Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
BMC Biotechnol ; 9: 88, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19835613

RESUMO

BACKGROUND: Sequencing of the human genome has led to most genes being available in BAC or PAC vectors. However, limited functional information has been assigned to most of these genes. Techniques for the manipulation and transfer of complete functional units on large DNA fragments into human cells are crucial for the analysis of complete genes in their natural genomic context. One limitation of the functional studies using these vectors is the low transfection frequency. RESULTS: We have constructed a shuttle vector, pPAC7, which contains both the EBNA-1 gene and oriP from the Epstein-Barr virus allowing stable maintenance of PAC clones in the nucleus of human cells. The pPAC7 vector also contains the EGFP reporter gene, which allows direct monitoring of the presence of PAC constructs in transfected cells, and the Bsr-cassette that allows highly efficient and rapid selection in mammalian cells by use of blasticidin. Positive selection for recombinant PAC clones is obtained in pPAC7 because the cloning sites are located within the SacBII gene. We show regulated expression of the CDH3 gene carried as a 132 kb genomic insert cloned into pPAC7, demonstrating that the pPAC7 vector can be used for functional studies of genes in their natural genomic context. Furthermore, the results from the transfection of a range of pPAC7 based constructs into two human cell lines suggest that the transfection efficiencies are not only dependent on construct size. CONCLUSION: The shuttle vector pPAC7 can be used to transfer large genomic constructs into human cells. The genes transferred could potentially contain all long-range regulatory elements, including their endogenous regulatory promoters. Introduction of complete genes in PACs into human cells would potentially allow complementation assays to identify or verify the function of genes affecting cellular phenotypes.


Assuntos
Cromossomos Artificiais Bacterianos , Antígenos Nucleares do Vírus Epstein-Barr/genética , Transfecção , Transgenes , Caderinas/genética , Linhagem Celular , Regulação da Expressão Gênica , Herpesvirus Humano 4/genética , Humanos
2.
Clin Cancer Res ; 12(13): 4055-61, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16818705

RESUMO

PURPOSE: The coagulation trigger tissue factor has been implicated in tumor growth, angiogenesis, and metastasis. In this study, we explore the effects of ex vivo and in vivo delivery of short interfering RNA (siRNA) targeting tissue factor on B16 melanoma colonization of the lung in a murine model for metastasis. The purposes of this work are to establish a noncytotoxic in vivo model for investigation of tissue factor function and provide preclinical assessment of the therapeutic potential of tissue factor siRNA for prevention of metastasis. EXPERIMENTAL DESIGN AND RESULTS: C57BL/6 mice were evaluated for pulmonary metastases following tail vein injection of B16 cells transfected with either active or inactive siRNA. Mice receiving cells transfected with active siRNA had significantly lower numbers of pulmonary tumors compared with mice injected with control cells (transfected with inactive siRNA). The average time point at which the mice started to exhibit tumor-associated stress was also increased significantly from 22 days for the control group to 27 days for the experimental group (P = 0.01). In a therapeutically more relevant model, where the siRNA was delivered i.p. and the cells (untransfected) by tail vein injection, an inhibitory effect on metastasis was observed when the siRNA treatment was initiated either before or at the time of cell injection. CONCLUSIONS: The results suggest that tissue factor has a crucial function in promoting lung tumor metastasis of blood-borne tumor cells in the early stages of the tumor take process and further suggest that treatment with tissue factor siRNA may become a viable clinical strategy for prevention of tumor metastasis.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Melanoma Experimental/tratamento farmacológico , RNA Interferente Pequeno/administração & dosagem , Tromboplastina/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Injeções Intravenosas , Injeções Subcutâneas , Neoplasias Pulmonares/patologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , RNA Interferente Pequeno/síntese química , RNA Interferente Pequeno/química
3.
Thromb Res ; 117(3): 343-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16378835

RESUMO

Tissue factor (TF) is a transmembrane glycoprotein that binds its zymogen cofactor, Factor VIIa (FVIIa) on the cell surface. Together (TF/FVIIa) they activate Factor X (FX) and Factor IX (FIX) and start the extrinsic pathway of blood coagulation. As such, the TF/FVIIa complex plays an important role in normal physiology as well as in thrombotic diseases such as unstable angina (UA), disseminated intravascular coagulation (DIC), and deep vein thrombosis (DVT). In addition to its function as an initiator of coagulation, TF/FVIIa plays an important role in inflammation. Expression of TF on the cell surface and its appearance as a soluble molecule are characteristic features of acute and chronic inflammation in conditions such as sepsis and atherosclerosis. Here we demonstrate that BCX-3607, a small molecule potent inhibitor of TF/FVIIa, reduces thrombus weight in an animal model of DVT. BCX-3607 also decreases the level of interleukin-6 (IL-6) in a LPS-stimulated mouse model of endotoxemia. Additionally, in vitro studies indicate that BCX-3607 blocks the generation of TF/FVIIa-induced IL-8 mRNA in human keratinocytes and reduces the TF/FVIIa-mediated generation of IL-6 and IL-8 in human umbilical vein endothelial cells (HUVEC). Therefore, BCX-3607 might block the TF/FVIIa-mediated coagulation and inflammation associated with pathological conditions.


Assuntos
Anti-Inflamatórios/farmacologia , Fator VIIa/antagonistas & inibidores , Fibrinolíticos/farmacologia , Piridinas/farmacologia , Tromboplastina/antagonistas & inibidores , Animais , Aterosclerose/tratamento farmacológico , Northern Blotting , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Endotoxemia/patologia , Humanos , Inflamação , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Queratinócitos/metabolismo , Lipopolissacarídeos/metabolismo , Masculino , Camundongos , Modelos Biológicos , Modelos Químicos , Tempo de Protrombina , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Sepse , Fatores de Tempo
4.
Biochim Biophys Acta ; 1734(3): 235-46, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15893958

RESUMO

The present study investigated the hepatic regulation of fatty acid metabolism in hTNFalpha transgenic mice. Reduced hepatic mRNA levels and activities of carnitine palmitoyltransferase-II (CPT-II) and mitochondrial HMG-CoA synthase were observed, accompanied by decreased fatty acid oxidation, fatty acyl-CoA oxidase and fatty acid synthase (FAS) activities and down-regulated gene expression of mitochondrial acetyl-CoA carboxylase 2 (ACC2). The mRNA levels of peroxisome proliferator-activated receptor alpha (PPARalpha) and PPARdelta were reduced. The hepatic fatty acid composition was altered, with increased amounts of saturated and polyunsaturated fatty acids. The relative amounts of Delta(9) desaturated fatty acids were decreased, as was Delta(9)desaturase mRNA. The CPT-I mRNA level remained unchanged. The PPARalpha targeted genes CPT-II and HMG-CoA synthase are potential regulators of mitochondrial fatty acid oxidation and ketogenesis in hTNFalpha transgenic mice, and the increased propionyl-CoA level found is a possible inhibitor of these processes. Reduced mitochondrial and peroxisomal fatty acid oxidation may explain the increased hepatic triglyceride level induced by TNFalpha. This is not due to de novo fatty acid synthesis as both FAS activity and gene expression of ACC2 were reduced.


Assuntos
Ácidos Graxos/metabolismo , Fígado/metabolismo , PPAR alfa/genética , Fator de Necrose Tumoral alfa/genética , Animais , Coenzima A Ligases/análise , Regulação para Baixo , Ácidos Graxos Dessaturases/metabolismo , Ácido Graxo Sintases/análise , Ácidos Graxos/análise , Ácidos Graxos/sangue , Ácidos Graxos Monoinsaturados/análise , Ácidos Graxos Monoinsaturados/sangue , Ácidos Graxos Monoinsaturados/metabolismo , Feminino , Hidroximetilglutaril-CoA Sintase , Cetoácidos/metabolismo , Fígado/química , Fígado/enzimologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Oxirredução , PPAR alfa/biossíntese , RNA Mensageiro/análise , Estearoil-CoA Dessaturase , Fator de Necrose Tumoral alfa/biossíntese
5.
Int J Cancer ; 112(6): 994-1002, 2004 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15386437

RESUMO

Tissue factor (TF) is the membrane receptor of the serine protease coagulation factor VIIa (FVIIa). Formation of the TF/FVIIa complex initiates the coagulation cascade. We used short hairpin RNA (shRNA)-mediated RNA interference to knock down TF expression in the human metastatic melanoma cell line LOX-L. After transfection with the shRNA construct, 3 stable clones with significantly downregulated TF expression were established. They exhibited decreased proliferation in vitro as determined by (14)C thymidine incorporation and soft agar assay. The in vivo metastatic potential was assessed in an experimental pulmonary metastasis model in which cells from different clones were injected into the tail vein of nude mice. The incidence of pulmonary tumors was significantly lower in mice receiving shRNA-expressing cells (33% +/- 15%) than in control mice injected with wild-type cells or cells stably transfected with empty expression vector (90% +/- 10%). The mice injected with TF-downregulated cells had markedly longer survival time (69 +/- 17 days) compared to the control mice (35.6 +/- 5 days; p = 0.03). Thus, reduction of TF levels in LOX-L cells significantly delayed and reduced lung tumor formation. As a first step in elucidating the molecular basis for this effect, we compared the global gene expression profile in TF-downregulated cells and control cells by using cDNA microarray analysis. Forty-four known human genes were found to be significantly upregulated (> 2-fold; p < 0.05) and 228 genes significantly downregulated (>or= 3-fold; p < 0.05) in TF-downregulated cells compared to control cells. The differentially expressed genes encode proteins functioning in transcription, translation, cell communication and cell growth/death. The results provide a basis for investigating molecular mechanisms underlying the effects of TF on the metastatic capacity of LOX-L melanoma cells.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma/metabolismo , Melanoma/secundário , Tromboplastina/metabolismo , Animais , Linhagem Celular Tumoral , DNA de Neoplasias/análise , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Incidência , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , RNA Neoplásico/metabolismo , Serina Endopeptidases/metabolismo , Taxa de Sobrevida , Tromboplastina/genética , Transfecção
6.
Eur J Immunol ; 34(8): 2191-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15259016

RESUMO

C-terminal Src kinase (Csk) controls the Src family kinase Lck, which is essential for T cell antigen receptor (TCR)-mediated signaling. For the first time, we here report the effects of acute elimination of Csk in Jurkat T cells and primary T cells using short interfering (si) RNA. In both cell types, 70-85% knockdown of Csk was achieved within 48 h. No alterations in surface expression of CD3, CD4 or CD8, or in Lck protein level were observed. Phosphorylation of Y505 in Lck was markedly reduced and a concomitant 4-5-fold increase in Lck Y394 phosphorylation was observed both in normal and Jurkat T cells. Kinase assays revealed 2-3-fold higher Lck activity. In Jurkat cells, basal levels of zeta chain phosphorylation were elevated, and spontaneous NFAT-AP-1 activation occurred, indicating aberrant Lck kinase activity. After TCR triggering, Csk knockdown cells revealed faster and stronger, but not sustained, phosphorylation of Lck Y394 and zeta chains compared to control. TCR-induced activation of NFAT-AP-1 and TCR/CD28-stimulated IL-2 secretion occurred at weaker stimuli and with augmented responses in Csk knockdown Jurkat and primary T cells, respectively. Altogether, these data suggest that acute elimination of Csk in T cells without evolution of compensatory mechanisms results in aberrant Lck activity and augmented TCR-stimulated responses.


Assuntos
Proteínas Tirosina Quinases/genética , Interferência de RNA/fisiologia , RNA Interferente Pequeno/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Proteína Tirosina Quinase CSK , Humanos , Células Jurkat , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Linfócitos T/enzimologia , Quinases da Família src
7.
Thromb Haemost ; 92(1): 13-22, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15213840

RESUMO

Binding of the coagulation protease factor VIIa to its receptor Tissue Factor (TF) induces intracellular signals in several cell types including HaCaT keratinocytes. TF belongs to the cytokine receptor family, but is most likely not alone in transferring the complete TF/FVIIa signal over the plasma membrane. The protease activated receptor PAR2 is involved in factor VIIa and factor Xa signal transduction. Our results indicate that the epidermal growth factor receptor (EGFR) and the proline rich tyrosine kinase 2 (PYK2) participate in TF/FVIIa signalling as formation of the TF/FVIIa complex increased the phosphorylation of these proteins. Both FVIIa protease activity and available TF were necessary for generation of the signal. Increased tyrosine phosphorylation of the EGFR was observed following TF/FVIIa complex formation on the cell surface. The EGFR kinase inhibitor tyrphostin AG1478 abrogated the TF/FVIIa-complex induced MAP kinase activation and mRNA increase of egr-1, heparin-binding EGF, and interleukin-8 following FVIIa addition. Using specific antibodies, increased phosphorylation of PYK2 tyrosine residues 402 and 580 was observed. The first site is the major autophosphorylation site and the docking site for Src family kinases. The second site is important for the kinase activity. The Src family kinase Yes and the tyrosine phosphatase SHP-2 were detected in immunoprecipitates using either anti-PYK2 or anti-EGFR antibodies. Their coprecipitation with EGFR increased in the presence of FVIIa. Moreover, the coprecipitation of EGFR and PYK2 increased with FVIIa stimulation. Together, these data suggest that EGFR, PYK2, Yes, and SHP-2 are involved in transduction of the TF/FVIIa signal possibly via transactivation of the EGF receptor.


Assuntos
Receptores ErbB/metabolismo , Fator VIIa/metabolismo , Proteínas Tirosina Quinases/metabolismo , Tromboplastina/metabolismo , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Fator de Crescimento Epidérmico/genética , Fator VIIa/farmacologia , Quinase 2 de Adesão Focal , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-8/genética , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Fosforilação , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/química , Quinazolinas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , Tirfostinas/farmacologia
8.
APMIS ; 112(1): 63-73, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14961977

RESUMO

Tumour necrosis factor (TNF) is involved in the pathogenesis of several diseases. In mice, human TNF signals only through p55, one of two murine TNF receptors. We here report a study of growth, viability and morphological alterations in transgenic mice expressing a low constitutive and tissue-restricted level of human TNF in vivo. The transgene was expressed solely in T cells. The transgenic mice showed a marked failure to thrive and a rapid cellular depletion in spleen and thymus. Slight fibrosis was seen in most tissues investigated, in addition to immature adipose tissue and irregular lymphocytic areas. Serum levels of hTNF were only slightly increased in the transgenic mice, enough, however, to cause an inflammatory reaction. All the symptoms were abrogated by an inhibitory hTNF antibody, demonstrating the essential role of hTNF in this phenotype. Transgenic mice constitute a multidimensional system allowing observation of disease processes over time in all tissues. The effects of hTNF were seen first and foremost in the lymphoid organs of the transgenic mice, verifying their cells as major targets at low levels of hTNF expression in the T-cell compartments. Chronic, low levels of TNF expression cause profound disturbances in lymphoid tissue development resulting in cachexia and premature death.


Assuntos
Linfonodos/patologia , Camundongos Transgênicos/crescimento & desenvolvimento , Baço/patologia , Timo/patologia , Fator de Necrose Tumoral alfa/genética , Animais , Apoptose/fisiologia , Western Blotting , Peso Corporal , Caspase 3 , Caspases/metabolismo , Feminino , Histocitoquímica , Linfonodos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Fator de Necrose Tumoral/metabolismo , Organismos Livres de Patógenos Específicos , Baço/metabolismo , Análise de Sobrevida , Timo/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
9.
Thromb Haemost ; 91(1): 28-37, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14691565

RESUMO

A novel mutation in the factor VII gene resulting in procoagulant activity of 7.5% and antigen levels of 23% is presented. Single-stranded conformational polymorphism and DNA sequencing analysis revealed heterozygous shifts, and mutations were detected in exons 5, 7 and 8. The mutant L204P in exon 7 was novel, while the common polymorphisms, H115H and R353Q, were located in exons 5 and 8, respectively. The molecular effect of the L204P mutation was characterized using recombinant mammalian expression in Chinese hamster ovary cells. Low levels (4 ng/ml) of secreted mutant protein were found in transiently transfected cells compared to wild-type factor VII (83 ng/ml). Metabolic labeling demonstrated that the rate of mutant protein synthesis was similar to that of wild-type FVII, and the mutant protein accumulated intracellularly with no signs of increased degradation during a four-hour chase. No interaction between secreted P204 protein and immobilized soluble tissue factor was detected using surface plasmon reso-nance. The activation rate of recombinant mutant FVII protein was strongly reduced compared to wild-type FVII. A 9-fold reduction in the rate of FX activation was detected whereas Km was nearly the same for wild-type and the mutant. This slow rate was caused by a correspondingly lowered rate of P204 activation. A synthetic peptide sequence comprising amino acids 177-206 blocked binding of FVIIa to the TF-chip, and the subsequent factor X activation with an IC(50) value of 0.5 micro M in a chromogenic factor Xa assay. Additionally, evaluation of the peptide by surface plasmon resonance analysis resulted in inhibition of complex formation with an apparent K(I) of 7 micro M.


Assuntos
Acetilcisteína/análogos & derivados , Fator VII/química , Fator VII/genética , Mutação , Acetilcisteína/química , Animais , Anticorpos Monoclonais/química , Encéfalo/metabolismo , Brefeldina A/química , Células CHO , Dicroísmo Circular , Cricetinae , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Precursores Enzimáticos/química , Éxons , Fator X/química , Fator Xa/química , Heterozigoto , Humanos , Concentração Inibidora 50 , Cinética , Modelos Moleculares , Peptídeos/química , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo Conformacional de Fita Simples , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Recombinantes/química , Mapeamento por Restrição , Análise de Sequência de DNA , Serina Endopeptidases/química , Ressonância de Plasmônio de Superfície , Fatores de Tempo , Transfecção
10.
Exp Cell Res ; 291(2): 299-312, 2003 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-14644153

RESUMO

We have dissected the molecular determinants involved in targeting the protein serine kinase PSKH1 to the endoplasmic reticulum (ER), the Golgi apparatus, and the plasma membrane (PM). Given this intracellular localization pattern, a potential role of PSKH1 in the secretory pathway was explored. The amino-terminal of PSKH1 revealed a striking similarity to the often acylated Src homology domain 4 (SH4)-harboring nonreceptor tyrosine kinases. Biochemical studies demonstrated that PSKH1 is myristoylated on glycine 2 and palmitoylated on cysteine 3. Dual amino-terminal acylation targets PSKH1 to Golgi as shown by colocalization with beta-COP and GM130, while nonpalmitoylated (myristoylated only) PSKH1 targets intracellular membranes colocalizing with protein disulphide isomerase (PDI, a marker for ER). Immunoelectron microscopy revealed that the dually acylated amino-terminal domain (in fusion with EGFP) was targeted to Golgi membranes as well as to the plasma membrane (PM), suggesting that the amino-terminal domain provides PSKH1 with membrane specificity dependent on its fatty acylation status. Subcellular fractionation by sucrose gradient analysis confirmed the impact of dual fatty acylation on endomembrane targeting, while cytosol and membrane fractioning revealed that myristoylation but not palmitoylation was required for general membrane association. A minimal region required for proper Golgi targeting of PSKH1 was identified within the first 29 amino acids. Expression of a PSKH1 mutant where the COOH-terminal kinase domain was swapped with green fluorescent protein and cysteine 3 was exchanged with serine resulted in disassembly of the Golgi apparatus as visualized by redistribution of beta-COP and GM130 to a diffuse cytoplasmic pattern, while leaving the tubulin skeleton intact. Our results suggest a structural and regulatory role of PSKH1 in maintenance of the Golgi apparatus, a key organelle within the secretory pathway.


Assuntos
Complexo de Golgi/metabolismo , Mutação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Acilação , Animais , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Endossomos/metabolismo , Endossomos/ultraestrutura , Ácidos Graxos , Vetores Genéticos , Complexo de Golgi/ultraestrutura , Humanos , Membranas Intracelulares/metabolismo , Microscopia Eletrônica , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Transfecção , Vesículas Transportadoras/metabolismo
11.
Nucleic Acids Res ; 30(23): 5301-9, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12466556

RESUMO

Small nuclear ribonucleoprotein particles (snRNPs) and non-snRNP splicing factors containing a serine/arginine-rich domain (SR proteins) concentrate in splicing factor compartments (SFCs) within the nucleus of interphase cells. Nuclear SFCs are considered mainly as storage sites for splicing factors, supplying splicing factors to active genes. The mechanisms controlling the interaction of the various spliceosome constituents, and the dynamic nature of the SFCs, are still poorly understood. We show here that endogenous PSKH1, a previously cloned kinase, is located in SFCs. Migration of PSKH1-FLAG into SFCs is enhanced during co-expression of T7-tagged ASF/SF2 as well as other members of the SR protein family, but not by two other non-SR nuclear proteins serving as controls. Similar to the SR protein kinase family, overexpression of PSKH1 led to reorganization of co-expressed T7-SC35 and T7-ASF/SF2 into a more diffuse nuclear pattern. This redistribution was not dependent on PSKH1 kinase activity. Different from the SR protein kinases, the SFC-associating features of PSKH1 were located within its catalytic kinase domain and within its C-terminus. Although no direct interaction was observed between PSKH1 and any of the SR proteins tested in pull-down or yeast two-hybrid assays, forced expression of PSKH1-FLAG was shown to stimulate distal splicing of an E1A minigene in HeLa cells. Moreover, a GST-ASF/SF2 fusion was not phosphorylated by PSKH1, suggesting an indirect mechanism of action on SR proteins. Our data suggest a mutual relationship between PSKH1 and SR proteins, as they are able to target PSKH1 into SFCs, while forced PSKH1 expression modulates nuclear dynamics and the function of co-expressed splicing factors.


Assuntos
Núcleo Celular/química , Proteínas Serina-Treonina Quinases/análise , Splicing de RNA , Ribonucleoproteínas , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Domínio Catalítico , Compartimento Celular , Células Cultivadas , Células HeLa , Humanos , Proteínas Nucleares/análise , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Precursores de RNA/metabolismo , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina , Spliceossomos/metabolismo , Transcrição Gênica
12.
FASEB J ; 16(11): 1450-2, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12205044

RESUMO

Tumor necrosis factor a (TNF-alpha) is a pleiotropic cytokine involved in several diseases. Various effects of TNF-alpha are mediated by the induction of a cellular state consistent with oxidative stress. Glutathione (GSH) is a major redox-buffer of eukaryotic cells and is important in the defense against oxidative stress. We hypothesized that persistent TNF-alpha secretion could induce oxidative stress through modulation of GSH metabolism. This hypothesis was examined in a transgenic mouse model with low, persistent expression of human TNF-alpha in the T cell compartment. Major findings were i) marked tissue-specific changes in GSH redox status and GSH regulating enzymes, with the most pronounced changes in liver; ii) moderate changes in GSH metabolism and up-regulation of GSH-regulating enzymes were observed in lung and kidney from transgenic mice; and iii) liver, lung and kidney from transgenic mice had decreased levels of total glutathione, whereas splenic CD4+ and CD8+ T cells had a marked increase in oxidized glutathione as the major change. Oxidative stress induced by persistent low-grade exposure to TNF-alpha in transgenic mice appears to involve marked organ-specific alterations in glutathione redox status and glutathione-regulating enzymes with the most pronounced changes in the liver. These mice constitute a useful model for immunodeficiency syndromes and chronic inflammatory diseases involving pathogenic interaction between TNF-alpha and oxidative stress.


Assuntos
Glutationa/metabolismo , Estresse Oxidativo , Fator de Necrose Tumoral alfa/genética , Animais , Antioxidantes/metabolismo , Humanos , Rim/enzimologia , Rim/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Pulmão/enzimologia , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Oxirredução , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
13.
J Biol Chem ; 277(26): 23620-6, 2002 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-11973337

RESUMO

Tissue factor (TF), a transmembrane receptor for the serine protease coagulation factor VII(a) (FVIIa), is the main initiator of the coagulation cascade. Through incompletely elucidated mechanisms, TF serves additional functions in tumor-associated angiogenesis and metastasis. We have studied interleukin-8 (IL-8) as a possible link between TF-FVIIa complex formation and subsequent processes. Recombinant human FVIIa induced the up-regulation of both IL-8 mRNA and protein in a FVIIa dose- and time-dependent fashion. A neutralizing antibody to TF reduced this induction by 93 +/- 5%. Active site-inhibited FVIIa had no stimulatory effect and completely blocked that of FVIIa. This confirms that the increased IL-8 production was dependent on the formation of TF-FVIIa complexes and the proteolytic activity of FVIIa. The IL-8 promoter contains DNA binding sites for nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1). In response to FVIIa, the DNA binding activity of both NF-kappaB and AP-1 was enhanced in an electrophoretic mobility shift assay. In addition, the IL-8 promoter was transcriptionally activated both in a luciferase reporter system and a nuclear run-off assay. Moreover, IL-8 mRNA stability was significantly enhanced by FVIIa-induced activation of the mitogen-activated protein kinases ERK1/2 and p38. Taken together, TF-FVIIa signaling induced increased transcription as well as mRNA stabilization leading to the significant up-regulation of IL-8 protein synthesis.


Assuntos
Fator VIIa/farmacologia , Interleucina-8/biossíntese , Queratinócitos/metabolismo , Tromboplastina/fisiologia , Linhagem Celular , DNA/metabolismo , Humanos , Interleucina-8/genética , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/fisiologia , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno
14.
Oncogene ; 21(12): 1832-40, 2002 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-11896615

RESUMO

The presence of putative tumor-suppressor genes on chromosome 16q23.2-24.1 has been suggested by LOH analysis in several cancer types. This region overlaps with the fragile site FRA16D and the region of homozygous deletions found in several cancer types. The candidate gene WWOX/FOR has been mapped within this region. The mouse homologue of the WWOX protein has been defined as an apoptogenic protein and an essential partner of p53 in cell death, supporting WWOX as a tumor suppressor gene candidate. We performed an expression study of the WWOX/FOR gene in a series of human breast tumors and breast cancer cell lines, and detected reduced expression of the WWOX/FOR transcript in a series of breast cancer cells. Furthermore, identification of two distinct alternative WWOX transcripts expressed at high levels in human tumors suggests an involvement of the WWOX gene in breast cancer progression.


Assuntos
Neoplasias da Mama/genética , Proteínas de Transporte/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes Supressores de Tumor , Proteínas de Neoplasias/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Processamento Alternativo/genética , Sequência de Bases , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Transporte/metabolismo , Cromossomos Humanos Par 16/genética , Ilhas de CpG , Primers do DNA/química , Feminino , Deleção de Genes , Humanos , Perda de Heterozigosidade , Repetições de Microssatélites , Pessoa de Meia-Idade , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA