Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Kidney Int ; 86(4): 726-37, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24850152

RESUMO

The murine cell surface protein Crry (complement receptor 1-related protein/gene y) is a key complement regulator with similar activities to human membrane cofactor protein (MCP) and decay-accelerating factor. MCP has a critical role in preventing complement-mediated tissue injury and its mutation has been implicated in several human kidney diseases. The study of Crry in mice has relevance to understanding MCP activity in human diseases; however, such efforts have been hampered by the embryonic lethality phenotype of Crry gene knockout. Here we used a conditional gene-targeting approach and deleted Crry from the mouse proximal tubular epithelial cells where Crry is prominently expressed. Absence of Crry from proximal tubular epithelial cells resulted in spontaneous C3 deposition on the basolateral surface but no apparent renal disease in unchallenged mice. However, mice deficient in Crry on proximal tubular epithelial cells developed exacerbated renal injury when subjected to renal ischemia-reperfusion, showing increased blood urea nitrogen levels, higher tubular injury scores, more tubular epithelial cell apoptosis, and inflammatory infiltrates. Renal ischemia-reperfusion injury in the Crry conditional knockout mice was prevented by blocking C3 and C5 activation using an anti-properdin or anti-C5 monoclonal antibody (mAb), respectively. Thus, Crry has a critical role in protecting proximal tubular epithelial cells during ischemia-reperfusion challenge. Our results highlight the latent risk for inflammatory kidney injury associated with defects in membrane complement regulators.


Assuntos
Predisposição Genética para Doença , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Animais , Anticorpos Monoclonais/farmacologia , Apoptose , Nitrogênio da Ureia Sanguínea , Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Complemento C5/imunologia , Complemento C5/metabolismo , Células Epiteliais , Técnicas de Inativação de Genes , Túbulos Renais Proximais , Camundongos , Camundongos Knockout , Fagócitos , Properdina/imunologia , Receptores de Complemento 3b
2.
J Am Soc Nephrol ; 24(1): 53-65, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23204401

RESUMO

Factor H (fH) and properdin both modulate complement; however, fH inhibits activation, and properdin promotes activation of the alternative pathway of complement. Mutations in fH associate with several human kidney diseases, but whether inhibiting properdin would be beneficial in these diseases is unknown. Here, we found that either genetic or pharmacological blockade of properdin, which we expected to be therapeutic, converted the mild C3 GN of an fH-mutant mouse to a lethal C3 GN with features of human dense deposit disease. We attributed this phenotypic change to a differential effect of properdin on the dynamics of alternative pathway complement activation in the fluid phase and the cell surface in the fH-mutant mice. Thus, in fH mutation-related C3 glomerulopathy, additional factors that impact the activation of the alternative pathway of complement critically determine the nature and severity of kidney pathology. These results show that therapeutic manipulation of the complement system requires rigorous disease-specific target validation.


Assuntos
Glomerulonefrite Membranoproliferativa/genética , Nefropatias/genética , Properdina/deficiência , Animais , Complemento C3/metabolismo , Fator H do Complemento/deficiência , Fator H do Complemento/genética , Via Alternativa do Complemento , Modelos Animais de Doenças , Glomerulonefrite Membranoproliferativa/metabolismo , Glomerulonefrite Membranoproliferativa/patologia , Doenças da Deficiência Hereditária de Complemento , Humanos , Glomérulos Renais/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Mutação
3.
J Am Soc Nephrol ; 24(1): 43-52, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23184055

RESUMO

Complement factor H (CFH) is a negative regulator of the alternative pathway of complement, and properdin is the sole positive regulator. CFH-deficient mice (CFH(-/-)) develop uncontrolled C3 activation and spontaneous renal disease characterized by accumulation of C3 along the glomerular basement membrane, but the role of properdin in the pathophysiology is unknown. Here, we studied mice deficient in both CFH and properdin (CFH(-/-).P(-/-)). Although CFH(-/-) mice had plasma depleted of both C3 and C5, CFH(-/-).P(-/-) animals exhibited depletion of C3 predominantly, recapitulating the plasma complement profile observed in humans with properdin-independent C3 nephritic factors. Glomerular inflammation, thickening of the capillary wall, and glomerular C3 staining were significantly increased in CFH(-/-).P(-/-) compared with CFH(-/-) mice. We previously reported that exogenous CFH ameliorates C3 staining of the glomerular basement membrane and triggers the appearance of mesangial C3 deposits in CFH(-/-) mice; here, we show that these effects require properdin. In summary, during uncontrolled activation of C3 driven by complete CFH deficiency, properdin influences the intraglomerular localization of C3, suggesting that therapeutic inhibition of properdin would be detrimental in this setting.


Assuntos
Glomerulonefrite/etiologia , Nefropatias/complicações , Properdina/deficiência , Animais , Complemento C3/metabolismo , Fator H do Complemento/deficiência , Glomerulonefrite/metabolismo , Doenças da Deficiência Hereditária de Complemento , Humanos , Glomérulos Renais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
4.
Nephrology (Carlton) ; 15(7): 663-75, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21040161

RESUMO

Complement is a part of the body's innate immune system that helps defend the host from microbial infection. It is tightly controlled by a number of cell surface and fluid-phase proteins so that under normal circumstances injury to autologous tissues is avoided. In many pathological settings, such as when the complement regulatory mechanisms are dysfunctional or overwhelmed, complement attack of autologous tissues can occur with severe, sometimes life-threatening consequences. The kidney appears to be particularly vulnerable to complement-mediated inflammatory injury and many kidney pathologies have been linked to abnormal complement activation. Clinical and experimental studies have shown that complement attack can be a primary cause in rare, genetically predisposed kidney diseases or a significant contributor to kidney injury caused by other etiological factors. Here we provide a brief review of recent advances on the activation and regulation of the complement system in kidney disease, with a particular emphasis on the relevance of complement regulatory proteins.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Nefropatias/imunologia , Rim/imunologia , Animais , Humanos , Imunidade Inata , Nefropatias/terapia , Transdução de Sinais
5.
Mol Immunol ; 56(3): 191-8, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23816404

RESUMO

The plasma protein properdin is the only known positive regulator of complement activation. Although regarded as an initiator of the alternative pathway of complement activation at the time of its discovery more than a half century ago, the role and mechanism of action of properdin in the complement cascade has undergone significant conceptual evolution since then. Despite the long history of research on properdin, however, new insight and unexpected findings on the role of properdin in complement activation, pathogen infection and host tissue injury are still being revealed by ongoing investigations. In this article, we provide a brief review on recent studies that shed new light on properdin biology, focusing on the following three topics: (1) its role as a pattern recognition molecule to direct and trigger complement activation, (2) its context-dependent requirement in complement activation on foreign and host cell surfaces, and (3) its involvement in alternative pathway complement-mediated immune disorders and considerations of properdin as a potential therapeutic target in human diseases.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Properdina/metabolismo , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Autoimunidade/genética , Autoimunidade/imunologia , Via Alternativa do Complemento/imunologia , Humanos , Properdina/genética
6.
J Lipid Res ; 50(6): 1047-56, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19151335

RESUMO

Thromboxane A(2) (TxA(2)), the principle product of platelet COX-1-dependent arachidonic acid metabolism, directs multiple pro-atherogenic processes via its receptor, TP. Oxidative challenge offsets TP degradation, a key component in limiting TxA(2)'s actions. Following TP activation, we observed cellular reactive oxygen species (ROS) generation coincident with increased TP expression. We examined the link between TP-evoked ROS and TP regulation. TP expression was augmented in TPalpha-transfected cells treated with a TxA(2) analog [1S-1alpha,2beta(5Z),3alpha(1E,3R*),4alpha]]-7-[3-(3-hydroxy-4-(4'-iodophenoxy)-1-butenyl)-7-oxabicyclo-[2.2.1]heptan-2-yl]-5-heptenoic acid (IBOP). This was reduced with a cellular antioxidant, N-acetyl cysteine, or two distinct NADPH oxidase inhibitors, diphenyleneiodonium and apocynin. Homologous upregulation of the native TP was also reduced in apocynin-treated aortic smooth muscle cells (ASMCs) and was absent in ASMCs lacking an NADPH oxidase subunit (p47(-/-)). TP transcription was not increased in IBOP-treated cells, indicating a posttranscriptional mechanism. IBOP induced translocation of TPalpha to the Golgi and reduced degradation of the immature form of the receptor. These data are consistent with a ROS-dependent mechanism whereby TP activation enhanced TP stability early in posttranscriptional biogenesis. Given the significant role played by TP and ROS in perturbed cardiovascular function, the convergence of TP on ROS-generating pathways for regulation of TxA(2)-dependent events may be critical for cardiovascular disease.


Assuntos
Espécies Reativas de Oxigênio/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Animais , Transporte Biológico Ativo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular , Células Cultivadas , Retículo Endoplasmático/metabolismo , Ácidos Graxos Insaturados/farmacologia , Complexo de Golgi/metabolismo , Humanos , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , NADPH Oxidases/deficiência , NADPH Oxidases/genética , Processamento Pós-Transcricional do RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tromboxano A2/metabolismo , Transfecção , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA