadas en la zona basal y en nidos vasculares. Estas proceden de células madre originarias acantonadas nichos perivasculares de la zona basal y posiblemente también a partir de células madre de medula ósea. Células madre y ovario: Existen datos que abogan por la existencia de ovulogénesis en los mamíferos adultos, incluida la mujer, a expensas de células madre. En algún trabajo se admite la posibilidad de lograr descendencia a partir ovocitos logrados con cultivos de células madre


OBJECTIVE: Literature review of stem cells implication in reproduction. RESULTS: Stem cells and endometrium: Cyclic endometrium regeneration originates from stem cells quartered in basal zone and vascular nodes and possibly from bone marrow stem cells. Stem cells and ovary: There are data advocating for the existence of ovulogenesis from stem cells in mammarian adults, including women. Some studies do even consider possibility of obtaining offspring from ovocytes coming from stem cells culture


Assuntos
Feminino , Humanos , Masculino , Células-Tronco/citologia , Células-Tronco/patologia , Reprodução/genética , Endométrio/anormalidades , Endométrio/citologia , Infarto do Miocárdio/diagnóstico , Doença de Parkinson/diagnóstico , Infertilidade/genética , Ductos Paramesonéfricos/citologia , Células Matadoras Naturais/patologia , Células-Tronco/classificação , Células-Tronco/fisiologia , Reprodução/fisiologia , Endométrio/metabolismo , Endométrio/patologia , Infarto do Miocárdio/metabolismo , Doença de Parkinson/complicações , Infertilidade/metabolismo , Ductos Paramesonéfricos/anormalidades , Células Matadoras Naturais/citologia
14.
Sex Dev ; 8(5): 281-96, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25033758

RESUMO

The Müllerian duct (MD) forms the female reproductive tract (FRT) consisting of the oviducts, uterus, cervix, and upper vagina. FRT function is vital to fertility, providing the site of fertilization, embryo implantation and fetal development. Developmental defects in the formation and diseases of the FRT, including cancer and endometriosis, are prevalent in humans and can result in infertility and death. Furthermore, because the MDs are initially formed regardless of genotypic sex, mesenchymal to epithelial signaling is required in males to mediate MD regression and prevents the development of MD-derived organs. In males, defects in MD regression result in the retention of FRT organs and have been described in several human syndromes. Although to date not reported in humans, ectopic activation of MD regression signaling components in females can result in aplasia of the FRT. Clearly, MD development is important to human health; however, the molecular mechanisms remain largely undetermined. Molecular genetics studies of human diseases and mouse models have provided new insights into molecular signaling during MD development, regression and differentiation. This review will provide an overview of MD development and important genes and signaling mechanisms involved.


Assuntos
Genitália Feminina/embriologia , Genitália Feminina/metabolismo , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/metabolismo , Animais , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genitália Feminina/citologia , Humanos , Masculino , Ductos Paramesonéfricos/citologia
15.
Poult Sci ; 93(4): 953-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24706973

RESUMO

Sex of birds is genetically determined by the inheritance of sex chromosomes (ZZ for male and ZW for female), and the Z-linked gene named doublesex and mab-3 related transcription factor 1 (DMRT1) is a candidate sex-determining gene in avian species. However, the mechanisms underlying sex determination in birds are not yet understood, and the expression patterns of the DMRT1 protein in urogenital tissues have not been identified. In the current study, we used immunohistochemistry to investigate the detailed expression patterns of the DMRT1 protein in the urogenital systems (including Müllerian ducts) in male and female chicken embryos throughout embryonic development. Gonadal somatic cells in the male indifferent gonads showed stronger expressions of DMRT1 compared with those in the female indifferent gonads well before the presumptive period of the sex determination, and Sertoli cells forming testicular cords expressed DMRT1 in the testes after sex determination. Germ cells expressed DMRT1 equally in males and females after sex determination. The expression was continuous in males, but in females it gradually disappeared from the germ cells in the central part of the cortex of the left ovary toward both edges. The DMRT1 was also detected in the tubal ridge, which is a precursor of the Müllerian duct, and at the mesenchyme and outermost coelomic epithelium of the Müllerian duct in both sexes. Strong expression was observed in the males, but it was restricted to coelomic epithelium after the regression of the duct started. Thus, we observed the detailed spatiotemporal expression patterns of DMRT1 in the developing chicken urogenital systems throughout embryonic development, suggesting its various roles in the development of urogenital tissues in the chicken embryo.


Assuntos
Embrião de Galinha/embriologia , Galinhas/genética , Ductos Paramesonéfricos/embriologia , Ovário/embriologia , Testículo/embriologia , Fatores de Transcrição/genética , Animais , Feminino , Imuno-Histoquímica/veterinária , Masculino , Ductos Paramesonéfricos/citologia , Ductos Paramesonéfricos/metabolismo , Ovário/citologia , Ovário/metabolismo , Processos de Determinação Sexual , Testículo/citologia , Testículo/metabolismo , Fatores de Transcrição/metabolismo
16.
Dev Dyn ; 243(8): 1037-45, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24753105

RESUMO

BACKGROUND: The noncanonical Wnt receptor and tyrosine kinase Ror2 has been associated with recessive Robinow syndrome (RRS) and dominant brachydactyly type B1. The phenotypes of mouse mutants implicate Ror2 in the development of the heart, lungs, bone, and craniofacial structures, which are affected in RRS. Following a recently identified role of Ror2 in the migration of mouse primordial germ cells, we extensively characterized its expression throughout the fetal internal reproductive system and the postnatal ductal system. RESULTS: We show that Ror2 gene products are present in the germ cells and somatic cells of the testis and the ovary of both the mouse and human fetus. In reproductive tract structures, we find that Ror2 is expressed in the mesonephros, developing Wolffian and Müllerian ducts, and later in their derivatives, the epididymal epithelium and uterine epithelium. CONCLUSIONS: This study sets the stage to explore function for this tyrosine kinase receptor in novel regions of expression in the developing reproductive system in both mouse and human.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteínas Wnt/metabolismo , Animais , Feminino , Células Germinativas/citologia , Células da Granulosa , Humanos , Masculino , Camundongos , Ductos Paramesonéfricos/citologia , Ovário/citologia , Proteínas Proto-Oncogênicas/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Células de Sertoli/citologia , Testículo/citologia , Proteínas Wnt/genética , Proteína Wnt-5a , Ductos Mesonéfricos/citologia
18.
Biochem Biophys Res Commun ; 414(1): 175-80, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21945936

RESUMO

Adenosine is an important modulator of neuronal survival and differentiation in the CNS. Our previous work showed that nucleoside transporters (NTs) are present in cultures of chick retinal cells, but little is known about the mechanisms regulating adenosine transport in these cultures. Our aim in the present work was to study the participation of the adenosine metabolism as well as the ERK pathway on adenosine uptake in different types of retinal cultures (mixed and purified glial cultures). Kinetic analysis in both cultures revealed that the uptake reached equilibrium after 30 min and presented two components. Incubation of cultures with S-(p-nitrobenzyl)-6-thioinosine (NBTI) or dipyridamole, different inhibitors of equilibrative nucleoside transporters (ENTs), produced a significant and concentration-dependent uptake reduction in both cultures. However, while dipyridamole presented similar maximal inhibitory effects in both cultures (although in different concentrations), the inhibition by NBTI was smaller in glial cultures than in mixed cultures, suggesting the presence of different transporters. Moreover, pre-incubation of [(3)H]-adenosine with adenosine deaminase (ADA) or adenosine kinase (ADK) inhibition with iodotubercidin promoted significant uptake inhibition in both cultures, indicating that the uptake is predominantly for adenosine and not inosine, and that taken up adenosine is preferentially directed to the synthesis of adenine nucleotides. In both cultures, the MEK inhibitors PD98059 or UO126, but not the inactive analog U0124, induced a significant and concentration-dependent uptake decrease. We have not observed any change in adenosine metabolism induced by MEK inhibitors, suggesting that this pathway is mediating a direct effect on NTs. Our results show the expression of different NTs in retinal cells in culture and that the activity of these transporters can be regulated by the ERK pathway or metabolic enzymes such as ADK which are then potential targets for regulation of Ado levels in normal or pathological conditions.


Assuntos
Adenosina/metabolismo , Proteínas de Transporte de Nucleosídeo Equilibrativas/metabolismo , Sistema de Sinalização das MAP Quinases , Neuroglia/metabolismo , Neurônios/metabolismo , Retina/metabolismo , Adenosina/antagonistas & inibidores , Animais , Butadienos/farmacologia , Células Cultivadas , Embrião de Galinha , Flavonoides/farmacologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Ductos Paramesonéfricos/citologia , Nitrilas/farmacologia , Inibidores de Proteínas Quinases/farmacologia
19.
Differentiation ; 82(3): 117-26, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21612855

RESUMO

In mammals, the female reproductive tract (FRT) develops from a pair of paramesonephric or Müllerian ducts (MDs), which arise from coelomic epithelial cells of mesodermal origin. During development, the MDs undergo a dynamic morphogenetic transformation from simple tubes consisting of homogeneous epithelium and surrounding mesenchyme into several distinct organs namely the oviduct, uterus, cervix and vagina. Following the formation of anatomically distinctive organs, the uniform MD epithelium (MDE) differentiates into diverse epithelial cell types with unique morphology and functions in each organ. Classic tissue recombination studies, in which the epithelium and mesenchyme isolated from the newborn mouse FRT were recombined, have established that the organ specific epithelial cell fate of MDE is dictated by the underlying mesenchyme. The tissue recombination studies have also demonstrated that there is a narrow developmental window for the epithelial cell fate determination in MD-derived organs. Accordingly, the developmental plasticity of epithelial cells is mostly lost in mature FRT. If the signaling that controls epithelial differentiation is disrupted at the critical developmental stage, the cell fate of MD-derived epithelial tissues will be permanently altered and can result in epithelial lesions in adult life. A disruption of signaling that maintains epithelial cell fate can also cause epithelial lesions in the FRT. In this review, the pathogenesis of cervical/vaginal adenoses and uterine squamous metaplasia is discussed as examples of such incidences.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Ductos Paramesonéfricos/citologia , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patologia , Animais , Colo do Útero/anormalidades , Colo do Útero/citologia , Colo do Útero/embriologia , Dietilestilbestrol/efeitos adversos , Células Epiteliais/metabolismo , Estrogênios não Esteroides/efeitos adversos , Feminino , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Metaplasia , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/metabolismo , Organogênese , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia , Útero/citologia , Útero/embriologia , Vagina/anormalidades , Vagina/citologia , Vagina/embriologia
20.
Reproduction ; 139(4): 749-57, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20089665

RESUMO

The somatic (Sertoli cell (SC), Leydig cell (LC), and peritubular myoid (PTM) cell) cells play key roles in development of the fetal testis. We established monolayer cultures from second trimester human testes and investigated the pattern of expression of cell-lineage characteristic mRNAs. Expression of some SC-associated genes (SRY, SOX9, WT1, GATA4, and SF1) was detectable up to and including passage 3 (P3), while others (anti-Müllerian hormone; desert hedgehog) present prior to dissociation were not expressed in the cultured cells. Transcripts encoding the androgen receptor were expressed but addition of dihydrotestosterone (DHT) had no impact on expression of mRNAs expressed in SC or LC. Total concentrations of mRNAs encoding smooth muscle actin (ACTA2) and desmin increased from P1 to P3; an increasing proportion of the cells in the cultures were immunopositive for ACTA2 consistent with proliferation/differentiation of PTM cells. In conclusion, somatic cell monolayer cultures were established from human fetal testes; these cultures could form the basis for future studies based on isolation of purified populations of somatic cells and manipulation of gene expression that is difficult to achieve with organ culture systems. Our results suggest that fetal SC do not maintain a fully differentiated phenotype in vitro, yet PTM (ACTA2 positive) cells readily adapt to monolayer culture conditions in the presence of DHT. This culture system provides an opportunity to study the impact of regulatory factors on gene expression in PTM cells, a population thought to play a key role in mediating androgen action within the developing testis.


Assuntos
Androgênios/farmacologia , Proliferação de Células/efeitos dos fármacos , Feto/citologia , Células de Sertoli/citologia , Células de Sertoli/fisiologia , Testículo/embriologia , Biomarcadores/análise , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Feminino , Feto/fisiologia , Humanos , Masculino , Ductos Paramesonéfricos/citologia , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/fisiologia , Especificidade de Órgãos , Gravidez , Segundo Trimestre da Gravidez , Testículo/citologia , Testículo/fisiologia , Estudos de Validação como Assunto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...