Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
J Cell Biol ; 166(6): 901-12, 2004 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-15353548

RESUMO

Actopaxin is an actin and paxillin binding protein that localizes to focal adhesions. It regulates cell spreading and is phosphorylated during mitosis. Herein, we identify a role for actopaxin phosphorylation in cell spreading and migration. Stable clones of U2OS cells expressing actopaxin wild-type (WT), nonphosphorylatable, and phosphomimetic mutants were developed to evaluate actopaxin function. All proteins targeted to focal adhesions, however the nonphosphorylatable mutant inhibited spreading whereas the phosphomimetic mutant cells spread more efficiently than WT cells. Endogenous and WT actopaxin, but not the nonphosphorylatable mutant, were phosphorylated in vivo during cell adhesion/spreading. Expression of the nonphosphorylatable actopaxin mutant significantly reduced cell migration, whereas expression of the phosphomimetic increased cell migration in scrape wound and Boyden chamber migration assays. In vitro kinase assays demonstrate that extracellular signal-regulated protein kinase phosphorylates actopaxin, and treatment of U2OS cells with the MEK1 inhibitor UO126 inhibited adhesion-induced phosphorylation of actopaxin and also inhibited cell migration.


Assuntos
Movimento Celular/efeitos dos fármacos , Proteínas dos Microfilamentos/metabolismo , Actinina , Sequência de Aminoácidos , Butadienos/farmacologia , Linhagem Celular Tumoral , Tamanho Celular/genética , Células Clonais , Inibidores Enzimáticos/farmacologia , Adesões Focais/metabolismo , Humanos , Cinética , Proteínas dos Microfilamentos/química , Mutação , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Estrutura Terciária de Proteína
2.
Biochem Biophys Res Commun ; 321(4): 1024-31, 2004 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-15358131

RESUMO

Growth differentiation factor 3 (GDF-3) is structurally a bone morphogenetic protein/growth differentiation factor subfamily member of the TGF-beta superfamily. GDF-3 exhibits highest level of expression in white fat tissue in mice and is greatly induced by high fat diet if fat metabolic pathway is blocked. To identify its biological function, GDF-3 was overexpressed in mice by adenovirus mediated gene transfer. Mice transduced with GDF-3 displayed profound weight gain when fed with high fat diet. The phenotypes included greatly expanded adipose tissue mass, increased body adiposity, highly hypertrophic adipocytes, hepatic steatosis, and elevated plasma leptin. GDF-3 stimulated peroxisome proliferator activated receptor expression in adipocytes, a master nuclear receptor that controls adipogenesis. However, GDF-3 was not involved in blood glucose homeostasis or insulin resistance, a condition associated with obesity. In contrast, similar phenotypes were not observed in GDF-3 mice fed with normal chow, indicating that GDF-3 is only active under high lipid load. Thus, GDF-3 is a new non-diabetic adipogenic factor tightly coupled with fat metabolism.


Assuntos
Tecido Adiposo/crescimento & desenvolvimento , Gorduras na Dieta/administração & dosagem , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/fisiologia , Animais , Sequência de Bases , Glicemia/metabolismo , Tamanho Celular/genética , Tamanho Celular/fisiologia , Células Cultivadas , Citocinas/genética , Citocinas/fisiologia , DNA/genética , Fígado Gorduroso/etiologia , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Expressão Gênica , Fator 3 de Diferenciação de Crescimento , Humanos , Insulina/sangue , Peptídeos e Proteínas de Sinalização Intercelular/genética , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Transdução Genética , Aumento de Peso/genética , Aumento de Peso/fisiologia
3.
Curr Biol ; 14(16): 1436-50, 2004 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-15324660

RESUMO

BACKGROUND: 14-3-3 proteins are abundant and conserved polypeptides that mediate the cellular effects of basophilic protein kinases through their ability to bind specific peptide motifs phosphorylated on serine or threonine. RESULTS: We have used mass spectrometry to analyze proteins that associate with 14-3-3 isoforms in HEK293 cells. This identified 170 unique 14-3-3-associated proteins, which show only modest overlap with previous 14-3-3 binding partners isolated by affinity chromatography. To explore this large set of proteins, we developed a domain-based hierarchical clustering technique that distinguishes structurally and functionally related subsets of 14-3-3 target proteins. This analysis revealed a large group of 14-3-3 binding partners that regulate cytoskeletal architecture. Inhibition of 14-3-3 phosphoprotein recognition in vivo indicates the general importance of such interactions in cellular morphology and membrane dynamics. Using tandem proteomic and biochemical approaches, we identify a phospho-dependent 14-3-3 binding site on the A kinase anchoring protein (AKAP)-Lbc, a guanine nucleotide exchange factor (GEF) for the Rho GTPase. 14-3-3 binding to AKAP-Lbc, induced by PKA, suppresses Rho activation in vivo. CONCLUSION: 14-3-3 proteins can potentially engage around 0.6% of the human proteome. Domain-based clustering has identified specific subsets of 14-3-3 targets, including numerous proteins involved in the dynamic control of cell architecture. This notion has been validated by the broad inhibition of 14-3-3 phosphorylation-dependent binding in vivo and by the specific analysis of AKAP-Lbc, a RhoGEF that is controlled by its interaction with 14-3-3.


Assuntos
Citoesqueleto/fisiologia , Estrutura Terciária de Proteína/fisiologia , Proteínas/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas 14-3-3 , Actinas/fisiologia , Animais , Diferenciação Celular/genética , Tamanho Celular/genética , Células Cultivadas , Análise por Conglomerados , Biologia Computacional , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoesqueleto/genética , Primers do DNA , DNA Complementar/genética , Cães , Imunofluorescência , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Espectrometria de Massas , Camundongos , Fosforilação , Testes de Precipitina , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Proteômica/métodos , Fatores de Troca de Nucleotídeo Guanina Rho , Transfecção
4.
Cancer Res ; 64(16): 5659-63, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15313904

RESUMO

We have established a primary pancreatic duct epithelial cell culture (PDEC) system to investigate the relationship between oncogenic activation of K-ras and pancreatic ductal tumorigenesis. We have found that the acute introduction of physiological levels of oncogenic K-ras (K-rasV12) into quiescent PDECs stimulates S-phase entry and induces a pronounced increase in cell size. Both effects are dependent on the functional integrity of the phosphatidylinositol 3'-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway. In addition, K-rasV12 promotes the loss of epithelial E-cadherin and the gain of mesenchymal N-cadherin in PDEC. Our observations indicate that the oncogenic activation of K-ras is sufficient to elicit mitogenic and morphogenic responses in pancreatic ductal cells and hence is likely to play an instructive role in the initiation of pancreatic ductal adenocarcinoma.


Assuntos
Carcinoma Ductal Pancreático/genética , Transformação Celular Neoplásica/genética , Genes ras/genética , Ductos Pancreáticos/fisiologia , Neoplasias Pancreáticas/genética , Animais , Carcinoma Ductal Pancreático/patologia , Ciclo Celular/genética , Divisão Celular/genética , Tamanho Celular/genética , Transformação Celular Neoplásica/patologia , Células Cultivadas , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica , Masculino , Mutação , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/patologia , Fosfatidilinositol 3-Quinases/fisiologia , Ratos , Ratos Endogâmicos F344
5.
Mol Cell Neurosci ; 26(3): 450-7, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15234349

RESUMO

Many cholinergic parasympathetic and enteric neurons require neurturin signaling through GDNF family receptor GFRalpha2 for target innervation. Since a distinct minority of sympathetic neurons are cholinergic, we examined whether GFRalpha2 is important for their development. We detected GFRalpha2 in neonatal sympathetic cholinergic neurons and neurturin mRNA in their target tissues, sweat glands in footpads, and periosteum. Lack of GFRalpha2 in mice did not affect the number of sympathetic cholinergic neurons, but their soma size was decreased in comparison to wild types. In adult and in 3-week-old GFRalpha2 knockout mice, the density of sympathetic cholinergic innervation was reduced by 50-70% in the sweat glands, and was completely absent in the periosteum. Sympathetic noradrenergic innervation of blood vessels in the footpads was unchanged. The density of sympathetic axons in sweat glands was unaffected at postnatal day P4 reflecting successful growth into the target area. Our results indicate that the cholinergic subpopulation of sympathetic neurons requires GFRalpha2 signaling for soma size and for growth or maintenance of target innervation. Thus, neurturin may be a general target-derived innervation factor for postganglionic cholinergic neurons in all parts of the autonomic nervous system.


Assuntos
Fibras Adrenérgicas/fisiologia , Fibras Colinérgicas/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Animais , Tamanho Celular/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Periósteo/citologia , Periósteo/inervação , Periósteo/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/fisiologia , Glândulas Sudoríparas/citologia , Glândulas Sudoríparas/inervação , Glândulas Sudoríparas/metabolismo
6.
Exp Cell Res ; 298(2): 624-31, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15265708

RESUMO

The coxsackie and adenovirus receptor (CAR) is a key factor in adenoviral cancer gene therapy. Reduced expression of CAR during progression of prostate and bladder cancer has been reported. In embryonic development and tissue differentiation, CAR is also differentially expressed. This study suggests a role of CAR expression in cell adhesion and cell motility of human cancer cells. Stable CAR-expressing clones from E-cadherin-deficient A2780 ovarian and CaSki cervical cancer cells with originally low and high CAR expression levels, respectively, were established. CAR reexpression in otherwise singularly growing A2780 parental cells resulted in formation of cell-cell contacts and aggregation in cell clusters. CAR overexpression in cell adhesion-forming CaSki cells did not result in morphological changes. Migration of the A2780 CAR clones was strongly reduced as characterized by using spread-off assays. Using migration chambers, formation of satellite colonies was reduced by 97% in CAR-expressing A2780 cell clones and by 23% in CAR-expressing CaSki cell clones. Parental A2780 and CaSki cells selected for high migratory ability by using migration chambers expressed endogenous CAR on lower levels associated with lower adenoviral transduction efficiency. Our data suggest CAR as a new inhibitory factor for cancer cell migration.


Assuntos
Movimento Celular/genética , Metástase Neoplásica/fisiopatologia , Neoplasias/metabolismo , Receptores Virais/metabolismo , Adenoviridae/genética , Caderinas/genética , Caderinas/metabolismo , Adesão Celular/genética , Agregação Celular/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Tamanho Celular/genética , Células Clonais/citologia , Células Clonais/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Vetores Genéticos/genética , Humanos , Metástase Neoplásica/genética , Metástase Neoplásica/prevenção & controle , Neoplasias/genética , Receptores Virais/genética , Transdução Genética
7.
J Chem Neuroanat ; 27(4): 237-46, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15261330

RESUMO

In order to furnish a combined model of relevance to human inclusion-body myopathy and Alzheimer's disease, transgenic mice expressing human betaAPP-C99 in skeletal muscle and brain under the control of the cytomegalovirus/beta-actin promoter were produced (Tg13592). These transgenic mice develop Abeta deposits in muscles but not in brain. Cell metabolic activity was analyzed in brain regions and muscle by cytochrome oxidase (CO) histochemistry, the terminal enzyme of the electron transport chain. By comparison to age-matched controls of the C57BL/6 strain, CO activity was selectively increased in dark skeletal muscle fibers of Tg13592 mice. In addition, only increases in CO activity were obtained in those brain regions where a significant difference appeared. The CO activity of Tg13592 mice was elevated in several thalamic nuclei, including laterodorsal, ventromedial, and midline as well as submedial, intralaminar, and reticular. In contrast, the groups did not differ in most cortical regions, except for prefrontal, secondary motor, and auditory cortices, and in most brainstem regions, except for cerebellar (fastigial and interpositus) nuclei and related areas (red and lateral vestibular nuclei). No variation in cell density and surface area appeared in conjunction with these enzymatic alterations. The overproduction of betaAPP-C99 fragments in brain without (amyloidosis did not appear to affect the metabolic activity of structures particularly vulnerable in Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Encéfalo/enzimologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Metabolismo Energético/genética , Músculo Esquelético/enzimologia , Fragmentos de Peptídeos/biossíntese , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Encéfalo/citologia , Contagem de Células , Tamanho Celular/genética , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Miosite de Corpos de Inclusão/genética , Miosite de Corpos de Inclusão/metabolismo , Neurônios/citologia , Neurônios/enzimologia , Fragmentos de Peptídeos/genética , Tálamo/citologia , Tálamo/enzimologia , Regulação para Cima/genética
8.
Glia ; 47(2): 180-8, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15185396

RESUMO

Individuals with tuberous sclerosis complex (TSC) develop central nervous system abnormalities that may reflect astrocyte dysfunction. In an effort to model astrocyte dysfunction in TSC, we generated mice lacking Tsc1 (hamartin) expression in astrocytes and demonstrated that Tsc1-null astrocytes exhibit abnormalities in contact inhibition growth arrest. In this study, we demonstrate that hamartin-deficient astrocytes are also defective in cell size regulation. We show that the increase in Tsc1-null astrocyte size is associated with increased activation of the S6-kinase pathway. In keeping with recent reports that the hamartin/tuberin complex may regulate Rheb and downstream S6K activation, we demonstrate that expression of either Rheb or S6K in primary astrocytes results in increased S6 pathway activation, and that inhibition of Rheb activity in Tsc1-deficient astrocytes using either pharmacologic or genetic strategies markedly reduces S6 activation. Collectively, these observations suggest that TSC inactivation in astrocytes results in defective cell size regulation associated with dysregulated Rheb/mTOR/S6K pathway activity.


Assuntos
Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuropeptídeos/metabolismo , Proteínas/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Esclerose Tuberosa/metabolismo , Animais , Astrócitos/patologia , Diferenciação Celular/genética , Tamanho Celular/genética , Células Cultivadas , Sistema Nervoso Central/fisiopatologia , Inibição de Contato/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Inibidores Enzimáticos/farmacologia , Camundongos , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/antagonistas & inibidores , Proteínas Monoméricas de Ligação ao GTP/genética , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/genética , Inibidores de Proteínas Quinases , Proteínas Quinases/metabolismo , Proteínas/genética , Interferência de RNA , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR , Transfecção , Esclerose Tuberosa/genética , Esclerose Tuberosa/fisiopatologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor , Regulação para Cima/genética
9.
Cell ; 117(7): 899-913, 2004 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-15210111

RESUMO

Cyclin-dependent kinase (CDK) activity initiates the eukaryotic cell division cycle by turning on a suite of gene expression in late G1 phase. In metazoans, CDK-dependent phosphorylation of the retinoblastoma tumor suppressor protein (Rb) alleviates repression of E2F and thereby activates G1/S transcription. However, in yeast, an analogous G1 phase target of CDK activity has remained elusive. Here we show that the cell size regulator Whi5 inhibits G1/S transcription and that this inhibition is relieved by CDK-mediated phosphorylation. Deletion of WHI5 bypasses the requirement for upstream activators of the G1/S transcription factors SBF/MBF and thereby accelerates the G1/S transition. Whi5 is recruited to G1/S promoter elements via its interaction with SBF/MBF in vivo and in vitro. In late G1 phase, CDK-dependent phosphorylation dissociates Whi5 from SBF and drives Whi5 out of the nucleus. Elimination of CDK activity at the end of mitosis allows Whi5 to reenter the nucleus to again repress G1/S transcription. These findings harmonize G1/S control in eukaryotes.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Proteínas Repressoras/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Anticorpos Monoclonais/metabolismo , Núcleo Celular/metabolismo , Tamanho Celular/genética , Cromatina/metabolismo , Cruzamentos Genéticos , Epistasia Genética , Fase G1 , Deleção de Genes , Regulação Fúngica da Expressão Gênica , Genes Fúngicos , Genes Reguladores , Modelos Biológicos , Fosforilação , Testes de Precipitina , Regiões Promotoras Genéticas , Proteínas/análise , RNA/análise , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Fase S , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Especificidade por Substrato , Transcrição Gênica
10.
J Neuropathol Exp Neurol ; 63(5): 429-40, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15198122

RESUMO

Down syndrome is the most frequent genetic cause of mental retardation, having an incidence of 1 in 700 live births. In the present study we used a transgenic mouse in vivo library consisting of 4 yeast artificial chromosome (YAC) transgenic mouse lines, each bearing a different fragment of the Down syndrome critical region 1 (DCR-1), implicated in brain abnormalities characterizing this pathology. The 152F7 fragment, in addition to genes also located on the other DCR-1 fragments, bears the DYRK1A gene, encoding for a serine-threonine kinase. The neurobehavioral analysis of these mouse lines showed that DYRK1A overexpressing 152F7 mice but not the other lines display learning impairment and hyperactivity during development. Additionally, 152F7 mice display increased brain weight and neuronal size. At a biochemical level we found DYRK1A overexpression associated with a development-dependent increase in phosphorylation of the transcription factor FKHR and with high levels of cyclin B1, suggesting for the first time in vivo a correlation between DYRK1A overexpression and cell cycle protein alteration. In addition, we found an altered phosphorylation of transcription factors of CREB family. Our findings support a role of DYRK1A overexpression in the neuronal abnormalities seen in Down syndrome and suggest that this pathology is linked to altered levels of proteins involved in the regulation of cell cycle.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Síndrome de Down/genética , Biblioteca Genômica , Proteínas Musculares/metabolismo , Malformações do Sistema Nervoso/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas , Animais , Proteínas de Ciclo Celular/genética , Tamanho Celular/genética , Aberrações Cromossômicas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ciclina B/metabolismo , Ciclina B1 , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Síndrome de Down/metabolismo , Síndrome de Down/fisiopatologia , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Humanos , Hipercinese/genética , Hipercinese/metabolismo , Hipercinese/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular , Deficiências da Aprendizagem/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Musculares/genética , Mutação/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/fisiopatologia , Tamanho do Órgão/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/genética , Quinases Dyrk
11.
J Cell Biol ; 165(4): 483-91, 2004 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15148306

RESUMO

The process of vascular smooth muscle cell (vSMC) differentiation is critical to embryonic angiogenesis. However, despite its importance, the vSMC differentiation program remains largely undefined. Murine gene disruption studies have identified several gene products that are necessary for vSMC differentiation, but these methodologies cannot establish whether or not a factor is sufficient to initiate the differentiation program. A gain-of-function system consisting of normal vSMC progenitor cells would serve as a useful complement to whole animal loss-of-function studies. We use such a system here, namely freshly isolated rat neural crest stem cells (NCSCs), to show that activation of the calcineurin signaling pathway is sufficient to drive these cells toward a smooth muscle fate. In addition, we present data suggesting that transforming growth factor (TGF)-beta1, which also causes NCSCs to differentiate into smooth muscle, activates calcineurin signaling in NCSCs, leading to a model in which activation of calcineurin signaling is the mechanism by which TGF-beta1 causes SMC differentiation in these cells.


Assuntos
Calcineurina/metabolismo , Diferenciação Celular/genética , Músculo Liso Vascular/embriologia , Neovascularização Fisiológica/genética , Crista Neural/metabolismo , Proteínas Nucleares , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Calcineurina/genética , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Tamanho Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Biológicos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Mutação/genética , Fatores de Transcrição NFATC , Neovascularização Fisiológica/efeitos dos fármacos , Crista Neural/citologia , Crista Neural/efeitos dos fármacos , Fenótipo , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Regulação para Cima/genética
12.
J Immunol ; 172(10): 6020-9, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15128785

RESUMO

B-1 cells, distinguishable from conventional B-2 cells by their cell surface marker, anatomical location, and self-replenishing activity, play an important role in innate immune responses. B-1 cells constitutively express the IL-5R alpha-chain (IL-5Ralpha) and give rise to Ab-producing cells in response to various stimuli, including IL-5 and LPS. Here we report that the IL-5/IL-5R system plays an important role in maintaining the number and the cell size as well as the functions of mature B-1 cells. The administration of anti-IL-5 mAb into wild-type mice, T cell-depleted mice, or mast cell-depleted mice resulted in reduction in the total number and cell size of B-1 cells to an extent similar to that of IL-5Ralpha-deficient (IL-5Ralpha(-/-)) mice. Cell transfer experiments have demonstrated that B-1 cell survival in wild-type mice and homeostatic proliferation in recombination-activating gene 2-deficient mice are impaired in the absence of IL-5Ralpha. IL-5 stimulation of wild-type B-1 cells, but not IL-5Ralpha(-/-) B-1 cells, enhances CD40 expression and augments IgM and IgG production after stimulation with anti-CD40 mAb. Enhanced IgA production in feces induced by the oral administration of LPS was not observed in IL-5Ralpha(-/-) mice. Our results illuminate the role of IL-5 in the homeostatic proliferation and survival of mature B-1 cells and in IgA production in the mucosal tissues.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Homeostase/imunologia , Imunoglobulinas/biossíntese , Interleucina-5/fisiologia , Animais , Anticorpos Monoclonais/administração & dosagem , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/patologia , Antígenos CD40/biossíntese , Antígenos CD40/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Divisão Celular/genética , Divisão Celular/imunologia , Tamanho Celular/genética , Tamanho Celular/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Homeostase/genética , Deficiência de IgA/genética , Deficiência de IgA/imunologia , Injeções Intraperitoneais , Interleucina-5/biossíntese , Interleucina-5/imunologia , Lipopolissacarídeos/administração & dosagem , Linfopenia/imunologia , Linfopenia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Receptores de Interleucina/deficiência , Receptores de Interleucina/genética , Receptores de Interleucina/fisiologia , Receptores de Interleucina-5 , Linfócitos T/patologia
13.
Dev Cell ; 6(4): 483-95, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15068789

RESUMO

Commitment of stem cells to different lineages is regulated by many cues in the local tissue microenvironment. Here we demonstrate that cell shape regulates commitment of human mesenchymal stem cells (hMSCs) to adipocyte or osteoblast fate. hMSCs allowed to adhere, flatten, and spread underwent osteogenesis, while unspread, round cells became adipocytes. Cell shape regulated the switch in lineage commitment by modulating endogenous RhoA activity. Expressing dominant-negative RhoA committed hMSCs to become adipocytes, while constitutively active RhoA caused osteogenesis. However, the RhoA-mediated adipogenesis or osteogenesis was conditional on a round or spread shape, respectively, while constitutive activation of the RhoA effector, ROCK, induced osteogenesis independent of cell shape. This RhoA-ROCK commitment signal required actin-myosin-generated tension. These studies demonstrate that mechanical cues experienced in developmental and adult contexts, embodied by cell shape, cytoskeletal tension, and RhoA signaling, are integral to the commitment of stem cell fate.


Assuntos
Linhagem da Célula/genética , Citoesqueleto/metabolismo , Células-Tronco/metabolismo , Proteína rhoA de Ligação ao GTP/deficiência , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Adipócitos/enzimologia , Adipócitos/ultraestrutura , Comunicação Celular/genética , Contagem de Células , Diferenciação Celular/genética , Tamanho Celular/genética , Células Cultivadas , Citoesqueleto/ultraestrutura , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mesoderma/enzimologia , Mesoderma/ultraestrutura , Mutação/genética , Miosinas/genética , Miosinas/metabolismo , Osteoblastos/enzimologia , Osteoblastos/ultraestrutura , Osteogênese/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Células-Tronco/ultraestrutura , Estresse Mecânico , Quinases Associadas a rho , Proteína rhoA de Ligação ao GTP/genética
14.
Eukaryot Cell ; 3(2): 430-46, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15075273

RESUMO

To identify new nonessential genes that affect genome integrity, we completed a screening for diploid mutant Saccharomyces cerevisiae strains that are sensitive to ionizing radiation (IR) and found 62 new genes that confer resistance. Along with those previously reported (Bennett et al., Nat. Genet. 29:426-434, 2001), these genes bring to 169 the total number of new IR resistance genes identified. Through the use of existing genetic and proteomic databases, many of these genes were found to interact in a damage response network with the transcription factor Ccr4, a core component of the CCR4-NOT and RNA polymerase-associated factor 1 (PAF1)-CDC73 transcription complexes. Deletions of individual members of these two complexes render cells sensitive to the lethal effects of IR as diploids, but not as haploids, indicating that the diploid G1 cell population is radiosensitive. Consistent with a role in G1, diploid ccr4Delta cells irradiated in G1 show enhanced lethality compared to cells exposed as a synchronous G2 population. In addition, a prolonged RAD9-dependent G1 arrest occurred following IR of ccr4Delta cells and CCR4 is a member of the RAD9 epistasis group, thus confirming a role for CCR4 in checkpoint control. Moreover, ccr4Delta cells that transit S phase in the presence of the replication inhibitor hydroxyurea (HU) undergo prolonged cell cycle arrest at G2 followed by cellular lysis. This S-phase replication defect is separate from that seen for rad52 mutants, since rad52Delta ccr4Delta cells show increased sensitivity to HU compared to rad52Delta or ccr4Delta mutants alone. These results indicate that cell cycle transition through G1 and S phases is CCR4 dependent following radiation or replication stress.


Assuntos
Ribonucleases/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/efeitos da radiação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Tamanho Celular/genética , RNA Helicases DEAD-box , Replicação do DNA/fisiologia , Diploide , Fase G1/genética , Deleção de Genes , Regulação Fúngica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Homeostase/genética , Interfase/genética , Fatores Matadores de Levedura , Micotoxinas/metabolismo , Proteína Quinase C/genética , Proteína Quinase C/fisiologia , RNA Helicases/genética , RNA Helicases/fisiologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Tolerância a Radiação/genética , Radiação Ionizante , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Ribonucleases/genética , Fase S/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
15.
Acta Cytol ; 48(2): 243-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15085761

RESUMO

BACKGROUND: Pulmonary papillary adenoma is a benign pulmonary neoplasm. Previously pulmonary papillary adenoma was described in terms of immunohistochemistry and ultrastructure. However, there are no previous reports describing the cytologic characteristics of pulmonary papillary adenoma. CASE: A 50-year-old male was admitted for evaluation of a coin lesion in the left upper lung field. Radiologic images showed a solid, round tumor approximately 25 mm in diameter in the left upper lung. Transbronchial needle aspiration biopsy (TBNA) was performed, and small numbers of atypical cells were collected. Adenocarcinoma was suggested clinically, and left upper segmentectomy was performed. The histologic diagnosis was pulmonary papillary adenoma. Imprint cytology of the cut surface of the tumor showed tumor cells arranged in sheets that contained scant or vesicular cytoplasm. The nuclei were oval or round, without obvious anisokaryosis, and their chromatin was fine, without hyperchromasia. Cytologically, the nuclei of the tumor cells in the imprint specimen (38.70 +/- 8.69 microns 2) were uniform in size and similar to the atypical cells in the TBNA specimen (38.29 +/- 11.56 microns 2) but significantly larger than the nuclei of the bronchial cells (23.61 +/- 5.98 microns 2) (P < .0001). CONCLUSION: The cytologic appearance of pulmonary papillary adenoma was characterized morphologically and morphometrically. The possibility of cytodiagnosis by TBNA was suggested.


Assuntos
Adenoma/patologia , Brônquios/patologia , Neoplasias Pulmonares/patologia , Adenoma/diagnóstico por imagem , Adenoma/genética , Biomarcadores Tumorais/metabolismo , Biópsia por Agulha Fina , Brônquios/metabolismo , Núcleo Celular/genética , Núcleo Celular/patologia , Tamanho Celular/genética , Aberrações Cromossômicas , Citoplasma/patologia , Diagnóstico Diferencial , Humanos , Imuno-Histoquímica , Cariometria , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/genética , Masculino , Pessoa de Meia-Idade , Proteína A Associada a Surfactante Pulmonar/metabolismo , Radiografia
16.
Blood ; 103(10): 3751-9, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-14764529

RESUMO

Lineage-marker depleted (Lin(-)) murine bone marrow cells expressing stem cell antigen 1 (Sca-1) were sorted on the basis of stem cell factor receptor (c-kit) expression to obtain Lin(-)Sca(+)Kit(+) or Lin(-)Sca(+)Kit(-) cells. Lin(-)Sca(+)Kit(-) cells have a markedly greater chemotactic response to stromal derived factor-1 (SDF-1). Using a novel fluorescent stain, we show that both populations generate similar levels of a key messenger, phosphatidylinositol 3,4,5 trisphosphate (PIP(3)), in response to SDF-1. Differences in motile behavior may therefore lie downstream of phosphatidylinositol 3-kinase (PI3-kinase) activation at the level of cytoskeleton regulation. The 2 cell populations were compared using 2-dimensional difference gel electrophoresis (2D-DIGE), with a maleimide CyDye fluorescent protein labeling technique that has enhanced sensitivity for low abundance samples. Comparative proteomic analysis of Cy3- and Cy5-labeled protein samples allows relative quantification of protein spots present in both cell populations; of these, 73% were common. Key protein differences were adseverin and gelsolin, actin micro-filament splicing proteins, regulated by Rac, downstream of PI3-kinase activation. Adseverin was shown to be acetylated, a novel modification for this protein. Differences in major regulators of cell shape and motility between the 2 populations can explain the differential response to SDF-1.


Assuntos
Quimiotaxia/genética , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Proteômica , Proteínas Proto-Oncogênicas c-kit/análise , Acetilação , Animais , Células da Medula Óssea , Tamanho Celular/genética , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacologia , Quimiotaxia/efeitos dos fármacos , Gelsolina/análise , Células-Tronco Hematopoéticas/química , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/análise , Proteínas dos Microfilamentos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/análise , Proteínas/análise , Proteínas/genética
17.
Brain Res Bull ; 62(4): 335-43, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14709348

RESUMO

Morphological features of interneuronal adaptation to an altered, more complex neuronal architecture have been investigated in p21H-Ras(Val12) transgenic mice. This transgenic strain serves as a model for studying the morphogenetic role of the G-protein p21Ras on cortical principal neurons. We have recently demonstrated that postmitotic expression of constitutively active p21H-Ras(Val12) in the neocortical pyramidal cell population results in increased size and dendritic complexity of the affected neurons, leading to an enlarged cortical volume. Interneurons do not express the transgene and are therefore excluded from direct, intrinsic p21H-Ras(Val12) effects. In the present study, immunolabelling of gamma-amino-butyric-acid (GABA), and of the calcium-binding proteins parvalbumin, calbindin and calretinin revealed that in the transgenic mice local circuit neurons are not increased in either somal size or number and their main morphological characteristics are preserved. However, the dendritic arbour of interneurons was found to be extended, at least in the vertical dimension, to follow the cortical expansion. Immunostaining for the vesicular GABA transporter revealed a denser inhibitory innervation of p21H-Ras(Val12)-expressing pyramidal cell perikarya than in those of wild-type animals, while the overall density of inhibitory axon terminals within the cortex was decreased in the transgenic animals as a consequence of cortical expansion. The findings of the present study demonstrate the morphogenetic capacity of interneurons for adapting to morphological alterations of principal neurons in the cerebral cortex.


Assuntos
Adaptação Fisiológica/genética , Interneurônios/citologia , Neocórtex/citologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Células Piramidais/citologia , Animais , Tamanho Celular/genética , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neocórtex/metabolismo , Células Piramidais/metabolismo
18.
J Cell Biol ; 164(1): 133-44, 2004 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-14699089

RESUMO

Remodelling of the plasma membrane cytoarchitecture is crucial for the regulation of epithelial cell adhesion and permeability. In Madin-Darby canine kidney cells, the protein AHNAK relocates from the cytosol to the cytosolic surface of the plasma membrane during the formation of cell-cell contacts and the development of epithelial polarity. This targeting is reversible and regulated by Ca(2+)-dependent cell-cell adhesion. At the plasma membrane, AHNAK associates as a multimeric complex with actin and the annexin 2/S100A10 complex. The S100A10 subunit serves to mediate the interaction between annexin 2 and the COOH-terminal regulatory domain of AHNAK. Down-regulation of both annexin 2 and S100A10 using an annexin 2-specific small interfering RNA inhibits the association of AHNAK with plasma membrane. In Madin-Darby canine kidney cells, down-regulation of AHNAK using AHNAK-specific small interfering RNA prevents cortical actin cytoskeleton reorganization required to support cell height. We propose that the interaction of AHNAK with the annexin 2/S100A10 regulates cortical actin cytoskeleton organization and cell membrane cytoarchitecture.


Assuntos
Anexina A2/metabolismo , Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas S100/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animais , Anexina A2/antagonistas & inibidores , Anexina A2/genética , Adesão Celular/genética , Comunicação Celular/genética , Linhagem Celular Tumoral , Membrana Celular/ultraestrutura , Polaridade Celular/genética , Tamanho Celular/genética , Citosol/metabolismo , Citosol/ultraestrutura , Cães , Regulação para Baixo/genética , Células Epiteliais/ultraestrutura , Humanos , Junções Intercelulares/metabolismo , Junções Intercelulares/ultraestrutura , Substâncias Macromoleculares , Estrutura Terciária de Proteína/genética , RNA Interferente Pequeno
19.
J Neurosci Res ; 74(6): 868-74, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14648591

RESUMO

The small G protein p21Ras is a key signal transducer mediating cellular growth and proliferation responses to extracellular stimuli. We investigated by electron microscopy the effects of augmented p21Ras activity on neuronal processes and microtubule arrangement in vivo. We used transgenic mice with a neuron-specific overexpression of p21H-RasVal12, which starts postnatally around Day 15. Axonal and dendritic diameters and the numerical density of dendritic microtubules were analyzed at postnatal Day 12 before the onset of transgene expression and in adult mice. In adult transgenic mice, calibers of both axons (corpus callosum) and dendrites (layers II/III of somatosensory cortex) were enlarged by about 57% and 79%, respectively. The increase in dendritic calibers was associated with an increment in the amount of microtubules. Even in dendrites of equivalent diameters, the number of microtubules was higher in transgenic mice compared to that in wild-type mice suggesting an elevated microtubule density. Changes in process diameters or microtubule density were not observed at postnatal Day 12 before relevant transcription of transgenic p21H-RasVal12. The present results extend previous findings on neuronal hypertrophy as a consequence of p21H-RasVal12 expression and suggest a profound influence on the dendritic microtubule network.


Assuntos
Axônios/metabolismo , Dendritos/metabolismo , Regulação da Expressão Gênica/fisiologia , Microtúbulos/metabolismo , Neurônios/metabolismo , Proteína Oncogênica p21(ras)/biossíntese , Proteína Oncogênica p21(ras)/genética , Transgenes/genética , Animais , Axônios/ultraestrutura , Tamanho Celular/genética , Corpo Caloso/metabolismo , Corpo Caloso/ultraestrutura , Dendritos/genética , Dendritos/ultraestrutura , Camundongos , Camundongos Transgênicos , Microtúbulos/genética , Microtúbulos/ultraestrutura , Neurônios/ultraestrutura , Valina/genética
20.
J Neurol Sci ; 216(1): 135-41, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14607315

RESUMO

Oxidative stress is thought to be a major contributor to the progress of the Parkinson's Disease (PD) because of the high vulnerability of dopaminergic cells against oxidative stress. The present work demonstrates that with the expression of the baculovirus p35 gene, PC12 cells could gain a high resistance against oxidative toxicants, hydrogen peroxide (H(2)O(2)) and 6-hydroxydopamine (6-OHDA). The DNA fragmentation analysis showed that PC12 cells underwent apoptosis after exposure to H(2)O(2) or 6-OHDA, while PP35 cells, a p35-expressing PC12 cell line, did not. Flow cytometric analysis showed that treatment with 150 microM H(2)O(2) or 120 microM 6-OHDA for 24 h caused 52.86% or 66.36% apoptotic cell, respectively, in PC 12 cells, but only 4.26% or 5.80% in PP35 cells. The cell viability measured by 3-(4,5-dimethylthiazal-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay indicated that H(2)O(2) and 6-OHDA induced a dose-dependent cell death on PC12 cells that were greatly remitted on PP35 cells. The viability of PP35 cells was even stronger than that of PC12 cells protected by glial cell line deprived neurotrophic factor (GDNF). The surviving PP35 cells remained normal cell morphology and showed positive with tyrosine hydroxylase (TH) immunocytochemical staining. These results indicate that baculovirus p35 gene possesses remarkable ability to rescue PC12 cells from death in experimental paradigms associated with oxidative stress.


Assuntos
Morte Celular/genética , Resistência a Medicamentos/genética , Estresse Oxidativo/genética , Doença de Parkinson/genética , Proteínas Virais/genética , Animais , Morte Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Tamanho Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Peróxido de Hidrogênio/farmacologia , Proteínas Inibidoras de Apoptose , Fatores de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotoxinas/farmacologia , Oxidantes/farmacologia , Oxidopamina/farmacologia , Células PC12 , Doença de Parkinson/metabolismo , Doença de Parkinson/terapia , Ratos , Transfecção , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA