Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mech Dev ; 148: 18-39, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28433748

RESUMO

Epithelial structures are foundational for tissue organization in all metazoans. Sheets of epithelial cells form lateral adhesive junctions and acquire apico-basal polarity perpendicular to the surface of the sheet. Genetic analyses in the insect model, Drosophila melanogaster, have greatly advanced our understanding of how epithelial organization is established, and how it is modulated during tissue morphogenesis. Major insights into collective cell migrations have come from analyses of morphogenetic movements within the adult follicular epithelium that cooperates with female germ cells to build a mature egg. Epithelial follicle cells progress through tightly choreographed phases of proliferation, patterning, reorganization and migrations, before they differentiate to form the elaborate structures of the eggshell. Distinct structural domains are organized by differential adhesion, within which lateral junctions are remodeled to further shape the organized epithelia. During collective cell migrations, adhesive interactions mediate supracellular organization of planar polarized macromolecules, and facilitate crawling over the basement membrane or traction against adjacent cell surfaces. Comparative studies with other insects are revealing the diversification of morphogenetic movements for elaboration of epithelial structures. This review surveys the repertoire of follicle cell morphogenesis, to highlight the coordination of epithelial plasticity with progressive differentiation of a secretory epithelium. Technological advances will keep this tissue at the leading edge for interrogating the precise spatiotemporal regulation of normal epithelial reorganization events, and provide a framework for understanding pathological tissue dysplasia.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Epitélio/crescimento & desenvolvimento , Morfogênese/genética , Óvulo/crescimento & desenvolvimento , Animais , Diferenciação Celular/genética , Movimento Celular/genética , Drosophila melanogaster/genética , Feminino , Oogênese/genética , Folículo Ovariano/crescimento & desenvolvimento
2.
Stem Cell Rev Rep ; 11(6): 813-25, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26323255

RESUMO

The Drosophila adult midgut contains intestinal stem cells that support homeostasis and repair. We show here that the leucine zipper protein Bunched and the adaptor protein Madm are novel regulators of intestinal stem cells. MARCM mutant clonal analysis and cell type specific RNAi revealed that Bunched and Madm were required within intestinal stem cells for proliferation. Transgenic expression of a tagged Bunched showed a cytoplasmic localization in midgut precursors, and the addition of a nuclear localization signal to Bunched reduced its function to cooperate with Madm to increase intestinal stem cell proliferation. Furthermore, the elevated cell growth and 4EBP phosphorylation phenotypes induced by loss of Tuberous Sclerosis Complex or overexpression of Rheb were suppressed by the loss of Bunched or Madm. Therefore, while the mammalian homolog of Bunched, TSC-22, is able to regulate transcription and suppress cancer cell proliferation, our data suggest the model that Bunched and Madm functionally interact with the TOR pathway in the cytoplasm to regulate the growth and subsequent division of intestinal stem cells.


Assuntos
Proliferação de Células/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Células-Tronco/citologia , Esclerose Tuberosa/genética , Proteínas Supressoras de Tumor/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Drosophila , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/metabolismo , Intestinos/citologia , Proteínas Monoméricas de Ligação ao GTP/biossíntese , Neuropeptídeos/biossíntese , Interferência de RNA , RNA Interferente Pequeno/genética , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Transdução de Sinais , Células-Tronco/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/metabolismo , Proteínas Supressoras de Tumor/metabolismo
3.
Genes Dev ; 22(18): 2578-90, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18794353

RESUMO

Morphogens are secreted signaling molecules that form concentration gradients and control cell fate in developing tissues. During development, it is essential that morphogen range is strictly regulated in order for correct cell type specification to occur. One of the best characterized morphogens is Drosophila Decapentaplegic (Dpp), a BMP signaling molecule that patterns the dorsal ectoderm of the embryo by activating the Mad and Medea (Med) transcription factors. We demonstrate that there is a spatial and temporal expansion of the expression patterns of Dpp target genes in SUMO pathway mutant embryos. We identify Med as the primary SUMOylation target in the Dpp pathway, and show that failure to SUMOylate Med leads to the increased Dpp signaling range observed in the SUMO pathway mutant embryos. Med is SUMO modified in the nucleus, and we provide evidence that SUMOylation triggers Med nuclear export. Hence, Med SUMOylation provides a mechanism by which nuclei can continue to monitor the presence of extracellular Dpp signal to activate target gene expression for an appropriate duration. Overall, our results identify an unusual strategy for regulating morphogen range that, rather than impacting on the morphogen itself, targets an intracellular transducer.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Transdução de Sinais , Proteína Smad4/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Mutação , Processamento de Proteína Pós-Traducional
4.
Proc Natl Acad Sci U S A ; 105(14): 5414-9, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18375761

RESUMO

TSC22D1, which encodes transforming growth factor beta-stimulated clone 22 (TSC-22), is thought to be a tumor suppressor because its expression is lost in many glioblastoma, salivary gland, and prostate cancers. TSC-22 is the founding member of the TSC-22/DIP/Bun family of leucine zipper transcription factors; its functions have not been investigated in a multicellular environment. Genetic studies in the model organism Drosophila melanogaster often provide fundamental insights into mechanisms disrupted in carcinogenesis, because of the strong evolutionary conservation of molecular mechanisms between flies and humans. Whereas humans and mice have four TSC-22 domain genes with numerous isoforms, Drosophila has only one TSC-22 domain gene, bunched (bun), which encodes both large and small protein isoforms. Surprisingly, Drosophila Bun proteins promote cellular growth and proliferation in ovarian follicle cells. Loss of both large isoforms has the strongest phenotypes, including increased apoptosis. Cultured S2 cells depleted for large Bun isoforms show increased apoptosis and less frequent cell division, with decreased cell size. Altogether, these data indicate that Drosophila TSC-22/DIP/Bun proteins are necessary for cellular growth, proliferation, and survival both in culture and in an epithelial context. Previous work demonstrated that bun prevents recruitment of epithelial cells to a migratory fate and, thus, maintains epithelial organization. We speculate that reduced TSC22D1 expression generally reduces cellular fitness and only contributes to carcinogenesis in specific tissue environments.


Assuntos
Proliferação de Células , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/química , Proteínas Supressoras de Tumor/fisiologia , Animais , Crescimento Celular , Sobrevivência Celular , Células Epiteliais/citologia
5.
Semin Cell Dev Biol ; 19(3): 271-82, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18304845

RESUMO

Epithelial morphogenesis is important for organogenesis and pivotal for carcinogenesis, but mechanisms that control it are poorly understood. The Drosophila follicular epithelium is a genetically tractable model to understand these mechanisms in vivo. This epithelium of follicle cells encases germline cells to create an egg. In this review, we summarize progress toward understanding mechanisms that maintain the epithelium or permit migrations essential for oogenesis. Cell-cell communication is important, but the same signals are used repeatedly to control distinct events. Understanding intrinsic mechanisms that alter responses to developmental signals will be important to understand regulation of cell shape and organization.


Assuntos
Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Morfogênese , Folículo Ovariano/citologia , Óvulo/citologia , Animais , Divisão Celular , Feminino , Óvulo/ultraestrutura , Células-Tronco/citologia
6.
BMC Dev Biol ; 8: 10, 2008 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-18226226

RESUMO

BACKGROUND: Transforming Growth Factor-beta1 stimulated clone-22 (TSC-22) is assumed to act as a negative growth regulator and tumor suppressor. TSC-22 belongs to a family of putative transcription factors encoded by four distinct loci in mammals. Possible redundancy among the members of the TSC-22/Dip/Bun protein family complicates a genetic analysis. In Drosophila, all proteins homologous to the TSC-22/Dip/Bun family members are derived from a single locus called bunched (bun). RESULTS: We have identified bun in an unbiased genetic screen for growth regulators in Drosophila. Rather unexpectedly, bun mutations result in a growth deficit. Under standard conditions, only the long protein isoform BunA - but not the short isoforms BunB and BunC - is essential and affects growth. Whereas reducing bunA function diminishes cell number and cell size, overexpression of the short isoforms BunB and BunC antagonizes bunA function. CONCLUSION: Our findings establish a growth-promoting function of Drosophila BunA. Since the published studies on mammalian systems have largely neglected the long TSC-22 protein version, we hypothesize that the long TSC-22 protein is a functional homolog of BunA in growth regulation, and that it is antagonized by the short TSC-22 protein.


Assuntos
Divisão Celular/genética , Drosophila melanogaster/genética , Genes de Insetos , Fator de Crescimento Transformador beta1/genética , Alelos , Animais , Contagem de Células , Tamanho Celular , Drosophila melanogaster/crescimento & desenvolvimento , Genes Reguladores , Genótipo , Mutação
7.
J Biol Chem ; 278(9): 7431-8, 2003 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-12468551

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) and transforming growth factor-beta (TGF-beta) are key regulators of epithelial cell biology. However, the molecular mechanisms by which either pathway induces growth inhibition and differentiation are incompletely understood. We have identified transforming growth factor-simulated clone-22 (TSC-22) as a target gene of both pathways in intestinal epithelial cells. TSC-22 is member of a family of leucine zipper containing transcription factors with repressor activity. Although little is known regarding its function in mammals, the Drosophila homolog of TSC-22, bunched, plays an essential role in fly development. The ability of PPARgamma to induce TSC-22 was not dependent on an intact TGF-beta1 signaling pathway and was specific for the gamma isoform. Localization studies revealed that TSC-22 mRNA is enriched in the postmitotic epithelial compartment of the normal human colon. Cells transfected with wild-type TSC-22 exhibited reduced growth rates and increased levels of p21 compared with vector-transfected cells. Furthermore, transfection with a dominant negative TSC-22 in which both repressor domains were deleted was able to reverse the p21 induction and growth inhibition caused by activation of either the PPARgamma or TGF-beta pathways. These results place TSC-22 as an important downstream component of PPARgamma and TGF-beta signaling during intestinal epithelial cell differentiation.


Assuntos
Células Epiteliais/citologia , Intestinos/citologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Repressoras/biossíntese , Tiazolidinedionas , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Northern Blotting , Western Blotting , Células COS , Diferenciação Celular , Divisão Celular , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , DNA/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Genes Dominantes , Vetores Genéticos , Humanos , Hibridização In Situ , Proteínas de Filamentos Intermediários/metabolismo , Queratina-20 , Ligantes , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Testes de Precipitina , Estrutura Terciária de Proteína , Rosiglitazona , Transdução de Sinais , Tiazóis/farmacologia , Fatores de Tempo , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA