Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 296: 100488, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33662399

RESUMO

Differentiation of mesenchymal stem cells into adipocyte requires coordination of external stimuli and depends upon the functionality of the primary cilium. The Rab8 small GTPases are regulators of intracellular transport of membrane-bound structural and signaling cargo. However, the physiological contribution of the intrinsic trafficking network controlled by Rab8 to mesenchymal tissue differentiation has not been fully defined in vivo and in primary tissue cultures. Here, we show that mouse embryonic fibroblasts (MEFs) lacking Rab8 have severely impaired adipocyte differentiation in vivo and ex vivo. Immunofluorescent localization and biochemical analyses of Rab8a-deficient, Rab8b-deficient, and Rab8a and Rab8b double-deficient MEFs revealed that Rab8 controls the Lrp6 vesicular compartment, clearance of basal signalosome, traffic of frizzled two receptor, and thereby a proper attenuation of Wnt signaling in differentiating MEFs. Upon induction of adipogenesis program, Rab8a- and Rab8b-deficient MEFs exhibited severely defective lipid-droplet formation and abnormal cilia morphology, despite overall intact cilia growth and ciliary cargo transport. Our results suggest that intracellular Rab8 traffic regulates induction of adipogenesis via proper positioning of Wnt receptors for signaling control in mesenchymal cells.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Via de Sinalização Wnt , Proteínas rab de Ligação ao GTP/metabolismo , Adipogenia/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Cílios/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Proteínas rab de Ligação ao GTP/genética
2.
Sci Signal ; 11(511)2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29295957

RESUMO

Asymmetric cell division results in two distinctly fated daughter cells. A molecular hallmark of asymmetric division is the unequal partitioning of cell fate determinants. We have previously established that growth factor signaling promotes protein depalmitoylation to foster polarized protein localization, which, in turn, drives migration and metastasis. We report protein palmitoylation as a key mechanism for the asymmetric partitioning of the cell fate determinants Numb and ß-catenin through the activity of the depalmitoylating enzyme APT1. Using point mutations, we showed that specific palmitoylated residues on Numb were required for its asymmetric localization. By live-cell imaging, we showed that reciprocal interactions between APT1 and the Rho family GTPase CDC42 promoted the asymmetric localization of Numb and ß-catenin to the plasma membrane. This, in turn, restricted Notch- or Wnt-responsive transcriptional activity to one daughter cell. Moreover, we showed that altering APT1 abundance changed the transcriptional signatures of MDA-MB-231 triple receptor-negative breast cancer cells, similar to changes in Notch and ß-catenin-mediated Wnt signaling. We also showed that loss of APT1 depleted a specific subpopulation of tumorigenic cells in colony formation assays. Together, our findings suggest that APT1-mediated depalmitoylation is a major mechanism of asymmetric cell division that maintains Notch- and Wnt-associated protein dynamics, gene expression, and cellular functions.


Assuntos
Divisão Celular Assimétrica/genética , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Notch/metabolismo , Tioléster Hidrolases/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , beta Catenina/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Feminino , Humanos , Lipoilação , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Mutação Puntual , Receptores Notch/genética , Tioléster Hidrolases/genética , Neoplasias de Mama Triplo Negativas/genética , Via de Sinalização Wnt , beta Catenina/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
3.
Mol Cell ; 62(3): 385-396, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-27153536

RESUMO

Inappropriate activation of the receptor tyrosine kinase EGFR contributes to a variety of human malignancies. Here we show a mechanism to induce vulnerability to an existing first line treatment for EGFR-driven cancers. We find that inhibiting the palmitoyltransferase DHHC20 creates a dependence on EGFR signaling for cancer cell survival. The loss of palmitoylation increases sustained EGFR signal activation and sensitizes cells to EGFR tyrosine kinase inhibition. Our work shows that the reversible modification of EGFR with palmitate "pins" the unstructured C-terminal tail to the plasma membrane, impeding EGFR activation. We identify by mass spectrometry palmitoylated cysteine residues within the C-terminal tail where mutation of the cysteine residues to alanine is sufficient to activate EGFR signaling promoting cell migration and transformation. Our results reveal that the targeting of a peripheral modulator of EGFR signaling, DHHC20, causes a loss of signal regulation and susceptibility to EGFR inhibitor-induced cell death.


Assuntos
Aciltransferases/metabolismo , Neoplasias da Mama/enzimologia , Receptores ErbB/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Aciltransferases/genética , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Morte Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cisteína , Endocitose , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/química , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/genética , Proteína Adaptadora GRB2/metabolismo , Gefitinibe , Células HEK293 , Humanos , Lipoilação , Espectrometria de Massas , Camundongos , Mutação , Células NIH 3T3 , Conformação Proteica , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Proteólise , Quinazolinas/farmacologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Transfecção
4.
Development ; 142(12): 2147-62, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26015543

RESUMO

Communication between stem and niche supporting cells maintains the homeostasis of adult tissues. Wnt signaling is a crucial regulator of the stem cell niche, but the mechanism that governs Wnt ligand delivery in this compartment has not been fully investigated. We identified that Wnt secretion is partly dependent on Rab8a-mediated anterograde transport of Gpr177 (wntless), a Wnt-specific transmembrane transporter. Gpr177 binds to Rab8a, depletion of which compromises Gpr177 traffic, thereby weakening the secretion of multiple Wnts. Analyses of generic Wnt/ß-catenin targets in Rab8a knockout mouse intestinal crypts indicate reduced signaling activities; maturation of Paneth cells - a Wnt-dependent cell type - is severely affected. Rab8a knockout crypts show an expansion of Lgr5(+) and Hopx(+) cells in vivo. However, in vitro, the knockout enteroids exhibit significantly weakened growth that can be partly restored by exogenous Wnts or Gsk3ß inhibitors. Immunogold labeling and surface protein isolation identified decreased plasma membrane localization of Gpr177 in Rab8a knockout Paneth cells and fibroblasts. Upon stimulation by exogenous Wnts, Rab8a-deficient cells show ligand-induced Lrp6 phosphorylation and transcriptional reporter activation. Rab8a thus controls Wnt delivery in producing cells and is crucial for Paneth cell maturation. Our data highlight the profound tissue plasticity that occurs in response to stress induced by depletion of a stem cell niche signal.


Assuntos
Celulas de Paneth/citologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/citologia , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Proteínas de Homeodomínio/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Receptores Acoplados a Proteínas G/metabolismo , Ativação Transcricional , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Proteínas rab de Ligação ao GTP/genética
5.
J Biol Chem ; 289(46): 32030-32043, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25271168

RESUMO

Rab11a has been conceived as a prominent regulatory component of the recycling endosome, which acts as a nexus in the endo- and exocytotic networks. The precise in vivo role of Rab11a in mouse embryonic development is unknown. We globally ablated Rab11a and examined the phenotypic and molecular outcomes in Rab11a(null) blastocysts and mouse embryonic fibroblasts. Using multiple trafficking assays and complementation analyses, we determined, among multiple important membrane-associated and soluble cargos, the critical contribution of Rab11a vesicular traffic to the secretion of multiple soluble MMPs. Rab11a(null) embryos were able to properly form normal blastocysts but died at peri-implantation stages. Our data suggest that Rab11a critically controls mouse blastocyst development and soluble matrix metalloproteinase secretion.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Metaloproteinases da Matriz/metabolismo , Proteínas rab de Ligação ao GTP/fisiologia , Fosfatase Alcalina/metabolismo , Alelos , Animais , Blastocisto/citologia , Desenvolvimento Embrionário , Feminino , Fibroblastos/citologia , Genoma , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Gravidez , Prenhez , Transferrina/metabolismo , Proteínas rab de Ligação ao GTP/genética
6.
EMBO J ; 33(17): 1882-95, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25063677

RESUMO

Compartmentalization of Toll-like receptors (TLRs) in intestinal epithelial cells (IECs) regulates distinct immune responses to microbes; however, the specific cellular machinery that controls this mechanism has not been fully identified. Here we provide genetic evidences that the recycling endosomal compartment in enterocytes maintains a homeostatic TLR9 intracellular distribution, supporting mucosal tolerance to normal microbiota. Genetic ablation of a recycling endosome resident small GTPase, Rab11a, a gene adjacent to a Crohn's disease risk locus, in mouse IECs and in Drosophila midgut caused epithelial cell-intrinsic cytokine production, inflammatory bowel phenotype, and early mortality. Unlike wild-type controls, germ-free Rab11a-deficient mouse intestines failed to tolerate the intraluminal stimulation of microbial agonists. Thus, Rab11a endosome controls intestinal host-microbial homeostasis at least partially via sorting TLRs.


Assuntos
Proteínas de Drosophila/metabolismo , Endossomos/metabolismo , Enterócitos/imunologia , Enterócitos/microbiologia , Microbiota/imunologia , Receptor Toll-Like 9/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Endossomos/imunologia , Deleção de Genes , Homeostase , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Receptor Toll-Like 9/imunologia , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/imunologia
7.
J Clin Invest ; 122(3): 1052-65, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22354172

RESUMO

The constant self renewal and differentiation of adult intestinal stem cells maintains a functional intestinal mucosa for a lifetime. However, the molecular mechanisms that regulate intestinal stem cell division and epithelial homeostasis are largely undefined. We report here that the small GTPases Cdc42 and Rab8a are critical regulators of these processes in mice. Conditional ablation of Cdc42 in the mouse intestinal epithelium resulted in the formation of large intracellular vacuolar structures containing microvilli (microvillus inclusion bodies) in epithelial enterocytes, a phenotype reminiscent of human microvillus inclusion disease (MVID), a devastating congenital intestinal disorder that results in severe nutrient deprivation. Further analysis revealed that Cdc42-deficient stem cells had cell division defects, reduced capacity for clonal expansion and differentiation into Paneth cells, and increased apoptosis. Cdc42 deficiency impaired Rab8a activation and its association with multiple effectors, and prevented trafficking of Rab8a vesicles to the midbody. This impeded cytokinesis, triggering crypt apoptosis and disrupting epithelial morphogenesis. Rab8a was also required for Cdc42-GTP activity in the intestinal epithelium, where continued cell division takes place. Furthermore, mice haploinsufficient for both Cdc42 and Rab8a in the intestine demonstrated abnormal crypt morphogenesis and epithelial transporter physiology, further supporting their functional interaction. These data suggest that defects of the stem cell niche can cause MVID. This hypothesis represents a conceptual departure from the conventional view of this disease, which has focused on the affected enterocytes, and suggests stem cell-based approaches could be beneficial to infants with this often lethal condition.


Assuntos
Regulação da Expressão Gênica , Intestinos/citologia , Células-Tronco/citologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Transporte Biológico , Ciclo Celular , Diferenciação Celular , Sobrevivência Celular , Células HeLa , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Modelos Biológicos , Modelos Genéticos , Fenótipo
8.
Front Biol (Beijing) ; 7(6): 587-593, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23439944

RESUMO

Throughout the animal kingdom, Wnt-triggered signal transduction pathways play fundamental roles in embryonic development and tissue homeostasis. Wnt proteins are modified as glycolipoproteins and are secreted into the extracellular environment as morphogens. Recent studies on the intracellular trafficking of Wnt proteins demonstrate multiple layers of regulation along its secretory pathway. These findings have propelled a great deal of interest among researchers to further investigate the molecular mechanisms that control the release of Wnts and hence the level of Wnt signaling. This review is dedicated to Wntless, a putative G-protein coupled receptor that transports Wnts intracellularly for secretion. Here, we highlight the conclusions drawn from the most recent cellular, molecular and genetic studies that affirm the role of Wntless in the secretion of Wnt proteins.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...