Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(21): 8190-5, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22566636

RESUMO

The xenoestrogen bisphenol A (BPA) used in the manufacturing of various plastics and resins for food packaging and consumer products has been shown to produce numerous endocrine and developmental effects in rodents. Exposure to low doses of BPA during fetal mammary gland development resulted in significant alterations in the gland's morphology that varied from subtle ones observed during the exposure period to precancerous and cancerous lesions manifested in adulthood. This study assessed the effects of BPA on fetal mammary gland development in nonhuman primates. Pregnant rhesus monkeys were fed 400 µg of BPA per kg of body weight daily from gestational day 100 to term, which resulted in 0.68 ± 0.312 ng of unconjugated BPA per mL of maternal serum, a level comparable to that found in humans. At birth, the mammary glands of female offspring were removed for morphological analysis. Morphological parameters similar to those shown to be affected in rodents exposed prenatally to BPA were measured in whole-mounted glands; estrogen receptor (ER) α and ß expression were assessed in paraffin sections. Student's t tests for equality of means were used to assess differences between exposed and unexposed groups. The density of mammary buds was significantly increased in BPA-exposed monkeys, and the overall development of their mammary gland was more advanced compared with unexposed monkeys. No significant differences were observed in ER expression. Altogether, gestational exposure to the estrogen-mimic BPA altered the developing mammary glands of female nonhuman primates in a comparable manner to that observed in rodents.


Assuntos
Poluentes Ocupacionais do Ar/toxicidade , Disruptores Endócrinos/toxicidade , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/efeitos dos fármacos , Fenóis/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Poluentes Ocupacionais do Ar/sangue , Animais , Compostos Benzidrílicos , Epitélio/efeitos dos fármacos , Epitélio/patologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Macaca mulatta , Glândulas Mamárias Animais/metabolismo , Ovário/anormalidades , Ovário/efeitos dos fármacos , Fenóis/sangue , Gravidez
2.
Anim Genet ; 45(6): 871-3, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25204440

RESUMO

Supernumerary teats represent a common abnormality of the bovine udder. A genome-wide association study was performed based on the proportion of the occurrence of supernumerary teats in the daughters of 1097 Holstein bulls. The heritability of caudal supernumerary teats without mammary gland in this study was 0.604. The largest proportion of the heritability was attributable to BTA 20. The strongest evidence for association was with five SNPs on chromosome 20, referred to as a QTL. The mode of inheritance at this QTL was dominant. These findings reveal that the occurrence of caudal supernumerary teats without mammary gland in Holstein cattle is influenced by a QTL on chromosome 20 and a polygenic part. The data support the high potential of the SNPs in the QTL region as markers for breeding against caudal supernumerary teats.


Assuntos
Doenças Mamárias/veterinária , Bovinos/genética , Glândulas Mamárias Animais/anormalidades , Mamilos/anormalidades , Locos de Características Quantitativas , Animais , Doenças Mamárias/genética , Cruzamento , Doenças dos Bovinos/genética , Feminino , Estudos de Associação Genética/veterinária , Padrões de Herança , Masculino , Polimorfismo de Nucleotídeo Único
3.
Biochem J ; 449(2): 401-13, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23075222

RESUMO

Genetically modified mice mimicking ODDD (oculodentodigital dysplasia), a disease characterized by reduced Cx43 (connexin 43)-mediated gap junctional intercellular communication, represent an in vivo model to assess the role of Cx43 in mammary gland development and function. We previously reported that severely compromised Cx43 function delayed mammary gland development and impaired milk ejection in mice that harboured a G60S Cx43 mutant, yet there are no reports of lactation defects in ODDD patients. To address this further, we obtained a second mouse model of ODDD expressing an I130T Cx43 mutant to assess whether a mutant with partial gap junction channel activity would be sufficient to retain mammary gland development and function. The results of the present study show that virgin Cx43I130T/+ mice exhibited a temporary delay in ductal elongation at 4 weeks. In addition, Cx43I130T/+ mice develop smaller mammary glands at parturition due to reduced cell proliferation despite similar overall gland architecture. Distinct from Cx43G60S/+ mice, Cx43I130T/+ mice adequately produce and deliver milk to pups, suggesting that milk ejection is unaffected. Thus the present study suggests that a loss-of-function mutant of Cx43 with partial gap junction channel coupling conductance results in a less severe mammary gland phenotype, which may partially explain the lack of reported lactation defects associated with ODDD patients.


Assuntos
Conexina 43/genética , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/metabolismo , Mutação Puntual , Animais , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Conexina 43/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Feminino , Deformidades Congênitas do Pé/genética , Deformidades Congênitas do Pé/metabolismo , Deformidades Congênitas do Pé/patologia , Junções Comunicantes/metabolismo , Junções Comunicantes/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lactação/efeitos dos fármacos , Lactação/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Ocitocina/farmacologia , Gravidez , Índice de Gravidade de Doença , Sindactilia/genética , Sindactilia/metabolismo , Sindactilia/patologia , Anormalidades Dentárias/genética , Anormalidades Dentárias/metabolismo , Anormalidades Dentárias/patologia
4.
Anim Genet ; 44(2): 139-48, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22690698

RESUMO

The inverted teat defect is the most common disorder of the mammary complex in pigs. It is characterized by the failure of teats to protrude from the udder surface, preventing normal milk flow and thus limiting the rearing capacity and increasing the risk of mastitis. The inverted teat defect is a liability trait with a complex mode of inheritance. We previously identified QTL for inverted teats. As a complementary approach that integrates map-based efforts to identify candidate genes for the inverted teat defect with function-driven expression analysis, application-specific microarrays were constructed that cover 1525 transcripts mapping in QTL regions on pig chromosomes 2, 3, 4, 6 and 11. About 950 transcripts were expressed in epithelial and mesenchymal teat tissue. The expression of three categories of teats was compared: normal teats of both non-affected and affected animals and inverted teats of affected animals. In epithelium and mesenchyme, 62 and 24 genes respectively were significantly differentially expressed (DE). The majority of biofunctions to which a significant number of DE genes were assigned are related to the following: (1) cell maintenance, proliferation, differentiation and replacement; (2) organismal, organ and tissue development; or (3) genetic information and nucleic acid processing. Moreover, the DE genes belong almost exclusively to canonical pathways related to signaling rather than metabolic pathways. This is in line with findings obtained by genome-wide catalogue microarrays. This study adds another piece to the puzzle of the etiology of inverted teats by indicating that causal genetic variation leading to the disorder is likely among the genes encoding for members of the signaling cascades of growth factors.


Assuntos
Glândulas Mamárias Animais/anormalidades , Locos de Características Quantitativas/genética , Transdução de Sinais/genética , Suínos/anormalidades , Suínos/genética , Animais , Mapeamento Cromossômico/veterinária , Primers do DNA/genética , Feminino , Perfilação da Expressão Gênica/veterinária , Estudos de Associação Genética/veterinária , Padrões de Herança/genética , Análise em Microsséries/veterinária , Reação em Cadeia da Polimerase em Tempo Real/veterinária
5.
Nat Genet ; 24(4): 391-5, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10742104

RESUMO

The composite structure of the mammalian skull, which forms predominantly via intramembranous ossification, requires precise pre- and post-natal growth regulation of individual calvarial elements. Disturbances of this process frequently cause severe clinical manifestations in humans. Enhanced DNA binding by a mutant MSX2 homeodomain results in a gain of function and produces craniosynostosis in humans. Here we show that Msx2-deficient mice have defects of skull ossification and persistent calvarial foramen. This phenotype results from defective proliferation of osteoprogenitors at the osteogenic front during calvarial morphogenesis, and closely resembles that associated with human MSX2 haploinsufficiency in parietal foramina (PFM). Msx2-/- mice also have defects in endochondral bone formation. In the axial and appendicular skeleton, post-natal deficits in Pth/Pthrp receptor (Pthr) signalling and in expression of marker genes for bone differentiation indicate that Msx2 is required for both chondrogenesis and osteogenesis. Consistent with phenotypes associated with PFM, Msx2-mutant mice also display defective tooth, hair follicle and mammary gland development, and seizures, the latter accompanied by abnormal development of the cerebellum. Most Msx2-mutant phenotypes, including calvarial defects, are enhanced by genetic combination with Msx1 loss of function, indicating that Msx gene dosage can modify expression of the PFM phenotype. Our results provide a developmental basis for PFM and demonstrate that Msx2 is essential at multiple sites during organogenesis.


Assuntos
Desenvolvimento Ósseo/genética , Osso e Ossos/anormalidades , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Displasia Ectodérmica/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Animais , Osso e Ossos/patologia , Cartilagem/anormalidades , Cartilagem/patologia , Diferenciação Celular , Divisão Celular , Cerebelo/anormalidades , Cerebelo/patologia , Condrócitos/citologia , Displasia Ectodérmica/patologia , Folículo Piloso/anormalidades , Folículo Piloso/patologia , Proteínas de Homeodomínio , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , Fenótipo , Convulsões/genética , Anormalidades Dentárias/genética , Anormalidades Dentárias/patologia
6.
Biochem Biophys Res Commun ; 424(4): 710-6, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22800760

RESUMO

Estrogen receptor alpha (ERα) is a nuclear receptor that regulates a range of physiological processes in response to estrogens. In order to study its biological role, we generated a floxed ERα mouse line that can be used to knock out ERα in selected tissues by using the Cre/LoxP system. In this study, we established a new ERα knockout mouse line by crossing the floxed ERα mice with Cre deleter mice. Here we show that genetic disruption of the ERα gene in all tissues results in sterility in both male and female mice. Histological examination of uterus and ovaries revealed a dramatically atrophic uterus and hemorrhagic cysts in the ovary. These results suggest that infertility in female mice is the result of functional defects of the reproductive tract. Moreover, female knockout mice are hyperglycemic, develop obesity and at the age of 4 months the body weight of these mice was more than 20% higher compared to wild type littermates and this difference increased over time. Our results demonstrate that ERα is necessary for reproductive tract development and has important functions as a regulator of metabolism in females.


Assuntos
Receptor alfa de Estrogênio/genética , Infertilidade Feminina/genética , Infertilidade Masculina/genética , Animais , Peso Corporal/genética , Corpo Lúteo/anormalidades , Feminino , Integrases , Masculino , Glândulas Mamárias Animais/anormalidades , Camundongos , Camundongos Knockout , Ovário/anormalidades , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/patologia , Útero/anormalidades
7.
Anim Genet ; 43(6): 689-95, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22497297

RESUMO

Supernumerary teats (hyperthelia, SNTs) are a common abnormality of the bovine udder with a medium to high heritability and a postulated oligogenic or polygenic inheritance pattern. SNTs not only negatively affect machine milking ability but also act as a reservoir for bacteria. A genome-wide association study was carried out to identify genes involved in the development of SNTs in the dual-purpose Fleckvieh breed. A total of 2467 progeny-tested bulls were genotyped at 43 698 single nucleotide polymorphisms, and daughter yield deviations (DYDs) for 'udder clearness' (UC) were used as high-heritability phenotypes. Massive structuring of the study population was accounted for by principal components analysis-based and mixed model-based approaches. Four loci on BTA5, BTA6, BTA11 and BTA17 were significantly associated with the UC DYD. Three associated regions contain genes of the highly conserved Wnt signalling pathway. The four QTL together account for 10.7% of the variance of the UC DYD, whereas the major fraction of the DYD variance is attributable to chromosomes with no identified QTL. Our results support both an oligogenic and a polygenic inheritance pattern of SNTs in cattle. The identified candidate genes permit insights into the genetic architecture of teat malformations in cattle and provide clues to unravel the molecular mechanisms of mammary gland alterations in cattle and other species.


Assuntos
Bovinos/anormalidades , Bovinos/genética , Mapeamento Cromossômico/veterinária , Glândulas Mamárias Animais/anormalidades , Locos de Características Quantitativas/genética , Animais , Cromossomos de Mamíferos , Feminino , Variação Genética , Estudo de Associação Genômica Ampla/veterinária , Genótipo , Polimorfismo de Nucleotídeo Único , Via de Sinalização Wnt/genética
8.
Proc Natl Acad Sci U S A ; 106(26): 10696-701, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19541653

RESUMO

PACT is a double-stranded RNA-binding protein that also binds and activates the latent protein kinase, PKR, which plays a major role in cellular antiviral defense in mammals. For evaluating PACT's contribution to the innate immune system, Pact(-/-) mice have been generated; these mice exhibit notable developmental abnormalities including microtia, with craniofacial, ear, and hearing defects. Here we report that, in addition, Pact(-/-) mice had smaller body size and fertility defects, both of which were caused by defective pituitary functions. Pact(-/-) mice exhibited anterior pituitary lobe (AL) hypoplasia, which developed postnatally, when the second phase of pituitary expansion occurs. Among the 5 cell types in AL, the numbers of corticotrophs, gonadotrophs, and somatotrophs were equally decreased in Pact(-/-) mice with a greater impact on lactotrophs and a lesser impact on thyrotrophs. PACT mRNA and protein were highly expressed in the pituitary of wild-type (Wt) mice during the postnatal wave of AL proliferation, the same period in which the hypoplasia developed in Pact(-/-) mice. During this time, the pituitaries of Pact(-/-) mice did not exhibit significantly increased apoptosis compared with Wt mice but showed a decrease in cell proliferation. The inhibition of cell proliferation observed in vivo could be recapitulated in vitro in GH3 somato/lactotroph and LbetaT2 gonadotroph cell lines; knockdown of PACT expression with siRNA diminished the rate of proliferation of these cells. Our study revealed a physiologically significant role for PACT in cell proliferation and an essential role of a dsRNA-binding protein in mammalian pituitary expansion.


Assuntos
Proliferação de Células , Hipófise/metabolismo , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Tamanho Corporal/genética , Tamanho Corporal/fisiologia , Peso Corporal/genética , Peso Corporal/fisiologia , Linhagem Celular , Feminino , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Infertilidade/genética , Infertilidade/fisiopatologia , Masculino , Glândulas Mamárias Animais/anormalidades , Camundongos , Camundongos Knockout , Ovário/anormalidades , Hipófise/patologia , Gravidez , Interferência de RNA , Proteínas de Ligação a RNA/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
9.
Anal Biochem ; 412(1): 92-5, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21255551

RESUMO

CRE-loxP-mediated inactivation and activation of genes in mouse mammary epithelium have been widely used to study genetic pathways in normal development and neoplastic transformation in vivo. In 1997, we generated three distinct mouse lines carrying an identical MMTV-Cre transgene (lines A, D, and F). Because the presence of CRE recombinase can adversely affect the physiology of nonmammary cells, we explored whether transgenic females display lactational defects. Whereas dams from line D nurse their pups and display overtly normal mammary development, line A shows some impairment during lactation and females from line F completely fail to nurse their litters. The ability to nurse a litter correlates with the extent of alveolar development and differentiation. This study demonstrates the importance of including appropriate "Cre-only" controls and provides guidelines to avoid problems in data interpretation.


Assuntos
Integrases/genética , Glândulas Mamárias Animais/metabolismo , Vírus do Tumor Mamário do Camundongo/genética , Transgenes , Animais , Animais Geneticamente Modificados , Feminino , Deleção de Genes , Integrases/metabolismo , Lactação , Glândulas Mamárias Animais/anormalidades , Vírus do Tumor Mamário do Camundongo/metabolismo , Camundongos , Camundongos Transgênicos/genética
10.
Mol Cell Endocrinol ; 538: 111465, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34597725

RESUMO

Growth Hormone (GH) plays crucial roles in mammary gland development and growth, and its upregulation has been associated with breast cancer promotion and/or progression. To ascertain how high GH levels could promote mammary tissue oncogenic transformation, morphological characteristics and the expression of receptors involved in mammary growth, development and cancer, and of mitogenic mediators were analyzed in the mammary gland of virgin adult transgenic mice that overexpress GH. Whole mounting and histologic analysis evidenced that transgenic mice exhibit increased epithelial ductal elongation and enlarged ducts along with deficient branching and reduced number of alveolar structures compared to wild type mice. The number of differentiated alveolar structures was diminished in transgenic mice while the amount of terminal end buds (TEBs) did not differ between both groups of mice. GH, insulin-like growth factor 1 (IGF1) and GH receptor mRNA levels were augmented in GH-overexpressing mice breast tissue, as well as IGF1 receptor protein content. However, GH receptor protein levels were decreased in transgenic mice. Fundamental receptors for breast growth and development like progesterone receptor and epidermal growth factor receptor were also increased in mammary tissue from transgenic animals. In turn, the levels of the proliferation marker Ki67, cFOS and Cyclin D1 were increased in GH-overexpressing mice, while cJUN expression was decreased and cMYC did not vary. In conclusion, prolonged exposure to high GH levels induces morphological and molecular alterations in the mammary gland that affects its normal development. While these effects would not be tumorigenic per se, they might predispose to oncogenic transformation.


Assuntos
Proteínas de Transporte/genética , Hormônio do Crescimento/genética , Fator de Crescimento Insulin-Like I/genética , Glândulas Mamárias Animais/anormalidades , Animais , Animais Geneticamente Modificados , Biomarcadores/metabolismo , Feminino , Hormônio do Crescimento/metabolismo , Glândulas Mamárias Animais/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo
11.
FASEB J ; 23(10): 3482-93, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19584305

RESUMO

The adult, virgin mammary gland is a highly organized branched ductal network comprising two major cell types: myoepithelial and luminal epithelial cells. To study the role and mechanism of focal adhesion kinase (FAK)-mediated signaling in mammary gland development and differentiation, we used a conditional Fak-knockout mammary epithelial cell (MEC) transplantation model. Conditional Cre recombinase (Cre)-mediated Fak deletion in primary cultured MECs isolated from FAK(lox/lox)/Rosa26Cre-ERT2 donor mice caused loss of FAK in all mammary cells. Transplantation of Fak-knockout MECs in a cleared mammary fat pad of immune-deficient recipient mice resulted in development of new but dilated virgin ducts with a disrupted myoepithelial and luminal epithelial cell multilayer and aberrant ductal morphogenesis during pregnancy. In the absence of FAK, MECs spread poorly, showed enhanced Rho kinase (ROCK)-mediated cytoskeletal contractility, and failed to respond to receptor-mediated cytoskeletal remodeling. Likewise, FAK deficiency fully inhibited branching morphogenesis of mammary gland organoids in a ROCK-dependent manner. Altogether these data suggest a model in which FAK coordinates contractile forces in MECs to maintain the bilayered cellular organization of myoepithelial and luminal epithelial cells in ducts, thus allowing proper mammary gland development and function.


Assuntos
Quinase 1 de Adesão Focal/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Morfogênese , Quinases Associadas a rho/metabolismo , Animais , Células Epiteliais/enzimologia , Células Epiteliais/fisiologia , Feminino , Quinase 1 de Adesão Focal/genética , Deleção de Genes , Lactação/genética , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/fisiologia , Camundongos , Camundongos Knockout , Morfogênese/genética , Gravidez
12.
Mol Cell Biol ; 27(14): 5120-7, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17485440

RESUMO

To investigate in vivo roles of a murine hypothalamic homeobox gene, Bsx, we generated and analyzed two mutant alleles, Bsx(DeltaHD) and Bsx(lacZ). Bsx(DeltaHD) lacks the homeodomain, and Bsx(lacZ) is an insertion of a lacZ reporter gene. Bsx-lacZ expression was detected in the hypothalamus and pineal gland and reiterates Bsx expression. Bsx homozygous mutant mice were born at the expected Mendelian ratio, but their growth was impaired. Offspring from Bsx homozygous mutant females exhibited a low survival rate due to a nursing defect. Mammary glands of the mutant females developed normally during pregnancy; however, they involuted quickly after parturition. These results demonstrate that Bsx is required for postnatal growth and maintenance of lactating mammary glands. Thus, mouse Bsx is likely involved in systemic control of suppression of apoptosis of postpartum mammary epithelial cells.


Assuntos
Encéfalo/metabolismo , Comportamento Alimentar/fisiologia , Genes Homeobox , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/embriologia , Feminino , Transtornos do Crescimento/patologia , Homozigoto , Masculino , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Mutantes , Especificidade de Órgãos , Gravidez , beta-Galactosidase/metabolismo
13.
Exp Mol Pathol ; 89(1): 9-19, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20399205

RESUMO

Caveolin-1 (Cav-1) is a major structural protein of caveolae, specialized plasma membrane invaginations that are involved in a cell-specific fashion in diverse cell activities such as molecular transport, cell adhesion, and signal transduction. In normal adult mammals, Cav-1 expression is abundant in mesenchyme-derived cells but relatively low in epithelial parenchyma. However, epithelial Cav-1 overexpression is associated with development and/or progression of many carcinomas. In this study, we generated and characterized a transgenic mouse model of Cav-1 overexpression under the control of a mouse mammary tumor virus (MMTV) long terminal-repeat promoter, which is predominantly expressed in specific epithelial cells. The MMTVcav-1(+) transgenic mice were fertile, and females bore litters of normal size with no obvious developmental abnormalities. However, by age 11months, the MMTVcav-1(+) mice demonstrated overtly different phenotypes in multiple exocrine organs when compared with their nontransgenic MMTVcav-1(-) littermates. Cav-1 overexpression in MMTVcav-1(+) mice produced organ-specific abnormalities, including hypotrophy of mammary glandular epithelia, bronchiolar epithelial hyperplasia and atypia, mucous-cell hyperplasia in salivary glands, elongated hair follicles and dermal thickening in the skin, and reduced accumulation of enzymogen granules in pancreatic acinar cells. In addition, the MMTVcav-1(+) transgenic mice tended to have a greater incidence of malignant tumors, including lung and liver carcinomas and lymphoma, than their MMTVcav-1(-) littermates. Our results indicate that Cav-1 overexpression causes organ-specific, age-related epithelial disorders and suggest the potential for increased susceptibility to carcinogenesis.


Assuntos
Anormalidades Múltiplas/genética , Caveolina 1/genética , Epitélio/patologia , Glândulas Exócrinas/anormalidades , Regulação da Expressão Gênica/fisiologia , Vírus do Tumor Mamário do Camundongo/genética , Regiões Promotoras Genéticas/genética , Animais , Células Cultivadas , Epitélio/metabolismo , Feminino , Hiperplasia , Incidência , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Masculino , Glândulas Mamárias Animais/anormalidades , Camundongos , Camundongos Transgênicos , Pele/metabolismo , Pele/patologia
14.
Dev Cell ; 2(5): 643-53, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12015971

RESUMO

Hair follicle morphogenesis is initiated by a dermal signal that induces the development of placodes in the overlying epithelium. To determine whether WNT signals are required for initiation of follicular development, we ectopically expressed Dickkopf 1, a potent diffusible inhibitor of WNT action, in the skin of transgenic mice. This produced a complete failure of placode formation prior to morphological or molecular signs of differentiation, and blocked tooth and mammary gland development before the bud stage. This phenotype indicates that activation of WNT signaling in the skin precedes, and is required for, localized expression of regulatory genes and initiation of hair follicle placode formation.


Assuntos
Folículo Piloso/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas/fisiologia , Transativadores , Proteínas de Peixe-Zebra , Animais , Sequência de Bases , Diferenciação Celular , Divisão Celular , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA/genética , Receptor Edar , Células Epidérmicas , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Cabelo/anormalidades , Folículo Piloso/anormalidades , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intercelular , Fator 1 de Ligação ao Facilitador Linfoide , Glândulas Mamárias Animais/anormalidades , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Proteínas/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , Receptores da Ectodisplasina , Receptores do Fator de Necrose Tumoral , Transdução de Sinais , Fenômenos Fisiológicos da Pele , Anormalidades Dentárias/genética , Fatores de Transcrição/genética , Regulação para Cima , Proteínas Wnt , beta Catenina
15.
Dev Cell ; 4(3): 371-82, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12636918

RESUMO

Netrin-1 and its receptors play an essential role patterning the nervous system by guiding neurons and axons to their targets. To explore whether netrin-1 organizes nonneural tissues, we examined its role in mammary gland morphogenesis. Netrin-1 is expressed in prelumenal cells, and its receptor neogenin is expressed in a complementary pattern in adjacent cap cells of terminal end buds (TEBs). We discovered that loss of either gene results in disorganized TEBs characterized by exaggerated subcapsular spaces, breaks in basal lamina, dissociated cap cells, and an increased influx of cap cells into the prelumenal compartment. Cell aggregation assays demonstrate that neogenin mediates netrin-1-dependent cell clustering. Thus, netrin-1 appears to act locally through neogenin to stabilize the multipotent progenitor (cap) cell layer during mammary gland development. Our results suggest that netrin-1 and its receptor neogenin provide an adhesive, rather than a guidance, function during nonneural organogenesis.


Assuntos
Adesão Celular/genética , Diferenciação Celular/genética , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/anormalidades , Proteínas de Membrana/deficiência , Fatores de Crescimento Neural/deficiência , Células-Tronco/metabolismo , Actinas/metabolismo , Animais , Apoptose/fisiologia , Membrana Basal/metabolismo , Membrana Basal/ultraestrutura , Caderinas/metabolismo , Comunicação Celular/genética , Células Cultivadas , Células Epiteliais/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Laminina/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Nus , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Fatores de Crescimento Neural/genética , Netrina-1 , Transplante de Células-Tronco , Células-Tronco/citologia , Proteínas Supressoras de Tumor
16.
Mol Cell Biol ; 26(15): 5797-808, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847332

RESUMO

The mammary gland consists of a branched ductal system comprised of milk-producing epithelial cells that form ductile tubules surrounded by a myoepithelial cell layer that provides contractility required for milk ejection. Myoepithelial cells bear a striking resemblance to smooth muscle cells, but they are derived from a different embryonic cell lineage, and little is known of the mechanisms that control their differentiation. Members of the myocardin family of transcriptional coactivators cooperate with serum response factor to activate smooth muscle gene expression. We show that female mice homozygous for a loss-of-function mutation of the myocardin-related transcription factor A (MRTF-A) gene are unable to effectively nurse their offspring due to a failure in maintenance of the differentiated state of mammary myoepithelial cells during lactation, resulting in apoptosis of this cell population, a consequent inability to release milk, and premature involution. The phenotype of MRTF-A mutant mice reveals a specific and essential role for MRTF-A in mammary myoepithelial cell differentiation and points to commonalities in the transcriptional mechanisms that control differentiation of smooth muscle and myoepithelial cells.


Assuntos
Células Epiteliais/fisiologia , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Animais , Animais Recém-Nascidos , Peso Corporal , Diferenciação Celular , Células Epiteliais/citologia , Feminino , Marcação In Situ das Extremidades Cortadas , Lactação , Glândulas Mamárias Animais/anormalidades , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Gravidez , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Mol Cell Biol ; 26(15): 5809-26, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847333

RESUMO

Transcription of immediate-early genes--as well as multiple genes affecting muscle function, cytoskeletal integrity, apoptosis control, and wound healing/angiogenesis--is regulated by serum response factor (Srf). Extracellular signals regulate Srf in part via a pathway involving megakaryoblastic leukemia 1 (Mkl1, also known as myocardin-related transcription factor A [Mrtf-a]), which coactivates Srf-responsive genes downstream of Rho GTPases. Here we investigate Mkl1 function using gene targeting and show the protein to be essential for the physiologic preparation of the mammary gland during pregnancy and the maintenance of lactation. Lack of Mkl1 causes premature involution and impairs expression of Srf-dependent genes in the mammary myoepithelial cells, which control milk ejection following oxytocin-induced contraction. Despite the importance of Srf in multiple transcriptional pathways and widespread Mkl1 expression, the spectrum of abnormalities associated with Mkl1 absence appears surprisingly restricted.


Assuntos
Lactação/fisiologia , Glândulas Mamárias Animais/anatomia & histologia , Glândulas Mamárias Animais/fisiologia , Transativadores/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Criança , Insuficiência de Crescimento , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Marcação de Genes , Coração/anatomia & histologia , Coração/embriologia , Humanos , Lactente , Leucemia Megacarioblástica Aguda , Masculino , Glândulas Mamárias Animais/anormalidades , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Leite , Miócitos Cardíacos/patologia , Miócitos Cardíacos/ultraestrutura , Análise de Sequência com Séries de Oligonucleotídeos , Ocitocina/metabolismo , Gravidez , Prolactina/metabolismo , Fator de Transcrição STAT3 , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transativadores/genética
18.
Mol Cell Biol ; 26(17): 6571-83, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16914740

RESUMO

Although the essential involvement of the progesterone receptor (PR) in female reproductive tissues is firmly established, the coregulators preferentially enlisted by PR to mediate its physiological effects have yet to be fully delineated. To further dissect the roles of members of the steroid receptor coactivator (SRC)/p160 family in PR-mediated reproductive processes in vivo, state-of-the-art cre-loxP engineering strategies were employed to generate a mouse model (PR(Cre/+) SRC-2(flox/flox)) in which SRC-2 function was abrogated only in cell lineages that express the PR. Fertility tests revealed that while ovarian activity was normal, PR(Cre/+) SRC-2(flox/flox) mouse uterine function was severely compromised. Absence of SRC-2 in PR-positive uterine cells was shown to contribute to an early block in embryo implantation, a phenotype not shared by SRC-1 or -3 knockout mice. In addition, histological and molecular analyses revealed an inability of the PR(Cre/+) SRC-2(flox/flox) mouse uterus to undergo the necessary cellular and molecular changes that precede complete P-induced decidual progression. Moreover, removal of SRC-1 in the PR(Cre/+) SRC-2(flox/flox) mouse uterus resulted in the absence of a decidual response, confirming that uterine SRC-2 and -1 cooperate in P-initiated transcriptional programs which lead to full decidualization. In the case of the mammary gland, whole-mount and histological analysis disclosed the absence of significant ductal side branching and alveologenesis in the hormone-treated PR(Cre/+) SRC-2(flox/flox) mammary gland, reinforcing an important role for SRC-2 in cellular proliferative changes that require PR. We conclude that SRC-2 is appropriated by PR in a subset of transcriptional cascades obligate for normal uterine and mammary morphogenesis and function.


Assuntos
Glândulas Mamárias Animais/embriologia , Morfogênese , Coativador 2 de Receptor Nuclear/metabolismo , Progesterona/farmacologia , Útero/efeitos dos fármacos , Útero/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Implantação do Embrião , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Estrogênios/farmacologia , Feminino , Infertilidade Feminina , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Camundongos Knockout , Ovulação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Receptores de Progesterona/metabolismo , Útero/anormalidades , Útero/citologia
19.
J Anim Breed Genet ; 126(3): 237-41, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19646152

RESUMO

Parathyroid hormone-like hormone gene (PTHLH) and its receptor, parathyroid hormone/ parathyroid hormone-like hormone receptor 1 (PTHR1), play a role in epithelial mesenchymal interactions during growth and differentiation of different tissues and anatomic structures, including teats. Therefore, PTHLH and PTHR1 were evaluated as functional candidate genes for their effects on number and shape of teats in pigs. In particular, focus was on the occurrence and number of inverted teats, the most frequent and economically relevant teat developmental defect in pigs. For this purpose, association and linkage of the PTHLH gene and the PTHR1 gene with inverted teat defect and the total number of teats and inverted teats were studied in an experimental Duroc and Berlin Miniature pig (DUMI) population. Polymorphism C1819T of PTHR1 was significantly associated with inverted teat phenotype (p = 0.014), total number of teats (p = 0.047) and was close to significance with the number of inverted teats (p = 0.078). Polymorphism C375T of PTHLH was close to significance with the inverted teat phenotype (p = 0.122) and showed no significant association with the total number of teats (p = 0.621) and the number of inverted teats (p = 0.256) in the DUMI population. Association analyses were also performed for combined effects of PTHLH and PTHR1 in order to address potential interaction, however, revealed no indication of effects of interaction. The function, position and the association shown here promote PTHR1 as a candidate gene for number of teats and in particular for affection by and number of inverted teats.


Assuntos
Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/anatomia & histologia , Proteína Relacionada ao Hormônio Paratireóideo/genética , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Sus scrofa/genética , Animais , Feminino , Regulação da Expressão Gênica , Genótipo , Glândulas Mamárias Animais/fisiologia , Especificidade de Órgãos , Polimorfismo Genético , Sus scrofa/anormalidades , Sus scrofa/anatomia & histologia
20.
Oncogene ; 38(3): 360-374, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30093634

RESUMO

The role of the tumour-suppressor miR-34 family in breast physiology and in mammary stem cells (MaSCs) is largely unknown. Here, we revealed that miR-34 family, and miR-34a in particular, is implicated in mammary epithelium homoeostasis. Expression of miR-34a occurs upon luminal commitment and differentiation and serves to inhibit the expansion of the pool of MaSCs and early progenitor cells, likely in a p53-independent fashion. Mutant mice (miR34-KO) and loss-of-function approaches revealed two separate functions of miR-34a, controlling both proliferation and fate commitment in mammary progenitors by modulating several pathways involved in epithelial cell plasticity and luminal-to-basal conversion. In particular, miR-34a acts as endogenous inhibitor of the Wnt/beta-catenin signalling pathway, targeting up to nine upstream regulators at the same time, thus modulating the expansion of the MaSCs/early progenitor pool. These multiple roles of miR-34a are maintained in a model of human breast cancer, in which chronic expression of miR-34a in triple-negative mesenchymal-like cells (enriched in cancer stem cells-CSCs) could promote a luminal-like differentiation programme, restrict the CSC pool, and inhibit tumour propagation. Hence, activation of miR-34a-dependent programmes could provide a therapeutic opportunity for the subset of breast cancers, which are rich in CSCs and respond poorly to conventional therapies.


Assuntos
Neoplasias da Mama/patologia , Glândulas Mamárias Animais/citologia , MicroRNAs/fisiologia , RNA Neoplásico/fisiologia , Animais , Neoplasias da Mama/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Autorrenovação Celular/fisiologia , Células Epiteliais/metabolismo , Feminino , Humanos , Glândulas Mamárias Animais/anormalidades , Glândulas Mamárias Animais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Knockout , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Esferoides Celulares , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA