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.
Biochem J ; 413(2): 251-9, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18402552

RESUMO

HAI-1 [HGF (hepatocyte growth factor) activator inhibitor-1] is a Kunitz-type transmembrane serine protease inhibitor that forms inhibitor complexes with the trypsin-like serine protease, matriptase. HAI-1 is essential for mouse placental development and embryo survival and together with matriptase it is a key regulator of carcinogenesis. HAI-1 is expressed in polarized epithelial cells, which have the plasma membrane divided by tight junctions into an apical and a basolateral domain. In the present study we show that HAI-1 at steady-state is mainly located on the basolateral membrane of both Madin-Darby canine kidney cells and mammary gland epithelial cells. After biosynthesis, HAI-1 is exocytosed mainly to the basolateral plasma membrane from where 15% of the HAI-1 molecules are proteolytically cleaved and released into the basolateral medium. The remaining membrane-associated HAI-1 is endocytosed and then recycles between the basolateral plasma membrane and endosomes for hours until it is transcytosed to the apical plasma membrane. Minor amounts of HAI-1 present at the apical plasma membrane are proteolytically cleaved and released into the apical medium. Full-length membrane-bound HAI-1 has a half-life of 1.5 h and is eventually degraded in the lysosomes, whereas proteolytically released HAI-1 is more stable. HAI-1 is co-localized with its cognate protease, matriptase, at the basolateral plasma membrane. We suggest that HAI-1, in addition to its protease inhibitory function, plays a role in transporting matriptase as a matriptase-HAI-1 complex from the basolateral plama membrane to the apical plasma membrane, as matriptase is known to interact with prostasin, located at the apical plasma membrane.


Assuntos
Regulação da Expressão Gênica , Glicoproteínas de Membrana/fisiologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Cães , Endocitose , Exocitose , Lisossomos/metabolismo , Glândulas Mamárias Animais/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Transporte Proteico , Proteínas Secretadas Inibidoras de Proteinases , Serina Endopeptidases/metabolismo
2.
Development ; 133(17): 3485-94, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16887827

RESUMO

The Nuclear Factor-kappaB (NF-kappaB) family of transcription factors are ubiquitously expressed and control a wide range of cellular responses, including apoptosis, proliferation, differentiation, inflammation and immunity. Here, we investigated the function of the NF-kappaB upstream regulator IkappaB kinase 2/beta (IKK2) in apoptosis regulation in the normal physiological setting of regressing mammary gland. Conditional deletion of the gene encoding IKK2 resulted, surprisingly, in delayed apoptosis and remodelling, and abrogation of caspase 3 cleavage. This failure to induce involution was associated with reduced expression, within 24 hours of involution, of the death receptor (DR) ligand TNF and its receptor TNFR1, which are known NF-kappaB targets. This was associated with elevated levels of active AKT and phosphorylated FOXO3a. Furthermore, we show that expression of TWEAK, another DR ligand, is dramatically downregulated, even in heterozygous IKK2 mammary glands. Unlike other DR ligands, the TWEAK promoter has six consensus FOXO-binding sites, further suggesting that it is differentially regulated. Interestingly, a cleaved form of TWEAK is upregulated during involution. This unexpected function of the IKK2/NF-kappaB pathway as a regulator of TWEAK expression and inducer of apoptosis has significant consequences for future therapeutic approaches for cancer and inflammatory diseases.


Assuntos
Quinase I-kappa B/fisiologia , Fatores de Necrose Tumoral/metabolismo , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Citocina TWEAK , Células Epiteliais/metabolismo , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Quinase I-kappa B/genética , Ligantes , Glândulas Mamárias Animais/citologia , Camundongos , Regiões Promotoras Genéticas , Transdução de Sinais , Fatores de Necrose Tumoral/genética
3.
BMC Cancer ; 6: 176, 2006 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-16820046

RESUMO

BACKGROUND: It has recently been shown that overexpression of the serine protease, matriptase, in transgenic mice causes a dramatically increased frequency of carcinoma formation. Overexpression of HAI-1 and matriptase together changed the frequency of carcinoma formation to normal. This suggests that the ratio of matriptase to HAI-1 influences the malignant progression. The aim of this study has been to determine the ratio of matriptase to HAI-1 mRNA expression in affected and normal tissue from individuals with colorectal cancer adenomas and carcinomas as well as in healthy individuals, in order to determine at which stages a dysregulated ratio of matriptase/HAI-1 mRNA is present during carcinogenesis. METHODS: Using quantitative RT-PCR, we have determined the mRNA levels for matriptase and HAI-1 in colorectal cancer tissue (n = 9), severe dysplasia (n = 15), mild/moderate dysplasia (n = 21) and in normal tissue from the same individuals. In addition, corresponding tissue was examined from healthy volunteers (n = 10). Matriptase and HAI-1 mRNA levels were normalized to beta-actin. RESULTS: Matriptase mRNA level was lower in carcinomas compared to normal tissue from healthy individuals (p < 0.01). In accordance with this, the matriptase mRNA level was also lower in adenomas/carcinomas combined as compared to their adjacent normal tissue (p < 0.01). HAI-1 mRNA levels in both normal and affected tissue from individuals with severe dysplasia or carcinomas and in affected tissue with mild/moderate dysplasia were all significantly lower than mRNA levels observed in corresponding tissue from healthy control individuals. HAI-1 mRNA was lower in carcinomas as compared to normal tissue from healthy individuals (p < 0.001). HAI-1 mRNA levels were significantly lower in tissue displaying mild/moderate (p < 0.001) and severe (p < 0.01) dysplasia compared to normal tissue from the same patients. Both adenomas and carcinomas displayed a significantly different matriptase/HAI-1 mRNA ratio than corresponding normal tissue from healthy control individuals (p < 0.05). In addition statistically significant difference (p < 0.001) could be observed between mild/moderate and severe adenomas and their adjacent normal tissue. CONCLUSION: Our results show that dysregulation of the matriptase/HAI-1 mRNA ratio occurs early during carcinogenesis. Future studies are required to clarify whether the dysregulated matriptase/HAI-1 ratio was causing the malignant progression or is a consequence of the same.


Assuntos
Adenoma/metabolismo , Carcinoma/metabolismo , Neoplasias Colorretais/metabolismo , Glicoproteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Adenoma/patologia , Carcinoma/patologia , Estudos de Casos e Controles , Transformação Celular Neoplásica , Neoplasias Colorretais/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Glicoproteínas de Membrana/análise , Pessoa de Meia-Idade , Proteínas Secretadas Inibidoras de Proteinases , RNA Mensageiro/análise , Serina Endopeptidases/análise
4.
Differentiation ; 74(5): 254-64, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16759291

RESUMO

Epithelial-to-mesenchymal transition (EMT) is an essential embryogenic and developmental process, characterized by altered cellular morphology, loss of cell adhesion, and gain of migratory ability. Dysregulation of this process has been implicated in tumorigenesis, mediating the acquisition of migratory and invasive phenotypes by tumor cells. Mammary epithelial cells provide an excellent model in which to study the process, being derived from mammary gland tissue that utilizes EMT to facilitate branching morphogenesis through which the developing gland migrates into and invades the fat pad. Inappropriate EMT has been heavily implicated in the progression of ductal hyperplasia and mammary tumor metastasis. We examined the morphological and molecular changes of three murine mammary epithelial cell lines following EMT induction. EMT was induced in the EpH-4 and NMuMG cell lines by transforming growth factor (TGF)-beta1 but not by ethanol, while the KIM-2 cell line was partially resistant to TGF-beta1 but responded fully to ethanol. The response to EMT-inducing reagent was shown to be critically dependent on the time of treatment, with confluent cells failing to respond. Timelapse photography identified increased motility during wound healing in cells pre-treated with EMT-inducing reagent compared with untreated controls. Furthermore, EMT conferred resistance to UV-induced apoptosis. Our data indicate that evaluation of characteristics other than loss and gain of phenotypic markers may be of benefit when assessing EMT, and contribute to the evidence suggesting that inappropriate EMT facilitates the acquisition of resistance to apoptosis, a key characteristic required for tumor survival.


Assuntos
Apoptose , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/citologia , Glândulas Mamárias Animais/citologia , Mesoderma/citologia , Animais , Western Blotting , Linhagem Celular , Etanol/farmacologia , Imunofluorescência , Camundongos , Fator de Transcrição STAT5/metabolismo , Cicatrização
5.
Cell Cycle ; 4(7): 897-900, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15970662

RESUMO

Phosphoinositide 3-kinases (PI3Ks) have been grouped into three major classes that have different substrate specificities. Class IA PI3Ks consist of a catalytic and a regulatory subunit and have multiple isoforms that arise from different subunit combinations. The role of two of the small regulatory subunits, p55alpha and p50alpha, is poorly understood. We have now identified a novel function for these subunits and have shown that their expression is specifically induced in the involuting mouse mammary gland where they are involved in the downregulation of PI3K signalling and Akt/PKB activity. This abrogation of survival signalling thru Akt/PKB and its downstream targets is essential for the induction of apoptosis. The switch from lactation to involution is associated with activation of the transcription factor Stat3, by the cytokine LIF. Stat3 is essential for the induction of apoptosis and, in the absence of Stat3 or LIF, expression of the p55alpha and p50alpha subunits is abrogated. Surprisingly, Stat3 is a direct regulator of p55alpha and p50alpha expression, as demonstrated using ChIP assays, and therefore these subunits are not splicing isoforms as previously thought. An important implication of our results is that the p55alpha and p50alpha small regulatory subunits are regulated independently of the larger p85alpha subunit, and have an essential role in Stat3-mediated apoptosis in mammary gland.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Animais , Regulação Enzimológica da Expressão Gênica , Janus Quinase 1 , Modelos Biológicos , Fosfatidilinositol 3-Quinases/química , Transdução de Sinais
6.
Nat Cell Biol ; 7(4): 392-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15793565

RESUMO

Physiological apoptosis is induced by a switch from survival to death signalling. Dysregulation of this process is frequently associated with cancer. A powerful model for this apoptotic switch is mammary gland involution, during which redundant milk-producing epithelial cells undergo apoptosis. Signal transducer and activator of transcription 3 (Stat3) is an essential mediator of this switch but the mechanism has not yet been defined. Stat3-dependent cell death during involution can be blocked by activation of Akt/protein kinase B (PKB), a downstream effector of the phosphoinositide-3-OH kinase (PI(3)K) pathway. Here we show that expression of the PI(3)K regulatory subunits p55alpha and p50alpha is induced by Stat3 during involution. In the absence of Stat3 in vivo, upregulation of p55alpha and p50alpha is abrogated, levels of activated Akt are sustained and apoptosis is prevented. Chromatin immunoprecipitation assays show that Stat3 binds directly to the p55alpha and p50alpha promoters in vivo. Overexpression of either p55alpha or p50alpha reduces levels of activated Akt. We propose a novel mechanism in which Stat3 regulates apoptosis by inducing expression of distinct PI(3)K regulatory subunits to downregulate PI(3)K-Akt-mediated survival signalling.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT3 , Transdução de Sinais/fisiologia
7.
Development ; 130(15): 3459-68, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12810593

RESUMO

STAT3 is the key mediator of apoptosis in mammary gland. We demonstrate here that LIF is the physiological activator of STAT3, because in involuting mammary glands of Lif(-/-) mice, pSTAT3 is absent and the STAT3 target, C/EBPdelta, is not upregulated. Similar to Stat3 knockouts, Lif(-/-) mammary glands exhibit delayed involution, reduced apoptosis and elevated levels of p53. Significantly, Lif(-/-) glands display precocious development during pregnancy, when pSTAT3 is not normally detected. We show that pERK1/2 is significantly reduced in Lif(-/-) glands at this time, suggesting that at this stage LIF mediates its effects through pERK1/2. Inhibition of LIF-mediated ERK1/2 phosphorylation potentiates the proapoptotic effects of STAT3. LIF therefore signals alternately through ERK1/2, then STAT3, to regulate mammary growth and apoptosis.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Inibidores do Crescimento/metabolismo , Interleucina-6 , Linfocinas/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas do Leite , Transativadores/metabolismo , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Caseínas/biossíntese , Caseínas/genética , Contactinas , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Feminino , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Fator Inibidor de Leucemia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia , Linfocinas/deficiência , Linfocinas/genética , MAP Quinase Quinase 1 , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/embriologia , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Moléculas de Adesão de Célula Nervosa/biossíntese , Moléculas de Adesão de Célula Nervosa/genética , Nitrilas/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Citocinas/biossíntese , Receptores de Citocinas/genética , Receptores de OSM-LIF , Receptores de Progesterona/metabolismo , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Transativadores/biossíntese , Transativadores/genética
8.
J Gene Med ; 5(6): 528-38, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12797118

RESUMO

BACKGROUND: Huntington's disease (HD) is an inherited neurodegenerative disorder which is caused by an expansion of a CAG repeat sequence in the HD gene. The repeat encodes an expanded polyglutamine tract in the protein huntingtin. The still unknown pathological mechanisms leading to death of specific neurons in the brains of HD patients correlate with the expression of mutant huntingtin. Therefore, we have studied whether mutant huntingtin expression can be downregulated by antisense technique. METHODS: NT2 precursor cells and differentiated postmitotic NT2-N neurons, respectively, were transfected with plasmid constructs containing exon 1 of the HD gene with expanded CAG repeats in frame with the reporter protein EGFP. The transfected cell cultures were treated with a phosphorothioated antisense oligonucleotide (PS-ASHD/20+) or a control oligonucleotide either by cotransfection or by addition to the culture medium. RESULTS: Expression of the fusion protein containing the mutant huntingtin fragment resulted in diffuse green fluorescence in the cytoplasm and formation of aggregates in some of the NT2 cells and NT2-N neurons. We obtained antisense sequence-specific inhibition of expression of the fusion protein and/or suppression of the aggregate formation in both cell types. In the NT2 cells the antisense effect was dependent on the way of administration of the oligo. CONCLUSIONS: The PS-antisense oligo is effective in downregulation of mutant huntingtin, and the reduction of aggregate formation is a sensitive biological marker. The findings suggest that antisense knockdown of huntingtin could be a useful strategy for treatment of HD, and could also be suitable for studies of the normal and pathological function of huntingtin in different cellular model systems.


Assuntos
Regulação para Baixo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Oligodesoxirribonucleotídeos Antissenso/genética , Linhagem Celular Tumoral , Vetores Genéticos , Proteínas de Fluorescência Verde , Humanos , Proteína Huntingtina , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/metabolismo , Neurônios/química , Proteínas Nucleares/metabolismo , Peptídeos/análise , Peptídeos/metabolismo , Plasmídeos/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...