Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
FASEB J ; 36(11): e22599, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36250902

RESUMO

Emerging evidence suggest that C3aR plays important roles in homeostasis, host defense and disease. Although it is known that C3aR is protective in several models of acute bacterial infections, the role for C3aR in chronic infection is largely unknown. Here we show that C3aR is protective in experimental chronic pyelonephritis. Global C3aR deficient (C3ar-/- ) mice had higher renal bacterial load, more pronounced renal histological lesions, increased renal apoptotic cell accumulation, tissue inflammation and extracellular matrix deposition following renal infection with uropathogenic E. coli (UPEC) strain IH11128, compared to WT control mice. Myeloid C3aR deficient (Lyz2-C3ar-/- ) mice exhibited a similar disease phenotype to global C3ar-/- mice. Pharmacological treatment with a C3aR agonist reduced disease severity in experimental chronic pyelonephritis. Furthermore, macrophages of C3ar-/- mice exhibited impaired ability to phagocytose UPEC. Our data clearly demonstrate a protective role for C3aR against experimental chronic pyelonephritis, macrophage C3aR plays a major role in the protection, and C3aR is necessary for phagocytosis of UPEC by macrophages. Our observation that C3aR agonist curtailed the pathology suggests a therapeutic potential for activation of C3aR in chronic infection.


Assuntos
Infecções por Escherichia coli , Pielonefrite , Receptores de Complemento , Animais , Camundongos , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/patologia , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Rim/microbiologia , Rim/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Pielonefrite/imunologia , Pielonefrite/microbiologia , Pielonefrite/patologia , Pielonefrite/prevenção & controle , Escherichia coli Uropatogênica/patogenicidade , Receptores de Complemento/agonistas , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Receptores de Complemento/imunologia , Matriz Extracelular/metabolismo
2.
Nat Commun ; 12(1): 7172, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34887405

RESUMO

Complement receptor of immunoglobulin superfamily (CRIg) is expressed on liver macrophages and directly binds complement component C3b or Gram-positive bacteria to mediate phagocytosis. CRIg plays important roles in several immune-mediated diseases, but it is not clear how its pathogen recognition and phagocytic functions maintain homeostasis and prevent disease. We previously associated cytolysin-positive Enterococcus faecalis with severity of alcohol-related liver disease. Here, we demonstrate that CRIg is reduced in liver tissues from patients with alcohol-related liver disease. CRIg-deficient mice developed more severe ethanol-induced liver disease than wild-type mice; disease severity was reduced with loss of toll-like receptor 2. CRIg-deficient mice were less efficient than wild-type mice at clearing Gram-positive bacteria such as Enterococcus faecalis that had translocated from gut to liver. Administration of the soluble extracellular domain CRIg-Ig protein protected mice from ethanol-induced steatohepatitis. Our findings indicate that ethanol impairs hepatic clearance of translocated pathobionts, via decreased hepatic CRIg, which facilitates progression of liver disease.


Assuntos
Enterococcus faecalis/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , Hepatopatias Alcoólicas/imunologia , Macrófagos/imunologia , Receptores de Complemento 3b/imunologia , Receptores de Complemento/imunologia , Animais , Translocação Bacteriana , Complemento C3b/imunologia , Enterococcus faecalis/fisiologia , Etanol/efeitos adversos , Feminino , Trato Gastrointestinal/microbiologia , Infecções por Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/microbiologia , Hepatopatias Alcoólicas/etiologia , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Receptores de Complemento 3b/genética
3.
Biochem Biophys Res Commun ; 516(3): 858-865, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31266632

RESUMO

The innate immune response contributes to hepatic steatosis and nonalcoholic fatty liver disease (NAFLD). However, the pathogenic mechanism of NAFLD is still poorly understood. The costimulatory molecule V-set and immunoglobulin domain-containing protein-4 (Vsig4), which is exclusively expressed on macrophages, shows significant role in regulating macrophage-mediated inflammation. Here, we attempted to explore if Vsig4 expression was involved in high fat diet (HFD)-induced NAFLD. The results indicated that Vsig4 expression was markedly down-regulated in fatty livers of NAFLD patients and obese mice. Vsig4 knockout accelerated HFD-induced metabolic dysfunction. In addition, the loss of Vsig4 significantly promoted insulin resistance and lipid deposition in liver samples of HFD-challenged mice. Furthermore, HFD-induced inflammation was apparently accelerated in Vsig4 knockout mice by further activating nuclear factor-κB (NF-κB) signaling pathway. Also, Vsig4 deficient mice exhibited greater collagen accumulation in hepatic samples in HFD-challenged mice compared to the WT mice, which was through promoting transforming growth factor-ß1 (TGFß1) signaling. Importantly, we found that lipopolysaccharide (LPS)- or TGFß1-stimulated inflammation and fibrosis in primary hepatocytes and hepatic stellate cells, respectively, were markedly exacerbated by co-culture with condition medium from bone marrow-derived macrophages (BMDMs) with Vsig4 deficiency. Finally, transplantation of bone marrow cells from control mice to Vsig4-knockout mice restored the severity of steatosis, inflammation and fibrosis after HFD feeding. Therefore, loss of Vsig4 accelerated the severity of lipid deposition, fibrosis and the inflammatory response. Vsig4 could be a therapeutic target for NAFLD treatment.


Assuntos
Cirrose Hepática/genética , Macrófagos/imunologia , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/genética , Receptores de Complemento/genética , Animais , Transplante de Medula Óssea , Colágeno/genética , Colágeno/imunologia , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica , Células Estreladas do Fígado/imunologia , Células Estreladas do Fígado/patologia , Hepatócitos/imunologia , Hepatócitos/patologia , Humanos , Imunidade Inata , Inflamação , Resistência à Insulina , Cirrose Hepática/etiologia , Cirrose Hepática/imunologia , Cirrose Hepática/terapia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/terapia , Obesidade/etiologia , Obesidade/patologia , Obesidade/terapia , Cultura Primária de Células , Receptores de Complemento/deficiência , Receptores de Complemento/imunologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/imunologia
4.
Rheumatology (Oxford) ; 57(8): 1400-1407, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29718374

RESUMO

Objectives: The aim was to describe the clinical characteristics and epidemiology of hypocomplementaemic urticarial vasculitis (HUV; anti-C1q vasculitis) in two geographically defined areas of Sweden. Methods: In the health-care districts surrounding Skåne University Hospital (mean population 950 560) and Linköping University Hospital (mean population 428 503), all incident cases of HUV residing within the study areas at the onset of disease were identified during the years 2000-15. The diagnosis of HUV was confirmed by review of medical records. Only patients meeting the proposed diagnostic HUV criteria and/or the 2012 Chapel Hill consensus definitions in combination with an ever-positive anti-C1q antibody test were included. Results: Sixteen patients (14 females) were identified during the study period. The median (interquartile range) age at diagnosis was 51 (40.7-56.7) years. Median (interquartile range) time of follow-up from diagnosis to 31 December 2015, or death, was 94 (46.5-136.2) months. The most frequent manifestations at diagnosis were urticaria (100%), arthritis (88%), followed by biopsy-proven glomerulonephritis (19%), episcleritis/scleritis (19%) and recurrent abdominal pain (13%). The annual incidence rate per million inhabitants was estimated as 0.7 (95% CI: 0.4, 1.1). Sixty-three per cent suffered from pulmonary disease at the last follow-up. Two patients died during the follow-up period. One patient underwent lung transplantation, and two patients proceeded to end-stage renal disease. The point prevalence on 31 December 2015 was 9.5/million (95% CI: 4.5, 14.5). Conclusion: Hypocomplementaemic urticarial vasculitis constitutes a rare, but not always benign condition. Renal and lung manifestations were severe in some cases, highlighting the need for careful screening and monitoring of this potentially serious condition.


Assuntos
Glicoproteínas de Membrana/deficiência , Vigilância da População , Receptores de Complemento/deficiência , Urticária/epidemiologia , Vasculite/epidemiologia , Adulto , Idoso , Feminino , Seguimentos , Humanos , Incidência , Masculino , Glicoproteínas de Membrana/sangue , Pessoa de Meia-Idade , Receptores de Complemento/sangue , Estudos Retrospectivos , Suécia/epidemiologia , Urticária/sangue , Urticária/complicações , Vasculite/sangue , Vasculite/etiologia
5.
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
6.
Nat Commun ; 8(1): 1322, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29109438

RESUMO

Exacerbation of macrophage-mediated inflammation contributes to pathogenesis of various inflammatory diseases, but the immunometabolic programs underlying regulation of macrophage activation are unclear. Here we show that V-set immunoglobulin-domain-containing 4 (VSIG4), a B7 family-related protein that is expressed by resting macrophages, inhibits macrophage activation in response to lipopolysaccharide. Vsig4 -/- mice are susceptible to high-fat diet-caused obesity and murine hepatitis virus strain-3 (MHV-3)-induced fulminant hepatitis due to excessive macrophage-dependent inflammation. VSIG4 activates the PI3K/Akt-STAT3 pathway, leading to pyruvate dehydrogenase kinase-2 (PDK2) upregulation and subsequent phosphorylation of pyruvate dehydrogenase, which results in reduction in pyruvate/acetyl-CoA conversion, mitochondrial reactive oxygen species secretion, and macrophage inhibition. Conversely, interruption of Vsig4 or Pdk2 promotes inflammation. Forced expression of Vsig4 in mice ameliorates MHV-3-induced viral fulminant hepatitis. These data show that VSIG4 negatively regulates macrophage activation by reprogramming mitochondrial pyruvate metabolism.


Assuntos
Ativação de Macrófagos/fisiologia , Ácido Pirúvico/metabolismo , Receptores de Complemento/metabolismo , Animais , Infecções por Coronavirus/etiologia , Dieta Hiperlipídica/efeitos adversos , Células HEK293 , Hepatite Viral Animal/etiologia , Humanos , Inflamação/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Vírus da Hepatite Murina , Obesidade/etiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Células THP-1
7.
J Immunol ; 196(11): 4783-92, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27183625

RESUMO

The complement peptide C3a is a key component of the innate immune system and a major fragment produced following complement activation. We used a murine model of melanoma (B16-F0) to identify a hitherto unknown role for C3a-C3aR signaling in promoting tumor growth. The results show that the development and growth of B16-F0 melanomas is retarded in mice lacking C3aR, whereas growth of established melanomas can be arrested by C3aR antagonism. Flow cytometric analysis showed alterations in tumor-infiltrating leukocytes in the absence of C3aR. Specifically, neutrophils and CD4(+) T lymphocyte subpopulations were increased, whereas macrophages were reduced. The central role of neutrophils was confirmed by depletion experiments that reversed the tumor inhibitory effects observed in C3aR-deficient mice and returned tumor-infiltrating CD4(+) T cells to control levels. Analysis of the tumor microenvironment showed upregulation of inflammatory genes that may contribute to the enhanced antitumor response observed in C3aR-deficient mice. C3aR deficiency/inhibition was also protective in murine models of BRAF(V600E) mutant melanoma and colon and breast cancer, suggesting a tumor-promoting role for C3aR signaling in a range of tumor types. We propose that C3aR activation alters the tumor inflammatory milieu, thereby promoting tumor growth. Therapeutic inhibition of C3aR may therefore be an effective means to trigger an antitumor response in melanoma and other cancers.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Carcinogênese/imunologia , Melanoma/imunologia , Melanoma/patologia , Neutrófilos/imunologia , Receptores de Complemento/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Células Cultivadas , Feminino , Melanoma/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/patologia , Receptores de Complemento/deficiência
8.
J Neuroinflammation ; 13: 23, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26822321

RESUMO

BACKGROUND: The complement system is becoming increasingly recognized as a key participant in many neurodegenerative diseases of the brain. Complement-deficient animals exhibit reduced neuroinflammation. METHODS: In the present study, we administered intracerebroventricularly lipopolysaccharide (LPS) to mimic local infection of the brain and investigated the role of key complement component C3 in brain vasculature endothelial activation and leukocyte recruitment. The degree of neutrophil infiltration was determined by esterase staining. Leukocyte-endothelial interactions were measured using intravital microscopy. Cerebral endothelial activation was evaluated using real-time PCR and Western blotting. RESULTS: Neutrophil infiltration into the brain cortex and hippocampus was significantly reduced in C3(-/-) mice and C3aR(-/-) mice but not in C6(-/-) mice. We detected markedly attenuated leukocyte-endothelial interactions in the brain microvasculature of C3(-/-) mice. Accordingly, in response to LPS administration, the brain microvasculature in these mice had decreased expression of P-selectin, E-selectin, intercellular cell adhesion molecule 1 (ICAM-1), and vascular cell adhesion molecule 1 (VCAM-1). Depletion of C3 from the circulation also caused reduction in VCAM-1 and E-selectin expression and leukocyte recruitment, suggesting that C3 in the circulation contributed to brain endothelial activation. Furthermore, C3(-/-) mice exhibited decreased leukocyte recruitment into the brain upon tumor necrosis factor-α (TNF-α) stimulation. C3a activated the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) and induced the upregulation of VCAM-1 and ICAM-1 expression in murine primary cerebral endothelial cells in vitro. CONCLUSIONS: Our study provides the first evidence that C3a plays a critical role in cerebral endothelial activation and leukocyte recruitment during inflammation in the brain.


Assuntos
Encéfalo/citologia , Complemento C3a/metabolismo , Células Endoteliais/fisiologia , Leucócitos/fisiologia , Infiltração de Neutrófilos/fisiologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiologia , Encéfalo/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Complemento C3a/genética , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Injeções Intraventriculares , Leucócitos/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microvasos/efeitos dos fármacos , Microvasos/fisiologia , NF-kappa B/metabolismo , Infiltração de Neutrófilos/efeitos dos fármacos , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Fatores de Tempo
9.
Mucosal Immunol ; 8(4): 874-85, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25465103

RESUMO

Exposure to cigarette smoke can initiate sterile inflammatory responses in the lung and activate myeloid dendritic cells (mDCs) that induce differentiation of T helper type 1 (Th1) and Th17 cells in the emphysematous lungs. Consumption of complement proteins increases in acute inflammation, but the contribution of complement protein 3 (C3) to chronic cigarette smoke-induced immune responses in the lung is not clear. Here, we show that following chronic exposure to cigarette smoke, C3-deficient (C3(-/-)) mice develop less emphysema and have fewer CD11b(+)CD11c(+) mDCs infiltrating the lungs as compared with wild-type mice. Proteolytic cleavage of C3 by neutrophil elastase releases C3a, which in turn increases the expression of its receptor (C3aR) on lung mDCs. Mice deficient in the C3aR (C3ar(-/-)) partially phenocopy the attenuated responses to chronic smoke observed in C3(-/-) mice. Consistent with a role for C3 in emphysema, C3 and its active fragments are deposited on the lung tissue of smokers with emphysema, and smoke-exposed mice. Together, these findings suggest a critical role for C3a through autocrine/paracrine induction of C3aR in the pathogenesis of cigarette smoke-induced sterile inflammation and provide new therapeutic targets for the treatment of emphysema.


Assuntos
Enfisema/etiologia , Enfisema/metabolismo , Receptores de Complemento/metabolismo , Fumar/efeitos adversos , Animais , Comunicação Autócrina , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Ativação do Complemento , Complemento C3/genética , Complemento C3/imunologia , Complemento C3/metabolismo , Complemento C3a/imunologia , Complemento C3a/metabolismo , Modelos Animais de Doenças , Enfisema/diagnóstico , Regulação da Expressão Gênica , Humanos , Elastase de Leucócito/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Metaloproteinase 12 da Matriz/metabolismo , Camundongos Knockout , Comunicação Parácrina , Proteólise , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Transdução de Sinais
10.
J Immunol ; 193(3): 1278-89, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24981453

RESUMO

Listeria monocytogenes is a Gram-positive intracellular bacterium that is acquired through tainted food and may lead to systemic infection and possible death. Despite the importance of the innate immune system in fighting L. monocytogenes infection, little is known about the role of complement and its activation products, including the potent C3a anaphylatoxin. In a model of systemic L. monocytogenes infection, we show that mice lacking the receptor for C3a (C3aR(-/-)) are significantly more sensitive to infection compared with wild-type mice, as demonstrated by decreased survival, increased bacterial burden, and increased damage to their livers and spleens. The inability of the C3aR(-/-) mice to clear the bacterial infection was not caused by defective macrophages or by a reduction in cytokines/chemokines known to be critical in the host response to L. monocytogenes, including IFN-γ and TNF-α. Instead, TUNEL staining, together with Fas, active caspase-3, and Bcl-2 expression data, indicates that the increased susceptibility of C3aR(-/-) mice to L. monocytogenes infection was largely caused by increased L. monocytogenes-induced apoptosis of myeloid and lymphoid cells in the spleen that are required for ultimate clearance of L. monocytogenes, including neutrophils, macrophages, dendritic cells, and T cells. These findings reveal an unexpected function of C3a/C3aR signaling during the host immune response that suppresses Fas expression and caspase-3 activity while increasing Bcl-2 expression, thereby providing protection to both myeloid and lymphoid cells against L. monocytogenes-induced apoptosis.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Apoptose/imunologia , Complemento C3a/metabolismo , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/patologia , Receptores de Complemento/fisiologia , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Caspase 3/metabolismo , Inibidores de Caspase/farmacologia , Complemento C3a/imunologia , Modelos Animais de Doenças , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Listeria monocytogenes/patogenicidade , Listeriose/genética , Linfócitos/imunologia , Linfócitos/metabolismo , Linfócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Regulação para Cima/genética , Regulação para Cima/imunologia , Receptor fas/antagonistas & inibidores , Receptor fas/biossíntese
11.
Infect Immun ; 82(1): 371-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24191300

RESUMO

The host immune response plays an important role in the onset and progression of cerebral malaria (CM). The complement system is an essential component of the innate immune response to malaria, and its activation generates the anaphylatoxin C5a. To test the hypothesis that C5a signaling contributes to the pathogenesis of CM, we investigated a causal role for the C5a receptors C5aR and C5L2 in a mouse model of experimental CM (ECM) induced by Plasmodium berghei ANKA infection, and using a case-control design, we examined levels of C5a in plasma samples from Ugandan children presenting with CM or uncomplicated malaria (UM). In the ECM model, C5aR(-/-) mice displayed significantly improved survival compared to their wild-type (WT) counterparts (P = 0.004), whereas C5L2(-/-) mice showed no difference in survival from WT mice. Improved survival in C5aR(-/-) mice was associated with reduced levels of the proinflammatory cytokines tumor necrosis factor (TNF) and gamma interferon (IFN-γ) and the chemokine, monocyte chemoattractant protein 1 (MCP-1) (CCL2). Furthermore, endothelial integrity was enhanced, as demonstrated by increased levels of angiopoietin-1, decreased levels of angiopoietin-2 and soluble ICAM-1, and decreased Evans blue extravasation into brain parenchyma. In the case-control study, the median levels of C5a at presentation were significantly higher in children with CM versus those in children with UM (43.7 versus 22.4 ng/ml; P < 0.001). These findings demonstrate that C5a is dysregulated in human CM and contributes to the pathogenesis of ECM via C5aR-dependent inflammation and endothelial dysfunction.


Assuntos
Complemento C5a/imunologia , Malária Cerebral/imunologia , Receptores de Quimiocinas/imunologia , Receptores de Complemento/imunologia , Animais , Estudos de Casos e Controles , Criança , Pré-Escolar , Complemento C5a/deficiência , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor da Anafilatoxina C5a , Receptores de Complemento/deficiência , Receptores de Concanavalina A
12.
J Immunol ; 191(3): 1006-10, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23817414

RESUMO

The complement system is a potent component of the innate immune response, promoting inflammation and orchestrating defense against pathogens. However, dysregulation of complement is critical to several autoimmune and inflammatory syndromes. Elevated expression of the proinflammatory cytokine IL-1ß is often linked to such diseases. In this study, we reveal the mechanistic link between complement and IL-1ß secretion using murine dendritic cells. IL-1ß secretion occurs following intracellular caspase-1 activation by inflammasomes. We show that complement elicits secretion of both IL-1ß and IL-18 in vitro and in vivo via the NLRP3 inflammasome. This effect depends on the inflammasome components NLRP3 and ASC, as well as caspase-1 activity. Interestingly, sublethal complement membrane attack complex formation, but not the anaphylatoxins C3a and C5a, activated the NLRP3 inflammasome in vivo. These findings provide insight into the molecular processes underlying complement-mediated inflammation and highlight the possibility of targeting IL-1ß to control complement-induced disease and pathological inflammation.


Assuntos
Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Células Dendríticas/imunologia , Inflamassomos/imunologia , Interleucina-1beta/imunologia , Animais , Células da Medula Óssea , Proteínas de Transporte/genética , Células Cultivadas , Complemento C6/deficiência , Complemento C6/genética , Proteínas do Sistema Complemento/imunologia , Células Dendríticas/metabolismo , Ativação Enzimática , Inflamação/imunologia , Interleucina-18/biossíntese , Interleucina-18/imunologia , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/genética , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Transdução de Sinais
13.
FASEB J ; 27(9): 3797-804, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23737250

RESUMO

Complement is an essential component of inflammation that plays a role in ischemic brain injury. Recent reports demonstrate novel functions of complement in normal and diseased CNS, such as regulation of neurogenesis and synapse elimination. Here, we examined the role of complement-derived peptide C3a in unilateral hypoxia-ischemia (HI), a model of neonatal HI encephalopathy. HI injury was induced at postnatal day 9 (P9), and loss of hippocampal tissue was determined on P31. We compared WT mice with transgenic mice expressing C3a under the control of glial fibrillary acidic protein promoter, which express biologically active C3a only in CNS and without the requirement of a priori complement activation. Further, we injected C3a peptide into the lateral cerebral ventricle of mice lacking the C3a receptor (C3aR) and WT mice and assessed HI-induced memory impairment 41 d later. We found that HI-induced tissue loss in C3a overexpressing mice was reduced by 50% compared with WT mice. C3a peptide injected 1 h after HI protected WT but not C3aR-deficient mice against HI-induced memory impairment. Thus, C3a acting through its canonical receptor ameliorates behavioral deficits after HI injury, and C3aR is a novel therapeutic target for the treatment of neonatal HI encephalopathy.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Receptores de Complemento/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/metabolismo , Complemento C3a/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/patologia , Imuno-Histoquímica , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , RNA Mensageiro/genética , Receptores de Complemento/deficiência , Receptores de Complemento/genética
14.
J Exp Med ; 210(2): 257-68, 2013 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-23382542

RESUMO

Thymus-derived (natural) CD4(+) FoxP3(+) regulatory T cells (nT reg cells) are required for immune homeostasis and self-tolerance, but must be stringently controlled to permit expansion of protective immunity. Previous findings linking signals transmitted through T cell-expressed C5a receptor (C5aR) and C3a receptor (C3aR) to activation, differentiation, and expansion of conventional CD4(+)CD25(-) T cells (T conv cells), raised the possibility that C3aR/C5aR signaling on nT reg cells could physiologically modulate nT reg cell function and thereby further impact the induced strength of T cell immune responses. In this study, we demonstrate that nT reg cells express C3aR and C5aR, and that signaling through these receptors inhibits nT reg cell function. Genetic and pharmacological blockade of C3aR/C5aR signal transduction in nT reg cells augments in vitro and in vivo suppression, abrogates autoimmune colitis, and prolongs allogeneic skin graft survival. Mechanisms involve C3a/C5a-induced phosphorylation of AKT and, as a consequence, phosphorylation of the transcription factor Foxo1, which results in lowered nT reg cell Foxp3 expression. The documentation that C3a/C3aR and C5a/C5aR modulate nT reg cell function via controlling Foxp3 expression suggests targeting this pathway could be exploited to manipulate pathogenic or protective T cell responses.


Assuntos
Receptor da Anafilatoxina C5a/metabolismo , Receptores de Complemento/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Doenças Autoimunes/prevenção & controle , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Rejeição de Enxerto/prevenção & controle , Imunidade Inata , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/genética , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/imunologia , Transplante de Pele/imunologia , Transplante de Pele/patologia , Linfócitos T Reguladores/metabolismo , Transplante Homólogo
15.
J Neuroinflammation ; 9: 220, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22989354

RESUMO

BACKGROUND: The neuronal microtubule-associated protein tau becomes hyperphosphorylated and forms aggregates in tauopathies but the processes leading to this pathological hallmark are not understood. Because tauopathies are accompanied by neuroinflammation and the complement cascade forms a key innate immune pathway, we asked whether the complement system has a role in the development of tau pathology. FINDINGS: We tested this hypothesis in two mouse models, which expressed either a central inhibitor of complement or lacked an inhibitor of the terminal complement pathway. Complement receptor-related gene/protein y is the natural inhibitor of the central complement component C3 in rodents. Expressing a soluble variant (sCrry) reduced the number of phospho-tau (AT8 epitope) positive neurons in the brain stem, cerebellum, cortex, and hippocampus of aged P301L mutant tau/sCrry double-transgenic mice compared with tau single-transgenic littermates (JNPL3 line). CD59a is the major inhibitor of formation of the membrane attack complex in mice. Intrahippocampal injection of adeno-associated virus encoding mutant human P301L tau into Cd59a-/- mice resulted in increased numbers of AT8-positive cells compared with wild-type controls. This was accompanied by neuronal and synaptic loss and reduced dendritic integrity. CONCLUSIONS: Our data in two independent mouse models with genetic changes in key regulators of the complement system support the hypothesis that the terminal pathway has an active role in the development of tau pathology. We propose that inhibition of the terminal pathway may be beneficial in tauopathies.


Assuntos
Antígenos CD59/metabolismo , Doenças Neurodegenerativas/metabolismo , Receptores de Complemento/deficiência , Transdução de Sinais/genética , Tauopatias/patologia , Animais , Encéfalo/patologia , Antígenos CD59/genética , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Humanos , Leucina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/genética , Neurônios/metabolismo , Fosforilação/genética , Prolina/genética , Receptores de Complemento/genética , Tauopatias/genética , Proteínas tau/genética , Proteínas tau/metabolismo
16.
J Immunol ; 185(5): 3086-94, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20675597

RESUMO

Congenital and acquired deficiencies of complement regulatory proteins are associated with pathologic complement activation in several renal diseases. To elucidate the mechanisms by which renal tubular epithelial cells (TECs) control the complement system, we examined the expression of complement regulatory proteins by the cells. We found that Crry is the only membrane-bound complement regulator expressed by murine TECs, and its expression is concentrated on the basolateral surface. Consistent with the polarized localization of Crry, less complement activation was observed when the basolateral surface of TECs was exposed to serum than when the apical surface was exposed. Furthermore, greater complement activation occurred when the basolateral surface of TECs from Crry(-/-)fB(-/-) mice was exposed to normal serum compared with TECs from wild-type mice. Complement activation on the apical and basolateral surfaces was also greater when factor H, an alternative pathway regulatory protein found in serum, was blocked from interacting with the cells. Finally, we injected Crry(-/-)fB(-/-) and Crry(+/+)fB(-/-) mice with purified factor B (an essential protein of the alternative pathway). Spontaneous complement activation was seen on the tubules of Crry(-/-)fB(-/-) mice after injection with factor B, and the mice developed acute tubular injury. These studies indicate that factor H and Crry regulate complement activation on the basolateral surface of TECs and that factor H regulates complement activation on the apical surface. However, congenital deficiency of Crry or reduced expression of the protein on the basolateral surface of injured cells permits spontaneous complement activation and tubular injury.


Assuntos
Fator H do Complemento/fisiologia , Proteínas Inativadoras do Complemento/fisiologia , Células Epiteliais/imunologia , Túbulos Renais/imunologia , Receptores de Complemento/fisiologia , Animais , Células Cultivadas , Fator H do Complemento/biossíntese , Fator H do Complemento/deficiência , Proteínas Inativadoras do Complemento/deficiência , Via Alternativa do Complemento/imunologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/deficiência , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica/imunologia , Receptores de Complemento/biossíntese , Receptores de Complemento/deficiência , Receptores de Complemento 3b
17.
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
18.
Blood ; 113(12): 2684-94, 2009 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-19136662

RESUMO

Although complement lysis is frequently used for the purification of lymphocyte subpopulations in vitro, how lymphocytes escape complement attack in vivo has not been clearly delineated. Here, we show that conditional gene targeting of a murine membrane complement regulator Crry on thymocytes led to complement-dependent peripheral T-cell lymphopenia. Notably, despite evidence of hypersensitivity to complement attack, Crry-deficient T cells escaped complement injury and developed normally in the thymus, because of low intrathymic complement activity. Crry-deficient T cells were eliminated in the periphery by a C3- and macrophage-mediated but C5-independent mechanism. Thus, Crry is essential for mature T-cell survival in the periphery but not for lymphogenesis in the thymus. The observation that the thymus is a complement-privileged site may have implications for complement-based antitumor therapies.


Assuntos
Complemento C3/imunologia , Linfopenia/genética , Receptores de Complemento/fisiologia , Subpopulações de Linfócitos T/citologia , Timo/citologia , Animais , Anticorpos Monoclonais/imunologia , Autoanticorpos/imunologia , Deleção Clonal , Via Clássica do Complemento , Marcação de Genes , Imunoglobulina G/imunologia , Linfopenia/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Opsonizantes/imunologia , Especificidade de Órgãos , RNA Mensageiro/biossíntese , Quimera por Radiação , Receptores de Complemento/biossíntese , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Receptores de Complemento 3b , Organismos Livres de Patógenos Específicos , Subpopulações de Linfócitos T/imunologia
19.
J Immunol ; 177(8): 4953-6, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17015675

RESUMO

Gene therapy is an attractive means to replace a deficient or defective protein. Using a murine retroviral vector, we provide an example of reconstituting a C regulator by neonatal in vivo gene transfer. The fusion gene containing the mouse C receptor 1-related gene/protein y (Crry) and a single chain Ab fragment with specificity for mouse glycophorin A was placed under transcriptional control of a liver-specific promoter. Shortly after birth, Crry KO mice were injected with the retroviral vectors. Protein expression progressively increased over the next 6-8 wk after which an equilibrium was established. Coating levels on RBCs were obtained that inhibited C activation similar to wild-type cells and remained constant for > 1 year. Thus, gene therapy with targeted regulators represents a treatment option to provide a long-term and sustained protein supply for the site-specific blockade of undesirable complement activation.


Assuntos
Terapia Genética/métodos , Receptores de Complemento/deficiência , Animais , Ativação do Complemento/efeitos dos fármacos , Eritrócitos/imunologia , Vetores Genéticos/uso terapêutico , Glicoforinas/imunologia , Fragmentos de Imunoglobulinas/administração & dosagem , Fragmentos de Imunoglobulinas/genética , Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , Receptores de Complemento/genética , Receptores de Complemento 3b , Proteínas Recombinantes de Fusão/administração & dosagem , Retroviridae
20.
J Immunol ; 177(8): 5558-66, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17015743

RESUMO

Decay-accelerating factor (DAF) and complement receptor 1-related gene/protein y (Crry) are two membrane-bound complement regulators on murine erythrocytes that inhibit C3/C5 convertases. Previously, we found that Crry- but not DAF-deficient erythrocytes were susceptible to alternative pathway complement-mediated elimination in vivo. To determine whether it is a unique activity or a higher level expression of Crry makes it indispensable on murine erythrocytes, we over-expressed DAF on Crry-deficient (Crry(-/-)) erythrocytes by retroviral vector-mediated DAF gene transduction of bone marrow stem cells. DAF retrovirus-transduced erythrocytes expressed 846 +/- 127 DAF molecules/cell (DAF(high)) compared with 249 +/- 94 DAF molecules/cell (DAF(low)) and 774 +/- 135 Crry molecules/cell on control mouse erythrocytes. DAF(high)-Crry(-/-) erythrocytes were significantly more resistant than either DAF(low)-Crry(-/-), DAF(-/-) -Crry(+/+) or wild-type erythrocytes to classical pathway complement-mediated C3 deposition in vitro. Furthermore, increased DAF expression rescued Crry(-/-) erythrocytes from acute alternative pathway complement attack in vivo. Notably, long term monitoring revealed that DAF(high)-Crry(-/-) erythrocytes were still more susceptible than wild-type erythrocytes to complement-mediated elimination as they had a shorter half-life in complement-sufficient mice but survived equally well in complement-deficient mice. These results suggest that both a high level expression and a more potent anti-alternative pathway complement activity of Crry contributed to its indispensable role on murine erythrocytes. Additionally, they demonstrate the feasibility of using stem cell gene therapy to correct membrane complement regulator deficiency on blood cells in vivo.


Assuntos
Antígenos CD55/administração & dosagem , Via Alternativa do Complemento , Eritrócitos , Receptores de Complemento/deficiência , Células-Tronco/metabolismo , Transdução Genética/métodos , Animais , Células da Medula Óssea , Antígenos CD55/uso terapêutico , Eritrócitos/imunologia , Eritrócitos/metabolismo , Eritrócitos/patologia , Terapia Genética/métodos , Meia-Vida , Camundongos , Camundongos Knockout , Receptores de Complemento/imunologia , Receptores de Complemento 3b , Retroviridae
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA