Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Nature ; 543(7643): 108-112, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28225753

RESUMEN

Gaucher disease is caused by mutations in GBA1, which encodes the lysosomal enzyme glucocerebrosidase (GCase). GBA1 mutations drive extensive accumulation of glucosylceramide (GC) in multiple innate and adaptive immune cells in the spleen, liver, lung and bone marrow, often leading to chronic inflammation. The mechanisms that connect excess GC to tissue inflammation remain unknown. Here we show that activation of complement C5a and C5a receptor 1 (C5aR1) controls GC accumulation and the inflammatory response in experimental and clinical Gaucher disease. Marked local and systemic complement activation occurred in GCase-deficient mice or after pharmacological inhibition of GCase and was associated with GC storage, tissue inflammation and proinflammatory cytokine production. Whereas all GCase-inhibited mice died within 4-5 weeks, mice deficient in both GCase and C5aR1, and wild-type mice in which GCase and C5aR were pharmacologically inhibited, were protected from these adverse effects and consequently survived. In mice and humans, GCase deficiency was associated with strong formation of complement-activating GC-specific IgG autoantibodies, leading to complement activation and C5a generation. Subsequent C5aR1 activation controlled UDP-glucose ceramide glucosyltransferase production, thereby tipping the balance between GC formation and degradation. Thus, extensive GC storage induces complement-activating IgG autoantibodies that drive a pathway of C5a generation and C5aR1 activation that fuels a cycle of cellular GC accumulation, innate and adaptive immune cell recruitment and activation in Gaucher disease. As enzyme replacement and substrate reduction therapies are expensive and still associated with inflammation, increased risk of cancer and Parkinson disease, targeting C5aR1 may serve as a treatment option for patients with Gaucher disease and, possibly, other lysosomal storage diseases.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Enfermedad de Gaucher/inmunología , Enfermedad de Gaucher/patología , Glucosilceramidas/inmunología , Glucosilceramidas/metabolismo , Inflamación/inmunología , Inflamación/patología , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Autoanticuerpos/inmunología , Activación de Complemento , Complemento C5a/biosíntesis , Complemento C5a/inmunología , Proteínas del Sistema Complemento/biosíntesis , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Enfermedad de Gaucher/metabolismo , Enfermedad de Gaucher/prevención & control , Glucosilceramidasa/antagonistas & inhibidores , Glucosilceramidasa/deficiencia , Glucosilceramidasa/genética , Glucosiltransferasas/biosíntesis , Glucosiltransferasas/metabolismo , Humanos , Inmunoglobulina G/inmunología , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Ratones , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología
2.
Circ Res ; 122(7): 970-983, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29437833

RESUMEN

RATIONALE: Inflammation and immunity play crucial roles in the development of hypertension. Complement activation-mediated innate immune response is involved in the regulation of hypertension and target-organ damage. However, whether complement-mediated T-cell functions could regulate blood pressure elevation in hypertension is still unclear. OBJECTIVE: We aim to determine whether C3aR (complement component 3a receptor) and C5aR (complement component 5a receptor) could regulate blood pressure via modulating regulatory T cells (Tregs). METHODS AND RESULTS: We showed that angiotensin II (Ang II)-induced hypertension resulted in an elevated expression of C3aR and C5aR in Foxp3 (forkhead box P3)+ Tregs. By using C3aR and C5aR DKO (double knockout) mice, we showed that C3aR and C5aR deficiency together strikingly decreased both systolic and diastolic blood pressure in response to Ang II compared with WT (wild type), single C3aR-deficient (C3aR-/-), or C5aR-deficient (C5aR-/-) mice. Flow cytometric analysis showed that Ang II-induced Treg reduction in the kidney and blood was also blocked in DKO mice. Histological analysis indicated that renal and vascular structure remodeling and damage after Ang II treatment were attenuated in DKO mice compared with WT mice. In vitro, Ang II was able to stimulate C3aR and C5aR expression in cultured CD4+CD25+ natural Tregs. CD3 and CD28 antibody stimuli downregulated Foxp3 expression in WT but not DKO Tregs. More important, depletion of Tregs with CD25 antibody abolished the protective effects against Ang II-induced hypertension and target-organ damage in DKO mice. Adoptive transfer of DKO Tregs showed much more profound protective effects against Ang II-induced hypertension than WT Treg transfer. Furthermore, we demonstrated that C5aR expression in Foxp3+ Tregs was higher in hypertensive patients compared with normotensive individuals. CONCLUSIONS: C3aR and C5aR DKO-mediated Treg function prevents Ang II-induced hypertension and target-organ damage. Targeting C3aR and C5aR in Tregs specifically may be an alternative novel approach for hypertension treatment.


Asunto(s)
Hipertensión/inmunología , Receptor de Anafilatoxina C5a/deficiencia , Receptores de Complemento 3b/deficiencia , Linfocitos T Reguladores/inmunología , Angiotensina II/toxicidad , Animales , Células Cultivadas , Hipertensión/etiología , Hipertensión/genética , Masculino , Ratones , Ratones Endogámicos BALB C
3.
Allergy ; 74(4): 767-779, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30341777

RESUMEN

BACKGROUND: Food-induced anaphylaxis is a serious allergic reaction caused by Fcε-receptor activation on mast cells (MCs). The exact mechanisms breaking oral tolerance and the effector pathways driving food allergy remain elusive. As complement is activated in food-induced anaphylaxis, we aimed to assess the role of C5a in disease pathogenesis. METHODS: Oral antigen-induced food-induced anaphylaxis was induced in BALB/c wild-type (wt) and C5ar1-/- mice. Readouts included diarrhea development, changes in rectal temperature, hematocrit, antigen-specific serum IgE, MCPT-1, and intestinal MC numbers, as well as FcεR1-mediated MC functions including C5a receptor 1 (C5aR1) regulation. Further, histamine-mediated hypothermia and regulation of endothelial tight junctions were determined. RESULTS: Repeated oral OVA challenge resulted in diarrhea, hypothermia, increased hematocrit, high OVA-specific serum IgE, and MCPT-1 levels in wt mice. Male C5ar1-/- mice were completely whereas female C5ar1-/- mice were partially protected from anaphylaxis development. Serum MCPT-1 levels were reduced gender-independent, whereas IgE levels were reduced in male but not in female C5ar1-/- mice. Mechanistically, IgE-mediated degranulation and IL-6 production from C5ar1-/- BMMCs of both sexes were significantly reduced. Importantly, FcεR1 cross-linking strongly upregulated C5aR1 MC expression in vitro and in vivo. Finally, C5ar1-/- male mice were largely protected from histamine-induced hypovolemic shock, which was associated with protection from histamine-induced barrier dysfunction in vitro following C5aR targeting. CONCLUSIONS: Our findings identify C5aR1 activation as an important driver of IgE-mediated food allergy through regulation of allergen-specific IgE production, FcεR1-mediated MC degranulation, and histamine-driven effector functions preferentially in male mice.


Asunto(s)
Hipersensibilidad a los Alimentos/etiología , Inmunoglobulina E/sangre , Receptor de Anafilatoxina C5a/genética , Factores Sexuales , Anafilaxia , Animales , Degranulación de la Célula , Quimasas/sangre , Femenino , Masculino , Mastocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptores de IgE/inmunología
4.
J Immunol ; 197(6): 2353-61, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27521340

RESUMEN

There is accumulating evidence during sepsis that cardiomyocyte (CM) homeostasis is compromised, resulting in cardiac dysfunction. An important role for complement in these outcomes is now demonstrated. Addition of C5a to electrically paced CMs caused prolonged elevations of intracellular Ca(2+) concentrations during diastole, together with the appearance of spontaneous Ca(2+) transients. In polymicrobial sepsis in mice, we found that three key homeostasis-regulating proteins in CMs were reduced: Na(+)/K(+)-ATPase, which is vital for effective action potentials in CMs, and two intracellular Ca(2+) concentration regulatory proteins, that is, sarcoplasmic/endoplasmic reticulum calcium ATPase 2 and the Na(+)/Ca(2+) exchanger. Sepsis caused reduced mRNA levels and reductions in protein concentrations in CMs for all three proteins. The absence of either C5a receptor mitigated sepsis-induced reductions in the three regulatory proteins. Absence of either C5a receptor (C5aR1 or C5aR2) diminished development of defective systolic and diastolic echocardiographic/Doppler parameters developing in the heart (cardiac output, left ventricular stroke volume, isovolumic relaxation, E' septal annulus, E/E' septal annulus, left ventricular diastolic volume). We also found in CMs from septic mice the presence of defective current densities for Ik1, l-type calcium channel, and Na(+)/Ca(2+) exchanger. These defects were accentuated in the copresence of C5a. These data suggest complement-related mechanisms responsible for development of cardiac dysfunction during sepsis.


Asunto(s)
Coinfección/inmunología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Sepsis/inmunología , Sepsis/fisiopatología , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/inmunología , Coinfección/microbiología , Coinfección/fisiopatología , Complemento C5a/inmunología , Citoplasma/química , Citoplasma/metabolismo , Corazón/fisiopatología , Ratones , Miocitos Cardíacos/microbiología , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/inmunología , Receptor de Anafilatoxina C5a/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/inmunología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Sepsis/complicaciones
5.
Kidney Int ; 91(6): 1386-1397, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28139294

RESUMEN

C3 glomerulopathy is a potentially life-threatening disease of the kidney caused by dysregulated alternative pathway complement activation. The specific complement mediator(s) responsible for kidney injury in C3 glomerulopathy are yet to be defined and no specific therapy is currently available. We previously developed a mouse model of lethal C3 glomerulopathy with factor H and properdin gene double mutations. Therefore, we used this model to examine the role of C5 and C5a receptor (C5aR) in the pathogenesis of the disease. Disease severity in these factor H/properdin double-mutant mice was found to be correlated with plasma C5 levels, and prophylactic anti-C5 mAb therapy was effective in preventing lethal C3 glomerulopathy. When given to these double-mutant mice that had already developed active disease with severe proteinuria, anti-C5 mAb treatment also prevented death in half of the mice. Deficiency of C5aR significantly reduced disease severity, suggesting that C5aR-mediated inflammation contributed to C3 glomerulopathy. Thus, C5 and C5aR have a critical role in C3 glomerulopathy. Hence, early intervention targeting these pathways may be an effective therapeutic strategy for patients with C3 glomerulopathy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Complemento C3/metabolismo , Complemento C5/antagonistas & inhibidores , Inactivadores del Complemento/farmacología , Vía Alternativa del Complemento/efectos de los fármacos , Glomerulonefritis/prevención & control , Riñón/efectos de los fármacos , Insuficiencia Renal/prevención & control , Animales , Complemento C3/inmunología , Complemento C5/inmunología , Complemento C5/metabolismo , Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Glomerulonefritis/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Properdina/deficiencia , Properdina/genética , Proteinuria/inmunología , Proteinuria/metabolismo , Proteinuria/prevención & control , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Insuficiencia Renal/genética , Insuficiencia Renal/inmunología , Insuficiencia Renal/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
6.
J Biol Chem ; 290(17): 10667-76, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25739439

RESUMEN

Complement 5a (C5a), a potent immune mediator generated by complement activation, promotes tumor growth; however, its role in tumor metastasis remains unclear. We demonstrate that C5a contributes to tumor metastases by modulating tumor inflammation in hepatic metastases of colon cancer. Colon cancer cell lines generate C5a under serum-free conditions, and C5a levels increase over time in a murine syngeneic colon cancer hepatic metastasis model. Furthermore, in the absence of C5a receptor or upon pharmacological inhibition of C5a production with an anti-C5 monoclonal antibody, tumor metastasis is severely impaired. A lack of C5a receptor in colon cancer metastatic foci reduces the infiltration of macrophages, neutrophils, and dendritic cells, and the role for C5a receptor on these cells were further verified by bone marrow transplantation experiments. Moreover, C5a signaling increases the expression of the chemokine monocyte chemoattractant protein-1 and the anti-inflammatory molecules arginase-1, interleukin 10, and transforming growth factor ß, but is inversely correlated with the expression of pro-inflammatory molecules, which suggests a mechanism for the role of C5a in the inflammatory microenvironment required for tumor metastasis. Our results indicate a new and potentially promising therapeutic application of complement C5a inhibitor for the treatment of malignant tumors.


Asunto(s)
Quimiocina CCL2/metabolismo , Neoplasias del Colon/inmunología , Complemento C5a/metabolismo , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/secundario , Animales , Línea Celular Tumoral , Neoplasias del Colon/patología , Activación de Complemento , Femenino , Inflamación/inmunología , Inflamación/patología , Neoplasias Hepáticas Experimentales/patología , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/patología , Infiltración Neutrófila , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Microambiente Tumoral/inmunología
7.
Mediators Inflamm ; 2016: 1340156, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27382187

RESUMEN

C5a is an inflammatory mediator generated by complement activation that positively regulates various arms of immune defense, including Toll-like receptor 4 (TLR4) signaling. The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is activated by pathogen products and cellular/tissue damage products and is a major contributor of IL-1ß. In this study, we investigate whether C5a modulates lipopolysaccharide- (LPS-) induced NLRP3 inflammasome activation in myeloid cells. Appearance of plasma IL-1ß during endotoxemia was reduced in C5aR1(-/-) mice when compared to wild-type mice. In vitro, C5a significantly enhanced LPS-induced production of IL-1ß in bone marrow Ly6C-high inflammatory monocytes, accompanied by augmented intracellular pro-IL-1ß expression. This effect was abolished during p38 blockade by SB 203580 and in the absence of C5aR1. Conversely, C5a suppressed LPS-induced macrophage production of IL-1ß, which was accompanied by attenuated levels of pro-IL-1ß, NLRP3, and caspase-1 expression. C5a's suppressive effects were negated during phosphoinositide 3-kinase (PI3K) inhibition by wortmannin but were largely preserved in the absence of C5aR1. Thus, C5a bidirectionally amplifies TLR4-mediated NLRP3 inflammasome activation in monocytes while suppressing this pathway in macrophages. However, as C5aR1 deficiency attenuates the IL-1ß response to LPS challenge in vivo, our results suggest overall that C5a augments physiologic inflammasome responses.


Asunto(s)
Inflamasomas/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Animales , Western Blotting , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Complemento C5a/metabolismo , Complemento C5a/farmacología , Modelos Animales de Enfermedad , Endotoxemia/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inflamasomas/efectos de los fármacos , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Biochim Biophys Acta ; 1842(1): 107-15, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24184716

RESUMEN

Premature babies are particularly vulnerable to brain injury. In this study we focus on cortical brain damage associated with long-term cognitive, behavioral, attentional or socialization deficits in children born preterm. Using a mouse model of preterm birth (PTB), we demonstrated that complement component C5a contributes to fetal cortical brain injury. Disruption of cortical dendritic and axonal cytoarchitecture was observed in PTB-mice. Fetuses deficient in C5aR (-/-) did not show cortical brain damage. Treatment with antibody anti-C5, that prevents generation of C5a, also prevented cortical fetal brain injury in PTB-mice. C5a also showed a detrimental effect on fetal cortical neuron development and survival in vitro. Increased glutamate release was observed in cortical neurons in culture exposed to C5a. Blockade of C5aR prevented glutamate increase and restored neurons dendritic and axonal growth and survival. Similarly, increased glutamate levels - measured by (1)HMRS - were observed in vivo in PTB-fetuses compared to age-matched controls. The blockade of glutamate receptors prevented C5a-induced abnormal growth and increased cell death in isolated fetal cortical neurons. Simvastatin and pravastatin prevented cortical fetal brain developmental and metabolic abnormalities -in vivo and in vitro. Neuroprotective effects of statins were mediated by Akt/PKB signaling pathways. This study shows that complement activation plays a crucial role in cortical fetal brain injury in PTL and suggests that complement inhibitors and statins might be good therapeutic options to improve neonatal outcomes in preterm birth.


Asunto(s)
Corteza Cerebral/metabolismo , Activación de Complemento , Complemento C5a/metabolismo , Neuronas/metabolismo , Nacimiento Prematuro/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Células Cultivadas , Corteza Cerebral/anomalías , Corteza Cerebral/efectos de los fármacos , Complemento C5a/antagonistas & inhibidores , Complemento C5a/genética , Femenino , Feto , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Recién Nacido , Ratones , Modelos Animales , Neuronas/efectos de los fármacos , Neuronas/patología , Pravastatina/farmacología , Embarazo , Nacimiento Prematuro/genética , Nacimiento Prematuro/patología , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Transducción de Señal , Simvastatina/farmacología
9.
Arterioscler Thromb Vasc Biol ; 34(2): 313-20, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24311376

RESUMEN

OBJECTIVE: Transfusion of aged blood has been associated with increased morbidity and mortality in critically ill patients. During storage, erythrocytes release increasing numbers of microvesicles (red blood cell-derived microvesicles [RBC-MV]). We hypothesized that RBC-MV mediate some of the deleterious effects of aged blood transfusions. APPROACH AND RESULTS: We established a murine transfusion model using RBC-MV purified from aged mouse erythrocytes. Injection of RBC-MV into healthy mice had no effect. However, they aggravated pulmonary leukocyte sequestration and peripheral blood leukopenia induced by lipopolysaccharides. Lipopolysaccharide-induced proinflammatory cytokines were significantly increased in plasma after RBC-MV injection. These effects were not seen in C5aR-deficient mice. In vitro, RBC-MV bound C3 fragments after incubation with plasma but failed to bind immunoglobulins, C1q, or mannose-binding lectin. Preventing thrombin generation inhibited complement activation in vitro and in vivo and reversed the proinflammatory effects of RBC-MV in lipopolysaccharide-primed mice. Finally, the RBC-MV-induced phenotype was recapitulated using phosphatidylserine-expressing liposomes, suggesting that surface expression of phosphatidylserine by RBC-MV was mechanistically involved. CONCLUSIONS: These results point toward a thrombin-dependent mechanism of complement activation by RBC-MV independent of the classical, lectin, or alternative pathway. Besides identifying RBC-MV as potential mediators of transfusion-related morbidity, our findings may be relevant for other inflammatory disorders involving intravascular microvesicle release, for example, sickle cell disease or thrombotic microangiopathy.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Activación de Complemento , Transfusión de Eritrocitos/efectos adversos , Eritrocitos/inmunología , Inflamación/inmunología , Trombina/inmunología , Animales , Micropartículas Derivadas de Células/metabolismo , Complemento C3/inmunología , Complemento C3/metabolismo , Eritrocitos/metabolismo , Genotipo , Inflamación/sangre , Inflamación/inducido químicamente , Leucopenia/sangre , Leucopenia/inmunología , Lipopolisacáridos , Liposomas , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Infiltración Neutrófila , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Fenotipo , Fosfatidilserinas/inmunología , Fosfatidilserinas/metabolismo , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Trombina/metabolismo , Factores de Tiempo
10.
J Immunol ; 191(3): 1006-10, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23817414

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Células Dendríticas/inmunología , Inflamasomas/inmunología , Interleucina-1beta/inmunología , Animales , Células de la Médula Ósea , Proteínas Portadoras/genética , Células Cultivadas , Complemento C6/deficiencia , Complemento C6/genética , Proteínas del Sistema Complemento/inmunología , Células Dendríticas/metabolismo , Activación Enzimática , Inflamación/inmunología , Interleucina-18/biosíntesis , Interleucina-18/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Transducción de Señal
11.
J Immunol ; 191(1): 178-86, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23709683

RESUMEN

The protective immunity induced by the whole-killed parasite vaccine against malarial blood-stage infection is dependent on the CD4(+) T cell response. However, the mechanism underlying this robust CD4(+) T cell response elicited by the whole-killed parasite vaccine is still largely unknown. In this study, we observe that immunization with Plasmodium yoelii-parasitized RBC lysate activates complement C5 and generates C5a. However, the protective efficacy against P. yoelii 17XL challenge is considerably reduced, and the malaria-specific CD4(+) T cell activation and memory T cell differentiation are largely suppressed in the C5aR-deficient (C5aR(-/-)) mice. An adoptive transfer assay demonstrates that the reduced protection of C5aR(-/-) mice is closely associated with the severely impaired CD4(+) T cell response. This is further confirmed by the fact that administration of C5aR antagonist significantly reduces the protective efficacy of the immunized B cell-deficient mice. Further study indicates that the defective CD4(+) T cell response in C5aR(-/-) mice is unlikely involved in the expansion of CD4(+)CD25(+)Foxp3(+) T cells, but strongly linked to a defect in dendritic cell (DC) maturation and the ability to allostimulate CD4(+) T cells. These results demonstrate that C5aR signaling is essential for the optimal induction of the malaria-specific CD4(+) T cell response by the whole-killed parasite vaccine through modulation of DCs function, which provides us with new clues to design an effective blood-stage subunit vaccine and helps us to understand the mechanism by which the T cell response is regulated by the complement system.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Malaria/patología , Plasmodium yoelii/inmunología , Receptor de Anafilatoxina C5a/fisiología , Transducción de Señal/inmunología , Animales , Linfocitos T CD4-Positivos/parasitología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Memoria Inmunológica/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Malaria/parasitología , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/sangre , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/patogenicidad , Receptor de Anafilatoxina C5a/sangre , Receptor de Anafilatoxina C5a/deficiencia , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/sangre
12.
J Am Soc Nephrol ; 25(2): 225-31, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24179165

RESUMEN

Necrotizing and crescentic GN (NCGN) with a paucity of glomerular immunoglobulin deposits is associated with ANCA. The most common ANCA target antigens are myeloperoxidase (MPO) and proteinase 3. In a manner that requires activation of the alternative complement pathway, passive transfer of antibodies to mouse MPO (anti-MPO) induces a mouse model of ANCA NCGN that closely mimics human disease. Here, we confirm the importance of C5aR/CD88 in the mediation of anti-MPO-induced NCGN and report that C6 is not required. We further demonstrate that deficiency of C5a-like receptor (C5L2) has the reverse effect of C5aR/CD88 deficiency and results in more severe disease, indicating that C5aR/CD88 engagement enhances inflammation and C5L2 engagement suppresses inflammation. Oral administration of CCX168, a small molecule antagonist of human C5aR/CD88, ameliorated anti-MPO-induced NCGN in mice expressing human C5aR/CD88. These observations suggest that blockade of C5aR/CD88 might have therapeutic benefit in patients with ANCA-associated vasculitis and GN.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/prevención & control , Autoantígenos/inmunología , Glomerulonefritis/prevención & control , Peroxidasa/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Administración Oral , Animales , Complemento C6/inmunología , Vía Alternativa del Complemento , Relación Dosis-Respuesta a Droga , Técnicas de Sustitución del Gen , Glomerulonefritis/complicaciones , Glomerulonefritis/inmunología , Hematuria/etiología , Hematuria/prevención & control , Humanos , Inmunización Pasiva , Leucocitos , Errores Innatos del Metabolismo/complicaciones , Errores Innatos del Metabolismo/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/deficiencia , Proteinuria/etiología , Proteinuria/prevención & control , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores de Quimiocina/fisiología , Proteínas Recombinantes de Fusión , Orina/citología
13.
J Immunol ; 186(2): 869-77, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149611

RESUMEN

The C5a anaphylatoxin receptor (C5aR; CD88) is activated as part of the complement cascade and exerts important inflammatory, antimicrobial, and regulatory functions, at least in part, via crosstalk with TLRs. However, the periodontal pathogen Porphyromonas gingivalis can control C5aR activation by generating C5a through its own C5 convertase-like enzymatic activity. In this paper, we show that P. gingivalis uses this mechanism to proactively and selectively inhibit TLR2-induced IL-12p70, whereas the same pathogen-instigated C5aR-TLR2 crosstalk upregulates other inflammatory and bone-resorptive cytokines (IL-1ß, IL-6, and TNF-α). In vivo, the ability of P. gingivalis to manipulate TLR2 activation via the C5a-C5aR axis allowed it to escape IL-12p70-dependent immune clearance and to cause inflammatory bone loss in a murine model of experimental periodontitis. In the latter regard, C5aR-deficient or TLR2-deficient mice were both resistant to periodontal bone loss, in stark contrast with wild-type control mice, which is consistent with the interdependent interactions of C5aR and TLR2 in P. gingivalis immune evasion and induction of bone-resorptive cytokines. In conclusion, P. gingivalis targets C5aR to promote its adaptive fitness and cause periodontal disease. Given the current availability of safe and effective C5aR antagonists, pharmacological blockade of C5aR could act therapeutically in human periodontitis and reduce associated systemic risks.


Asunto(s)
Pérdida de Hueso Alveolar/inmunología , Pérdida de Hueso Alveolar/microbiología , Interleucina-12/antagonistas & inhibidores , Interleucina-12/fisiología , Periodontitis/inmunología , Periodontitis/microbiología , Porphyromonas gingivalis/inmunología , Receptor de Anafilatoxina C5a/fisiología , Pérdida de Hueso Alveolar/metabolismo , Animales , Modelos Animales de Enfermedad , Evasión Inmune/genética , Evasión Inmune/inmunología , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Interleucina-10/fisiología , Interleucina-12/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Periodontitis/metabolismo , Porphyromonas gingivalis/crecimiento & desarrollo , Receptor Cross-Talk/inmunología , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/fisiología
14.
Eur J Immunol ; 40(3): 710-21, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20017191

RESUMEN

C5a is a proinflammatory mediator that has recently been shown to regulate adaptive immune responses. Here we demonstrate that C5a receptor (C5aR) signaling in DC affects the development of Treg and Th17 cells. Genetic ablation or pharmacological targeting of the C5aR in spleen-derived DC results in increased production of TGF-beta leading to de novo differentiation of Foxp3(+) Treg within 12 h after co-incubation with CD4(+) T cells from DO11.10/RAG2(-/-) mice. Stimulation of C5aR(-/-) DC with OVA and TLR2 ligand Pam(3)CSK(4) increased TGF-beta production and induced high levels of IL-6 and IL-23 but only minor amounts of IL-12 leading to differentiation of Th cells producing IL-17A and IL-21. Th17 differentiation was also found in vivo after adoptive transfer of CD4(+) Th cell into C5aR(-/-) mice immunized with OVA and Pam(3)CSK(4). The altered cytokine production of C5aR(-/-) DC was associated with low steady state MHC class II expression and an impaired ability to upregulate CD86 and CD40 in response to TLR2. Our data suggest critical roles for C5aR in Treg and Th17-cell differentiation through regulation of DC function.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Receptor de Anafilatoxina C5a/inmunología , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Reguladores/citología , Animales , Separación Celular , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interleucina-17/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Receptor de Anafilatoxina C5a/deficiencia , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
15.
Blood ; 114(5): 1005-15, 2009 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-19491392

RESUMEN

Toll-like receptors (TLRs) and complement are 2 major components of innate immunity that provide a first-line host defense and shape the adaptive immune responses. We show here that coincidental activation of complement and several TLRs in mice led to the synergistic production of serum factors that promoted T-helper cell 17 (Th17) differentiation from anti-CD3/CD28 or antigen-stimulated T cells. Although multiple TLR-triggered cytokines were regulated by complement, Th17 cell-promoting activity in the serum was correlated with interleukin (IL)-6 induction, and antibody neutralization of IL-6 abrogated the complement effect. By using both in vitro and in vivo approaches, we examined in more detail the mechanism and physiologic implication of complement/TLR4 interaction on Th17-cell differentiation. We found that the complement effect required C5a receptor, was evident at physiologically relevant levels of C5a, and could be demonstrated in cultured peritoneal macrophages as well as in the setting of antigen immunization. Importantly, despite an inhibitory effect of complement on IL-23 production, complement-promoted Th17 cells were functionally competent in causing autoimmunity in an adoptive transfer model of experimental autoimmune encephalomyelitis. Collectively, these data establish a link between complement/TLR interaction and Th17-cell differentiation and provide new insight into the mechanism of action of complement in autoimmunity.


Asunto(s)
Activación de Complemento , Inflamación/inmunología , Interleucina-6/fisiología , Linfocitos T Colaboradores-Inductores/citología , Receptores Toll-Like/fisiología , Traslado Adoptivo , Animales , Antígenos CD55/genética , Células Cultivadas/citología , Células Cultivadas/metabolismo , Complemento C5a/fisiología , Complemento C5a/uso terapéutico , Citocinas/sangre , Citocinas/fisiología , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/inmunología , Inmunidad Innata , Inmunización , Inflamación/inducido químicamente , Interleucina-6/biosíntesis , Interleucina-6/sangre , Interleucina-6/deficiencia , Interleucina-6/genética , Linfopoyesis , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/fisiología , Proteínas Recombinantes/uso terapéutico , Organismos Libres de Patógenos Específicos , Linfocitos T Colaboradores-Inductores/clasificación , Linfocitos T Colaboradores-Inductores/inmunología , Receptor Toll-Like 4/fisiología
16.
J Immunol ; 183(5): 2921-31, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19648268

RESUMEN

It is known that C3 is required for optimal expansion of T cells during acute viral infections. However, it is not yet determined whether T cell responses to intracellular bacterial infections require C3. Therefore, we have investigated the requirement for C3 to elicit potent T cell responses to Listeria monocytogenes (LM). We show that expansion of Ag-specific CD8 and CD4 T cells during a primary response to LM was markedly reduced in the absence of C3 activity. Further studies indicated that, unlike in an influenza virus infection, the regulation of LM-specific T cell responses by C3 might not involve the downstream effector C5a. Moreover, reduced T cell responses to LM was not linked to defective maturation of dendritic cells or developmental anomalies in the peripheral T cell compartment of C3-deficient mice. Experiments involving adoptive transfer of C3-deficient CD8 T cells into the C3-sufficient environment of wild-type mice showed that these T cells do not have intrinsic proliferative defects, and a paracrine source of C3 will suffice for clonal expansion of CD8 T cells in vivo. However, stimulation of purified C3-deficient CD8 T cells by plastic-immobilized anti-CD3 showed that C3 promotes T cell proliferation directly, independent of its effects on APC. On the basis of these findings, we propose that diminished T cell responses to LM in C3-deficient mice might be at least in part due to lack of direct effects of C3 on T cells. These studies have furthered our understanding of C3-mediated regulation of T cell immunity to intracellular pathogens.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Proliferación Celular , Complemento C3/fisiología , Listeriosis/inmunología , Listeriosis/patología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Complemento C3/deficiencia , Complemento C3/genética , Listeria monocytogenes/inmunología , Listeriosis/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/fisiología
17.
J Am Soc Nephrol ; 21(8): 1344-53, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20651167

RESUMEN

Interaction between C5a, a product of complement activation, and its receptor (C5aR) upregulates antigen-specific T cell responses by modulating the activation of antigen-presenting cells and T cells. Whether this C5a-C5aR interaction contributes to the immune responses that promote renal allograft rejection is unknown. Here, we found that deficiency of C5aR in both graft and recipient reduced allospecific T cell responses and prolonged renal allograft survival. In addition, lack of C5aR impaired the function of donor and recipient antigen-presenting cells and inhibited the response of recipient T cells to allostimulation. Furthermore, deficiency of C5aR in both graft and recipient reduced early inflammation in the grafts, with less cellular infiltration around the vessels and fewer F4/80 positive cells in the peritubular interstitium. These data demonstrate that C5aR is critical for a full adaptive immune response and mediates renal allograft rejection. Engagement of C5aR on dendritic cells and T cells modulates their function, enhancing allospecific T cell responses that lead to allograft rejection. Targeting C5a signaling may have therapeutic potential for T cell-mediated graft rejection.


Asunto(s)
Supervivencia de Injerto , Trasplante de Riñón , Receptor de Anafilatoxina C5a/deficiencia , Animales , Supervivencia de Injerto/inmunología , Trasplante de Riñón/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Factores de Tiempo
18.
Sci Immunol ; 6(66): eabf2489, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34932384

RESUMEN

While serum-circulating complement destroys invading pathogens, intracellularly active complement, termed the "complosome," functions as a vital orchestrator of cell-metabolic events underlying T cell effector responses. Whether intracellular complement is also nonredundant for the activity of myeloid immune cells is currently unknown. Here, we show that monocytes and macrophages constitutively express complement component (C) 5 and generate autocrine C5a via formation of an intracellular C5 convertase. Cholesterol crystal sensing by macrophages induced C5aR1 signaling on mitochondrial membranes, which shifted ATP production via reverse electron chain flux toward reactive oxygen species generation and anaerobic glycolysis to favor IL-1ß production, both at the transcriptional level and processing of pro­IL-1ß. Consequently, atherosclerosis-prone mice lacking macrophage-specific C5ar1 had ameliorated cardiovascular disease on a high-cholesterol diet. Conversely, inflammatory gene signatures and IL-1ß produced by cells in unstable atherosclerotic plaques of patients were normalized by a specific cell-permeable C5aR1 antagonist. Deficiency of the macrophage cell-autonomous C5 system also protected mice from crystal nephropathy mediated by folic acid. These data demonstrate the unexpected intracellular formation of a C5 convertase and identify C5aR1 as a direct modulator of mitochondrial function and inflammatory output from myeloid cells. Together, these findings suggest that the complosome is a contributor to the biologic processes underlying sterile inflammation and indicate that targeting this system could be beneficial in macrophage-dependent diseases, such as atherosclerosis.


Asunto(s)
Inflamación/inmunología , Interleucina-1beta/biosíntesis , Macrófagos/inmunología , Receptor de Anafilatoxina C5a/inmunología , Animales , Línea Celular , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia
19.
Nat Commun ; 12(1): 3352, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099640

RESUMEN

Platelets contribute to the regulation of tissue neovascularization, although the specific factors underlying this function are unknown. Here, we identified the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) on platelets as a negative regulatory mechanism of vessel formation. We showed that platelets expressing C5aR1 exert an inhibitory effect on endothelial cell functions such as migration and 2D and 3D tube formation. Growth factor- and hypoxia-driven vascularization was markedly increased in C5ar1-/- mice. Platelet-specific deletion of C5aR1 resulted in a proangiogenic phenotype with increased collateralization, capillarization and improved pericyte coverage. Mechanistically, we found that C5a induced preferential release of CXC chemokine ligand 4 (CXCL4, PF4) from platelets as an important antiangiogenic paracrine effector molecule. Interfering with the C5aR1-CXCL4 axis reversed the antiangiogenic effect of platelets both in vitro and in vivo.In conclusion, we identified a mechanism for the control of tissue neovascularization through C5a/C5aR1 axis activation in platelets and subsequent induction of the antiangiogenic factor CXCL4.


Asunto(s)
Plaquetas/metabolismo , Factor Plaquetario 4/metabolismo , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Inductores de la Angiogénesis , Animales , Activación de Complemento , Complemento C5a , Inflamación , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptores CXCR3/genética , Transducción de Señal
20.
Sci Rep ; 11(1): 2158, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33495506

RESUMEN

Singular blockade of C5a in experimental models of sepsis is known to confer protection by rescuing lethality and decreasing pro-inflammatory responses. However, the role of inhibiting C5a has not been evaluated in the context of sterile systemic inflammatory responses, like polytrauma and hemorrhagic shock (PT + HS). In our presented study, a novel and highly specific C5a L-aptamer, NoxD21, was used to block C5a activity in an experimental murine model of PT + HS. The aim of the study was to assess early modulation of inflammatory responses and lung damage 4 h after PT + HS induction. NoxD21-treated PT + HS mice displayed greater polymorphonuclear cell recruitment in the lung, increased pro-inflammatory cytokine levels in the bronchoalveolar lavage fluids (BALF) and reduced myeloperoxidase levels within the lung tissue. An in vitro model of the alveolar-capillary barrier was established to confirm these in vivo observations. Treatment with a polytrauma cocktail induced barrier damage only after 16 h, and NoxD21 treatment in vitro did not rescue this effect. Furthermore, to test the exact role of both the cognate receptors of C5a (C5aR1 and C5aR2), experimental PT + HS was induced in C5aR1 knockout (C5aR1 KO) and C5aR2 KO mice. Following 4 h of PT + HS, C5aR2 KO mice had significantly reduced IL-6 and IL-17 levels in the BALF without significant lung damage, and both, C5aR1 KO and C5aR2 KO PT + HS animals displayed reduced MPO levels within the lungs. In conclusion, the C5aR2 could be a putative driver of early local inflammatory responses in the lung after PT + HS.


Asunto(s)
Complemento C5a/metabolismo , Inflamación/patología , Pulmón/patología , Traumatismo Múltiple/complicaciones , Traumatismo Múltiple/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Choque Hemorrágico/complicaciones , Choque Hemorrágico/metabolismo , Adulto , Animales , Aptámeros de Péptidos/farmacología , Biomarcadores/metabolismo , Líquido del Lavado Bronquioalveolar , Capilares/patología , Línea Celular , Quimiotaxis/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Alveolos Pulmonares/patología , Receptor de Anafilatoxina C5a/deficiencia
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda