Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 8(1): 2078, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29233958

RESUMO

Regeneration of skeletal muscle following injury is accompanied by transient inflammation. Here we show that complement is activated in skeletal muscle injury and plays a key role during regeneration. Genetic ablation of complement C3 or its inactivation with Cobra Venom Factor (CVF) result in impaired muscle regeneration following cardiotoxin-induced injury in mice. The effect of complement in muscle regeneration is mediated by the alternative pathway and C3a receptor (C3aR) signaling, as deletion of Cfb, a key alternative pathway component, or C3aR leads to impaired regeneration and reduced monocyte/macrophage infiltration. Monocytes from C3aR-deficient mice express a reduced level of adhesion molecules, cytokines and genes associated with antigen processing and presentation. Exogenous administration of recombinant CCL5 to C3aR-deficient mice rescues the defects in inflammatory cell recruitment and regeneration. These findings reveal an important role of complement C3a in skeletal muscle regeneration, and suggest that manipulating complement system may produce therapeutic benefit in muscle injury and regeneration.


Assuntos
Complemento C3a/fisiologia , Inflamação/imunologia , Monócitos/fisiologia , Músculo Esquelético/fisiologia , Receptores de Complemento/fisiologia , Regeneração/imunologia , Animais , Transplante de Medula Óssea , Cardiotoxinas/toxicidade , Movimento Celular/fisiologia , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimera/fisiologia , Complemento C3a/antagonistas & inibidores , Via Alternativa do Complemento/fisiologia , Modelos Animais de Doenças , Venenos Elapídicos/farmacologia , Humanos , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Receptores de Complemento/deficiência , Regeneração/efeitos dos fármacos , Transdução de Sinais/fisiologia , Cicatrização/fisiologia
2.
Nat Commun ; 8(1): 351, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28839129

RESUMO

Complement C3a is an important protein in innate and adaptive immunity, but its specific roles in vivo remain uncertain because C3a degrades rapidly to form the C3a-desArg protein, which does not bind to the C3a receptor and is indistinguishable from C3a using antibodies. Here we develop the most potent, stable and highly selective small molecule modulators of C3a receptor, using a heterocyclic hinge to switch between agonist and antagonist ligand conformations. This enables characterization of C3 areceptor-selective pro- vs. anti-inflammatory actions in human mast cells and macrophages, and in rats. A C3a receptor-selective agonist induces acute rat paw inflammation by first degranulating mast cells before activating macrophages and neutrophils. An orally administered C3a receptor-selective antagonist inhibits mast cell degranulation, thereby blocking recruitment and activation of macrophages and neutrophils, expression of inflammatory mediators and inflammation in a rat paw edema model. These novel tools reveal the mechanism of C3a-induced inflammation and provide new insights to complement-based medicines.Complement C3a is an important protein in innate and adaptive immunity, but its roles in vivo are unclear. Here the authors develop novel chemical agonists and antagonists for the C3a receptor, and show that they modulate mast cell degranulation and inflammation in a rat paw edema model.


Assuntos
Complemento C3a/fisiologia , Imunidade Inata/genética , Receptores de Complemento/química , Animais , Antiasmáticos/farmacologia , Degranulação Celular/efeitos dos fármacos , Células Cultivadas , Complemento C3a/genética , Complemento C3a/metabolismo , Cromolina Sódica/farmacologia , Humanos , Ligantes , Macrófagos/imunologia , Masculino , Mastócitos/imunologia , Neutrófilos/imunologia , Conformação Proteica , Ratos , Ratos Wistar , Receptores de Complemento/agonistas , Receptores de Complemento/antagonistas & inibidores
4.
Dev Cell ; 21(6): 1026-37, 2011 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-22118769

RESUMO

Collective cell migration is a mode of movement crucial for morphogenesis and cancer metastasis. However, little is known about how migratory cells coordinate collectively. Here we show that mutual cell-cell attraction (named here coattraction) is required to maintain cohesive clusters of migrating mesenchymal cells. Coattraction can counterbalance the natural tendency of cells to disperse via mechanisms such as contact inhibition and epithelial-to-mesenchymal transition. Neural crest cells are coattracted via the complement fragment C3a and its receptor C3aR, revealing an unexpected role of complement proteins in early vertebrate development. Loss of coattraction disrupts collective and coordinated movements of these cells. We propose that coattraction and contact inhibition act in concert to allow cell collectives to self-organize and respond efficiently to external signals, such as chemoattractants and repellents.


Assuntos
Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Complemento C3a/fisiologia , Animais , Adesão Celular/fisiologia , Fatores Quimiotáticos/genética , Fatores Quimiotáticos/fisiologia , Complemento C3a/genética , Transição Epitelial-Mesenquimal/fisiologia , Modelos Neurológicos , Dados de Sequência Molecular , Células-Tronco Multipotentes/fisiologia , Crista Neural/citologia , Crista Neural/embriologia , Células-Tronco Neurais/fisiologia , Receptores de Complemento/genética , Receptores de Complemento/fisiologia , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiologia , Xenopus laevis/embriologia , Xenopus laevis/genética , Xenopus laevis/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia
5.
J Cell Biochem ; 112(9): 2594-605, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21598302

RESUMO

There is a tight interaction of the bone and the immune system. However, little is known about the relevance of the complement system, an important part of innate immunity and a crucial trigger for inflammation. The aim of this study was, therefore, to investigate the presence and function of complement in bone cells including osteoblasts, mesenchymal stem cells (MSC), and osteoclasts. qRT-PCR and immunostaining revealed that the central complement receptors C3aR and C5aR, complement C3 and C5, and membrane-bound regulatory proteins CD46, CD55, and CD59 were expressed in human MSC, osteoblasts, and osteoclasts. Furthermore, osteoblasts and particularly osteoclasts were able to activate complement by cleaving C5 to its active form C5a as measured by ELISA. Both C3a and C5a alone were unable to trigger the release of inflammatory cytokines interleukin (IL)-6 and IL-8 from osteoblasts. However, co-stimulation with the pro-inflammatory cytokine IL-1ß significantly induced IL-6 and IL-8 expression as well as the expression of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG) indicating that complement may modulate the inflammatory response of osteoblastic cells in a pro-inflammatory environment as well as osteoblast-osteoclast interaction. While C3a and C5a did not affect osteogenic differentiation, osteoclastogenesis was significantly induced even in the absence of RANKL and macrophage-colony stimulating factor (M-CSF) suggesting that complement could directly regulate osteoclast formation. It can therefore be proposed that complement may enhance the inflammatory response of osteoblasts and increase osteoclast formation, particularly in a pro-inflammatory environment, for example, during bone healing or in inflammatory bone disorders.


Assuntos
Complemento C3a/farmacologia , Complemento C5a/farmacologia , Interleucina-1beta/farmacologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Adulto , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciação Celular , Células Cultivadas , Complemento C3a/metabolismo , Complemento C3a/fisiologia , Complemento C5a/metabolismo , Complemento C5a/fisiologia , Expressão Gênica , Humanos , Inflamação , Interleucina-1beta/fisiologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Microscopia de Fluorescência , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Osteoprotegerina/metabolismo , Proteólise , Ligante RANK/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Adulto Jovem
6.
Leukemia ; 23(8): 1455-61, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19357704

RESUMO

We reported that complement (C) becomes activated and cleaved in bone marrow during preconditioning for hematopoietic transplantation and the third C component (C3) cleavage fragments, C3a and (desArg)C3a, increase responsiveness of hematopoietic stem/progenitor cells (HSPCs) to stromal-derived factor-1 (SDF-1). We also showed that this homing-promoting effect is not C3a receptor (C3aR) dependent. Herein, we report our new observation that transplantation of C3aR(-/-) HSPCs into lethally irradiated recipients results in: (1) approximately 5-7 day delay in recovery of platelets and leukocytes; (2) decrease in formation of day 12 colony-forming units-spleen; and (3) decrease in the number of donor-derived CFU-granulocyte-macrophage progenitors detectable in the bone marrow cavities at day 16 after transplantation. In agreement with the murine data, blockage of C3aR on human umbilical cord blood CD34(+) cells by C3aR antagonist SB290157 impairs their engraftment in non-obese diabetic/severe combined immunodeficient mice. However, HSPCs from C3aR(-/-) mice stimulated by C3a still better responded to SDF-1 gradient, after exposure to C3a, they secrete less matrix metalloprotease-9 and show impaired adhesion to stroma cells. We conclude that C3a, in addition to enhancing responsiveness of HSPCs to SDF-1 gradient in a C3aR independent manner, may also directly modulate HSPC homing by augmenting C3aR-mediated secretion of matrix metalloprotease-9 and cell adhesion.


Assuntos
Medula Óssea/patologia , Movimento Celular/fisiologia , Complemento C3a/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Receptores de Complemento/fisiologia , Animais , Adesão Celular/fisiologia , Ensaio de Unidades Formadoras de Colônias , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/enzimologia , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/patologia , Síndromes de Imunodeficiência/cirurgia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Quimera por Radiação , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Transplante Heterólogo
7.
Int Immunopharmacol ; 9(6): 800-6, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19285573

RESUMO

In the rat, C5a infusion mediates well-defined effects including hypotension and neutropenia. Conversely, the comparative effect of C3a in the rat is not yet defined. In the current study, we have investigated C3a receptor (C3aR) activation in the rat, using recombinant human C3a, the C3aR agonist WWGKKYRASKLGLAR, which is a C-terminal analogue of C3a, and a nonpeptide C3aR antagonist SB-290157, as pharmacological tools. In vitro, C3a and WWGKKYRASKLGLAR selectively bound to C3aRs and induced degranulation of C3aR-transfected RBL-2H3 cells. C3a or WWGKKYRASKLGLAR-induced degranulation was dose-dependently antagonized in a surmountable fashion by the nonpeptide C3aR antagonist. Intravenous infusion of C3a and WWGKKYRASKLGLAR to rats induced a rapid, transient and concentration-dependent hypertensive response, which was mediated by C3aR-induced prostanoid release. C3a and WWGKKYRASKLGLAR caused a small drop in circulating neutrophils, but a rise in circulating neutrophils was evident after 90-120 min. In contrast to C3a, C5a infusion resulted in hypotension, and rapid and transient neutropenia. These results demonstrate that C3a and C5a mediate distinct effects on blood pressure and circulating polymorphonuclear leukocytes in the rat.


Assuntos
Pressão Sanguínea/fisiologia , Complemento C3a/fisiologia , Complemento C5a/fisiologia , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Compostos Benzidrílicos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Degranulação Celular , Linhagem Celular , Complemento C3a/agonistas , Complemento C3a/antagonistas & inibidores , Complemento C5a/agonistas , Complemento C5a/antagonistas & inibidores , Feminino , Humanos , Neutropenia/imunologia , Neutropenia/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Peptídeos/química , Peptídeos/farmacologia , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Proteínas Recombinantes/farmacologia
8.
J Immunol ; 182(6): 3827-36, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265162

RESUMO

Mesenchymal stem cells (MSCs) have a great potential for tissue repair, especially if they can be delivered efficiently to sites of tissue injury. Since complement activation occurs whenever there is tissue damage, the effects of the complement activation products C3a and C5a on MSCs were examined. Both C3a and C5a were chemoattractants for human bone marrow-derived MSCs, which expressed both the C3a receptor (C3aR) and the C5a receptor (C5aR; CD88) on the cell surface. Specific C3aR and C5aR inhibitors blocked the chemotactic response, as did pertussis toxin, indicating that the response was mediated by the known anaphylatoxin receptors in a G(i) activation-dependent fashion. While C5a causes strong and prolonged activation of various signaling pathways in many different cell types, the response observed with C3a is generally transient and weak. However, we show herein that in MSCs both C3a and C5a caused prolonged and robust ERK1/2 and Akt phosphorylation. Phospho-ERK1/2 was translocated to the nucleus in both C3a and C5a-stimulated MSCs, which was associated with subsequent phosphorylation of the transcription factor Elk, which could not be detected in other cell types stimulated with C3a. More surprisingly, the C3aR itself was translocated to the nucleus in C3a-stimulated MSCs, especially at low cell densities. Since nuclear activation/translocation of G protein-coupled receptors has been shown to induce long-term effects, this novel observation implies that C3a exerts far-reaching consequences on MSC biology. These results suggest that the anaphylatoxins C3a and C5a present in injured tissues contribute to the recruitment of MSCs and regulation of their behavior.


Assuntos
Fatores Quimiotáticos/fisiologia , Complemento C3a/fisiologia , Complemento C5a/fisiologia , Células-Tronco Mesenquimais/enzimologia , Células-Tronco Mesenquimais/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Linhagem Celular , Células Cultivadas , Quimiotaxia de Leucócito/imunologia , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor da Anafilatoxina C5a , Receptores de Complemento/biossíntese , Receptores de Complemento/genética , Fatores de Tempo
9.
Cardiovasc Res ; 76(2): 340-50, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17673191

RESUMO

OBJECTIVE: Mounting evidence suggests that activation of complement, an important constituent of innate immunity, contributes to atherosclerosis. Here we investigated the expression of complement components (CCs) in the setting of experimental and clinical hypercholesterolemia, a major risk factor for atherosclerosis, their effects on vascular smooth muscle cell (VSMC) and macrophage proliferation, and the underlying molecular mechanisms. METHODS: For this study we analyzed the mRNA and protein expression of several CCs in plasma and aorta of hypercholesterolemic atherosclerosis-prone apolipoprotein E-null mice (apoE-KO) and in plasma of normocholesterolemic subjects and familial hypercholesterolemia (FH) patients. We also carried out in vitro molecular studies to assess the role of CCs on the control of macrophage and VSMC proliferation. RESULTS: Fat-fed apoE-KO mice experiencing severe hypercholesterolemia (approximately 400 mg/dL), but not fat-fed wild-type controls with plasma cholesterol level<110 mg/dL, displayed in aortic tissue upregulation of several CC mRNAs, including C3, C4, C1s, and C1q. In apoE-KO mice, induction of C3 mRNA was already apparent two days after fat feeding when hypercholesterolemia was manifested yet atherosclerotic lesions were absent or incipient. Rapid C3 and C4 protein upregulation was also observed in the plasma of fat-fed apoE-KO mice, and FH patients exhibited higher plasmatic C3a, C4 gamma chain, C1s and C3c alpha chain protein levels than normocholesterolemic subjects. In vitro, C3 and C3a, but not C3a-desArg, C4 and C1q, promoted macrophage and VSMC proliferation through Gi protein-dependent activation of extracellular signal-regulated kinase 1/2 (ERK1/2). We also found that C3-enriched FH plasma evoked a stronger mitogenic response in macrophages than normocholesterolemic plasma, and treatment with anti-C3 antibodies eliminated this difference. CONCLUSIONS: Both experimental and clinical hypercholesterolemia coincides with a concerted activation of several CCs. However, only C3 and C3a elicited a mitogenic response in cultured VSMCs and macrophages through Gi protein-dependent ERK1/2 activation. Thus, excess of C3/C3a in hypercholesterolemic apoE-KO mice and FH patients may contribute to atheroma growth by promoting neointimal cell proliferation.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Hipercolesterolemia/etiologia , Macrófagos/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Sequência de Aminoácidos , Animais , Apolipoproteínas E/fisiologia , Proliferação de Células , Complemento C3/fisiologia , Complemento C3a/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular
10.
Cancer Res ; 67(15): 7421-30, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671212

RESUMO

Administration of a combination of yeast-derived beta-glucan with antitumor monoclonal antibodies (mAb) has significant therapeutic efficacy in a variety of syngeneic murine tumor models. We have now tested this strategy using human carcinomas implanted in immunocompromised severe combined immunodeficient mice. Combined immunotherapy was therapeutically effective in vivo against NCI-H23 human non-small-cell lung carcinomas, but this modality was surprisingly ineffective against SKOV-3 human ovarian carcinomas. Whereas NCI-H23 tumors responded to this combination therapy with increased intratumoral neutrophil infiltration and C5a production, these responses were lacking in treated SKOV-3 tumors. Further results suggested that SKOV-3 tumors were protected by up-regulation of the membrane complement regulatory protein CD55 (decay-accelerating factor). Blockade of CD55 in vitro led to enhanced deposition of C activation product C3b and increased cytotoxicity mediated by beta-glucan-primed neutrophils. In vivo, administration of anti-CD55 mAb along with beta-glucan and anti-Her-2/neu mAb caused tumor regression and greatly improved long-term survival in animals bearing the previously resistant SKOV-3 tumors. This was accompanied by increased intratumoral neutrophil accumulation and C5a production. We conclude that CD55 suppresses tumor killing by antitumor mAb plus beta-glucan therapy (and, perhaps, in other circumstances). These results suggest a critical role for CD55 to regulate iC3b and C5a release and in turn to influence the recruitment of beta-glucan-primed neutrophils eliciting killing activity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos CD55/metabolismo , Carcinoma Pulmonar de Células não Pequenas/terapia , Complemento C5a/fisiologia , Infiltração de Neutrófilos/imunologia , Neoplasias Ovarianas/terapia , beta-Glucanas/uso terapêutico , Animais , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Complemento C3a/fisiologia , Quimioterapia Combinada , Feminino , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Infiltração de Neutrófilos/genética , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Saccharomyces cerevisiae/metabolismo , Células Tumorais Cultivadas
11.
Am J Respir Crit Care Med ; 175(12): 1250-8, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17400733

RESUMO

RATIONALE: The factors that control the secretion of epithelial mucins are essential to understanding obstructive airway diseases such as asthma. Although the complement anaphylatoxin C3a and its receptor have been shown to promote many features of allergic lung inflammation, the contribution to mucin expression has not been elucidated. OBJECTIVES: To determine if the C3a receptor with its ligand regulates airway epithelial mucin production. METHODS: Mice deficient in the C3a receptor were examined in a model of allergic airway disease for the presence of goblet cells and the gel-forming secreted mucin Muc5ac. MEASUREMENTS AND MAIN RESULTS: Lungs from antigen-challenged C3a receptor-deficient mice revealed a dramatic decrease in goblet cells and Muc5ac compared with challenged wild-type control animals. These differences were dependent on C3a binding to its receptor since intranasal challenge with C3a induced the formation of goblet cells only in wild-type but not C3a receptor-deficient mice. Increased numbers of goblet cells were also found in C3a-stimulated RAG-1-deficient mice demonstrating a mechanism independent of T lymphocytes and Th2 cytokines, mediators which have been shown to regulate mucin expression. A direct physiological role for C3a in these models was further demonstrated in cultures of airway epithelial Clara cells, which not only express the C3a receptor but also produce Muc5ac in response to C3a. CONCLUSIONS: These studies identify a novel C3a receptor-dependent mechanism in the development of airway epithelial goblet cells and regulation of Muc5ac production and implicate C3a as a mediator of airway obstruction in asthma.


Assuntos
Asma/fisiopatologia , Complemento C3a/fisiologia , Mucinas/metabolismo , Células Th2/fisiologia , Animais , Asma/metabolismo , Células Cultivadas , Células Epiteliais/metabolismo , Interleucina-13/farmacologia , Pulmão/citologia , Camundongos , Camundongos Endogâmicos C57BL , Mucina-5AC
12.
Leukemia ; 21(5): 973-82, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17330096

RESUMO

We hypothesized that the third complement component (C3) cleavage fragments (C3a and (des-Arg)C3a) are involved in stress/inflammation-related thrombocytosis, and investigated their potential role in reactive thrombocytosis induced by bleeding. We found that platelet counts are lower in C3-deficient mice in response to excessive bleeding as compared to normal littermates and that C3a and (des-Arg)C3a enhance stromal-derived factor-1 (SDF-1)-dependent megakaryocyte (Megs) migration, adhesion and platelet shedding. At the molecular level, C3a stimulates in Megs MAPKp42/44 phosphorylation, and enhances incorporation of CXCR4 into membrane lipid rafts increasing the responsiveness of Megs to SDF-1. We found that perturbation of lipid raft formation by statins decreases SDF-1/C3a-dependent platelet production in vitro and in an in vivo model statins ameliorated post-bleeding thrombocytosis. Thus, inhibition of lipid raft formation could find potential clinical application as a means of ameliorating some forms of thrombocytosis.


Assuntos
Quimiocinas CXC/fisiologia , Complemento C3a/fisiologia , Hemorragia/fisiopatologia , Trombocitose/fisiopatologia , Trombopoese , Quimiocina CXCL12 , Humanos , Microdomínios da Membrana/fisiologia , Contagem de Plaquetas , Receptores CXCR4/fisiologia , Receptores de Complemento/análise , Transdução de Sinais
13.
J Immunol ; 178(3): 1819-28, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17237432

RESUMO

The complement system is one of the major ways by which the body detects injury to self cells, and the alternative pathway of complement is rapidly activated within the tubulointerstitium after renal ischemia/reperfusion (I/R). In the current study, we investigate the hypothesis that recognition of tubular injury by the complement system is a major mechanism by which the systemic inflammatory response is initiated. Gene array analysis of mouse kidney following I/R initially identified MIP-2 (CXCL2) and keratinocyte-derived chemokine (KC or CXCL1) as factors that are produced in a complement-dependent fashion. Using in situ hybridization, we next demonstrated that these factors are expressed in tubular epithelial cells of postischemic kidneys. Mouse proximal tubular epithelial cells (PTECs) in culture were then exposed to an intact alternative pathway and were found to rapidly produce both chemokines. Selective antagonism of the C3a receptor significantly attenuated production of MIP-2 and KC by PTECs, whereas C5a receptor antagonism and prevention of membrane attack complex (MAC) formation did not have a significant effect. Treatment of PTECs with an NF-kappaB inhibitor also prevented full expression of these factors in response to an intact alternative pathway. In summary, alternative pathway activation after renal I/R induces production of MIP-2 and KC by PTECs. This innate immune system thereby recognizes hypoxic injury and triggers a systemic inflammatory response through the generation of C3a and subsequent activation of the NF-kappaB system.


Assuntos
Quimiocinas CXC/biossíntese , Quimiocinas/genética , Complemento C3a/genética , Complemento C3a/fisiologia , Células Epiteliais/imunologia , Túbulos Renais/patologia , NF-kappa B/metabolismo , Traumatismo por Reperfusão/imunologia , Animais , Quimiocina CXCL1 , Quimiocina CXCL2 , Quimiocinas CXC/genética , Ativação do Complemento , Células Epiteliais/patologia , Perfilação da Expressão Gênica , Inflamação , Nefropatias , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão/patologia
14.
Microbiol Immunol ; 49(11): 981-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16301808

RESUMO

The contribution of complement activation to allergic asthma remains controversial. In order to elucidate the role played by the complement split products, anaphylatoxins C3a and C5a, we evaluated their effects on production of cysteinyl-leukotrienes (cysLTs) by human lung fragments following an anaphylactic reaction. The lung tissues obtained from two patients with lung cancer showed C5aR-, C5L2R-, and C3aR-mRNA expression. When the chopped lung fragments passively sensitized with human IgE were incubated with anti-human IgE antibody, a significant amount of cysLTs was generated in comparison with the control (without anti-IgE antibody). The co-addition of human C5a at doses of 0.1 to 10 ng/ml to the anti-IgE antibody potentiated cysLT production. The response was bell-shaped in distribution, significant, and peaked at a C5a concentration of 1 ng/ml. The co-addition of human C3a up to 1,000 ng/ml seemed to increase cysLT production, but not to any significant extent. A novel C5a receptor complementary peptide, acetylated peptide A, dose-dependently inhibited cysLT production by the human lung fragments following the anaphylactic reaction in the presence of 1 ng/ml C5a. However, this peptide did not inhibit cysLT production in the presence of 100 ng/ml C3a. It is suggested that the anaphylatoxin C5a potentiates cysLT production in human lung tissues and contributes to allergic inflammation in disorders such as asthma, thus acetylated peptide A may be useful for suppressing allergic inflammation in the lungs.


Assuntos
Anafilatoxinas/fisiologia , Complemento C3a/fisiologia , Complemento C5a/fisiologia , Cisteína/metabolismo , Imunoglobulina E/farmacologia , Leucotrienos/metabolismo , Pneumonia/imunologia , Anafilatoxinas/imunologia , Anticorpos Anti-Idiotípicos/farmacologia , Asma/imunologia , Asma/metabolismo , Complemento C3a/farmacologia , Complemento C5a/farmacologia , Humanos , Técnicas In Vitro , Peptídeos/farmacologia
15.
J Immunol ; 175(6): 3698-706, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16148115

RESUMO

Complement C3a promotes CXCL12-induced migration and engraftment of human and murine hemopoietic progenitor cells, suggesting a cross-influence between anaphylatoxin and chemokine axes. Here we have explored the underlying mechanism(s) of complement anaphylatoxin and chemokine cooperation. In addition to C3a, C3a-desArg and C4a but not C5a, are potent enhancers of CXCL12-induced chemotaxis of human and murine bone marrow (BM) stem/progenitor cells and B lineage cells. C3a enhancement of chemotaxis is chemokine specific because it is also observed for chemotaxis to CCL19 but not to CXCL13. The potentiating effect of C3a on CXCL12 is independent of the classical C3a receptor (C3aR). First, human BM CD34(+) and B lineage cells do not express C3aR by flow cytometry. Second, the competitive C3aR inhibitor SB290157 does not affect C3a-mediated enhancement of CXCL12-induced chemotaxis. Third, enhancement of chemotaxis of hemopoietic cells is also mediated by C3a-desArg, which does not bind to C3aR. Finally, C3a enhances CXCL12-induced chemotaxis of BM cells from C3aR knockout mice similar to BM cells from wild-type mice. Subsequent studies revealed that C3a increased the binding affinity of CXCL12 to human CXCR4(+)/C3aR(-), REH pro-B cells, which is compatible with a direct interaction between C3a and CXCL12. BM stromal cells were able to generate C3a, C3a-desArg, C4a, as well as CXCL12, suggesting that this pathway could function in vivo. Taken together, we demonstrate a C3a-CXCL12 interaction independent of the C3aR, which may provide a mechanism to modulate the function of CXCL12 in the BM microenvironment.


Assuntos
Quimiocinas CXC/fisiologia , Quimiotaxia , Complemento C3a/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Proteínas de Membrana/fisiologia , Receptores de Complemento/fisiologia , Anafilatoxinas/metabolismo , Anafilatoxinas/fisiologia , Animais , Linfócitos B/metabolismo , Medula Óssea , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas/metabolismo , Quimiocinas/fisiologia , Quimiocinas CXC/metabolismo , Complemento C3a/metabolismo , Humanos , Camundongos , Camundongos Knockout , Células Estromais/metabolismo
16.
J Immunol ; 175(3): 1947-55, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16034139

RESUMO

Signaling of the C3a anaphylatoxin through its G protein-coupled receptor, C3aR, is relevant in a variety of inflammatory diseases, but its role in lupus nephritis is undefined. In this study, we show that expression of C3aR was significantly increased in prediseased and diseased kidneys of MRL/lpr lupus mice compared with MRL/+ controls. To investigate the role of C3aR in experimental lupus, a small molecule antagonist of C3aR (C3aRa) was administered continuously to MRL/lpr mice from 13 to 19 wk of age. All 13 C3aRa-treated mice survived during the 6-wk treatment compared with 9 of 14 (64.3%) control animals given vehicle (p = 0.019). Relative to controls, C3aRa-treated animals were protected from renal disease as measured by albuminuria (p = 0.040) and blood urea nitrogen (p = 0.021). In addition, there were fewer neutrophils, monocytes, and apoptotic cells in the kidneys of C3aRa-treated mice. C3aRa treatment also led to reduced renal IL-1beta and RANTES mRNA and phosphorylated phosphatase and tensin homologue deleted on chromosome 10 protein, whereas the mass of phosphorylated protein kinase B/Akt was increased by C3aRa. Thus, C3aR antagonism significantly reduces renal disease in MRL/lpr mice, which further translates into prolonged survival. These data illustrate that C3aR is relevant in experimental lupus nephritis and may be a target for therapeutic intervention in the human disease.


Assuntos
Complemento C3a/fisiologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas de Membrana/fisiologia , Receptores de Complemento/fisiologia , Transdução de Sinais/imunologia , Regulação para Cima/imunologia , Animais , Anticorpos Antinucleares/biossíntese , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Arginina/administração & dosagem , Arginina/análogos & derivados , Compostos Benzidrílicos/administração & dosagem , Complemento C3a/metabolismo , Proteínas Inativadoras do Complemento/administração & dosagem , Técnica Indireta de Fluorescência para Anticorpo , Mediadores da Inflamação/metabolismo , Rim/efeitos dos fármacos , Rim/imunologia , Rim/metabolismo , Rim/patologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/mortalidade , Lúpus Eritematoso Sistêmico/patologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos MRL lpr , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação/efeitos dos fármacos , RNA Mensageiro/biossíntese , Receptores de Complemento/antagonistas & inibidores , Receptores de Complemento/biossíntese , Receptores de Complemento/genética , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima/genética
18.
J Immunol ; 174(11): 7050-6, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15905548

RESUMO

Intravenous and orally administered beta-glucans promote tumor regression and survival by priming granulocyte and macrophage C receptor 3 (CR3, iC3bR and CD11b/CD18) to trigger the cytotoxicity of tumor cells opsonized with iC3b via anti-tumor Abs. Despite evidence for priming of macrophage CR3 by oral beta-glucan in vivo, the current study in C57BL/6 and BALB/c mice showed that granulocytes were the essential killer cells in mAb- and oral beta-glucan-mediated tumor regression, because responses were absent in granulocyte-depleted mice. Among granulocytes, neutrophils were the major effector cells, because tumor regression did not occur when C5a-dependent chemotaxis was blocked with a C5aR antagonist, whereas tumor regression was normal in C3aR(-/-) mice. Neutrophil recruitment by C5a in vivo required amplification via leukotriene B(4), because both C5a-mediated leukocyte recruitment into the peritoneal cavity and tumor regression were suppressed in leukotriene B(4)R-deficient (BLT-1(-/-)) mice.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Quimiotaxia de Leucócito/imunologia , Complemento C5a/fisiologia , Leucotrieno B4/fisiologia , Linfoma/imunologia , Linfoma/terapia , Infiltração de Neutrófilos/imunologia , beta-Glucanas/uso terapêutico , Administração Oral , Animais , Linhagem Celular Tumoral , Quimiotaxia de Leucócito/genética , Complemento C3a/fisiologia , Granulócitos/citologia , Granulócitos/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Linfoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/genética , Receptores do Leucotrieno B4/deficiência , Receptores do Leucotrieno B4/genética , Receptores do Leucotrieno B4/fisiologia , beta-Glucanas/administração & dosagem
19.
FASEB J ; 19(7): 798-800, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15758041

RESUMO

Growing evidence suggests that anaphylatoxins, C3a and C5a, play important roles in innate immunity and may also participate in the pathogenesis of asthma. Previous studies with animal models and immunohistochemistry analysis of lung tissue indicated that anaphylatoxins may regulate airway hyperresponsiveness (AHR) in asthma via the activation of their cell surface G protein-coupled receptors (C3aR and C5aR) in airway smooth muscle (ASM) cells. Using RT-PCR, flow cytometry, and confocal microscopy, we made the surprising observation that while C3aR and C5aR were expressed in human mast cells, they were not present in cultured primary human or murine ASM cells. Furthermore, we could not detect C3aR in smooth muscle-positive cells of human trachea or bronchus. Interestingly, incubation of human mast cells with ASM cells, but not its culture supernatant, caused a significant enhancement of C3a-induced mast cell degranulation. Although stem cell factor (SCF) and its receptor c-kit are constitutively expressed on ASM cells and mast cells, respectively, neutralizing antibodies to SCF and c-kit failed to inhibit ASM cell-mediated enhancement of mast cell degranulation. However, dexamethasone-treated ASM cells were normal for cell surface SCF expression but were significantly less effective in enhancing C3a-induced mast cell degranulation when compared with untreated cells. These findings suggest that cell-cell interaction between ASM cells and mast cells, via a SCF-c-kit-independent but dexamethasone-sensitive mechanism, enhances C3a-induced mast cell degranulation, which likely regulates ASM function, thus contributing to the pathogenesis of asthma.


Assuntos
Degranulação Celular/fisiologia , Complemento C3a/fisiologia , Mastócitos/fisiologia , Músculo Liso/fisiologia , Sistema Respiratório/citologia , Animais , Linhagem Celular , Células Cultivadas , Técnicas de Cocultura , Complemento C3a/análise , Complemento C3a/farmacologia , Complemento C5a/farmacologia , Meios de Cultivo Condicionados , Dexametasona/farmacologia , Humanos , Imuno-Histoquímica , Antígeno de Macrófago 1/análise , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/fisiologia , Macrófagos/química , Macrófagos/fisiologia , Macrófagos Alveolares/química , Mastócitos/química , Camundongos , Músculo Liso/química , RNA Mensageiro/análise , Receptor da Anafilatoxina C5a/análise , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Traqueia/química , Traqueia/citologia , Transfecção
20.
J Biol Chem ; 278(13): 11123-9, 2003 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-12540846

RESUMO

The orphan receptor C5L2 has recently been described as a high affinity binding protein for complement fragments C5a and C3a that, unlike the previously described C5a receptor (CD88), couples only weakly to G(i)-like G proteins (Cain, S. A., and Monk, P. N. (2002) J. Biol. Chem. 277, 7165-7169). Here we demonstrate that C5L2 binds the metabolites of C4a and C3a, C4a des-Arg(77), and C3a des-Arg(77) (also known as the acylation-stimulating protein or ASP) at a site distinct from the C5a binding site. The binding of these metabolites to C5L2 does not stimulate the degranulation of transfected rat basophilic leukemia cells either through endogenous rat G proteins or when co-transfected with human G(alpha 16). C3a des-Arg(77)/ASP and C3a can potently stimulate triglyceride synthesis in human skin fibroblasts and 3T3-L1 preadipocytes. Here we show that both cell types and human adipose tissue express C5L2 mRNA and that the human fibroblasts express C5L2 protein at the cell surface. This is the first demonstration of the expression of C5L2 in cells that bind and respond to C3a des-Arg(77)/ASP and C3a. Thus C5L2, a promiscuous complement fragment-binding protein with a high affinity site that binds C3a des-Arg(77)/ASP, may mediate the acylation-stimulating properties of this peptide.


Assuntos
Proteínas Sanguíneas/metabolismo , Complemento C3a/análogos & derivados , Complemento C3a/metabolismo , Receptores de Quimiocinas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas Sanguíneas/fisiologia , Degranulação Celular/fisiologia , Linhagem Celular , Complemento C3a/fisiologia , Primers do DNA , Humanos , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Ratos , Receptor da Anafilatoxina C5a , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA