Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Br J Cancer ; 92(6): 1126-9, 2005 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-15756273

RESUMO

Mutations in LKB1 lead to Peutz-Jeghers syndrome (PJS). However, only a subset of PJS patients harbours LKB1 mutations. We performed a mutation analysis of three genes encoding novel LKB1-interacting proteins, BRG1, STRADalpha, and MO25alpha, in 28 LKB1-negative PJS patients. No disease-causing mutations were detected in the studied genes in PJS patients from different European populations.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Mutação , Proteínas Nucleares/genética , Síndrome de Peutz-Jeghers/genética , Fatores de Transcrição/genética , DNA Helicases , Humanos , Íntrons , Polimorfismo Genético
2.
Mol Biol Cell ; 12(12): 3987-99, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11739795

RESUMO

In normal cells, activation of cyclin-dependent kinases (cdks) requires binding to a cyclin and phosphorylation by the cdk-activating kinase (CAK). The Kaposi's sarcoma-associated herpesvirus encodes a protein with similarity to D-type cyclins. This KSHV-cyclin activates CDK6, alters its substrate specificity, and renders CDK6 insensitive to inhibition by the cdk inhibitor p16(INK4a). Here we investigate the regulation of the CDK6/KSHV-cyclin kinase with the use of purified proteins and a cell-based assay. We find that KSHV-cyclin can activate CDK6 independent of phosphorylation by CAK in vitro. In addition, CAK phosphorylation decreased the p16(INK4a) sensitivity of CDK6/KSHV-cyclin complexes. In cells, expression of CDK6 or to a lesser degree of a nonphosphorylatable CDK6(T177A) together with KSHV-cyclin induced apoptosis, indicating that CDK6 activation by KSHV-cyclin can proceed in the absence of phosphorylation by CAK in vivo. Coexpression of p16 partially protected cells from cell death. p16 and KSHV-cyclin can form a ternary complex with CDK6 that can be detected by binding assays as well as by conformational changes in CDK6. The Kaposi's sarcoma-associated herpesvirus has adopted a clever strategy to render cell cycle progression independent of mitogenic signals, cdk inhibition, or phosphorylation by CAK.


Assuntos
Apoptose/efeitos dos fármacos , Quinases Ciclina-Dependentes , Ciclinas/farmacologia , Herpesvirus Humano 8 , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/farmacologia , Quinase 6 Dependente de Ciclina , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Imunofluorescência , Humanos , Fosforilação/efeitos dos fármacos , Conformação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Células Tumorais Cultivadas , Quinase Ativadora de Quinase Dependente de Ciclina
3.
Science ; 293(5533): 1323-6, 2001 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-11509733

RESUMO

The LKB1 tumor suppressor gene, mutated in Peutz-Jeghers syndrome, encodes a serine/threonine kinase of unknown function. Here we show that mice with a targeted disruption of Lkb1 die at midgestation, with the embryos showing neural tube defects, mesenchymal cell death, and vascular abnormalities. Extraembryonic development was also severely affected; the mutant placentas exhibited defective labyrinth layer development and the fetal vessels failed to invade the placenta. These phenotypes were associated with tissue-specific deregulation of vascular endothelial growth factor (VEGF) expression, including a marked increase in the amount of VEGF messenger RNA. Moreover, VEGF production in cultured Lkb1(-/-) fibroblasts was elevated in both normoxic and hypoxic conditions. These findings place Lkb1 in the VEGF signaling pathway and suggest that the vascular defects accompanying Lkb1 loss are mediated at least in part by VEGF.


Assuntos
Vasos Sanguíneos/anormalidades , Embrião de Mamíferos/metabolismo , Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Linfocinas/genética , Linfocinas/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Fatores de Transcrição , Proteínas Quinases Ativadas por AMP , Animais , Vasos Sanguíneos/embriologia , Morte Celular , Hipóxia Celular , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário e Fetal , Endotélio Vascular/anormalidades , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Marcação de Genes , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Hibridização In Situ , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/anormalidades , Músculo Liso Vascular/citologia , Músculo Liso Vascular/embriologia , Defeitos do Tubo Neural/embriologia , Proteínas Nucleares/metabolismo , Fenótipo , Placenta/irrigação sanguínea , Placenta/embriologia , Placenta/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
4.
EMBO J ; 20(11): 2844-56, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11387217

RESUMO

The trimeric Cdk7-cyclin H-Mat1 complex comprises the kinase subunit of basal transcription factor TFIIH and has been shown to function as a cyclin-dependent kinase (Cdk)-activating kinase. Herein we report that disruption of the murine Mat1 gene leads to peri-implantation lethality coincident with depletion of maternal Mat1 protein. In culture, Mat1(-/-) blastocysts gave rise to viable post-mitotic trophoblast giant cells while mitotic lineages failed to proliferate and survive. In contrast to wild-type trophoblast giant cells, Mat1(-/-) cells exhibited a rapid arrest in endoreduplication, which was characterized by an inability to enter S phase. Additionally, Mat1(-/-) cells exhibited defects in phosphorylation of the C-terminal domain (CTD) of RNA polymerase II on both Ser5 and Ser2 of the heptapeptide repeat. Despite this, Mat1(-/-) cells demonstrated apparent transcriptional and translational integrity. These data indicate an essential role for Mat1 in progression through the endocycle and suggest that while Mat1 modulates CTD phosphorylation, it does not appear to be essential for RNA polymerase II-mediated transcription.


Assuntos
Blastocisto/citologia , Ciclo Celular/fisiologia , Quinases Ciclina-Dependentes , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Polimerase II/metabolismo , Fatores de Transcrição TFII , Animais , Sítios de Ligação , Blastocisto/fisiologia , Feminino , Morte Fetal , Genótipo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Fosforilação , Reação em Cadeia da Polimerase , Gravidez , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/deficiência , Sequências Repetitivas de Aminoácidos , Fase S , Serina , Fator de Transcrição TFIIH , Fatores de Transcrição/metabolismo , Transcrição Gênica , Trofoblastos/citologia , Quinase Ativadora de Quinase Dependente de Ciclina
5.
J Biol Chem ; 276(32): 30399-406, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11395493

RESUMO

Topoisomerase IIbeta-binding protein (TopBP1), a human protein with eight BRCT domains, is similar to Saccharomyces cerevisiae Dpb11 and Schizosaccharomyces pombe Cut5 checkpoint proteins and closely related to Drosophila Mus101. We show that human TopBP1 is required for DNA replication and that it interacts with DNA polymerase epsilon. In S phase TopBP1 colocalizes with Brca1 to foci that do not represent sites of ongoing DNA replication. Inhibition of DNA synthesis leads to relocalization of TopBP1 together with Brca1 to replication forks, suggesting a role in rescue of stalled forks. DNA damage induces formation of distinct TopBP1 foci that colocalize with Brca1 in S phase, but not in G(1) phase. We also show that TopBP1 interacts with the checkpoint protein hRad9. Thus, these results implicate TopBP1 in replication and checkpoint functions.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Dano ao DNA , Reparo do DNA , Replicação do DNA , Proteínas de Ligação a DNA , Proteínas de Drosophila , Proteínas de Saccharomyces cerevisiae , Proteínas de Schizosaccharomyces pombe , Transglutaminases , Animais , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Mapeamento Cromossômico , Clonagem Molecular , Citoplasma/metabolismo , DNA Polimerase II/metabolismo , DNA Complementar/metabolismo , Drosophila , Ecdisona/metabolismo , Proteínas Fúngicas/química , Células HeLa , Humanos , Hibridização in Situ Fluorescente , Camundongos , Proteínas Nucleares , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Fase S , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/metabolismo , Transdução de Sinais , Fatores de Tempo , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
6.
Mech Dev ; 101(1-2): 209-12, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11231077

RESUMO

Transforming growth factor-beta (TGF-beta) signaling is mediated from serine/threonine kinase receptors to transcriptional responses via Smad proteins. Here comparison of mRNA expression of Smad3-7 in mouse embryos (E9-E15) revealed developmentally regulated distinct expression patterns for Smad3, 4, 6, and 7. Smad3 was prominently expressed in the differentiating (from E10) central nervous system, but also in developing bones, branchial arches and epithelium of various tissues. Smad4 mostly showed ubiquitous expression, but in E15 embryos, a pronounced signal appeared in epithelial crypts of the gut. Inhibitory Smad6 and Smad7 were coexpressed at high levels in developing cardiovascular system from the earliest stages studied. In contrast, Smad6 was selectively expressed at high levels, e.g. in intramembranous bone whereas Smad7 was prominent in seminiferous tubules of the testis, demonstrating distinct expression of these genes in non-cardiovascular tissues.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais , Transativadores/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Animais , Sistema Cardiovascular/embriologia , Sistema Nervoso Central/embriologia , Sistema Digestório/embriologia , Embrião de Mamíferos/metabolismo , Etiquetas de Sequências Expressas , Masculino , Camundongos , RNA Mensageiro/metabolismo , Proteína Smad3 , Proteína Smad4 , Proteína Smad6 , Proteína Smad7 , Testículo/metabolismo , Fatores de Tempo , Distribuição Tecidual
7.
EMBO J ; 20(1-2): 82-90, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11226158

RESUMO

Activating phosphorylation of cyclin-dependent kinases (Cdks) is mediated by at least two structurally distinct types of Cdk-activating kinases (Caks): the trimeric Cdk7-cyclin H-Mat1 complex in metazoans and the single-subunit Cak1 in budding yeast. Fission yeast has both Cak types: Mcs6 is a Cdk7 ortholog and Csk1 a single-subunit kinase. Both phosphorylate Cdks in vitro and rescue a thermosensitive budding yeast CAK1 strain. However, this apparent redundancy is not observed in fission yeast in vivo. We have identified mutants that exhibit phenotypes attributable to defects in either Mcs6-activating phosphorylation or in Cdc2-activating phosphorylation. Mcs6, human Cdk7 and budding yeast Cak1 were all active as Caks for Cdc2 when expressed in fission yeast. Although Csk1 could activate Mcs6, it was unable to activate Cdc2. Biochemical experiments supported these genetic results: budding yeast Cak1 could bind and phosphorylate Cdc2 from fission yeast lysates, whereas fission yeast Csk1 could not. These results indicate that Mcs6 is the direct activator of Cdc2, and Csk1 only activates Mcs6. This demonstrates in vivo specificity in Cdk activation by Caks.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces/enzimologia , Clonagem Molecular , Ativação Enzimática , Teste de Complementação Genética , Glutationa Transferase/genética , Humanos , Fases de Leitura Aberta , Fenótipo , Fosforilação , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Recombinantes de Fusão/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/crescimento & desenvolvimento , Especificidade por Substrato , Quinase Ativadora de Quinase Dependente de Ciclina
8.
Nat Cell Biol ; 2(11): 819-25, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11056537

RESUMO

v-cyclin encoded by Kaposi's sarcoma herpesvirus/human herpesvirus 8 (KSHV or HHV8) associates with cellular cyclin-dependent kinase 6 (CDK6) to form a kinase complex that promotes cell-cycle progression, but can also induce apoptosis in cells with high levels of CDK6. Here we show that whereas HHV8-encoded v-Bcl-2 protects against this apoptosis, cellular Bcl-2 has lost its anti-apoptotic potential as a result of an inactivating phosphorylation in its unstructured loop region. Moreover, we identify Bcl-2 as a new substrate for v-cyclin-CDK6 in vitro, and show that it is present in a complex with CDK6 in cell lysates. A Bcl-2 mutant with a S70A S87A double substitution in the loop region is not phosphorylated and provides resistance to apoptosis, indicating that inactivation of Bcl-2 by v-cyclin-CDK6 may be required for the observed apoptosis. Furthermore, the identification of phosphorylated Bcl-2 in HHV8-positive Kaposi's sarcoma indicates that HHV8-mediated interference with host apoptotic signalling pathways may encourage the development of Kaposi's sarcoma.


Assuntos
Apoptose , Quinases Ciclina-Dependentes , Ciclinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Células COS , Extratos Celulares , Chlorocebus aethiops , Quinase 6 Dependente de Ciclina , Ciclinas/genética , Fase G2 , Glutationa Transferase/genética , Herpesvirus Humano 8/fisiologia , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitose , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Serina/metabolismo , Especificidade por Substrato , Células Tumorais Cultivadas , Proteínas Virais
9.
J Biol Chem ; 275(45): 34837-40, 2000 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-10958787

RESUMO

Cyclin-dependent kinase 7 (Cdk7) forms a trimeric complex with cyclin H and Mat1 to form the mammalian Cdk-activating kinase, CAK, as well as a part of the basal transcription factor TFIIH, where Cdk7 phosphorylates the C-terminal domain (CTD) of the large subunit of RNA polymerase II. Here, we report a novel interaction between Cdk7 and a histidine triad (HIT) family protein, Hint/PKCI-1. This interaction was initially observed in a yeast two-hybrid study and subsequently verified by co-immunoprecipitation and subcellular localization studies, where overexpression of Cdk7 leads to partial relocalization of Hint to the nucleus. The physical association is independent of cyclin H binding or Cdk7 kinase activity and is conserved between the related Sacharomyces cerevisiae CTD kinase Kin28 and the HIT protein Hnt1. Furthermore, combination of a disruption of HNT1 and a KIN28 temperature-sensitive allele in S. cerevisiae led to highly elongated cell morphology and reduced colony formation, indicating a genetic interaction between KIN28 and HNT1. The physical and genetic interactions of Hint and Hnt1 with Cdk7 and Kin28 suggest a role for this class of histidine triad proteins in the regulation of Cdk7 and Kin28 functions.


Assuntos
Quinases Ciclina-Dependentes , Histidina/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae , Alelos , Animais , Western Blotting , Núcleo Celular/metabolismo , Ciclina H , Ciclinas/metabolismo , Imunofluorescência , Glutationa Transferase/metabolismo , Humanos , Mutagênese , Fosforilação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/genética , Saccharomyces cerevisiae/enzimologia , Temperatura , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido , Quinase Ativadora de Quinase Dependente de Ciclina
10.
J Biol Chem ; 275(39): 30638-43, 2000 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-10913154

RESUMO

The cyclin-dependent kinase inhibitor p21 is required for a sustained G(2) arrest after activation of the DNA damage checkpoint. Here we have addressed the mechanism by which p21 can contribute to this arrest in G(2). We show that p21 blocks the activating phosphorylation of Cdc2 on Thr(161). p21 does not interfere with the dephosphorylation of two inhibitory phosphorylation sites on Cdc2, Thr(14) and Tyr(15), indicating that p21 targets a different event in Cdc2 activation as the well described DNA damage checkpoint pathway involving Chk1 and Cdc25C. Taken together our data show that a cell is equipped with at least two independent pathways to ensure efficient inhibition of Cdc2 activity in response to DNA damage, influencing both positive and negative regulatory phosphorylation events on Cdc2.


Assuntos
Proteína Quinase CDC2/metabolismo , Ciclina B/metabolismo , Ciclinas/metabolismo , Dano ao DNA , Fase G2 , Compartimento Celular , Núcleo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Modelos Biológicos , Fosforilação , Ligação Proteica , Treonina/metabolismo
11.
Nat Genet ; 25(3): 298-301, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10888877

RESUMO

Mulibrey nanism (for muscle-liver-brain-eye nanism, MUL; MIM 253250) is an autosomal recessive disorder that involves several tissues of mesodermal origin, implying a defect in a highly pleiotropic gene. Characteristic features include severe growth failure of prenatal onset and constrictive pericardium with consequent hepatomegaly. In addition, muscle hypotonia, J-shaped sella turcica, yellowish dots in the ocular fundi, typical dysmorphic features and hypoplasia of various endocrine glands causing hormonal deficiency are common. About 4% of MUL patients develop Wilms' tumour. MUL is enriched in the Finnish population, but is rare elsewhere. We previously assigned MUL to chromosome 17q22-q23 and constructed a physical contig over the critical MUL region. The region has now been further refined by haplotype analysis and new positional candidate genes have been localized. We identified a gene with four independent MUL-associated mutations that all cause a frameshift and predict a truncated protein. MUL is ubiquitously expressed and encodes a new member of the RING-B-box-Coiled-coil (RBCC) family of zinc-finger proteins, whose members are involved in diverse cellular functions such as developmental patterning and oncogenesis.


Assuntos
Cromossomos Humanos Par 17 , Nanismo/genética , Mutação da Fase de Leitura , Proteínas Nucleares/genética , Dedos de Zinco , Processamento Alternativo , Animais , Sequência de Bases , Mapeamento Cromossômico , Códon de Terminação , DNA Complementar , Humanos , Camundongos , Dados de Sequência Molecular , Ratos , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
12.
J Biol Chem ; 275(15): 11100-5, 2000 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-10753915

RESUMO

The PDZ-LIM family of proteins (Enigma/LMP-1, ENH, ZASP/Cypher, RIL, ALP, and CLP-36) has been suggested to act as adapters that direct LIM-binding proteins to the cytoskeleton. Most interactions of PDZ-LIM proteins with the cytoskeleton have been identified in striated muscle, where several PDZ-LIM proteins are predominantly expressed. By contrast, CLP-36 mRNA is expressed in several nonmuscle tissues, and here we demonstrate high expression of CLP-36 in epithelial cells by in situ hybridization analysis. Our subcellular localization studies indicate that in nonmuscle cells, CLP-36 protein localizes to actin stress fibers. This localization is mediated via the PDZ domain of CLP-36 that associates with the spectrin-like repeats of alpha-actinin. Interestingly, immunoprecipitation and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis indicate that both nonmuscle alpha-actinin-1 and alpha-actinin-4 form complexes with CLP-36. The high expression of alpha-actinin-4 in the colon, together with these results, suggests a specific function for the alpha-actinin-4-CLP-36 complex in the colonic epithelium. More generally, results presented here demonstrate that the association of PDZ-LIM proteins with the cytoskeleton extends to the actin stress fibers of nonmuscle cells.


Assuntos
Actinina/química , Proteínas de Homeodomínio/química , Proteínas dos Microfilamentos , Animais , Células COS , Citoesqueleto/química , Proteínas de Ligação a DNA/química , Proteínas de Homeodomínio/análise , Proteínas de Homeodomínio/biossíntese , Proteínas com Domínio LIM , Camundongos , Camundongos Endogâmicos CBA , Coelhos
13.
Cancer Res ; 59(19): 4984-9, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10519412

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) has a key etiological role in development of Kaposi's sarcoma (KS). v-Cyclin is a KSHV-encoded homologue to D-type cyclins that associates with cellular cyclin-dependent kinase 6 (CDK6). v-Cyclin promotes S-phase entry of quiescent cells and has been suggested to execute functions of both D- and E-type cyclins. In this study, expression of v-cyclin in cells with elevated levels of CDK6 led to apoptotic cell death after the cells entered S phase. The cell death required the kinase activity of CDK6 because cells expressing a kinase-deficient form of CDK6 did not undergo apoptosis upon v-cyclin expression. Studies on the mechanisms involved in this caspase-3-mediated apoptosis indicated that it was independent of cellular p53 or pRb status, and it was not suppressed by Bcl-2. In contrast, the KSHV-encoded v-Bcl-2 efficiently suppressed v-cyclin-/CDK6-induced apoptosis, demonstrating a marked difference in the antiapoptotic properties of c-Bcl-2 and v-Bcl-2. In KS lesions, high CDK6 expression was confined to a subset of cells, some of which displayed signs of apoptosis. These results suggest that v-cyclin may exert both growth-promoting and apoptotic functions in KS, depending on factors regulating CDK6 and v-Bcl-2 levels.


Assuntos
Apoptose/fisiologia , Quinases Ciclina-Dependentes , Ciclinas/genética , Ciclinas/metabolismo , Herpesvirus Humano 8/genética , Proteínas Serina-Treonina Quinases/metabolismo , Sarcoma de Kaposi/patologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias Ósseas , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Quinase 6 Dependente de Ciclina , Inibidores de Cisteína Proteinase/farmacologia , Inibidores Enzimáticos/farmacologia , Herpesvirus Humano 8/fisiologia , Humanos , Dados de Sequência Molecular , Oligopeptídeos/farmacologia , Osteossarcoma , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína do Retinoblastoma/metabolismo , Sarcoma de Kaposi/enzimologia , Estaurosporina/farmacologia , Células Tumorais Cultivadas , Proteínas Virais
14.
Proc Natl Acad Sci U S A ; 96(16): 9248-51, 1999 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-10430928

RESUMO

Germ-line mutations of LKB1 (STK11) lead to Peutz-Jeghers syndrome characterized by gastrointestinal polyps and cancer of different organ systems. The mutations lead to loss or severe impairment of Lkb1 serine/threonine kinase activity. Therefore LKB1 has been implicated as a tumor suppressor gene, but only a few mutations in the coding exons of LKB1 have been detected in sporadic tumors. Here, we have identified tumor cell lines with severely reduced mRNA levels and impaired Lkb1 kinase activity. Reintroducing Lkb1 into these cells suppressed cell growth. The Lkb1-mediated growth inhibition was caused by a G(1) cell cycle block and was not detected with several naturally occurring Lkb1 mutants. These results indicate that LKB1 has functional and specific growth-suppressing activity.


Assuntos
Ciclo Celular/fisiologia , Regulação Neoplásica da Expressão Gênica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transcrição Gênica , Células 3T3 , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Animais , Divisão Celular , Metilação de DNA , Éxons , Citometria de Fluxo , Fase G1/fisiologia , Células HeLa , Humanos , Melanoma , Camundongos , Mutação , Síndrome de Peutz-Jeghers/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Proteínas Recombinantes/metabolismo , Transfecção , Células Tumorais Cultivadas
15.
Mech Dev ; 83(1-2): 187-90, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10381580

RESUMO

Germ-line mutations of LKB1 and PTEN tumor suppressor genes underlie the phenotypically related Peutz-Jeghers syndrome (PJS) and Cowden disease (CD), respectively. To analyze possible developmental roles of PTEN and LKB1, we have studied their mRNA expression during mouse embryonic development (E7-17.5) by in situ hybridization. Ubiquitous expression of both genes during early stages (E7-11) became more restricted in later embryonic development (E15-19) where LKB1 and PTEN showed prominent overlapping expression in e.g. gastrointestinal tract and lung. In contrast, LKB1 was selectively expressed at high levels in testis and PTEN was prominently expressed in skin epithelium and underlying mesenchyme. These results indicate that LKB1 and PTEN display largely overlapping expression patterns during embryonic development. Moreover, a high expression of these genes was observed in the tissues and organs affected in PJS and CD patients and in PTEN+/- mice.


Assuntos
Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor , Proteínas Quinases Ativadas por AMP , Animais , Embrião de Mamíferos/anatomia & histologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Supressores de Tumor , Camundongos , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/análise , Distribuição Tecidual
16.
Hum Mol Genet ; 8(1): 45-51, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9887330

RESUMO

Germline mutations in LKB1 have been reported to underlie familial Peutz-Jeghers syndrome (PJS) with intestinal hamartomatous polyps and an elevated risk of various neoplasms. To investigate the prevalence of LKB1 germline mutations in PJS more generally, we studied samples from 33 unrelated PJS patients including eight non-familial sporadic patients, 20 familial patients and five patients with unknown family history. Nineteen germline mutations were identified, 12 (60%) in familial and four (50%) in sporadic cases. LKB1 mutations were not detected in 14 (42%) patients, indicating that the existence of additional minor PJS loci cannot be excluded. LKB1 is predicted to encode a serine/threonine kinase. To demonstrate the putative Lkb1 kinase function and to study the consequences of LKB1 mutations in PJS and sporadic tumors, we have analyzed the kinase activity of wild-type and mutant Lkb1 proteins. Interestingly, while most of the small deletions or missense mutations resulted in loss-of-function alleles, one missense mutation (G163D) previously identified in a sporadic testicular tumor demonstrated severely impaired but detectable kinase activity.


Assuntos
Mutação em Linhagem Germinativa , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/genética , Neoplasias Testiculares/genética , Quinases Proteína-Quinases Ativadas por AMP , Alelos , Sequência de Bases , DNA/genética , Primers do DNA/genética , Feminino , Humanos , Masculino , Síndrome de Peutz-Jeghers/enzimologia , Mutação Puntual , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Deleção de Sequência , Neoplasias Testiculares/enzimologia
17.
EMBO J ; 17(24): 7230-8, 1998 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9857180

RESUMO

Cell cycle progression is dependent on the sequential activity of cyclin-dependent kinases (CDKs). For full activity, CDKs require an activating phosphorylation of a conserved residue (corresponding to Thr160 in human CDK2) carried out by the CDK-activating kinase (CAK). Two distinct CAK kinases have been described: in budding yeast Saccharomyces cerevisiae, the Cak1/Civ1 kinase is responsible for CAK activity. In several other species including human, Xenopus, Drosophila and fission yeast Schizosaccharomyces pombe, CAK has been identified as a complex homologous to CDK7-cyclin H (Mcs6-Mcs2 in fission yeast). Here we identify the fission yeast Csk1 kinase as an in vivo activating kinase of the Mcs6-Mcs2 CAK defining Csk1 as a CAK-activating kinase (CAKAK).


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces/enzimologia , Sequência de Aminoácidos , Ciclo Celular , Sequência Conservada , Ciclina H , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Drosophila , Ativação Enzimática , Dosagem de Genes , Genes Supressores , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Quinase Ativadora de Quinase Dependente de Ciclina
18.
Oncogene ; 17(8): 1019-26, 1998 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-9747881

RESUMO

Specific genetic alterations affecting proto-oncogenes of the myc gene family are frequently detected in human lung cancer. Among 11 SCLC cell lines with L-myc gene amplification, four were found to have alteration of the RLF gene by Southern blot and RT-PCR analyses. One cell line, NCI-H378, contained aberrantly-sized L-myc-hybridizing bands by Southern and Northern blot hybridization but had no alteration of RLF. Some L-myc-hybridizing cDNAs from NCI-H378 contained a novel sequence with close homology to the cyclophilins joined to antisense L-myc exon 2 sequence. Full length cDNAs isolated from human skeletal muscle containing only the novel sequence identify open reading frames of 301 and 296 amino acids and differ in the C-terminal region by 22 and 17 amino acids. This gene, tentatively named PPIE (peptidyl-prolyl cis-trans isomerase E), has 83% amino acid identity with the central conserved region of cyclophilin A, is evolutionarily conserved by Southern blot, and exhibits differential tissue expression with highest levels found in muscle and brain. Co-amplification of PPIE was observed in seven of eleven L-myc amplified cell lines. Analysis of radiation hybrids suggests that the gene order is RLF-PPIE-L-myc on chromosome 1p and pulse-field gel electrophoresis localizes all three genes to an 800 megabase Mlu I fragment. The prognostic and functional consequences of PPIE gene amplification in SCLC can now be determined.


Assuntos
Carcinoma de Células Pequenas/genética , Ciclofilinas , Amplificação de Genes , Genes myc , Neoplasias Pulmonares/genética , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Carcinoma de Células Pequenas/metabolismo , Cromossomos Humanos Par 1/genética , DNA Complementar/química , DNA Complementar/isolamento & purificação , Eletroforese em Gel de Campo Pulsado , Humanos , Neoplasias Pulmonares/metabolismo , Dados de Sequência Molecular , Peptidilprolil Isomerase/biossíntese , Peptidilprolil Isomerase/química , Translocação Genética , Células Tumorais Cultivadas
19.
Biochem Biophys Res Commun ; 247(3): 616-9, 1998 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-9647742

RESUMO

The natural estrogen metabolite 2-methoxyestradiol (2ME) is anti-angiogenic in vivo and a strong growth inhibitor in vitro. The growth inhibition is due to mitotic arrest and apoptosis. These effects are reminiscent of those induced by taxol, and appear to be mediated by inhibition of microtubule dynamics. Here we have studied the cellular response to 2ME in regard to potential mediators of the observed cellular changes. 2ME treatment increases the insoluble polymerized fraction of cellular tubulin similar to taxol, and in contrast to the microtubule depolymerizing drugs such as colcemid and vincristine. This stabilization following 2ME treatment is accompanied by phosphorylation and inactivation of Bcl-2 increasing gradually from 2-24 hours. To study the pathway leading to Bcl-2 phosphorylation we analyzed Raf-1 and JNK/SAPK kinases, both of which have been reported to be involved in Bcl-2 inactivation. Our results indicate that Raf-1 is phosphorylated in response to 2ME, but this occurs later than Bcl-2 phosphorylation suggesting that Raf-1 is not directly phosphorylating Bcl-2. JNK/SAPK was activated rapidly after 2ME treatment. However, this activation was transient and returned to undetectable levels by 2 hours of treatment, demonstrating that JNK/SAPK is not directly phosphorylating Bcl-2. Taken together with previous results indicating that overexpression of JNK/SAPK leads to Bcl-2 phosphorylation, our results would support a model where JNK/SAPK is indirectly phosphorylating Bcl-2.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Estradiol/análogos & derivados , Proteínas Quinases Ativadas por Mitógeno , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , 2-Metoxiestradiol , Demecolcina/farmacologia , Estradiol/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas de Neoplasias/metabolismo , Paclitaxel/farmacologia , Fosforilação/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas , Vincristina/farmacologia
20.
Exp Cell Res ; 242(1): 211-21, 1998 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-9665818

RESUMO

The carboxyl-terminal domain (CTD) of the largest RNA polymerase II (pol II) subunit is a target for extensive phosphorylation in vivo. Using in vitro kinase assays it was found that several different protein kinases can phosphorylate the CTD including the transcription factor IIH-associated CDK-activating CDK7 kinase (R. Roy, J. P. Adamczewski, T. Seroz, W. Vermeulen, J. P. Tassan, L. Schaeffer, E. A. Nigg, J. H. Hoeijmakers, and J. M. Egly, 1994, Cell 79, 1093-1101). Here we report the colocalization of CDK7 and the phosphorylated form of CTD (phosphoCTD) to actively transcribing genes in intact salivary gland cells of Chironomus tentans. Following a heat-shock treatment, both CDK7 and pol II staining disappear from non-heat-shock genes concomitantly with the abolishment of transcriptional activity of these genes. In contrast, the actively transcribing heat-shock genes, manifested as chromosomal puff 5C on chromosome IV (IV-5C), stain intensely for phosphoCTD, but are devoid of CDK7. Furthermore, the staining of puff IV-5C with anti-PCTD antibodies was not detectably influenced by the TFIIH kinase and transcription inhibitor 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB). Following heat-shock treatment, the transcription of non-heat-shock genes was completely eliminated, while newly formed heat-shock gene transcripts emerged in a DRB-resistant manner. Thus, heat shock in these cells induces a rapid clearance of CDK7 from the non-heat-shock genes, indicating a lack of involvement of CDK7 in the induction and function of the heat-induced genes. The results taken together suggest the existence of heat-shock-specific CTD phosphorylation in living cells. This phosphorylation is resistant to DRB treatment, suggesting that not only phosphorylation but also transcription of heat-shock genes is DRB resistant and that CDK7 in heat shock cells is not associated with TFIIH.


Assuntos
Chironomidae/genética , Quinases Ciclina-Dependentes , Resposta ao Choque Térmico/genética , RNA Polimerase II/metabolismo , Transcrição Gênica/fisiologia , Animais , Chironomidae/enzimologia , Cromossomos/enzimologia , Diclororribofuranosilbenzimidazol/farmacologia , Genes de Insetos , Proteínas de Insetos/genética , Larva , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fosforilação , Proteínas Serina-Treonina Quinases/análise , RNA Polimerase II/análise , RNA Mensageiro/análise , Glândulas Salivares/citologia , Glândulas Salivares/enzimologia , Quinase Ativadora de Quinase Dependente de Ciclina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA