Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 439
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Dev Biol ; 446(2): 180-192, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30594504

RESUMO

The evolutionarily conserved transcription factor, Tbx18, is expressed in a dynamic pattern throughout embryonic and early postnatal life and plays crucial roles in the development of multiple organ systems. Previous studies have indicated that this dynamic function is controlled by an expansive regulatory structure, extending far upstream and downstream of the gene. With the goal of identifying elements that interact with the Tbx18 promoter in developing prostate, we coupled chromatin conformation capture (4C) and ATAC-seq from embryonic day 18.5 (E18.5) mouse urogenital sinus (UGS), where Tbx18 is highly expressed. The data revealed dozens of active chromatin elements distributed throughout a 1.5 million base pair topologically associating domain (TAD). To identify cell types contributing to this chromatin signal, we used lineage tracing methods with a Tbx18 Cre "knock-in" allele; these data show clearly that Tbx18-expressing precursors differentiate into wide array of cell types in multiple tissue compartments, most of which have not been previously reported. We also used a 209 kb Cre-expressing Tbx18 transgene, to partition enhancers for specific precursor types into two rough spatial domains. Within this central 209 kb compartment, we identified ECR1, previously described to regulate Tbx18 expression in ureter, as an active regulator of UGS expression. Together these data define the diverse fates of Tbx18+ precursors in prostate-associated tissues for the first time, and identify a highly active TAD controlling the gene's essential function in this tissue.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Próstata/metabolismo , Elementos Reguladores de Transcrição/genética , Proteínas com Domínio T/genética , Animais , Sítios de Ligação/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Cromatina/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Masculino , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Próstata/citologia , Próstata/embriologia , Proteínas com Domínio T/metabolismo , Sistema Urogenital/citologia , Sistema Urogenital/embriologia , Sistema Urogenital/metabolismo
2.
Biol Reprod ; 101(3): 602-616, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31058957

RESUMO

Alfred Jost's work in the 1940s laid the foundation of the current paradigm of sexual differentiation of reproductive tracts, which contends that testicular hormones drive the male patterning of reproductive tract system whereas the female phenotype arises by default. Once established, the sex-specific reproductive tracts undergo morphogenesis, giving rise to anatomically and functionally distinct tubular organs along the rostral-caudal axis. Impairment of sexual differentiation of reproductive tracts by genetic alteration and environmental exposure are the main causes of disorders of sex development, and infertility at adulthood. This review covers past and present work on sexual differentiation and morphogenesis of reproductive tracts, associated human disorders, and emerging technologies that have made impacts or could radically expand our knowledge in this field.


Assuntos
Pesquisa Biomédica , Genitália/embriologia , Morfogênese/fisiologia , Diferenciação Sexual/fisiologia , Adulto , Animais , Pesquisa Biomédica/história , Pesquisa Biomédica/métodos , Pesquisa Biomédica/tendências , Feminino , Regulação da Expressão Gênica no Desenvolvimento , História do Século XX , História do Século XXI , Humanos , Invenções , Masculino , Reprodução/genética , Sistema Urogenital/embriologia
3.
Development ; 142(10): 1893-908, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25968320

RESUMO

Malformation of the urogenital tract represents a considerable paediatric burden, with many defects affecting the lower urinary tract (LUT), genital tubercle and associated structures. Understanding the molecular basis of such defects frequently draws on murine models. However, human anatomical terms do not always superimpose on the mouse, and the lack of accurate and standardised nomenclature is hampering the utility of such animal models. We previously developed an anatomical ontology for the murine urogenital system. Here, we present a comprehensive update of this ontology pertaining to mouse LUT, genital tubercle and associated reproductive structures (E10.5 to adult). Ontology changes were based on recently published insights into the cellular and gross anatomy of these structures, and on new analyses of epithelial cell types present in the pelvic urethra and regions of the bladder. Ontology changes include new structures, tissue layers and cell types within the LUT, external genitalia and lower reproductive structures. Representative illustrations, detailed text descriptions and molecular markers that selectively label muscle, nerves/ganglia and epithelia of the lower urogenital system are also presented. The revised ontology will be an important tool for researchers studying urogenital development/malformation in mouse models and will improve our capacity to appropriately interpret these with respect to the human situation.


Assuntos
Sistema Urogenital/anatomia & histologia , Sistema Urogenital/embriologia , Animais , Camundongos , Modelos Animais , Uretra/anatomia & histologia , Uretra/embriologia , Bexiga Urinária/anatomia & histologia , Bexiga Urinária/embriologia , Sistema Urinário/anatomia & histologia , Sistema Urinário/embriologia
4.
J Anat ; 233(6): 724-739, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30294789

RESUMO

Subdivision of cloaca into urogenital and anorectal passages has remained controversial because of disagreements about the identity and role of the septum developing between both passages. This study aimed to clarify the development of the cloaca using a quantitative 3D morphological approach in human embryos of 4-10 post-fertilisation weeks. Embryos were visualised with Amira 3D-reconstruction and Cinema 4D-remodelling software. Distances between landmarks were computed with Amira3D software. Our main finding was a pronounced difference in growth between rapidly expanding central and ventral parts, and slowly or non-growing cranial and dorsal parts. The entrance of the Wolffian duct into the cloaca proved a stable landmark that remained linked to the position of vertebra S3. Suppressed growth in the cranial cloaca resulted in an apparent craniodorsal migration of the entrance of the Wolffian duct, while suppressed growth in the dorsal cloaca changed the entrance of the hindgut from cranial to dorsal on the cloaca. Transformation of this 'end-to-end' into an 'end-to-side' junction produced temporary 'lateral (Rathke's) folds'. The persistent difference in dorsoventral growth straightened the embryonic caudal body axis and concomitantly extended the frontally oriented 'urorectal (Tourneux's) septum' caudally between the ventral urogenital and dorsal anorectal parts of the cloaca. The dorsoventral growth difference also divided the cloacal membrane into a well-developed ventral urethral plate and a thin dorsal cloacal membrane proper, which ruptured at 6.5 weeks. The expansion of the pericloacal mesenchyme followed the dorsoventral growth difference and produced the genital tubercle. Dysregulation of dorsal cloacal development is probably an important cause of anorectal malformations: too little regressive development may result in anorectal agenesis, and too much regression in stenosis or atresia of the remaining part of the dorsal cloaca.


Assuntos
Cloaca/embriologia , Sistema Urogenital/embriologia , Embrião de Mamíferos , Humanos
5.
Environ Toxicol ; 31(12): 1740-1750, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26443714

RESUMO

Substances that mimic endogenous hormones may alter the cell signaling that govern prostate development and predispose it to developing lesions in adult and senile life. Bisphenol A is able to mimic estrogens, and studies have demonstrated that low levels of exposure to this compound have caused alterations during prostate development. The aim of this study was to describe the prostate development in both male and female neonatal gerbils in normal conditions and under exposure to BPA during intrauterine life, and also to analyze whether the effects of intrauterine exposure to BPA remain in adulthood. Morphological, stereological, three-dimensional reconstruction, and immunohistochemical methods were employed. The results demonstrated that in 1-day-old normal gerbils, the female paraurethral glands and the male ventral lobe are morphologically similar, although its tissue components-epithelial buds (EB), periurethral mesenchyme (PeM), paraurethral mesenchyme (PaM) or ventral mesenchymal pad (VMP), and smooth muscle (SM)-have presented different immunolabeling pattern for androgen receptor (AR), and for proliferating cell nuclear antigen (PCNA). Moreover, we observed a differential response of male and female prostate to intrauterine BPA exposure. In 1-day-old males, the intrauterine exposure to BPA caused a decrease of AR-positive cells in the PeM and SM, and a decrease of the proliferative status in the EB. In contrast, no morphological alterations were observed in ventral prostate of adult males. In 1-day-old females, BPA exposure promoted an increase of estrogen receptor alpha (ERα) positive cells in PeM and PaM, a decrease of AR-positive cells in EB and PeM, besides a reduction of cell proliferation in EB. Additionally, the adult female prostate of BPA-exposed animals presented an increase of AR- and PCNA-positive cells. These results suggest that the prostate of female gerbils were more susceptible to the intrauterine BPA effects, since they became more proliferative in adult life. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1740-1750, 2016.


Assuntos
Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Fenóis/toxicidade , Sistema Urogenital/efeitos dos fármacos , Fatores Etários , Animais , Animais Recém-Nascidos , Proliferação de Células/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Gerbillinae , Masculino , Exposição Materna/efeitos adversos , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Próstata/citologia , Próstata/efeitos dos fármacos , Próstata/embriologia , Próstata/crescimento & desenvolvimento , Receptores Androgênicos/metabolismo , Fatores Sexuais , Sistema Urogenital/citologia , Sistema Urogenital/embriologia , Sistema Urogenital/crescimento & desenvolvimento
6.
Genesis ; 53(6): 366-76, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25980463

RESUMO

The developing limb is a useful model for studying organogenesis and developmental processes. Although Cre alleles exist for conditional loss- or gain-of-function in limbs, Cre alleles targeting specific limb subdomains are desirable. Here we report on the generation of the Hoxa13:Cre line, in which the Cre gene is inserted in the endogenous Hoxa13 gene. We provide evidence that the Cre is active in embryonic tissues/regions where the endogenous Hoxa13 gene is expressed. Our results show that cells expressing Hoxa13 in developing limb buds contribute to the entire autopod (hand/feet) skeleton and validate Hoxa13 as a distal limb marker as far as the skeleton is concerned. In contrast, in the limb musculature, Cre-based fate mapping shows that almost all muscle masses of the zeugopod (forearm) and part of the triceps contain Hoxa13-expressing cells and/or their descendants. Besides the limb, the activity of the Cre is detectable in the urogenital system and the hindgut, primarily in the epithelium and smooth muscles. Together our data show that the Hoxa13:Cre allele is a useful tool for conditional gene manipulation in the urogenital system, posterior digestive tract, autopod and part of the limb musculature.


Assuntos
Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Botões de Extremidades/metabolismo , Sistema Urogenital/embriologia , Sistema Urogenital/metabolismo , Animais , Feminino , Membro Anterior/citologia , Membro Anterior/embriologia , Membro Anterior/metabolismo , Trato Gastrointestinal/citologia , Trato Gastrointestinal/embriologia , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Integrases/genética , Integrases/metabolismo , Botões de Extremidades/citologia , Botões de Extremidades/embriologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculos/citologia , Músculos/embriologia , Músculos/metabolismo , Organogênese/genética , Fatores de Tempo , Sistema Urogenital/citologia
7.
Dev Biol ; 395(2): 342-54, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25224227

RESUMO

The issues of whether and how some organs coordinate their size and shape with the blueprint of the embryo axis, while others appear to regulate their morphogenesis autonomously, remain poorly understood. Mutations in Ift144, encoding a component of the trafficking machinery of primary cilia assembly, result in a range of embryo patterning defects, affecting the limbs, skeleton and neural system. Here, we show that embryos of the mouse mutant Ift144(twt) develop gonads that are larger than wild-type. Investigation of the early patterning of the urogenital ridge revealed that the anterior-posterior domain of the gonad/mesonephros was extended at 10.5 dpc, with no change in the length of the metanephros. In XY embryos, this extension resulted in an increase in testis cord number. Moreover, we observed a concomitant extension of the trunk axis in both sexes, with no change in the length of the tail domain or somite number. Our findings support a model in which: (1) primary cilia regulate embryonic trunk elongation; (2) the length of the trunk axis determines the size of the urogenital ridges; and (3) the gonad domain is partitioned into a number of testis cords that depends on the available space, rather than being divided a predetermined number of times to generate a specific number of cords.


Assuntos
Padronização Corporal/fisiologia , Cílios/fisiologia , Modelos Biológicos , Proteínas/metabolismo , Tronco/embriologia , Sistema Urogenital/embriologia , Animais , Proteínas do Citoesqueleto , Feminino , Imunofluorescência , Processamento de Imagem Assistida por Computador , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Microscopia Confocal , Proteínas/genética , Reação em Cadeia da Polimerase em Tempo Real
8.
Dev Biol ; 392(2): 483-93, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24854998

RESUMO

The vertebrate T-box transcription factor gene Tbx18 performs a vital role in development of multiple organ systems. Tbx18 insufficiency manifests as recessive phenotypes in the upper urinary system, cardiac venous pole, inner ear, and axial skeleton; homozygous null mutant animals die perinatally. Here, we report a new regulatory mutation of Tbx18, a reciprocal translocation breaking 78kbp downstream of the gene. 12Gso homozygotes present urinary and vertebral defects very similar to those associated with Tbx18-null mutations, but 12Gso is clearly not a global null allele since homozygotes survive into adulthood. We show that 12Gso down-regulates Tbx18 expression in a manner that is both spatially- and temporally-specific; combined with other data, the mutation points particularly to the presence of an essential urogenital enhancer located near the translocation breakpoint site. In support of this hypothesis, we identify a distal enhancer element, ECR1, which is active in developing urogenital and other tissues; we propose that disruption of this element leads to premature loss of Tbx18 function in 12Gso mutant mice. These data reveal a long-range regulatory architecture extending far downstream of Tbx18, identify a novel and likely essential urogenital enhancer, and introduce a new tool for dissecting postnatal phenotypes associated with dysregulation of Tbx18.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Proteínas com Domínio T/metabolismo , Sistema Urogenital/embriologia , Azul Alciano , Animais , Antraquinonas , Sequência de Bases , Mapeamento Cromossômico , Primers do DNA/genética , Técnicas Histológicas , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de DNA , Proteínas com Domínio T/genética , Translocação Genética/genética , Sistema Urogenital/metabolismo
9.
Dev Biol ; 395(2): 209-17, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25261715

RESUMO

The mammalian urogenital sinus (UGS) develops in a sex specific manner, giving rise to the prostate in the male and the sinus vagina in the embryonic female. Androgens, produced by the embryonic testis, have been shown to be crucial to this process. In this study we show that retinoic acid signaling is required for the initial stages of bud development from the male UGS. Enzymes involved in retinoic acid synthesis are expressed in the UGS mesenchyme in a sex specific manner and addition of ligand to female tissue is able to induce prostate-like bud formation in the absence of androgens, albeit at reduced potency. Functional studies in mouse organ cultures that faithfully reproduce the initiation of prostate development indicate that one of the roles of retinoic acid signaling in the male is to inhibit the expression of Inhba, which encodes the ßA subunit of Activin, in the UGS mesenchyme. Through in vivo genetic analysis and culture studies we show that inhibition of Activin signaling in the female UGS leads to a similar phenotype to that of retinoic acid treatment, namely bud formation in the absence of androgens. Our data also reveals that both androgens and retinoic acid have extra independent roles to that of repressing Activin signaling in the development of the prostate during fetal stages. This study identifies a novel role for retinoic acid as a mesenchymal factor that acts together with androgens to determine the position and initiation of bud development in the male UGS epithelia.


Assuntos
Ativinas/metabolismo , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Tretinoína/metabolismo , Sistema Urogenital/embriologia , Ativinas/antagonistas & inibidores , Animais , Primers do DNA , Feminino , Imuno-Histoquímica , Hibridização In Situ , Subunidades beta de Inibinas/antagonistas & inibidores , Masculino , Camundongos , Próstata/embriologia , Reação em Cadeia da Polimerase em Tempo Real , Fatores Sexuais , Tretinoína/farmacologia , Sistema Urogenital/metabolismo , beta-Galactosidase
10.
Dev Biol ; 385(1): 41-51, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24479159

RESUMO

Anorectal malformation (ARM) is a common birth defect but the developmental history and the underlying molecular mechanism are poorly understood. Using murine genetic models, we report here that a signaling molecule Dickkopf-1 (Dkk1) is a critical regulator. The anorectal and genitourinary tracts are major derivatives of caudal hindgut, or the cloaca.Dkk1 is highly expressed in the dorsal peri-cloacal mesenchymal (dPCM) progenitors. We show that the deletion of Dkk1 causes the imperforate anus with rectourinary fistula. Mutant genital tubercles exhibit a preputial hypospadias phenotype and premature urethral canalization.Dkk1 mutants have an ectopic expansion of the dPCM tissue, which correlates with an aberrant increase of cell proliferation and survival. This ectopic tissue is detectable before the earliest sign of the anus formation, suggesting that it is most likely the primary or early cause of the defect. Deletion of Dkk1 results in an elevation of the Wnt/ß-catenin activity. Signaling molecules Shh, Fgf8 and Bmp4 are also upregulated. Furthermore, genetic hyperactivation of Wnt/ß-catenin signal pathway in the cloacal mesenchyme partially recapitulates Dkk1 mutant phenotypes. Together, these findings underscore the importance ofDKK1 in regulating behavior of dPCM progenitors, and suggest that formation of anus and urethral depends on Dkk1-mediated dynamic inhibition of the canonical Wnt/ß-catenin signal pathway.


Assuntos
Canal Anal/embriologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mesoderma/embriologia , Reto/embriologia , Sistema Urogenital/embriologia , Canal Anal/anormalidades , Animais , Malformações Anorretais , Anus Imperfurado/embriologia , Anus Imperfurado/genética , Proteína Morfogenética Óssea 4/biossíntese , Diferenciação Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Ativação Enzimática/genética , Fator 8 de Crescimento de Fibroblasto/biossíntese , Proteínas Hedgehog/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Reto/anormalidades , Células-Tronco , Regulação para Cima , Anormalidades Urogenitais/embriologia , Anormalidades Urogenitais/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
11.
Dev Biol ; 387(1): 64-72, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24394376

RESUMO

Defects of the ventral body wall are prevalent birth anomalies marked by deficiencies in body wall closure, hypoplasia of the abdominal musculature and multiple malformations across a gamut of organs. However, the mechanisms underlying ventral body wall defects remain elusive. Here, we investigated the role of Wnt signaling in ventral body wall development by inactivating Wls or ß-catenin in murine abdominal ectoderm. The loss of Wls in the ventral epithelium, which blocks the secretion of Wnt proteins, resulted in dysgenesis of ventral musculature and genito-urinary tract during embryonic development. Molecular analyses revealed that the dermis and myogenic differentiation in the underlying mesenchymal progenitor cells was perturbed by the loss of ectodermal Wls. The activity of the Wnt-Pitx2 axis was impaired in the ventral mesenchyme of the mutant body wall, which partially accounted for the defects in ventral musculature formation. In contrast, epithelial depletion of ß-catenin or Wnt5a did not resemble the body wall defects in the ectodermal Wls mutant. These findings indicate that ectodermal Wnt signaling instructs the underlying mesodermal specification and abdominal musculature formation during ventral body wall development, adding evidence to the theory that ectoderm-mesenchyme signaling is a potential unifying mechanism for the origin of ventral body wall defects.


Assuntos
Abdome/embriologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Desenvolvimento Muscular/genética , Receptores Acoplados a Proteínas G/fisiologia , Via de Sinalização Wnt/genética , beta Catenina/fisiologia , Abdome/crescimento & desenvolvimento , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Ectoderma/embriologia , Ectoderma/crescimento & desenvolvimento , Ectoderma/metabolismo , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mesoderma/embriologia , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Fatores de Transcrição/genética , Sistema Urogenital/embriologia , Sistema Urogenital/crescimento & desenvolvimento , Proteínas Wnt/deficiência , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a , beta Catenina/genética , Proteína Homeobox PITX2
12.
Biol Reprod ; 93(6): 138, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26510867

RESUMO

In mammals, the primary role of anti-Müllerian hormone (AMH) during development is the regression of Müllerian ducts in males. These structures otherwise develop into fallopian tubes, oviducts, and upper vagina, as in females. This highly conserved function is retained in birds and is supported by the high levels of AMH expression in developing testes. In mammals, AMH expression is controlled partly by the transcription factor, SOX9. However, in the chicken, AMH mRNA expression precedes that of SOX9 , leading to the view that AMH may lie upstream of SOX9 and play a more central role in avian testicular development. To help define the role of AMH in chicken gonad development, we suppressed AMH expression in chicken embryos using RNA interference. In males, AMH knockdown did not affect the expression of key testis pathway genes, and testis cords developed normally. However, a reduction in the size of the mesonephros and gonads was observed, a phenotype that was evident in both sexes. This growth defect occurred as a result of the reduced proliferative capacity of the cells of these tissues, and male gonads also had a significant reduction in germ cell numbers. These data suggest that although AMH does not directly contribute to testicular or ovarian differentiation, it is required in a sex-independent manner for proper cell proliferation and urogenital system growth.


Assuntos
Hormônio Antimülleriano/genética , Ovário/embriologia , Diferenciação Sexual/genética , Testículo/embriologia , Sistema Urogenital/embriologia , Animais , Hormônio Antimülleriano/metabolismo , Embrião de Galinha , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Ovário/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Testículo/metabolismo , Sistema Urogenital/metabolismo
13.
Proc Natl Acad Sci U S A ; 109(50): E3395-404, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23184966

RESUMO

Carcinomas most often result from the stepwise acquisition of genetic alterations within the epithelial compartment. The surrounding stroma can also play an important role in cancer initiation and progression. Given the rare frequencies of genetic events identified in cancer-associated stroma, it is likely that epigenetic changes in the tumor microenvironment could contribute to its tumor-promoting activity. We use Hmga2 (High-mobility group AT-hook 2) an epigenetic regulator, to modify prostate stromal cells, and demonstrate that perturbation of the microenvironment by stromal-specific overexpression of this chromatin remodeling protein alone is sufficient to induce dramatic hyperplasia and multifocal prostatic intraepithelial neoplasia lesions from adjacent naïve epithelial cells. Importantly, we find that this effect is predominantly mediated by increased Wnt/ß-catenin signaling. Enhancement of Hmga2-induced paracrine signaling by overexpression of androgen receptor in the stroma drives frank murine prostate adenocarcinoma in the adjacent epithelial tissues. Our findings provide compelling evidence for the critical contribution of epigenetic changes in stromal cells to multifocal tumorigenesis.


Assuntos
Epigênese Genética , Comunicação Parácrina , Neoplasias da Próstata/etiologia , Via de Sinalização Wnt , Adenocarcinoma/etiologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Hormônio-Dependentes/etiologia , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Próstata/crescimento & desenvolvimento , Próstata/metabolismo , Neoplasia Prostática Intraepitelial/etiologia , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Microambiente Tumoral , Sistema Urogenital/embriologia , Sistema Urogenital/metabolismo
14.
Dev Biol ; 380(1): 25-36, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23685333

RESUMO

The mammalian urogenital system derives from multipotent progenitor cells of different germinal tissues. The contribution of individual sub-populations to specific components of the mature system, and the spatiotemporal restriction of the respective lineages have remained poorly characterized. Here, we use comparative expression analysis to delineate sub-regions within the developing urogenital system that express the T-box transcription factor gene Tbx18. We show that Tbx18 is transiently expressed in the epithelial lining and the subjacent mesenchyme of the urogenital ridge. At the onset of metanephric development Tbx18 expression occurs in a band of mesenchyme in between the metanephros and the Wolffian duct but is subsequently restricted to the mesenchyme surrounding the distal ureter stalk. Genetic lineage tracing reveals that former Tbx18(+) cells of the urogenital ridge and the metanephric field contribute substantially to the adrenal glands and gonads, to the kidney stroma, the ureteric and the bladder mesenchyme. Loss of Tbx18 does not affect differentiation of the adrenal gland, the gonad, the bladder and the kidney. However, ureter differentiation is severely disturbed as the mesenchymal lineage adopts a stromal rather than a ureteric smooth muscle fate. DiI labeling and tissue recombination experiments show that the restriction of Tbx18 expression to the prospective ureteric mesenchyme does not reflect an active condensation process but is due to a specific loss of Tbx18 expression in the mesenchyme out of range of signals from the ureteric epithelium. These cells either contribute to the renal stroma or undergo apoptosis aiding in severing the ureter from its surrounding tissues. We show that Tbx18-deficient cells do not respond to epithelial signals suggesting that Tbx18 is required to prepattern the ureteric mesenchyme. Our study provides new insights into the molecular diversity of urogenital progenitor cells and helps to understand the specification of the ureteric mesenchymal sub-lineage.


Assuntos
Células Estromais/citologia , Proteínas com Domínio T/metabolismo , Sistema Urogenital/embriologia , Animais , Apoptose , Linhagem da Célula , Cruzamentos Genéticos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Hibridização In Situ , Rim/embriologia , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso/patologia , Técnicas de Cultura de Órgãos , Células-Tronco/citologia , Células Estromais/metabolismo , Proteínas com Domínio T/genética , Fatores de Tempo , Ureter/embriologia , Ureter/patologia
15.
J Biol Chem ; 288(26): 18811-24, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23661704

RESUMO

ADAMTS16 (a disintegrin and metalloproteinase with thrombospondin motifs) is a secreted mammalian metalloproteinase with unknown function. We report here that murine Adamts16 is co-expressed with the Wilms tumor protein, Wt1, in the developing glomeruli of embryonic kidneys. Adamts16 mRNA levels were significantly reduced upon transfection of embryonic murine kidney explants with Wt1 antisense vivo-morpholinos. Antisense knockdown of Adamts16 inhibited branching morphogenesis in kidney organ cultures. Adamts16 was detected by in situ mRNA hybridization and/or immunohistochemistry also in embryonic gonads and in spermatids and granulosa cells of adult testes and ovaries, respectively. Silencing of Wt1 by transfection with antisense vivo-morpholinos significantly increased Adamts16 mRNA in cultured embryonic XY gonads (11.5 and 12.5 days postconception), and reduced Adamts16 transcripts in XX gonads (12.5 and 13.5 days postconception). Three predicted Wt1 consensus motifs could be identified in the promoter and the 5'-untranslated region of the murine Adamts16 gene. Binding of Wt1 protein to these elements was verified by EMSA and ChIP. A firefly luciferase reporter gene under control of the Adamts16 promoter was activated ∼8-fold by transient co-transfection of human granulosa cells with a Wt1 expression construct. Gradual shortening of the 5'-flanking sequence successively reduced and eventually abrogated Adamts16 promoter activation by Wt1. These findings demonstrate that Wt1 differentially regulates the Adamts16 gene in XX and XY embryonic gonads. It is suggested that Adamts16 acts immediately downstream of Wt1 during murine urogenital development. We propose that Adamts16 is involved in branching morphogenesis of the kidneys in mice.


Assuntos
Proteínas ADAM/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Sistema Urogenital/embriologia , Proteínas WT1/metabolismo , Proteínas ADAM/metabolismo , Proteínas ADAMTS , Motivos de Aminoácidos , Animais , Feminino , Inativação Gênica , Células da Granulosa/citologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Células de Sertoli/citologia , Fatores de Tempo
16.
Pediatr Nephrol ; 29(4): 757-66, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24500705

RESUMO

Vesicoureteric reflux (VUR) is a common congenital urinary tract defect that predisposes children to recurrent kidney infections. Kidney infections can result in renal scarring or reflux nephropathy defined by the presence of chronic tubulo-interstitial inflammation and fibrosis that is a frequent cause of end-stage renal failure. The discovery of mouse models with VUR and with reflux nephropathy has provided new opportunities to understand the pathogenesis of these conditions and may provide insight on the genes and the associated phenotypes that need to be examined in human studies.


Assuntos
Nefropatias/etiologia , Sistema Urogenital/embriologia , Refluxo Vesicoureteral/complicações , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Refluxo Vesicoureteral/genética
17.
Magn Reson Imaging Clin N Am ; 32(3): 529-551, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38944439

RESUMO

Fetal MR imaging overcomes many of the technical barriers of ultrasound and is an important diagnostic tool for fetal genitourinary (GU) anomalies. It is suited for evaluation of GU anomalies because of the fluid-sensitive sequences and superior soft tissue contrast. Often GU malformations are part of a multisystem genetic or congenital condition, and imaging the entire fetus with MR adds additional clarity about the extent of disease. It adds confidence to diagnoses of renal agenesis, urinary tract dilation, cystic disease, and tumors. It is particularly useful to delineate anatomy in complex GU malformations. This additional information guides counseling.


Assuntos
Imageamento por Ressonância Magnética , Diagnóstico Pré-Natal , Anormalidades Urogenitais , Sistema Urogenital , Humanos , Imageamento por Ressonância Magnética/métodos , Anormalidades Urogenitais/diagnóstico por imagem , Diagnóstico Pré-Natal/métodos , Feminino , Gravidez , Sistema Urogenital/diagnóstico por imagem , Sistema Urogenital/embriologia
18.
Dev Biol ; 360(1): 186-94, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21968101

RESUMO

The evolutionarily conserved Six1-Eya1 transcription complex is central to mammalian organogenesis, and deletion of these genes in mice results in developmental anomalies of multiple organs that recapitulate human branchio-oto-renal (BOR) and DiGeorge syndromes. Here, we report that both Six1 and Eya1 are strongly expressed in the peri-cloacal mesenchyme (PCM) surrounding the cloaca, the terminal end of hindgut dilation. Six1 and Eya1 are absent from the intra-cloacal mesenchyme (ICM), a cell mass that divides the cloaca into dorsal hindgut and ventral urogenital sinus. Deletion of either or both Six1 and Eya1 genes results in a spectrum of genitourinary tract defects including persistent cloaca - hypoplastic perineum tissue between external urogenital and anorectal tracts; hypospadias - ectopic ventral positioning of the urethral orifice; and hypoplastic genitalia. Analyses of critical signaling molecules indicate normal expression of Shh in the cloaca and cloaca-derived endodermal epithelia. Using a Cre/loxP genetic fate mapping strategy, we demonstrate that Six1-positive PCM progenitors give rise to the most caudal structures of the body plan including the urogenital and anorectal complex, and the perineum region. Thus, Six1 and Eya1 are key regulators of both upper and lower urinary tract morphogenesis. Results from this study uncover essential roles of the PCM progenitors during genitourinary tract formation.


Assuntos
Cloaca/embriologia , Cloaca/metabolismo , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Sistema Urogenital/embriologia , Sistema Urogenital/metabolismo , Animais , Padronização Corporal , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proliferação de Células , Sobrevivência Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Modelos Biológicos , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Gravidez , Proteínas Tirosina Fosfatases/deficiência , Proteínas Tirosina Fosfatases/genética , Transdução de Sinais , Anormalidades Urogenitais/embriologia , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/metabolismo
19.
J Anat ; 220(4): 363-71, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22256858

RESUMO

Unresolved questions remain concerning the derivation of the vagina with respect to the relative contributions from the Müllerian ducts, the urogenital sinus, and the Wolffian ducts. Recent molecular and cellular studies in rodents have opened up a large gap between the level of understanding of vaginal development in mice and understanding of human vaginal development, which is based on histology. To compare the findings in mice with human vaginal development and to address this gap, we analysed molecular characteristics of the urogenital sinus, Wolffian ducts, and Müllerian ducts in 8-14-week-old human specimens using immunohistochemical methods. The monoclonal antibodies used were directed against cytokeratin (CK) 14, CK19, vimentin, laminin, p63, E-cadherin, caspase-3, Ki67, HOX A13, and BMP-4. The immunohistochemical analysis revealed that, during weeks 8-9, the epithelium of the Müllerian ducts became positive for p63 as p63-positive cells that originated from the sinus epithelium reached the caudal tip of the fused Müllerian ducts via the Wolffian ducts. The lumen of the fused Müllerian ducts was closed by an epithelial plug that contained both vimentin-positive and vimentin-negative cells. Subsequently, the resulting epithelial tube enlarged by proliferation of basal p63-positive cells. The first signs of squamous differentiation were detected during week 14, with the appearance of CK14-positive cells. According to our results, all three components, namely, the urogenital sinus, Wolffian ducts, and Müllerian ducts, interacted during the formation of the human vagina. The sinus epithelium provided p63-positive cells, the Wollfian ducts acted as a 'transporter', and the Müllerian ducts contributed the guiding structure for the vaginal anlagen. Epithelial differentiation began at the end of the period studied and extended in a caudo-cranial direction. The present study is one of the first to provide up-to-date molecular correlates for human vaginal development that can be compared with the results of cell biological studies in rodents.


Assuntos
Vagina/embriologia , Epitélio/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Ductos Paramesonéfricos/anatomia & histologia , Ductos Paramesonéfricos/embriologia , Ductos Paramesonéfricos/fisiologia , Sistema Urogenital/anatomia & histologia , Sistema Urogenital/embriologia , Sistema Urogenital/fisiologia , Ductos Mesonéfricos/anatomia & histologia , Ductos Mesonéfricos/embriologia , Ductos Mesonéfricos/fisiologia
20.
AJR Am J Roentgenol ; 199(5): W545-53, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23096198

RESUMO

OBJECTIVE: Congenital urinary anomalies may be symptomatic or encountered during imaging for other clinical indications. The array of abnormalities is related to the embryologic stage at the time of the developmental insult, and these abnormalities result in a spectrum of conditions ranging from insignificant to incompatible with life. CONCLUSION: Understanding the implications of common congenital urinary anomalies is the key to detecting associated anomalies, initiating therapy, and avoiding both complications and unnecessary intervention.


Assuntos
Diagnóstico por Imagem , Anormalidades Urogenitais/diagnóstico , Anormalidades Urogenitais/terapia , Diagnóstico Diferencial , Humanos , Sistema Urogenital/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA