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.
Bioorg Med Chem Lett ; 20(7): 2264-9, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20188554
2.
J Leukoc Biol ; 85(1): 132-5, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18945822

RESUMO

The TNF superfamily ligand, TNF-like weak inducer of apoptosis (TWEAK), regulates cellular responses ranging from proliferation to cell death in a manner highly dependent on the cell type and the microenvironmental context. We have shown previously that treatment of experimental autoimmune encephalomyelitis mice after the priming phase with neutralizing anti-TWEAK antibodies results in a reduction in the severity of the disease and leukocyte infiltration. To further characterize TWEAK/fibroblast growth factor-inducible 14-kDa protein (Fn14) involvement during multiple sclerosis (MS), we evaluated in MS patients and controls: TWEAK and Fn14 expression on PBMC and soluble TWEAK concentration in serum and cerebrospinal fluid (CSF). Thirty-six consecutive patients were enrolled, including 11 patients with relapsing-remitting MS, 11 with a clinical isolated syndrome suggestive of MS (CISSMS), and 14 controls with non-MS diseases. Intracellular TWEAK could be observed in lymphocytes and/or monocytes in all groups of patients. None of the 36 patients displayed TWEAK expression at the cell surface of lymphocytes. In contrast, 12 out of the 36 patients were positive for membrane TWEAK expression on their monocytes. Among these patients, eight were from the CISSMS group. Fn14 was not detected in PBMC. The soluble form of TWEAK is detectable in serum and CSF of patients, and TWEAK concentrations were not statistically different between the disease groups. We demonstrated for the first time that TWEAK is expressed at the cell surface of monocytes during MS, especially in the CISSMS group. Our results support the proposal that TWEAK could be a target for antibody therapy in MS.


Assuntos
Membrana Celular/metabolismo , Monócitos/metabolismo , Esclerose Múltipla/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/biossíntese , Adolescente , Adulto , Idoso , Citocina TWEAK , Feminino , Humanos , Linfócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Receptor de TWEAK , Fatores de Necrose Tumoral/sangue , Fatores de Necrose Tumoral/líquido cefalorraquidiano , Adulto Jovem
3.
J Neurol Sci ; 275(1-2): 117-20, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18793781

RESUMO

Stroke outcome is determined by delayed neuronal cell death and edema formation. TWEAK, a cytokine of the TNF superfamily, and its membrane receptor Fn14 promote ischemia-induced neuronal apoptosis and leakage of the blood-brain barrier. Both TWEAK and Fn14 are upregulated in experimental stroke models. In this study, we investigated whether TWEAK and Fn14 are upregulated in stroke patients. We measured serum concentrations of TWEAK in stroke patients and matched control subjects by ELISA. Expression of Fn14 in the brain was evaluated by real-time RT-PCR and immunohistochemistry. TWEAK serum concentrations were elevated in stroke patients. In autopsy samples, we found elevated mRNA levels of the receptor Fn14 and a trend towards higher TWEAK mRNA levels. In the infarcted and peri-infarct tissue immunostaining for Fn14 was enhanced. These data show that the cytokine TWEAK and its membrane receptor Fn14 are upregulated in stroke and suggest that they contribute to stroke outcome.


Assuntos
Encéfalo/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Acidente Vascular Cerebral , Fatores de Necrose Tumoral/sangue , Regulação para Cima/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Citocina TWEAK , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Lateralidade Funcional , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Receptor de TWEAK
4.
J Neuroimmunol ; 194(1-2): 97-106, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18207576

RESUMO

Inflammatory cytokines have been implicated in the pathology of multiple neurologic diseases, including multiple sclerosis. We examined the role of the TNF family member TWEAK in neuroinflammation. Cuprizone-fed mice undergo neuroinflammation and demyelination in the brain, but upon removal of cuprizone from the diet, inflammation is resolved and remyelination occurs. Using this model, we demonstrate that mice lacking TWEAK exhibit a significant delay in demyelination and microglial infiltration. During remyelination, mice lacking the TWEAK gene demonstrate only a marginal delay in remyelination. Thus, this study identifies a primary role of TWEAK in promoting neuroinflammation and exacerbating demyelination during cuprizone-induced damage.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Encefalite/fisiopatologia , Fatores de Necrose Tumoral/fisiologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Quelantes/toxicidade , Cobre , Cuprizona/toxicidade , Citocina TWEAK , Doenças Desmielinizantes/induzido quimicamente , Método Duplo-Cego , Encefalite/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Bainha de Mielina/fisiologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , RNA Mensageiro/biossíntese , Receptores do Fator de Necrose Tumoral/fisiologia , Receptor de TWEAK , Fatores de Necrose Tumoral/biossíntese , Fatores de Necrose Tumoral/deficiência , Fatores de Necrose Tumoral/genética
5.
Am J Respir Crit Care Med ; 177(1): 56-65, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17916809

RESUMO

RATIONALE: Transforming growth factor (TGF)-beta has a central role in driving many of the pathological processes that characterize pulmonary fibrosis. Inhibition of the integrin alpha(v)beta6, a key activator of TGF-beta in lung, is an attractive therapeutic strategy, as it may be possible to inhibit TGF-beta at sites of alpha(v)beta6 up-regulation without affecting other homeostatic roles of TGF-beta. OBJECTIVES: To analyze the expression of alpha(v)beta6 in human pulmonary fibrosis, and to functionally test the efficacy of therapeutic inhibition of alpha(v)beta6-mediated TGF-beta activation in murine bleomycin-induced pulmonary fibrosis. METHODS: Lung biopsies from patients with a diagnosis of systemic sclerosis or idiopathic pulmonary fibrosis were stained for alpha(v)beta6 expression. A range of concentrations of a monoclonal antibody that blocks alpha(v)beta6-mediated TGF-beta activation was evaluated in murine bleomycin-induced lung fibrosis. MEASUREMENTS AND MAIN RESULTS: Alpha(v)beta6 is overexpressed in human lung fibrosis within pneumocytes lining the alveolar ducts and alveoli. In the bleomycin model, alpha(v)beta6 antibody was effective in blocking pulmonary fibrosis. At high doses, there was increased expression of markers of inflammation and macrophage activation, consistent with the effects of TGF-beta inhibition in the lung. Low doses of antibody attenuated collagen expression without increasing alveolar inflammatory cell populations or macrophage activation markers. CONCLUSIONS: Partial inhibition of TGF-beta using alpha(v)beta6 integrin antibodies is effective in blocking murine pulmonary fibrosis without exacerbating inflammation. In addition, the elevated expression of alpha(v)beta6, an activator of the fibrogenic cytokine, TGF-beta, in human pulmonary fibrosis suggests that alpha(v)beta6 monoclonal antibodies could represent a promising new therapeutic strategy for treating pulmonary fibrosis.


Assuntos
Anticorpos Monoclonais/farmacologia , Modelos Animais de Doenças , Integrinas/antagonistas & inibidores , Fibrose Pulmonar/imunologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Antígenos de Neoplasias/fisiologia , Colágeno/metabolismo , Relação Dose-Resposta a Droga , Integrinas/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/imunologia , Fibrose Pulmonar/patologia , Fibrose Pulmonar/terapia , Escleroderma Sistêmico/imunologia , Escleroderma Sistêmico/patologia , Escleroderma Sistêmico/terapia , Fator de Crescimento Transformador beta/fisiologia
6.
Cytokine ; 40(1): 1-16, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17981048

RESUMO

First described as a weak apoptosis inducer, the TNF superfamily ligand TWEAK has since emerged as a cytokine that regulates multiple cellular responses, including proinflammatory activity, angiogenesis and cell proliferation, suggesting roles in inflammation and cancer. More recently TWEAK's ability to regulate progenitor cell fate was elucidated. Experiments using genetic overexpression and pathway inhibition or deficiency in mice indicate that TWEAK coordinates inflammatory and progenitor cell responses in settings of acute injury through its highly inducible receptor, FGF-inducible molecule 14 (Fn14), establishing the pathway's physiological role in facilitating acute tissue repair. In contrast, in chronic inflammatory disease models characterized by persistent TWEAK/Fn14 activation, TWEAK functions as a novel pathogenic mediator by amplifying inflammation, promoting tissue damage and potentially impeding endogenous repair mechanisms. Herein we aim not only to review the multifaceted functions of this emerging pathway, but also propose a conceptual framework for TWEAK/Fn14 pathway function in health and disease, supported by studies employing TWEAK and Fn14 deficient mice and anti-TWEAK blocking mAbs in acute injury and inflammatory disease settings. In addition to a perspective of the biology, we discuss potential therapeutic strategies targeting this pathway for the treatment of tissue injury, chronic inflammatory diseases and cancer.


Assuntos
Citocinas/fisiologia , Mediadores da Inflamação/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais/fisiologia , Fatores de Necrose Tumoral/fisiologia , Cicatrização/fisiologia , Animais , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Citocina TWEAK , Humanos , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores do Fator de Necrose Tumoral/genética , Receptor de TWEAK , Fatores de Necrose Tumoral/biossíntese , Cicatrização/genética
7.
Nat Med ; 13(10): 1228-33, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17906634

RESUMO

Demyelinating diseases, such as multiple sclerosis, are characterized by the loss of the myelin sheath around neurons, owing to inflammation and gliosis in the central nervous system (CNS). Current treatments therefore target anti-inflammatory mechanisms to impede or slow disease progression. The identification of a means to enhance axon myelination would present new therapeutic approaches to inhibit and possibly reverse disease progression. Previously, LRR and Ig domain-containing, Nogo receptor-interacting protein (LINGO-1) has been identified as an in vitro and in vivo negative regulator of oligodendrocyte differentiation and myelination. Here we show that loss of LINGO-1 function by Lingo1 gene knockout or by treatment with an antibody antagonist of LINGO-1 function leads to functional recovery from experimental autoimmune encephalomyelitis. This is reflected biologically by improved axonal integrity, as confirmed by magnetic resonance diffusion tensor imaging, and by newly formed myelin sheaths, as determined by electron microscopy. Antagonism of LINGO-1 or its pathway is therefore a promising approach for the treatment of demyelinating diseases of the CNS.


Assuntos
Axônios/fisiologia , Encefalomielite Autoimune Experimental/induzido quimicamente , Proteínas de Membrana/antagonistas & inibidores , Bainha de Mielina/fisiologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Traumatismos da Medula Espinal/terapia , Animais , Axônios/diagnóstico por imagem , Axônios/ultraestrutura , Encefalomielite Autoimune Experimental/patologia , Injeções Espinhais , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Proteínas da Mielina , Bainha de Mielina/ultraestrutura , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Associada a Mielina/farmacologia , Glicoproteína Mielina-Oligodendrócito , Proteínas do Tecido Nervoso/administração & dosagem , Proteínas do Tecido Nervoso/fisiologia , Ratos , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Tomografia Computadorizada por Raios X
8.
J Neurosci Res ; 85(16): 3535-44, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17803219

RESUMO

TWEAK/Fn14 signaling regulates progenitor cell proliferation, differentiation, and survival in multiple organ systems. This study examined the effects of TWEAK (tumor necrosis factor-like weak inducer of apoptosis) treatment on cultured mouse neural progenitor cells. The receptor for TWEAK is expressed by neural progenitor cells from the early embryonic stages through postnatal development. Although embryonic day 12 (E12) and postnatal day 1 (PN1) neural progenitor cells both express the receptor for TWEAK, TWEAK treatment of cultured E12 and PN1 progenitor cells resulted in age-dependent effects on proliferation and on neurite extension by neuronal progeny. TWEAK treatment did not alter proliferation of E12 neural progenitor cells but shifted PN1 progenitor cells toward cell-cycle phases G0 and G1 and reduced the rate at which they incorporated CldU. Conversely, the effects of TWEAK on axon elongation were more prominent in the earlier developmental stage. TWEAK induced extensive neurite outgrowth by the neuronal progeny of E12 but not PN1 progenitors. Treatment of the E12 progenitor cells with a TWEAK-neutralizing antibody repressed neurite extension, indicating that endogenous activation of this pathway may be required for neurite extension by the embryonic neuronal progeny. These studies indicate that TWEAK/Fn14 receptor activation exerts different effects on neural progenitor cells and their progeny depending on the developmental stage of the cells.


Assuntos
Envelhecimento/fisiologia , Encéfalo/embriologia , Neurônios/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Células-Tronco/metabolismo , Fatores de Necrose Tumoral/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos/farmacologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocina TWEAK , Regulação da Expressão Gênica no Desenvolvimento/genética , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/metabolismo , Cones de Crescimento/ultraestrutura , Camundongos , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/genética , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Receptor de TWEAK , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/farmacologia
9.
Am J Pathol ; 170(1): 110-25, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200187

RESUMO

The transforming growth factor (TGF)-beta-inducible integrin alpha v beta6 is preferentially expressed at sites of epithelial remodeling and has been shown to bind and activate latent precursor TGF-beta. Herein, we show that alpha v beta6 is overexpressed in human kidney epithelium in membranous glomerulonephritis, diabetes mellitus, IgA nephropathy, Goodpasture's syndrome, and Alport syndrome renal epithelium. To assess the potential regulatory role of alpha v beta6 in renal disease, we studied the effects of function-blocking alpha v beta6 monoclonal antibodies (mAbs) and genetic ablation of the beta6 subunit on kidney fibrosis in Col4A3-/- mice, a mouse model of Alport syndrome. Expression of alpha v beta6 in Alport mouse kidneys was observed primarily in cortical tubular epithelial cells and in correlation with the progression of fibrosis. Treatment with alpha v beta6-blocking mAbs inhibited accumulation of activated fibroblasts and deposition of interstitial collagen matrix. Similar inhibition of renal fibrosis was observed in beta6-deficient Alport mice. Transcript profiling of kidney tissues showed that alpha v beta6-blocking mAbs significantly inhibited disease-associated changes in expression of fibrotic and inflammatory mediators. Similar patterns of transcript modulation were produced with recombinant soluble TGF-beta RII treatment, suggesting shared regulatory functions of alpha v beta6 and TGF-beta. These findings demonstrate that alpha v beta6 can contribute to the regulation of renal fibrosis and suggest this integrin as a potential therapeutic target.


Assuntos
Antígenos de Neoplasias/biossíntese , Integrinas/biossíntese , Nefrite Hereditária/metabolismo , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Bloqueadores/farmacologia , Antígenos de Neoplasias/imunologia , Modelos Animais de Doenças , Matriz Extracelular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Humanos , Imuno-Histoquímica , Integrinas/antagonistas & inibidores , Integrinas/imunologia , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Knockout , Células NIH 3T3 , Nefrite Hereditária/tratamento farmacológico , Nefrite Hereditária/etiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima
10.
J Cereb Blood Flow Metab ; 27(3): 534-44, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16835630

RESUMO

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor superfamily. TWEAK acts via binding to a cell surface receptor named Fn14. To study the role of this cytokine in the regulation of the permeability of the neurovascular unit (NVU) during cerebral ischemia, TWEAK activity was inhibited in wild-type mice with a soluble Fn14-Fc decoy receptor administered either immediately or 1 h after middle cerebral artery occlusion (MCAO). Administration of Fn14-Fc decoy resulted in faster recovery of motor function and a 66.4%+/-10% decrease in Evans blue dye extravasation when treatment was administered immediately after MCAO and a 46.1%+/-13.1% decrease when animals were treated 1 h later (n=4, P<0.05). Genetic deficiency of Fn14 resulted in a 60%+/-12.8% decrease in the volume of the ischemic lesion (n=6, P<0.05), and a 87%+/-22% inhibition in Evans blue dye extravasation 48 h after the onset of the ischemic insult (n=6, P<0.005). Compared with control animals, treatment with Fn14-Fc decoy or genetic deficiency of Fn14 also resulted in a significant inhibition of nuclear factor-kappaB pathway activation, matrix metalloproteinase-9 activation and basement membrane laminin degradation after MCAO. These findings show that the cytokine TWEAK plays a role in the disruption of the structure of the NVU during cerebral ischemia and that TWEAK antagonism is a potential therapeutic strategy for acute cerebral ischemia.


Assuntos
Barreira Hematoencefálica/ultraestrutura , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Permeabilidade Capilar/fisiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/ultraestrutura , Barreira Hematoencefálica/metabolismo , Western Blotting , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Edema Encefálico , Isquemia Encefálica/etiologia , Citocina TWEAK , Ensaio de Desvio de Mobilidade Eletroforética , Células Endoteliais/ultraestrutura , Imunofluorescência , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Metaloproteinase 9 da Matriz , Camundongos , Microscopia Eletrônica de Transmissão , NF-kappa B/metabolismo , Recuperação de Função Fisiológica , Receptor de TWEAK
11.
Clin Immunol ; 117(1): 15-23, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16027043

RESUMO

TWEAK is a member of the TNF family, constitutively expressed in the central nervous system (CNS), with pro-inflammatory, proliferative or apoptotic effects depending upon cell types. Its receptor, Fn14, is expressed in CNS by endothelial cells, reactive astrocytes and neurons. We showed that TWEAK and Fn14 mRNA expression increased in spinal cord during experimental autoimmune encephalomyelitis (EAE). We investigated the role of TWEAK during EAE using neutralizing anti-TWEAK antibody in myelin oligodendrocyte glycoprotein (MOG) induced EAE in C57BL/6 mice. We observed a reduction of disease severity and leukocyte infiltration when mice were treated after the priming phase.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Encéfalo/efeitos dos fármacos , Proteínas de Transporte/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Medula Espinal/efeitos dos fármacos , Animais , Proteínas Reguladoras de Apoptose , Astrócitos/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Proteínas de Transporte/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citocina TWEAK , Encefalomielite Autoimune Experimental/patologia , Humanos , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/imunologia , Medula Espinal/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Fatores de Tempo , Fatores de Necrose Tumoral
12.
Oncogene ; 24(30): 4820-8, 2005 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-15856027

RESUMO

LMO4, a member of the LIM-only family of zinc-finger proteins, is overexpressed in a significant proportion of breast carcinomas and acts as a negative regulator of mammary epithelial differentiation. To delineate cell types within the developing mouse mammary gland that express Lmo4, we analysed different stages of mammopoiesis by immunohistochemistry. Lmo4 was found to be highly expressed in the proliferating cap cells of the terminal end bud and in the ductal and alveolar luminal cells of the mature mammary gland but was negligible or low in myoepithelial cells. To assess the physiological role of Lmo4 in the mammary gland, we generated conditionally targeted mice lacking Lmo4 in the mammary epithelium during pregnancy. Acute loss of Lmo4 in late pregnancy impaired lobuloalveolar development, accompanied by a two-fold reduction in the percentage of BrdU-positive cells. In contrast, germline loss of Lmo4 did not alter lobuloalveolar development arising from transplanted mammary anlagen, implying the existence of a compensatory mechanism in these knockout mice. Thus, the use of a conditional targeting strategy has revealed that Lmo4 is required for proper development of the mammary gland during pregnancy and indicated that Lmo4 acts as a positive regulator of alveolar epithelial proliferation.


Assuntos
Proteínas de Homeodomínio/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proliferação de Células , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas com Domínio LIM , Lactação , Glândulas Mamárias Animais/anatomia & histologia , Camundongos , Leite/metabolismo , Mutação/genética , Gravidez , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
13.
J Neurosci ; 24(38): 8237-44, 2004 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-15385607

RESUMO

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor (TNF) family of cytokines. It has proangiogenic and proinflammatory properties in vivo and induces cell death in tumor cell lines. TWEAK effects are mediated by the membrane receptor Fn14. In a systematic search for genes regulated in a murine stroke model with the tag-sequencing technique massively parallel signature sequencing, we have identified TWEAK as an induced gene. After 24 hr of focal cerebral ischemia in vivo or oxygen glucose deprivation in primary cortical neurons, both TWEAK and its receptor Fn14 were significantly upregulated. TWEAK induced cell death in primary neurons. Transfection of a nuclear factor (NF)-kappaB-luciferase fusion gene demonstrated that TWEAK stimulated transcriptional activity of NF-kappaB through Fn14 and the IkappaB kinase. Inhibition of NF-kappaB reduced TWEAK-stimulated neuronal cell death, suggesting that NF-kappaB mediates TWEAK-induced neurodegeneration at least in part. Intraperitoneal injection of a neutralizing anti-TWEAK antibody significantly reduced the infarct size after 48 hr of permanent cerebral ischemia. In summary, our data show that TWEAK induces neuronal cell death and is involved in neurodegeneration in vivo.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Degeneração Neural/metabolismo , Neurônios/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Anticorpos/administração & dosagem , Anticorpos/farmacologia , Proteínas Reguladoras de Apoptose , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Proteínas de Transporte/antagonistas & inibidores , Morte Celular/genética , Morte Celular/fisiologia , Células Cultivadas , Infarto Cerebral/genética , Infarto Cerebral/patologia , Infarto Cerebral/prevenção & controle , Citocina TWEAK , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Quinase I-kappa B , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , NF-kappa B/biossíntese , NF-kappa B/genética , Degeneração Neural/etiologia , Degeneração Neural/patologia , Neurônios/patologia , Proteínas Serina-Treonina Quinases/metabolismo , RNA/biossíntese , RNA/genética , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia , Receptor de TWEAK , Transfecção , Fatores de Necrose Tumoral , Regulação para Cima/genética
14.
Mol Cell Biol ; 24(5): 2074-82, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14966286

RESUMO

LMO4 belongs to a family of transcriptional regulators that comprises two zinc-binding LIM domains. LIM-only (LMO) proteins appear to function as docking sites for other factors, leading to the assembly of multiprotein complexes. The transcription factor Deaf-1/NUDR has been identified as one partner protein of LMO4. We have disrupted the Lmo4 and Deaf-1 genes in mice to define their biological function in vivo. All Lmo4 mutants died shortly after birth and showed defects within the presphenoid bone, with 50% of mice also exhibiting exencephaly. Homeotic transformations were observed in Lmo4-null embryos and newborn mice, but with incomplete penetrance. These included skeletal defects in cervical vertebrae and the rib cage. Furthermore, fusions of cranial nerves IX and X and defects in cranial nerve V were apparent in some Lmo4(-/-) and Lmo4(+/-) mice. Remarkably, Deaf-1 mutants displayed phenotypic abnormalities similar to those observed in Lmo4 mutants. These included exencephaly, transformation of cervical segments, and rib cage abnormalities. In contrast to Lmo4 nullizygous mice, nonexencephalic Deaf-1 mutants remained healthy. No defects in the sphenoid bone or cranial nerves were apparent. Thus, Lmo4 and Deaf-1 mutant mice exhibit overlapping as well as distinct phenotypes. Our data indicate an important role for these two transcriptional regulators in pathways affecting neural tube closure and skeletal patterning, most likely reflecting their presence in a functional complex in vivo.


Assuntos
Padronização Corporal , Sistema Nervoso Central/anormalidades , Sistema Nervoso Central/embriologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Animais Recém-Nascidos , Osso e Ossos/anormalidades , Sistema Nervoso Central/fisiologia , Nervos Cranianos/anormalidades , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/patologia , Embrião de Mamíferos/fisiologia , Marcação de Genes , Proteínas de Homeodomínio/genética , Proteínas com Domínio LIM , Camundongos , Camundongos Knockout , Morfogênese/fisiologia , Osso Esfenoide/anormalidades , Osso Esfenoide/embriologia , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...