Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood ; 117(7): 2284-95, 2011 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-21183689

RESUMO

The reticulon (Rtn) family of proteins are localized primarily to the endoplasmic reticulum (ER) of most cells. The Rtn-4 family, (aka Nogo) consists of 3 splice variants of a common gene called Rtn-4A, Rtn-4B, and Rtn-4C. Recently, we identified the Rtn-4B (Nogo-B) protein in endothelial and smooth muscle cells of the vessel wall, and showed that Nogo-B is a regulator of cell migration in vitro and vascular remodeling and angiogenesis in vivo. However, the role of Nogo-B in inflammation is still largely unknown. In the present study, we use 2 models of inflammation to show that endothelial Nogo-B regulates leukocyte transmigration and intercellular adhesion molecule-1 (ICAM-1)-dependent signaling. Mice lacking Nogo-A/B have a marked reduction in neutrophil and monocyte recruitment to sites of inflammation, while Nogo-A/B(-/-) mice engrafted with wild-type (WT) bone marrow still exhibit impaired inflammation compared with WT mice engrafted with Nogo-A/B(-/-) bone marrow, arguing for a critical role of host Nogo in this response. Using human leukocytes and endothelial cells, we show mechanistically that the silencing of Nogo-B with small interfering RNA (siRNA) impairs the transmigration of neutrophils and reduces ICAM-1-stimulated phosphorylation of vascular endothelial-cell cadherin (VE-cadherin). Our results reveal a novel role of endothelial Nogo-B in basic immune functions and provide a key link in the molecular network governing endothelial-cell regulation of diapedesis.


Assuntos
Inflamação/etiologia , Molécula 1 de Adesão Intercelular/fisiologia , Leucócitos/fisiologia , Proteínas da Mielina/fisiologia , Animais , Antígenos CD/fisiologia , Caderinas/fisiologia , Carragenina/toxicidade , Movimento Celular/fisiologia , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Quinase 2 de Adesão Focal/metabolismo , Humanos , Técnicas In Vitro , Inflamação/patologia , Inflamação/fisiopatologia , Leucócitos/patologia , Macrófagos/patologia , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Knockout , Monócitos/patologia , Monócitos/fisiologia , Proteínas da Mielina/antagonistas & inibidores , Proteínas da Mielina/deficiência , Proteínas da Mielina/genética , Neutrófilos/patologia , Neutrófilos/fisiologia , Proteínas Nogo , Fosforilação , RNA Interferente Pequeno/genética , Transdução de Sinais , Quinases da Família src/metabolismo
2.
Am J Pathol ; 177(6): 2765-73, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20971739

RESUMO

Nogo-B is a member of the reticulon family of proteins that has been implicated in diverse forms of vascular injury. Although Nogo-B is expressed in renal tissues, its localization and function in the kidney have not been examined. Here, we report that Nogo-B is expressed specifically in the epithelial cells of the distal nephron segments in the murine kidney. After unilateral ureteral obstruction (UUO) and ischemia/reperfusion, Nogo-B gene and protein levels increased dramatically in the kidney. This increase was driven in part by injury-induced de novo expression in proximal tubules. Examination of Nogo-B immunostaining in human biopsy specimens from patients with acute tubular necrosis showed similar increases in Nogo-B in cortical tubules. Mice genetically deficient in Nogo-A/B were indistinguishable from wild-type (WT) mice based on histological appearance and serum analyses. After UUO, there was a significant delay in recruitment of macrophages to the kidney in the Nogo-A/B-deficient mice. However, measurements of fibrosis, inflammatory gene expression, and histological damage were not significantly different from WT mice. Thus, Nogo-B is highly expressed in murine kidneys in response to experimental injuries and may serve as a marker of diverse forms of renal injury in tissues from mice and humans. Furthermore, Nogo-B may regulate macrophage recruitment after UUO, although it does not greatly affect the degree of tissue injury or fibrosis in this model.


Assuntos
Células Epiteliais/metabolismo , Túbulos Renais/metabolismo , Proteínas da Mielina/genética , Animais , Movimento Celular/genética , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Necrose do Córtex Renal/genética , Necrose do Córtex Renal/metabolismo , Necrose do Córtex Renal/patologia , Medula Renal/metabolismo , Medula Renal/patologia , Necrose Papilar Renal/genética , Necrose Papilar Renal/metabolismo , Necrose Papilar Renal/patologia , Túbulos Renais/patologia , Túbulos Renais/fisiologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas da Mielina/metabolismo , Proteínas Nogo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Obstrução Ureteral/complicações , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
3.
Proc Natl Acad Sci U S A ; 106(41): 17511-6, 2009 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-19805174

RESUMO

Blood vessel formation during ischemia and wound healing requires coordination of the inflammatory response with genes that regulate blood vessel assembly. Here we show that the reticulon family member 4B, aka Nogo-B, is upregulated in response to ischemia and is necessary for blood flow recovery secondary to ischemia and wound healing. Mice lacking Nogo-B exhibit reduced arteriogenesis and angiogenesis that are linked to a decrease in macrophage infiltration and inflammatory gene expression in vivo. Bone marrow-derived macrophages isolated from Nogo knock-out mice have reduced spreading and chemotaxis due to impaired Rac activation. Bone marrow reconstitution experiments show that Nogo in myeloid cells is necessary to promote macrophage homing and functional recovery after limb ischemia. Thus, endogenous Nogo coordinates macrophage-mediated inflammation with arteriogenesis, wound healing, and blood flow control.


Assuntos
Macrófagos/fisiologia , Proteínas da Mielina/farmacologia , Proteínas da Mielina/fisiologia , Animais , Velocidade do Fluxo Sanguíneo/fisiologia , Movimento Celular/fisiologia , Inflamação/prevenção & controle , Isquemia/fisiopatologia , Isquemia/prevenção & controle , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Proteínas da Mielina/deficiência , Proteínas da Mielina/genética , Proteínas Nogo , Fluxo Sanguíneo Regional/fisiologia , Regulação para Cima , Cicatrização/fisiologia
4.
Mol Ther ; 16(11): 1798-804, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18781142

RESUMO

Nogo-B was recently identified as a novel vascular marker; the normally high vascular expression of Nogo-B is rapidly lost following vascular injury. Here we assess the potential therapeutic effects of Ad-Nogo-B delivery to injured vessels in vivo. Nogo-B overexpression following Ad-Ng-B infection of vascular smooth muscle cells (VSMCs) was shown to block proliferation and migration in a dose-dependent manner in vitro. We next assessed the effects of Ad-Ng-B treatment on neointima formation in two in vivo models of acute vascular injury. Adventitial delivery of Ad-Ng-B to wire-injured murine femoral arteries led to a significant decrease in the intimal area [0.014 mm(2) versus 0.030 mm(2) (P = 0.049)] and the intima:media ratio [0.78 versus 1.67 (P = 0.038)] as compared to the effects of Ad-beta-Gal control virus at 21 days after injury. Similarly, lumenal delivery of Ad-Ng-B to porcine saphenous veins prior to carotid artery grafting significantly reduced the intimal area [2.87 mm(2) versus 7.44 mm(2) (P = 0.0007)] and the intima:media ratio [0.32 versus 0.55 (P = 0.0044)] as compared to the effects following the delivery of Ad- beta-Gal, at 28 days after grafting. Intimal VSMC proliferation was significantly reduced in both the murine and porcine disease models. Gene delivery of Nogo-B exerts a positive effect on vascular injury-induced remodeling and reduces neointimal development in two arterial and venous models of vascular injury.


Assuntos
Músculo Liso Vascular/metabolismo , Proteínas da Mielina/biossíntese , Túnica Íntima/metabolismo , Túnica Média/metabolismo , Adenoviridae/genética , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Artérias Carótidas/cirurgia , Proliferação de Células , Células Cultivadas , Quimiotaxia , Constrição Patológica/patologia , Constrição Patológica/prevenção & controle , Modelos Animais de Doenças , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Técnicas de Transferência de Genes , Vetores Genéticos , Oclusão de Enxerto Vascular/patologia , Oclusão de Enxerto Vascular/prevenção & controle , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Proteínas da Mielina/genética , Proteínas Nogo , Veia Safena/metabolismo , Veia Safena/patologia , Suínos , Túnica Íntima/patologia , Túnica Média/patologia
5.
J Biol Chem ; 277(43): 40397-402, 2002 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-12183462

RESUMO

Entry of most primary human immunodeficiency virus, type 1 (HIV-1) isolates into their target cells requires the cellular receptor CD4 and the G protein-coupled chemokine coreceptor CCR5. An acidic, tyrosine-rich, and tyrosine-sulfated domain of the CCR5 amino terminus plays a critical role in the ability of CCR5 to serve as an HIV-1 coreceptor, and tyrosine-sulfated peptides based on this region physically associate with the HIV-1 envelope glycoprotein gp120 and slow HIV-1 entry into CCR5-expressing cells. Here we show that the same tyrosine-sulfated peptides, but not their unsulfated analogs, can restore the HIV-1 coreceptor activity of a CCR5 variant lacking residues 2-17 of its amino terminus. Additionally, these sulfated peptides restored the ability of this CCR5 variant to mobilize calcium in response to the chemokines macrophage inflammatory factors 1alpha and 1beta. These observations show that a tyrosine-sulfated region of the CCR5 amino terminus can function independently to mediate association of chemokines and the HIV-1 envelope glycoprotein with the remaining domains of CCR5.


Assuntos
Peptídeos/metabolismo , Receptores CCR5/metabolismo , Sulfatos/metabolismo , Tirosina/metabolismo , Sequência de Aminoácidos , Cálcio/metabolismo , HIV-1/metabolismo , Dados de Sequência Molecular , Receptores CCR5/química , Receptores CCR5/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA