Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1832(8): 1313-25, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23563317

RESUMO

BACKGROUND: Impairments in skeletal muscle oxidative phenotype (OXPHEN) have been linked to the development of insulin resistance, metabolic inflexibility and progression of the metabolic syndrome and have been associated with progressive disability in diseases associated with chronic systemic inflammation. We previously showed that the inflammatory cytokine tumour necrosis factor-α (TNF-α) directly impairs muscle OXPHEN but underlying molecular mechanisms remained unknown. Interestingly, the inflammatory signalling pathway classical nuclear factor-κB (NF-κB) is activated in muscle in abovementioned disorders. Therefore, we hypothesised that muscle activation of classical NF-κB signalling is sufficient and required for inflammation-induced impairment of muscle OXPHEN. METHODS: Myotubes from mouse and human muscle cell lines were subjected to activation or blockade of the classical NF-κB pathway. In addition, wild-type and MISR (muscle-specific inhibition of classical NF-κB) mice were injected intra-muscularly with TNF-α. Markers and key regulators of muscle OXPHEN were investigated. RESULTS: Classical NF-κB activation diminished expression of oxidative phosphorylation (OXPHOS) sub-units, slow myosin heavy chain expression, activity of mitochondrial enzymes and potently reduced intra-cellular ATP levels. Accordingly, PGC-1/PPAR/NRF-1/Tfam signalling, the main pathway controlling muscle OXPHEN, was impaired upon classical NF-κB activation which required intact p65 trans-activation domains and depended on de novo gene transcription. Unlike wild-type myotubes, IκBα-SR myotubes (blocked classical NF-κB signalling) were refractory to TNF-α-induced impairments in OXPHEN and its regulation by the PGC-1/PPAR/NRF-1/Tfam cascade. In line with in vitro data, NF-κB blockade in vivo abrogated TNF-α-induced reductions in PGC-1α expression. CONCLUSION: Classical NF-κB activation impairs skeletal muscle OXPHEN.


Assuntos
Músculo Esquelético/metabolismo , NF-kappa B/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Humanos , Inflamação/genética , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , NF-kappa B/genética , Oxirredução , Fenótipo , Fosforilação , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
2.
Gene Ther ; 17(12): 1476-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20720575

RESUMO

The IκB kinase (IKKα, ß and the regulatory subunit IKKγ) complex regulates nuclear factor of κB (NF-κB) transcriptional activity, which is upregulated in many chronic inflammatory diseases. NF-κB signaling promotes inflammation and limits muscle regeneration in Duchenne muscular dystrophy (DMD), resulting in fibrotic and fatty tissue replacement of muscle that exacerbates the wasting process in dystrophic muscles. Here, we examined whether dominant-negative forms of IKKα (IKKα-dn) and IKKß (IKKß-dn) delivered by adeno-associated viral (AAV) vectors to the gastrocnemius (GAS) and tibialis anterior (TA) muscles of 1, 2 and 11-month-old mdx mice, a murine DMD model, block NF-κB activation and increase muscle regeneration. At 1 month post-treatment, the levels of nuclear NF-κB in locally treated muscle were decreased by gene transfer with either AAV-CMV-IKKα-dn or AAV-CMV-IKKß-dn, but not by IKK wild-type controls (IKKα and ß) or phosphate-buffered saline (PBS). Although treatment with AAV-IKKα-dn or AAV-IKKß-dn vectors had no significant effect on muscle regeneration in young mdx mice treated at 1 and 2 months of age and collected 1 month later, treatment of old (11 months) mdx with AAV-CMV-IKKα-dn or AAV-CMV-IKKß-dn significantly increased levels of muscle regeneration. In addition, there was a significant decrease in myofiber necrosis in the AAV-IKKα-dn- and AAV-IKKß-dn-treated mdx muscle in both young and old mice. These results demonstrate that inhibition of IKKα or IKKß in dystrophic muscle reduces the adverse effects of NF-κB signaling, resulting in a therapeutic effect. Moreover, these results clearly demonstrate the therapeutic benefits of inhibiting NF-κB activation by AAV gene transfer in dystrophic muscle to promote regeneration, particularly in older mdx mice, and block necrosis.


Assuntos
Dependovirus/genética , Terapia Genética , Quinase I-kappa B , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne , NF-kappa B , Animais , Núcleo Celular/enzimologia , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/enzimologia , Distrofia Muscular de Duchenne/terapia , NF-kappa B/genética , NF-kappa B/metabolismo , Regeneração/fisiologia , Transdução de Sinais/genética
3.
Surgeon ; 4(5): 273-83, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17009546

RESUMO

Skeletal muscle wasting is an important systemic manifestation of a wide range of diseases, including trauma, sepsis and cancer. The clinical consequences of muscle wasting undoubtedly include significant patient morbidity and worsened survival. Recently, there has been important progress in our understanding of the molecular mechanisms behind muscle wasting. In this review, the common systemic mediators, intracellular signalling pathways and effector mechanisms of skeletal muscle wasting are discussed with particular reference to different models of wasting and the development of novel therapeutic strategies.


Assuntos
Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Doenças Musculoesqueléticas/metabolismo , Síndrome de Emaciação/metabolismo , Animais , Metabolismo Energético , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Músculo Esquelético/enzimologia , Músculo Esquelético/fisiopatologia , Doenças Musculoesqueléticas/fisiopatologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Síndrome de Emaciação/fisiopatologia
4.
J Biol Chem ; 276(42): 39259-63, 2001 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-11500509

RESUMO

The activation of nuclear factor kappaB (NF-kappa B) plays a pivotal role in the regulation of tumor necrosis factor (TNF)-mediated apoptosis. However, little is known about the regulation of TNF-mediated apoptosis by other signaling pathways or growth factors. Here, unexpectedly, we found that bone morphogenetic protein (BMP)-2 and BMP-4 inhibited TNF-mediated apoptosis by inhibition of caspase-8 activation in C2C12 cells, a pluripotent mesenchymal cell line that has the potential to differentiate into osteoblasts depending on BMP stimulation. Utilizing both a trans-dominant IkappaBalpha inhibitor of NF-kappaB expressed in C2C12 cells and IkappaB kinase beta-deficient embryonic mouse fibroblast, we show that BMP-mediated survival was independent of NF-kappaB activation. Rather, the antiapoptotic activity of BMPs functioned through the Smad signaling pathway. Thus, these findings provide the first report of a BMP/Smad signaling pathway that can inhibit TNF-mediated apoptosis, independent of the prosurvival activity of NF-kappaB. Our results suggest that BMPs not only stimulate osteoblast differentiation but can also promote cell survival during the induction of bone formation, offering new insight into the biological functions of BMPs.


Assuntos
Apoptose , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas I-kappa B , NF-kappa B/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Fator de Crescimento Transformador beta , Fator de Necrose Tumoral alfa/metabolismo , Animais , Western Blotting , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Fibroblastos/metabolismo , Genes Dominantes , Genes Reporter , Humanos , Luciferases/metabolismo , Mesoderma/metabolismo , Camundongos , Inibidor de NF-kappaB alfa , Osteoblastos/metabolismo , Fosforilação , Ligação Proteica , Proteínas Smad , Proteína Smad5 , Azul Tripano/farmacologia
5.
J Biol Chem ; 276(25): 22382-7, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11297551

RESUMO

The sedative and anti-nausea drug thalidomide, which causes birth defects in humans, has been shown to have both anti-inflammatory and anti-oncogenic properties. The anti-inflammatory effect of thalidomide is associated with suppression of cytokine expression and the anti-oncogenic effect with inhibition of angiogenesis. It is presently unclear whether the teratogenic properties of thalidomide are connected in any way to the beneficial, anti-disease characteristics of this drug. The transcription factor NF-kappaB has been shown to be a key regulator of inflammatory genes such as tumor necrosis factor-alpha and interleukin-8. Inhibition of NF-kappaB is associated with reduced inflammation in animal models, such as those for rheumatoid arthritis. We show here that thalidomide can block NF-kappaB activation through a mechanism that involves the inhibition of activity of the IkappaB kinase. Consistent with the observed inhibition of NF-kappaB, thalidomide blocked the cytokine-induced expression of NF-kappaB-regulated genes such as those encoding interleukin-8, TRAF1, and c-IAP2. These data indicate that the therapeutic potential for thalidomide may be based on its ability to block NF-kappaB activation through suppression of IkappaB kinase activity.


Assuntos
Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Talidomida/farmacologia , Sequência de Bases , Sondas de DNA , Humanos , Quinase I-kappa B , Interleucina-1/antagonistas & inibidores , Interleucina-1/fisiologia , Interleucina-8/genética , Células Jurkat , NF-kappa B/biossíntese , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia
6.
J Cell Biol ; 152(1): 87-96, 2001 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-11149923

RESUMO

Wnt signaling plays a critical role in development and oncogenesis. Although significant progress has been made in understanding the downstream signaling cascade of Wnt signaling, little is known regarding Wnt signaling modification of the cell death machinery. Given that numerous oncogenes transform cells by providing cell survival function, we hypothesized that Wnt signaling may inhibit apoptosis. Here, we report that cells expressing Wnt-1 were resistant to cancer therapy-mediated apoptosis. Wnt-1 signaling inhibited the cytochrome c release and the subsequent caspase-9 activation induced by chemotherapeutic drugs, including both vincristine and vinblastine. Furthermore, we found that Wnt-1-mediated cell survival was dependent on the activation of beta-catenin/T cell factor (Tcf) transcription. Inhibition of beta-catenin/Tcf transcription by expression of the dominant-negative mutant of Tcf-4 blocked Wnt-1-mediated cell survival and rendered cells sensitive to apoptotic stimuli. These results provide the first demonstration that Wnt-1 inhibits cancer therapy-mediated apoptosis and suggests that Wnt-1 may exhibit its oncogenic potential through a mechanism of anti-apoptosis.


Assuntos
Apoptose , Proteínas do Citoesqueleto/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/fisiologia , Transativadores , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas de Peixe-Zebra , Animais , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Sobrevivência Celular , Neoplasias Colorretais , Grupo dos Citocromos c/metabolismo , Ativação Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Ratos , Fatores de Transcrição TCF , Proteína 2 Semelhante ao Fator 7 de Transcrição , Transcrição Gênica , Células Tumorais Cultivadas , Proteínas Wnt , Proteína Wnt1 , beta Catenina
7.
Science ; 289(5488): 2363-6, 2000 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-11009425

RESUMO

MyoD regulates skeletal muscle differentiation (SMD) and is essential for repair of damaged tissue. The transcription factor nuclear factor kappa B (NF-kappaB) is activated by the cytokine tumor necrosis factor (TNF), a mediator of skeletal muscle wasting in cachexia. Here, the role of NF-kappaB in cytokine-induced muscle degeneration was explored. In differentiating C2C12 myocytes, TNF-induced activation of NF-kappaB inhibited SMD by suppressing MyoD mRNA at the posttranscriptional level. In contrast, in differentiated myotubes, TNF plus interferon-gamma (IFN-gamma) signaling was required for NF-kappaB-dependent down-regulation of MyoD and dysfunction of skeletal myofibers. MyoD mRNA was also down-regulated by TNF and IFN-gamma expression in mouse muscle in vivo. These data elucidate a possible mechanism that may underlie the skeletal muscle decay in cachexia.


Assuntos
Caquexia/etiologia , Proteínas I-kappa B , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Proteína MyoD/genética , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células CHO , Caquexia/metabolismo , Caquexia/patologia , Diferenciação Celular , Linhagem Celular , Cricetinae , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Interferon gama/farmacologia , Interleucinas/farmacologia , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Músculo Esquelético/patologia , Proteína MyoD/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição RelA , Transcrição Gênica , Transfecção
8.
Oncogene ; 19(9): 1123-31, 2000 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-10713699

RESUMO

Members of the NF-kappa B/Rel transcription factor family have been shown recently to be required for cellular transformation by oncogenic Ras and by other oncoproteins and to suppress transformation-associated apoptosis. Furthermore, NF-kappa B has been shown to be activated by several oncoproteins including HER2/Neu, a receptor tyrosine kinase often expressed in human breast cancer. Human breast cancer cell lines, human breast tumors and normal adjacent tissue were analysed by gel mobility shift assay, immunoblotting of nuclear extracts and immunohistochemistry for activation of NF-kappa B. Furthermore, RNA levels for NF-kappa B-activated genes were analysed in order to determine if NF-kappa B is functionally active in human breast cancer. Our data indicate that the p65/RelA subunit of NF-kappa B is activated (i.e., nuclear) in breast cancer cell lines. However, breast tumors exhibit an absence or low level of nuclear p65/RelA but show activated c-Rel, p50 and p52 as compared to nontumorigenic adjacent tissue. Additionally, the I kappa B homolog Bcl-3, which functions to stimulate transcription with p50 or p52, was also activated in breast tumors. There was no apparent correlation between estrogen receptor status and levels of nuclear NF-kappa B complexes. Transcripts of NF-kappa B-regulated genes were found elevated in breast tumors, as compared to adjacent normal tissue, indicating functional NF-kappa B activity. These data suggest a potential role for a subset of NF-kappa B and I kappa B family proteins, particularly NF-kappa B/p52 and Bcl-3, in human breast cancer. Additionally, the activation of functional NF-kappa B in these tumors likely involves a signal transduction pathway distinct from that utilized by cytokines.


Assuntos
Neoplasias da Mama/metabolismo , NF-kappa B/metabolismo , NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteína 3 do Linfoma de Células B , Neoplasias da Mama/química , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Núcleo Celular/metabolismo , DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Imuno-Histoquímica , NF-kappa B/genética , Subunidade p50 de NF-kappa B , Subunidade p52 de NF-kappa B , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-rel/metabolismo , Fator de Transcrição RelA , Fatores de Transcrição , Células Tumorais Cultivadas
9.
Mol Cell Biol ; 20(5): 1626-38, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10669740

RESUMO

It is well established that cell survival signals stimulated by growth factors, cytokines, and oncoproteins are initiated by phosphoinositide 3-kinase (PI3K)- and Akt-dependent signal transduction pathways. Oncogenic Ras, an upstream activator of Akt, requires NF-kappaB to initiate transformation, at least partially through the ability of NF-kappaB to suppress transformation-associated apoptosis. In this study, we show that oncogenic H-Ras requires PI3K and Akt to stimulate the transcriptional activity of NF-kappaB. Activated forms of H-Ras and MEKK stimulate signals that result in nuclear translocation and DNA binding of NF-kappaB as well as stimulation of the NF-kappaB transactivation potential. In contrast, activated PI3K or Akt stimulates NF-kappaB-dependent transcription by stimulating transactivation domain 1 of the p65 subunit rather than inducing NF-kappaB nuclear translocation via IkappaB degradation. Inhibition of IkappaB kinase (IKK), using an IKKbeta dominant negative protein, demonstrated that activated Akt requires IKK to efficiently stimulate the transactivation domain of the p65 subunit of NF-kappaB. Inhibition of endogenous Akt activity sensitized cells to H-Ras(V12)-induced apoptosis, which was associated with a loss of NF-kappaB transcriptional activity. Finally, Akt-transformed cells were shown to require NF-kappaB to suppress the ability of etoposide to induce apoptosis. Our work demonstrates that, unlike activated Ras, which can stimulate parallel pathways to activate both DNA binding and the transcriptional activity of NF-kappaB, Akt stimulates NF-kappaB predominantly by upregulating of the transactivation potential of p65.


Assuntos
Apoptose/genética , NF-kappa B/genética , Proteínas Oncogênicas de Retroviridae/genética , Transdução de Sinais/genética , Células 3T3 , Animais , Camundongos , Proteína Oncogênica v-akt , Fosfatidilinositol 3-Quinases/genética , Proteínas Tirosina Quinases/genética , Fator de Transcrição RelA , Ativação Transcricional
10.
Mol Cell Biol ; 19(9): 5923-9, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10454539

RESUMO

Recent evidence indicates that the transcription factor NF-kappaB is a major effector of inducible antiapoptotic mechanisms. For example, it was shown that NF-kappaB activation suppresses the activation of caspase 8, the apical caspase in tumor necrosis factor (TNF) receptor family signaling cascades, through the transcriptional regulation of certain TRAF and IAP proteins. However, it was unknown whether NF-kappaB controls other key regulatory mechanisms in apoptosis. Here we show that NF-kappaB activation suppresses mitochondrial release of cytochrome c through the activation of the Bcl-2 family member A1/Bfl-1. The restoration of A1 in NF-kappaB null cells diminished TNF-induced apoptosis by reducing the release of proapoptotic cytochrome c from mitochondria. In addition, A1 potently inhibited etoposide-induced apoptosis by inhibiting the release of cytochrome c and by blocking caspase 3 activation. Our findings demonstrate that A1 is an important antiapoptotic gene controlled by NF-kappaB and establish that the prosurvival function of NF-kappaB can be manifested at multiple levels.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/fisiologia , NF-kappa B/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Apoptose/efeitos dos fármacos , Apoptose/genética , Sequência de Bases , Caspase 3 , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Sondas de DNA/genética , Ativação Enzimática , Etoposídeo/farmacologia , Expressão Gênica , Humanos , Antígenos de Histocompatibilidade Menor , Mitocôndrias/metabolismo , Proteínas/genética , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
11.
Mol Cell Biol ; 19(8): 5785-99, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10409765

RESUMO

Accumulating evidence implicates the transcription factor NF-kappaB as a positive mediator of cell growth, but the molecular mechanism(s) involved in this process remains largely unknown. Here we use both a skeletal muscle differentiation model and normal diploid fibroblasts to gain insight into how NF-kappaB regulates cell growth and differentiation. Results obtained with the C2C12 myoblast cell line demonstrate that NF-kappaB functions as an inhibitor of myogenic differentiation. Myoblasts generated to lack NF-kappaB activity displayed defects in cellular proliferation and cell cycle exit upon differentiation. An analysis of cell cycle markers revealed that NF-kappaB activates cyclin D1 expression, and the results showed that this regulatory pathway is one mechanism by which NF-kappaB inhibits myogenesis. NF-kappaB regulation of cyclin D1 occurs at the transcriptional level and is mediated by direct binding of NF-kappaB to multiple sites in the cyclin D1 promoter. Using diploid fibroblasts, we demonstrate that NF-kappaB is required to induce cyclin D1 expression and pRb hyperphosphorylation and promote G(1)-to-S progression. Consistent with results obtained with the C2C12 differentiation model, we show that NF-kappaB also promotes cell growth in embryonic fibroblasts, correlating with its regulation of cyclin D1. These data therefore identify cyclin D1 as an important transcriptional target of NF-kappaB and reveal a mechanism to explain how NF-kappaB is involved in the early phases of the cell cycle to regulate cell growth and differentiation.


Assuntos
Diferenciação Celular/genética , Divisão Celular/genética , Ciclina D1/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , NF-kappa B/fisiologia , Transcrição Gênica , Células 3T3/citologia , Células 3T3/metabolismo , Animais , Ciclo Celular/genética , Transformação Celular Neoplásica , Células Cultivadas , Sequência Consenso , Ciclina D1/genética , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/metabolismo , Fase G1 , Células HeLa/citologia , Células HeLa/metabolismo , Humanos , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Transfecção
12.
Am J Respir Cell Mol Biol ; 21(2): 238-45, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10423407

RESUMO

Current paradigms in cancer therapy suggest that activation of nuclear factor-kappaB (NF-kappaB) by a variety of stimuli, including some cytoreductive agents, may inhibit apoptosis. Thus, inhibiting NF-kappaB activation may sensitize cells to anticancer therapy, thereby providing a more effective treatment for certain cancers. E-1-deleted adenoviral (Ad) vectors encoding a "superrepressor" form of the NF-kappaB inhibitor IkappaBalpha (AdIkappaBalphaSR) or beta-galactosidase (AdLacZ) were tested alone and in combination with tumor necrosis factor-alpha (TNF-alpha) in lung cancer cells for sensitization of the cells to death. Following transduction with AdIkappaBalphaSR, lung cancer cells expressed IkappaBalphaSR in a dose-dependent manner. Probing nuclear extracts of lung cancer cells with NF-kappaB-sequence-specific oligonucleotides indicated that there was a minimal amount of NF-kappaB in the nucleus at baseline and an expected and dramatic increase in nuclear NF-kappaB following exposure of cells to TNF-alpha. Control E-1-deleted AdLacZ did not promote NF-kappaB activation. Importantly, AdIkappaBalphaSR-mediated gene transfer resulted in the complete block of nuclear translocation of NF-kappaB by specific binding of its p65/relA component with transgenic IkappaBalphaSR. At the cellular level, transduction with AdIkappaBalphaSR resulted in increased cytotoxicity in lung cancer cells as opposed to transduction with equivalent doses of AdLacZ. In addition, whereas the parental cells were resistant to TNF-alpha-mediated cytotoxicity, IkappaBalphaSR-transduced cells could be sensitized to TNF-alpha. Consequently, AdIkappaBalphaSR transduction followed by exposure to TNF-alpha uniformly resulted in the death of non-small-cell lung cancer cells. These data suggest that novel approaches incorporating IkappaBalpha gene therapy may have a role in the treatment of lung cancer.


Assuntos
Carcinoma de Células Escamosas/terapia , Proteínas de Ligação a DNA/genética , Técnicas de Transferência de Genes , Proteínas I-kappa B , Fator de Necrose Tumoral alfa/farmacologia , Adenoviridae/genética , Carcinoma de Células Escamosas/metabolismo , Morte Celular , Sobrevivência Celular , Relação Dose-Resposta a Droga , Vetores Genéticos , Humanos , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Transdução Genética , Transgenes , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
13.
J Biol Chem ; 272(39): 24117-20, 1997 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-9305855

RESUMO

Thrombin is a multifunctional serine protease whose activity is regulated in the extravasculature by an extracellular inhibitor, protease nexin-1. Because protease nexin-1 expression has been shown to be regulated during skeletal muscle cell differentiation, we reasoned that thrombin inactivation may be an important requirement for this developmental process. To test this hypothesis, we examined the effects of thrombin on differentiating C2C12 myoblasts. We report here that myogenesis, as scored by myotube formation, is considerably delayed by thrombin. This regulation correlated with delayed expression of myogenin and p21(CIP1/WAF1), both considered critical components of the skeletal muscle cell differentiation program. Regulation occurred at the RNA level, indicating that the effect of thrombin is either transcriptional or post-transcriptional. Furthermore, we present evidence suggesting that this regulation is mediated by the thrombin receptor. Although thrombin is mitogenic for certain cell types, we found that delay of myogenesis in C2C12 cells did not involve a mitogenic signal. Taken together, these results imply that inhibition of the serine protease thrombin may be required for proper progression through the myogenic differentiation program. The data point to potentially important roles that thrombin and protease nexin-1 may play during skeletal muscle development.


Assuntos
Ciclinas/genética , Regulação da Expressão Gênica/fisiologia , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Miogenina/genética , Trombina/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21 , Camundongos , Músculo Esquelético/metabolismo , Receptores de Trombina/fisiologia
14.
J Neurochem ; 67(2): 498-507, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8764573

RESUMO

Protease nexin-1 (PN-1 ) is a potent inhibitor of serine proteases in the extracellular environment. It is abundantly expressed in the nervous system, where it is thought to participate in local injury and repair processes. Although some information has been obtained regarding PN-1 gene structure, relatively little is known about the cis- and trans-acting factors that regulate its expression. Elucidation of these factors should provide a better understanding of PN-1 function during development and wound repair. In this report we describe the characterization of the human PN-1 promoter and identify regulatory domains and a transactivator mediating its transcriptional activity. The promoter is highly G/C rich proximal to the transcriptional start site. It exhibits tissue specificity and is negatively regulated by a silencer element upstream of position -480. A positive regulatory element was mapped between -199 and -45, which contains multiple putative Sp1 consensus binding sites. Electrophoretic mobility shift analysis confirmed that Sp1 specifically binds this region of the PN-1 promoter. DNase I foot-printing revealed six potential Sp1 binding sites between -103 and -56 that were protected by recombinant Sp1. Cotransfection experiments into the Sp1-deficient Drosophila SL2 cell line also showed that Sp1 activates PN-1 promoter activity in a dose-dependent fashion. Thus, our analysis demonstrates that activation of PN-11 transcription is regulated by Sp1 through G/C-rich cis-acting elements in the 5'proximal promoter region.


Assuntos
Regiões Promotoras Genéticas , Fator de Transcrição Sp1/genética , Precursor de Proteína beta-Amiloide , Animais , Sequência de Bases , Sítios de Ligação , Proteínas de Transporte/genética , Células Cultivadas , Primers do DNA/química , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Dados de Sequência Molecular , Nexinas de Proteases , Ratos , Receptores de Superfície Celular , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Serpina E2 , Transcrição Gênica , Ativação Transcricional
15.
J Biol Chem ; 268(25): 18966-74, 1993 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-8360185

RESUMO

Thrombin participates in several regulatory events following injury as a result of its effects on blood coagulation and cell migration, proliferation, and differentiation. Protease nexin-1 (PN-1) is a potent thrombin inhibitor in the extracellular environment. Since injury-related factors are known to regulate the synthesis and secretion of PN-1, the inhibitor may serve to modulate the actions of thrombin during injury. Here we report the molecular mechanisms that underlie this regulation. In normal human fibroblasts, interleukin-1 (IL-1) beta stimulated the synthesis and secretion of PN-1. The stimulation correlated with an increase in steady-state levels of PN-1 mRNA. Treatment of cells with both cycloheximide and IL-1 reduced the levels of PN-1 mRNA. Nuclear run-on assays indicated that IL-1 modestly increased the rate of PN-1 transcription. However, experiments with actinomycin D demonstrated that IL-1 significantly increased the half-life of the PN-1 mRNA. In contrast, dexamethasone (DXM) repressed the synthesis and secretion of PN-1 from fibroblasts. This effect correlated with a decrease in PN-1 mRNA. A sustained decrease in PN-1 mRNA was also seen when cells were treated with cycloheximide and DXM. In nuclear run-on assays, DXM functioned as a transcriptional repressor of PN-1 synthesis. Treatment of cells with actinomycin D showed that DXM did not affect mRNA stability. Thus, our experiments demonstrate that IL-1 and DXM, which function biologically in different fashions, regulate the synthesis of PN-1 by separate molecular mechanisms. While DXM directly regulates PN-1 at the level of transcription, IL-1 in the presence of ongoing protein synthesis regulates PN-1 production predominantly in a post-transcriptional fashion by increasing the half-life of the PN-1 mRNA.


Assuntos
Proteínas de Transporte/genética , Dexametasona/farmacologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Interleucina-1/farmacologia , Trombina/antagonistas & inibidores , Precursor de Proteína beta-Amiloide , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Cicloeximida/farmacologia , Citocinas/farmacologia , Dactinomicina/farmacologia , Estabilidade de Medicamentos , Matriz Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Humanos , Nexinas de Proteases , RNA Mensageiro/metabolismo , Receptores de Superfície Celular , Serpina E2 , Trombina/metabolismo , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...