Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Immunol ; 195(1): 246-56, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26026062

RESUMO

Previous work has shown conflicting roles for Tec family kinases in regulation of TLR-dependent signaling in myeloid cells. In the present study, we performed a detailed investigation of the role of the Tec kinases Btk and Tec kinases in regulating TLR signaling in several types of primary murine macrophages. We demonstrate that primary resident peritoneal macrophages deficient for Btk and Tec secrete less proinflammatory cytokines in response to TLR stimulation than do wild-type cells. In contrast, we found that bone marrow-derived and thioglycollate-elicited peritoneal macrophages deficient for Btk and Tec secrete more proinflammatory cytokines than do wild-type cells. We then compared the phosphoproteome regulated by Tec kinases and LPS in primary peritoneal and bone marrow-derived macrophages. From this analysis we determined that Tec kinases regulate different signaling programs in these cell types. In additional studies using bone marrow-derived macrophages, we found that Tec and Btk promote phosphorylation events necessary for immunoreceptor-mediated inhibition of TLR signaling. Taken together, our results are consistent with a model where Tec kinases (Btk, Tec, Bmx) are required for TLR-dependent signaling in many types of myeloid cells. However, our data also support a cell type-specific TLR inhibitory role for Btk and Tec that is mediated by immunoreceptor activation and signaling via PI3K.


Assuntos
Macrófagos/imunologia , Fosfoproteínas/imunologia , Proteínas Tirosina Quinases/imunologia , Tirosina Quinase da Agamaglobulinemia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , Cavidade Peritoneal/citologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Fosfoproteínas/genética , Fosforilação , Cultura Primária de Células , Proteínas Tirosina Quinases/genética , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
2.
Transl Res ; 249: 49-73, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35691544

RESUMO

As an anti-inflammatory strategy, MAPK-activated protein kinase-2 (MK2) inhibition can potentially avoid the clinical failures seen for direct p38 inhibitors, especially tachyphylaxis. CC-99677, a selective targeted covalent MK2 inhibitor, employs a rare chloropyrimidine that bonds to the sulfur of cysteine 140 in the ATP binding site via a nucleophilic aromatic substitutions (SNAr) mechanism. This irreversible mechanism translates biochemical potency to cells shown by potent inhibition of heat shock protein 27 (HSP27) phosphorylation in LPS-activated monocytic THP-1 cells. The cytokine inhibitory profile of CC-99677 differentiates it from known p38 inhibitors, potentially suppressing a p38 pathway inflammatory response while avoiding tachyphylaxis. Dosed orally, CC-99677 is efficacious in a rat model of ankylosing spondylitis. Single doses, 3 to 400 mg, in healthy human volunteers show linear pharmacokinetics and apparent sustained tumor necrosis factor-α inhibition, with a favorable safety profile. These results support further development of CC-99677 for autoimmune diseases like ankylosing spondylitis.


Assuntos
Doenças Autoimunes , Espondilite Anquilosante , Trifosfato de Adenosina , Animais , Anti-Inflamatórios , Doenças Autoimunes/tratamento farmacológico , Cisteína , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipopolissacarídeos , Proteínas Serina-Treonina Quinases , Ratos , Enxofre , Fator de Necrose Tumoral alfa , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
J Clin Invest ; 115(9): 2330-40, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16110324

RESUMO

Progenitor ("oval") cell expansion accompanies many forms of liver injury, including alcohol toxicity and submassive parenchymal necrosis as well as experimental injury models featuring blocked hepatocyte replication. Oval cells can potentially become either hepatocytes or biliary epithelial cells and may be critical to liver regeneration, particularly when hepatocyte replication is impaired. The regulation of oval cell proliferation is incompletely understood. Herein we present evidence that a TNF family member called TWEAK (TNF-like weak inducer of apoptosis) stimulates oval cell proliferation in mouse liver through its receptor Fn14. TWEAK has no effect on mature hepatocytes and thus appears to be selective for oval cells. Transgenic mice overexpressing TWEAK in hepatocytes exhibit periportal oval cell hyperplasia. A similar phenotype was obtained in adult wild-type mice, but not Fn14-null mice, by administering TWEAK-expressing adenovirus. Oval cell expansion induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) was significantly reduced in Fn14-null mice as well as in adult wild-type mice with a blocking anti-TWEAK mAb. Importantly, TWEAK stimulated the proliferation of an oval cell culture model. Finally, we show increased Fn14 expression in chronic hepatitis C and other human liver diseases relative to its expression in normal liver, which suggests a role for the TWEAK/Fn14 pathway in human liver injury. We conclude that TWEAK has a selective mitogenic effect for liver oval cells that distinguishes it from other previously described growth factors.


Assuntos
Proliferação de Células , Fígado/citologia , Fígado/metabolismo , Células-Tronco/fisiologia , Fatores de Necrose Tumoral/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Citocina TWEAK , Feminino , Humanos , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Células-Tronco/citologia , Receptor de TWEAK , Distribuição Tecidual , Transgenes , Fatores de Necrose Tumoral/genética
4.
Oncogene ; 24(16): 2613-24, 2005 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-15735761

RESUMO

Members of the tumor necrosis factor (TNF) superfamily regulate cell survival and proliferation and have been implicated in cancer. Tweak (TNF-related weak inducer of apoptosis) has pleiotropic biological functions including proapoptotic, proangiogenic and proinflammatory activities. We explored a role for Tweak in mammary gland transformation using a three-dimensional model culture system. Tweak stimulates a branching morphogenic phenotype, similar to that induced by pro-oncogenic factors, in Eph4 mammary epithelial cells cultured in matrigel. Increased proliferation and invasiveness are observed, with a concomitant inhibition of functional differentiation. Levels of matrix metalloproteinase-9 (MMP-9) are significantly increased following Tweak treatment. Notably, MMP inhibitors are sufficient to block the branching phenotype induced by Tweak. The capacity to promote proliferation, inhibit differentiation and induce invasion suggests a role for Tweak in mammary gland tumorigenesis. Consistent with this, we have observed elevated protein levels of the Tweak receptor, Fn14, in human breast tumor cell lines and xenograft models as well as in primary human breast tumors. Together, our results suggest that the Tweak/Fn14 pathway may be protumorigenic in human breast cancer.


Assuntos
Proteínas de Transporte/fisiologia , Glândulas Mamárias Humanas/citologia , Proteínas Reguladoras de Apoptose , Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Colágeno , Citocina TWEAK , Combinação de Medicamentos , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica , Laminina , Metaloproteinase 9 da Matriz/metabolismo , Modelos Biológicos , Morfogênese , Proteoglicanas , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor de TWEAK , Fatores de Necrose Tumoral
5.
AANA J ; 72(2): 129-32, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15098526

RESUMO

The purpose of this study was to determine if 4 mg of ondansetron and 12.5 mg of dolasetron were equally effective in preventing postoperative nausea and vomiting (PONV) in patients undergoing gynecological procedures. While the overall incidence of PONV appears to be 25% to 35%, the incidence among this patient population is considerably higher. Patients were assigned to 1 of 2 antiemetic treatment groups. Patients in group 1 received 4 mg of ondansetron at the end of surgery, while patients in group 2 received 12.5 mg of dolasetron at the end of surgery. Data collection occurred perioperatively and in the 24 hours following surgery. chi 2 determined there was no statistical difference between groups related to emesis in the postanesthesia care unit (PACU), emesis in the 24 hours following surgery, and side effects. Results of this study showed there was no statistically significant difference between 4 mg of ondansetron or 12.5 mg of dolasetron when administered at the end of surgery for preventing PONV in patients undergoing gynecological procedures. Given the cost difference between these 2 antiemetics, there is a potential for significant cost savings in this high-risk patient population.


Assuntos
Antieméticos/uso terapêutico , Procedimentos Cirúrgicos em Ginecologia/efeitos adversos , Indóis/uso terapêutico , Ondansetron/uso terapêutico , Náusea e Vômito Pós-Operatórios/tratamento farmacológico , Quinolizinas/uso terapêutico , Antieméticos/economia , Redução de Custos , Método Duplo-Cego , Custos de Medicamentos/estatística & dados numéricos , Feminino , Humanos , Incidência , Indóis/economia , Ondansetron/economia , Náusea e Vômito Pós-Operatórios/epidemiologia , Náusea e Vômito Pós-Operatórios/etiologia , Estudos Prospectivos , Quinolizinas/economia , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento
6.
Cancer Biol Ther ; 9(6): 437-45, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20061819

RESUMO

Integrin alpha6beta4 signaling interactions have been implicated in tumor progression, and beta4 expression has been linked to poor prognosis in certain breast cancer subtypes. We generated human antibodies to alpha6beta4 to further evaluate its role in tumor cell signaling. Biochemical characterization indicated these antibodies are specific for alpha6beta4, recognize distinct epitopes and have low nanomolar affinities for both human and murine protein. The antibodies demonstrated differing effects on alpha6beta4-mediated cellular adhesion, highlighting the existence of different functional epitopes on alpha6beta4. Interestingly however both antibodies blocked adhesion-independent growth in a panel of breast cancer cell lines. Antibody induced apoptosis and inhibition of phosphoinositide 3-kinase (PI3K) signaling were also observed within the context of matrix adhesion. Enhanced inhibitory effects were observed when the alpha6beta4 antibodies were used in combination with antibodies to epidermal growth factor receptor (EGFR) or erythoblastic leukemia viral oncogene homolog 2 (ErbB2). These findings illustrate a role for both the adhesive and signaling functions of alpha6beta4 in breast cancer cell survival. The antibodies and data generated herein advance our understanding of alpha6beta4 in regulating tumorigenic processes, and suggest that combination therapies involving alpha6beta4 may be therapeutically effective in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Integrina alfa6beta4/metabolismo , Integrina alfa6beta4/fisiologia , Anticorpos/metabolismo , Apoptose/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular , Receptores ErbB/metabolismo , Feminino , Humanos , Integrinas/metabolismo , Neoplasias/metabolismo , Transdução de Sinais
7.
Cell Commun Adhes ; 15(4): 317-31, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18979297

RESUMO

Integrin alpha6beta4-mediated adhesion interactions play key roles in keratinocyte and epithelial tumor cell biology. In order to evaluate how alpha6beta4 adhesion interactions contribute to these important cellular processes, the authors generated soluble versions of the integrin by recombinant expression of the subunit ectodomains fused to a human immunoglobulin G (IgG) Fc constant domain. Coexpression of the appropriate subunits enabled dimerization, secretion and purification of stable Fc-containing alpha6beta4 heterodimers. The soluble proteins exhibited the same metal ion and ligand dependency in their binding characteristics as intact alpha6beta4. Using these reagents in combination with anti-beta4 antibodies, the authors identified two distinct functional epitopes on the beta4 subunit. They demonstrated the involvement of one epitope in adhesion interactions and the other in regulating adhesion-independent growth in alpha6beta4-expressing tumor cell lines. The availability of these soluble integrin reagents and the data provided herein help to further delineate the structure-function relationships regulating alpha6beta4 signaling biology.


Assuntos
Integrina alfa6beta4/fisiologia , Integrina beta4/química , Animais , Anticorpos/metabolismo , Células CHO , Adesão Celular , Comunicação Celular , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Dimerização , Humanos , Integrina beta4/imunologia , Integrina beta4/fisiologia , Células K562 , Queratinócitos/citologia , Queratinócitos/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
8.
J Exp Med ; 205(5): 1049-62, 2008 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-18411337

RESUMO

T helper type 17 (Th17) cells play an important pathogenic function in autoimmune diseases; their regulation, however, is not well understood. We show that the expression of a tumor necrosis factor receptor family member, death receptor 3 (DR3; also known as TNFRSF25), is selectively elevated in Th17 cells, and that TL1A, its cognate ligand, can promote the proliferation of effector Th17 cells. To further investigate the role of the TL1A-DR3 pathway in Th17 regulation, we generated a TL1A-deficient mouse and found that TL1A(-/-) dendritic cells exhibited a reduced capacity in supporting Th17 differentiation and proliferation. Consistent with these data, TL1A(-/-) animals displayed decreased clinical severity in experimental autoimmune encephalomyelitis (EAE). Finally, we demonstrated that during EAE disease progression, TL1A was required for the optimal differentiation as well as effector function of Th17 cells. These observations thus establish an important role of the TL1A-DR3 pathway in promoting Th17 cell function and Th17-mediated autoimmune disease.


Assuntos
Doenças Autoimunes/imunologia , Encefalomielite Autoimune Experimental/imunologia , Antígeno HLA-DR3/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/deficiência , Animais , Encéfalo/imunologia , Encéfalo/patologia , Diferenciação Celular , Divisão Celular , Citocinas/metabolismo , Primers do DNA , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medula Espinal/imunologia , Medula Espinal/patologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
9.
J Immunol ; 176(3): 1889-98, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16424220

RESUMO

TNF-like weak inducer of apoptosis, or TWEAK, is a relatively new member of the TNF-ligand superfamily. Ligation of the TWEAK receptor Fn14 by TWEAK has proinflammatory effects on fibroblasts, synoviocytes, and endothelial cells. Several of the TWEAK-inducible cytokines are important in the pathogenesis of kidney diseases; however, whether TWEAK can induce a proinflammatory effect on kidney cells is not known. We found that murine mesangial cells express cell surface TWEAK receptor. TWEAK stimulation of mesangial cells led to a dose-dependent increase in CCL2/MCP-1, CCL5/RANTES, CXCL10/IFN-gamma-induced protein 10 kDa, and CXCL1/KC. The induced levels of chemokines were comparable to those found following mesangial cell exposure to potent proinflammatory stimuli such as TNF-alpha + IL-1beta. CXCL11/interferon-inducible T cell alpha chemoattractant, CXCR5, mucosal addressin cell adhesion molecule-1, and VCAM-1 were up-regulated by TWEAK as well. TWEAK stimulation of mesangial cells resulted in an increase in phosphorylated Ikappa-B, while pretreatment with an Ikappa-B phosphorylation inhibitor significantly blocked chemokine induction, implicating activation of the NF-kappaB signaling pathway in TWEAK-induced chemokine secretion. Importantly, the Fn14-mediated proinflammatory effects of TWEAK on kidney cells were confirmed using mesangial cells derived from Fn14-deficient mice and by injection in vivo of TWEAK into wild-type vs Fn14-deficient mice. Finally, TWEAK-induced chemokine secretion was prevented by treatment with novel murine anti-TWEAK Abs. We conclude that TWEAK induces mesangial cells to secrete proinflammatory chemokines, suggesting a prominent role for TWEAK in the pathogenesis of renal injury. Our results support Ab inhibition of TWEAK as a potential new approach for the treatment of chemokine-dependent inflammatory kidney diseases.


Assuntos
Mesângio Glomerular/metabolismo , Mediadores da Inflamação/fisiologia , Células Mesangiais/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Fatores de Necrose Tumoral/fisiologia , Animais , Anticorpos Monoclonais , Apoptose/imunologia , Linhagem Celular Transformada , Células Cultivadas , Quimiocinas/biossíntese , Citocina TWEAK , Proteínas I-kappa B/metabolismo , Mediadores da Inflamação/metabolismo , Interferon gama/fisiologia , Interleucina-1/fisiologia , Interleucina-6/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Inibidor de NF-kappaB alfa , Fosforilação , Receptores do Fator de Necrose Tumoral/genética , Receptor de TWEAK , Inibidores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/fisiologia , Fatores de Necrose Tumoral/imunologia , Fatores de Necrose Tumoral/metabolismo , Regulação para Cima/fisiologia
10.
J Immunol ; 177(4): 2610-20, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16888023

RESUMO

TNF-like weak inducer of apoptosis (TWEAK) is a TNF family member with pleiotropic effects on a variety of cell types, one of which is the induction of proinflammatory cytokines by synovial fibroblasts derived from rheumatoid arthritis (RA) patients. In this study, we report that the serum TWEAK level was dramatically elevated during mouse collagen-induced arthritis (CIA) and blocking TWEAK by a neutralizing mAb significantly reduced the clinical severity of CIA. Histological analyses also revealed that TWEAK inhibition diminished joint inflammation, synovial angiogenesis, as well as cartilage and bone erosion. Anti-TWEAK treatment proved efficacious when administered just before the disease onset but not during the priming phase of CIA. Consistent with this, TWEAK inhibition did not affect either cellular or humoral responses to collagen. In contrast, TWEAK inhibition significantly reduced serum levels of a panel of arthritogenic mediators, including chemokines such as MIP-1beta (CCL-4), lymphotactin (XCL-1), IFN-gamma-inducible protein 10 (IP-10) (CXCL-10), MCP-1 (CCL-2), and RANTES (CCL-5), as well as the matrix metalloprotease-9. Exploring the possible role of the TWEAK/Fn14 pathway in human RA pathogenesis, we showed that TWEAK can target human primary chondrocytes and osteoblast-like cells, in addition to synovial fibroblasts. We further demonstrated that TWEAK induced the production of matrix metalloproteases in human chondrocytes and potently inhibited chondrogenesis and osteogenesis using in vitro models. These results provide evidence for a novel cytokine pathway that contributes to joint tissue inflammation, angiogenesis, and damage, as well as may inhibit endogenous repair, suggesting that TWEAK may be a new therapeutic target for human RA.


Assuntos
Artrite Experimental/metabolismo , Artrite Experimental/patologia , Mediadores da Inflamação/fisiologia , Fatores de Necrose Tumoral/fisiologia , Animais , Apoptose/imunologia , Artrite Experimental/sangue , Células Cultivadas , Colágeno Tipo II/administração & dosagem , Citocina TWEAK , Adjuvante de Freund/administração & dosagem , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/sangue , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/fisiologia , Receptor de TWEAK , Inibidores do Fator de Necrose Tumoral , Fatores de Necrose Tumoral/biossíntese , Fatores de Necrose Tumoral/sangue
11.
EMBO J ; 25(24): 5826-39, 2006 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17124496

RESUMO

Inflammation participates in tissue repair through multiple mechanisms including directly regulating the cell fate of resident progenitor cells critical for successful regeneration. Upon surveying target cell types of the TNF ligand TWEAK, we observed that TWEAK binds to all progenitor cells of the mesenchymal lineage and induces NF-kappaB activation and the expression of pro-survival, pro-proliferative and homing receptor genes in the mesenchymal stem cells, suggesting that this pro-inflammatory cytokine may play an important role in controlling progenitor cell biology. We explored this potential using both the established C2C12 cell line and primary mouse muscle myoblasts, and demonstrated that TWEAK promoted their proliferation and inhibited their terminal differentiation. By generating mice deficient in the TWEAK receptor Fn14, we further showed that Fn14-deficient primary myoblasts displayed significantly reduced proliferative capacity and altered myotube formation. Following cardiotoxin injection, a known trigger for satellite cell-driven skeletal muscle regeneration, Fn14-deficient mice exhibited reduced inflammatory response and delayed muscle fiber regeneration compared with wild-type mice. These results indicate that the TWEAK/Fn14 pathway is a novel regulator of skeletal muscle precursor cells and illustrate an important mechanism by which inflammatory cytokines influence tissue regeneration and repair. Coupled with our recent demonstration that TWEAK potentiates liver progenitor cell proliferation, the expression of Fn14 on all mesenchymal lineage progenitor cells supports a broad involvement of this pathway in other tissue injury and disease settings.


Assuntos
Células-Tronco Mesenquimais/citologia , Músculo Esquelético/fisiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Regeneração , Fatores de Necrose Tumoral/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteínas Cardiotóxicas de Elapídeos/farmacologia , Citocina TWEAK , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Modelos Biológicos , Desenvolvimento Muscular/efeitos dos fármacos , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/genética , Regeneração/efeitos dos fármacos , Receptor de TWEAK , Fatores de Necrose Tumoral/genética
12.
J Cell Sci ; 115(Pt 2): 267-74, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11839778

RESUMO

Angiogenic regulators modulate endothelial cell functions, including proliferation, migration, secretion, and adhesion, through their action on endothelial cells or other cell types. TWEAK, a novel member of the tumor necrosis factor family, appears to be a pro-angiogenic agent on the basis of previous studies demonstrating its ability to induce interleukin-8 production by epithelial tumor lines, stimulate proliferation of human vascular cell types and neovascularization in rat corneas. Here, we further characterized the angiogenic potential of TWEAK, revealing a dual role for TWEAK as an angiogenic regulator. We demonstrate that TWEAK is a potent inducer of endothelial cell survival and cooperates with basic fibroblast growth factor to induce the proliferation and migration of human endothelial cells and morphogenesis of capillary lumens. In contrast, TWEAK antagonizes the morphogenic response of endothelial cells to vascular endothelial growth factor (VEGF) without inhibiting VEGF-induced survival or proliferation. Thus, our observations suggest that TWEAK may differentially regulate microvascular growth, remodeling and/or maintenance in vivo, depending upon the angiogenic context.


Assuntos
Inibidores da Angiogênese/metabolismo , Capilares/metabolismo , Proteínas de Transporte/metabolismo , Endotélio Vascular/metabolismo , Neovascularização Fisiológica/fisiologia , Proteínas Reguladoras de Apoptose , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Capilares/citologia , Capilares/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Citocina TWEAK , Fatores de Crescimento Endotelial/antagonistas & inibidores , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/agonistas , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Recém-Nascido , Linfocinas/antagonistas & inibidores , Linfocinas/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Fatores de Necrose Tumoral , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
13.
J Biol Chem ; 278(7): 5444-54, 2003 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-12466268

RESUMO

Tumor necrosis factor (TNF) ligand and receptor superfamily members play critical roles in diverse developmental and pathological settings. In search for novel TNF superfamily members, we identified a murine chromosomal locus that contains three new TNF receptor-related genes. Sequence alignments suggest that the ligand binding regions of these murine TNF receptor homologues, mTNFRH1, -2 and -3, are most homologous to those of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors. By using a number of in vitro ligand-receptor binding assays, we demonstrate that mTNFRH1 and -2, but not mTNFRH3, bind murine TRAIL, suggesting that they are indeed TRAIL receptors. This notion is further supported by our demonstration that both mTNFRH1:Fc and mTNFRH2:Fc fusion proteins inhibited mTRAIL-induced apoptosis of Jurkat cells. Unlike the only other known murine TRAIL receptor mTRAILR2, however, neither mTNFRH2 nor mTNFRH3 has a cytoplasmic region containing the well characterized death domain motif. Coupled with our observation that overexpression of mTNFRH1 and -2 in 293T cells neither induces apoptosis nor triggers NFkappaB activation, we propose that the mTnfrh1 and mTnfrh2 genes encode the first described murine decoy receptors for TRAIL, and we renamed them mDcTrailr1 and -r2, respectively. Interestingly, the overall sequence structures of mDcTRAILR1 and -R2 are quite distinct from those of the known human decoy TRAIL receptors, suggesting that the presence of TRAIL decoy receptors represents a more recent evolutionary event.


Assuntos
Glicoproteínas de Membrana/genética , Receptores do Fator de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/genética , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular , Mapeamento Cromossômico , Evolução Molecular , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Receptores do Fator de Necrose Tumoral/metabolismo , Análise de Sequência , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa