Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Blood ; 128(6): 862-5, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27297794

RESUMEN

The activating platelet collagen receptor glycoprotein VI (GPVI) is a promising antithrombotic target because of its central role in arterial thrombosis and its minor relevance for normal hemostasis. The receptor can be specifically targeted by antibodies and irreversibly downregulated in circulating platelets in vivo, resulting in long-term antithrombotic protection in mice. This GPVI immunodepletion predominantly occurs through ectodomain shedding, which is accompanied by a transient drop in peripheral platelet counts. Mechanistic studies on this targeted GPVI loss have been hampered because it cannot be reproduced in isolated platelets in vitro. Here we show that both the transient thrombocytopenia and GPVI ectodomain shedding depend on the Fc portion of the anti-GPVI antibody and its interaction with the inhibitory Fcγ receptor (FcγR)IIB. In wild-type, but not Fcgr2b(-/-) mice, anti-GPVI-opsonized platelets became transiently trapped in the liver followed by the appearance of the soluble GPVI ectodomain in the plasma. Depletion of Kupffer cells neither affected anti-GPVI-induced platelet accumulation nor GPVI shedding, demonstrating that the other major FcγRIIB-expressing cell type, liver sinusoidal endothelial cells, is required for both processes to occur. These results reveal a novel and unexpected function of hepatic FcγRIIB in the targeted downregulation of GPVI in vivo.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Plaquetas/inmunología , Hígado/inmunología , Glicoproteínas de Membrana Plaquetaria/inmunología , Receptores de IgG/inmunología , Animales , Ratones , Ratones Noqueados , Glicoproteínas de Membrana Plaquetaria/genética , Receptores de IgG/genética , Trombocitopenia/genética , Trombocitopenia/inmunología
2.
Eur J Immunol ; 45(7): 2143-53, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25912155

RESUMEN

Stromal interaction molecule 1 (STIM1)-dependent store operated calcium-entry (SOCE) through Orai1-mediated calcium (Ca(2+) ) influx is considered a major pathway of Ca(2+) signaling, serving T-cell, mast cell, and platelet responses. Here, we show that Orai1 is critical for neutrophil function. Orai1-deficient neutrophils present defects in fMLP and complement C5a-induced Ca(2+) influx and migration, although they respond normally to another chemoattractant, CXCL2. Up until now, no specific contribution of Orai1 independent from STIM1 or SOCE has been recognized in immune cells. Here, we observe that Orai1-deficient neutrophils exhibit normal STIM1-dependent SOCE and STIM1-deficient neutrophils respond to fMLP and C5a efficiently. Despite substantial cytokine production, Orai1(-/-) chimeric mice show impaired neutrophil recruitment in LPS-induced peritonitis. Moreover, Orai1 deficiency results in profoundly defective C5a-triggered neutrophil lung recruitment in hypersensitivity pneumonitis. Comparative evaluation of inflammation in Stim1(-/-) chimeras reveals a distinct pathogenic contribution of STIM1, including its involvement in IgG-induced C5a production. Our data establish Orai1 as key signal mediator of C5aR activation, contributing to inflammation by a STIM1-independent pathway of Ca(2+) -influx in neutrophils.


Asunto(s)
Canales de Calcio/inmunología , Complemento C5a/inmunología , Inflamación/inmunología , Infiltración Neutrófila/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína ORAI1 , Receptor de Anafilatoxina C5a/inmunología
3.
J Immunol ; 190(1): 324-33, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23225882

RESUMEN

Heterotrimeric G proteins of the Gα(i) family have been implicated in signaling pathways regulating cell migration in immune diseases. The Gα(i)-protein-coupled C5a receptor is a critical regulator of IgG FcR function in experimental models of immune complex (IC)-induced inflammation. By using mice deficient for Gα(i2) or Gα(i3), we show that Gα(i2) is necessary for neutrophil influx in skin and lung Arthus reactions and agonist-induced neutrophilia in the peritoneum, whereas Gα(i3) plays a less critical but variable role. Detailed analyses of the pulmonary IC-induced inflammatory response revealed several shared functions of Gα(i2) and Gα(i3), including mediating C5a anaphylatoxin receptor-induced activation of macrophages, involvement in alveolar production of chemokines, transition of neutrophils from bone marrow into blood, and modulation of CD11b and CD62L expression that account for neutrophil adhesion to endothelial cells. Interestingly, C5a-stimulated endothelial polymorphonuclear neutrophil transmigration, but not chemotaxis, is enhanced versus reduced in the absence of neutrophil Gα(i3) or Gα(i2), respectively, and knockdown of endothelial Gα(i2) caused decreased transmigration of wild-type neutrophils. These data demonstrate that Gα(i2) and Gα(i3) contribute to inflammation by redundant, overlapping, and Gα(i)-isoform-specific mechanisms, with Gα(i2) exhibiting unique functions in both neutrophils and endothelial cells that appear essential for polymorphonuclear neutrophil recruitment in IC disease.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2/fisiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Reacción de Arthus/genética , Reacción de Arthus/inmunología , Reacción de Arthus/patología , Adhesión Celular/genética , Adhesión Celular/inmunología , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Subunidad alfa de la Proteína de Unión al GTP Gi2/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Neutrófilos/inmunología , Neutrófilos/patología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
4.
J Immunol ; 189(2): 980-7, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22706085

RESUMEN

Various heterotrimeric G(i) proteins are considered to be involved in cell migration and effector function of immune cells. The underlying mechanisms, how they control the activation of myeloid effector cells, are not well understood. To elucidate isoform-redundant and -specific roles for Gα(i) proteins in these processes, we analyzed mice genetically deficient in Gα(i2) or Gα(i3). First, we show an altered distribution of tissue macrophages and blood monocytes in the absence of Gα(i2) but not Gα(i3). Gα(i2)-deficient but not wild-type or Gα(i3)-deficient mice exhibited reduced recruitment of macrophages in experimental models of thioglycollate-induced peritonitis and LPS-triggered lung injury. In contrast, genetic ablation of Gα(i2) had no effect on Gα(i)-dependent peritoneal cytokine production in vitro and the phagocytosis-promoting function of the Gα(i)-coupled C5a anaphylatoxin receptor by liver macrophages in vivo. Interestingly, actin rearrangement and CCL2- and C5a anaphylatoxin receptor-induced chemotaxis but not macrophage CCR2 and C5a anaphylatoxin receptor expression were reduced in the specific absence of Gα(i2). Furthermore, knockdown of Gα(i2) caused decreased cell migration and motility of RAW 264.7 cells, which was rescued by transfection of Gα(i2) but not Gα(i3). These results indicate that Gα(i2), albeit redundant to Gα(i3) in some macrophage activation processes, clearly exhibits a Gα(i) isoform-specific role in the regulation of macrophage migration.


Asunto(s)
Inhibición de Migración Celular/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Macrófagos/inmunología , Macrófagos/patología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Inhibición de Migración Celular/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/patología , Tioglicolatos/toxicidad
5.
J Clin Immunol ; 33(1): 288-96, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23007236

RESUMEN

PURPOSE: A poor antibody response of IgM and IgA antibodies upon vaccination with pneumococcal polysaccharides (PnPS) is discussed as independent risk factors for bronchiectasis in patients with antibody deficiency syndrome (ADS) receiving immunoglobulin replacement therapy. However, the kinetics of the specific IgM and IgA response to vaccination with multivalent pneumococcal polysaccharides requires a more detailed knowledge. In this study we aimed i) to develop a standardised multivalent PnPS-IgM and IgA-ELISA, and ii) to compare the sensitivity of the multivalent to the serotype specific antibody response, and iii) to determine the kinetics of the anti-PnPS IgM and IgA antibodies in healthy subjects. METHODS: We immunised n=20 healthy adults with a 23-valent PnPS vaccine (Pneumovax®). The kinetics of the 23-valent antibody response was assessed for 1 year with newly developed ELISAs for IgM and IgA isotypes, along with serotype specific responses. RESULTS: The IgA and IgM antibody response peaked at 2 and 3 weeks, respectively. IgM antibody levels remained at a plateau (above 80 % of peak response) for 3 months. After one year, specific antibody levels were still at about 30 % of the peak response. The 23-valent antibody response yielded significantly higher responder rates than assessment of single serotypes. CONCLUSION: Testing the IgM and IgA immune response to polysaccharide vaccination with a multivalent PnPS ELISA may be a feasible tool for assessment of the immune function in patient groups who receive IgG replacement therapy.


Asunto(s)
Cápsulas Bacterianas/inmunología , Inmunoglobulina A/biosíntesis , Inmunoglobulina M/biosíntesis , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/inmunología , Polisacáridos Bacterianos/inmunología , Adulto , Especificidad de Anticuerpos/inmunología , Cápsulas Bacterianas/metabolismo , Reacciones Cruzadas/inmunología , Humanos , Inmunoglobulina A/metabolismo , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/metabolismo , Persona de Mediana Edad , Vacunas Neumococicas/farmacocinética , Polisacáridos Bacterianos/administración & dosificación , Polisacáridos Bacterianos/farmacocinética , Adulto Joven
6.
J Immunol ; 187(6): 3218-29, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21844386

RESUMEN

Subepithelial immune complex deposition in glomerular disease causes local inflammation and proteinuria by podocyte disruption. A rat model of membranous nephropathy, the passive Heymann nephritis, suggests that Abs against specific podocyte Ags cause subepithelial deposit formation and podocyte foot process disruption. In this study, we present a mouse model in which a polyclonal sheep anti-mouse podocyte Ab caused subepithelial immune complex formation. Mice developed a nephrotic syndrome with severe edema, proteinuria, hypoalbuminemia, and elevated cholesterol and triglycerides. Development of proteinuria was biphasic: an initial protein loss was followed by a second massive increase of protein loss beginning at approximately day 10. By histology, podocytes were swollen. Electron microscopy revealed 60-80% podocyte foot process effacement and subepithelial deposits, but no disruption of the glomerular basement membrane. Nephrin and synaptopodin staining was severely disrupted, and podocyte number was reduced in anti-podocyte serum-treated mice, indicating severe podocyte damage. Immunohistochemistry detected the injected anti-podocyte Ab exclusively along the glomerular filtration barrier. Immunoelectron microscopy localized the Ab to podocyte foot processes and the glomerular basement membrane. Similarly, immunohistochemistry localized mouse IgG to the subepithelial space. The third complement component (C3) was detected in a linear staining pattern along the glomerular basement membrane and in the mesangial hinge region. However, C3-deficient mice were not protected from podocyte damage, indicating a complement-independent mechanism. Twenty proteins were identified as possible Ags to the sheep anti-podocyte serum by mass spectrometry. Together, these data establish a reproducible model of immune-mediated podocyte injury in mice with subepithelial immune complex formation.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Glomerulonefritis/inmunología , Síndrome Nefrótico/inmunología , Podocitos/inmunología , Animales , Complejo Antígeno-Anticuerpo/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/patología , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Ensayo de Inmunoadsorción Enzimática , Glomerulonefritis/patología , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Síndrome Nefrótico/patología , Podocitos/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Circulation ; 122(10): 1026-36, 2010 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-20733098

RESUMEN

BACKGROUND: Receptor binding of complement C5a leads to proinflammatory activation of many cell types, but the role of receptor-mediated action during arterial remodeling after injury has not been studied. In the present study, we examined the contribution of the C5a receptor (C5aR) to neointima formation in apolipoprotein E-deficient mice employing a C5aR antagonist (C5aRA) and a C5aR-blocking monoclonal antibody. METHODS AND RESULTS: Mice fed an atherogenic diet were subjected to wire-induced endothelial denudation of the carotid artery and treated with C5aRA and anti-C5aR-blocking monoclonal antibody or vehicle control. Compared with controls, neointima formation was significantly reduced in mice receiving C5aRA or anti-C5aR-blocking monoclonal antibody for 1 week but not for 3 weeks, attributable to an increased content of vascular smooth muscle cells, whereas a marked decrease in monocyte and neutrophil content was associated with reduced vascular cell adhesion molecule-1. As assessed by immunohistochemistry, reverse transcription polymerase chain reaction, and flow cytometry, C5aR was expressed in lesional and cultured vascular smooth muscle cells, upregulated by injury or tumor necrosis factor-alpha, and reduced by C5aRA. Plasma levels and neointimal plasminogen activator inhibitor-1 peaked 1 week after injury and were downregulated in C5aRA-treated mice. In vitro, C5a induced plasminogen activator inhibitor-1 expression in endothelial cells and vascular smooth muscle cells in a C5aRA-dependent manner, possibly accounting for higher vascular smooth muscle cell immigration. CONCLUSIONS: One-week treatment with C5aRA or anti-C5aR-blocking monoclonal antibody limited neointimal hyperplasia and inflammatory cell content and was associated with reduced vascular cell adhesion molecule-1 expression. However, treatment for 3 weeks failed to reduce but rather stabilized plaques, likely by reducing vascular plasminogen activator inhibitor-1 and increasing vascular smooth muscle cell migration.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Oligopéptidos/farmacología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Animales , Apolipoproteínas E/genética , Aterosclerosis/patología , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Complemento C5a/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Leucocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Serpina E2 , Serpinas/metabolismo , Túnica Íntima/efectos de los fármacos , Túnica Íntima/inmunología , Túnica Íntima/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Eur J Immunol ; 40(5): 1284-95, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20148421

RESUMEN

During asthma, lung DC capture and process antigens to initiate and maintain allergic Th2 cell responses to inhaled allergens. The aim of the study was to investigate whether allergen-specific IgG, generated during sensitization, can potentiate the acute airway inflammation through Fcgamma receptor (FcgammaR)-mediated antigen uptake and enhance antigen presentation resulting in augmented T-cell proliferation. We examined the impact of antigen presentation and T-cell stimulation on allergic airway hyperresponsiveness and inflammation using transgenic and gene-deficient mice. Both airway inflammation and eosinophilia in bronchoalveolar lavage fluid were markedly reduced in sensitized and challenged FcgammaR-deficient mice. Lung DC of WT, but not FcgammaR-deficient mice, induced increased antigen-specific CD4+ T-cell proliferation when pulsed with anti-OVA IgG immune complexes. Intranasal application of anti-OVA IgG immune complexes resulted in enhanced airway inflammation, eosinophilia and Th2 cytokine release, mediated through enhanced antigen-specific T-cell proliferation in vivo. Finally, antigen-specific IgG in the serum of sensitized mice led to a significant increase of antigen-specific CD4+ T-cell proliferation induced by WT, but not FcgammaR-deficient, lung DC. We conclude that FcgammaR-mediated enhanced antigen presentation and T-cell stimulation by lung DC has a significant impact on inflammatory responses following allergen challenge in asthma.


Asunto(s)
Presentación de Antígeno , Hiperreactividad Bronquial/inmunología , Células Dendríticas/inmunología , Inmunoglobulina G/inmunología , Pulmón/inmunología , Receptores de IgG/inmunología , Traslado Adoptivo , Animales , Asma/inmunología , Femenino , Inflamación , Pulmón/patología , Activación de Linfocitos , Linfocinas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética
9.
Blood ; 113(5): 1097-104, 2009 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18941110

RESUMEN

Fcgamma receptors (FcgammaRs) on mononuclear phagocytes trigger autoantibody and immune complex-induced diseases through coupling the self-reactive immunoglobulin G (IgG) response to innate effector pathways, such as phagocytosis, and the recruitment of inflammatory cells. FcRgamma-based activation is critical in the pathogenesis of these diseases, although the contribution of FcgammaR-mediated calcium signaling in autoimmune injury is unclear. Here we show that macrophages lacking the endoplasmic reticulum-resident calcium sensor, STIM1, cannot activate FcgammaR-induced Ca(2+) entry and phagocytosis. As a direct consequence, STIM1 deficiency results in resistance to experimental immune thrombocytopenia and anaphylaxis, autoimmune hemolytic anemia, and acute pneumonitis. These results establish STIM1 as a novel and essential component of FcgammaR activation and also indicate that inhibition of STIM1-dependent signaling might become a new strategy to prevent or treat IgG-dependent immunologic diseases.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Enfermedades Autoinmunes/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , Fagocitosis/inmunología , Receptores de IgG/inmunología , Animales , Complejo Antígeno-Anticuerpo/genética , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Calcio/inmunología , Canales de Calcio , Femenino , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Macrófagos/patología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Fagocitosis/genética , Receptores de IgG/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Molécula de Interacción Estromal 1
10.
Eur J Immunol ; 39(12): 3343-56, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19795417

RESUMEN

FcgammaRIV is a relatively new IgG Fc receptor (FcgammaR) that is reported to contribute to the pathogenesis of autoimmune diseases, although its specific role in relation to FcgammaRIII, complement and IgG2 subclasses remains uncertain. Here we define FcgammaRIV on macrophages as a receptor for soluble IgG2a/b complexes but not for cellular bound IgG2a and show that simultaneous activation of FcgammaRIV and FcgammaRIII is critical to mediate certain type II/III autoimmune responses. FcgammaRIII-deficient mice display compensatory enhanced FcgammaRIV expression, are protected from lung inflammation after deposition of IgG complexes, and show reduced sensitivity to IgG2a/b-mediated hemolytic anemia, indicating that increased FcgammaRIV alone is not sufficient to trigger these diseases in the absence of FcgammaRIII. Importantly, however, blockade of FcgammaRIV is also effective in inhibiting phagocytosis and cytokine production in IgG2b-induced anemia and acute lung injury, processes that display a further dependence on C5a anaphylatoxin receptor. Using gene deletion and functional inhibition studies, we found that FcgammaRIII and FcgammaRIV are each essential to trigger an FcRgamma-linker for activation of T-cell-dependent signal that drives C5a production in the Arthus reaction. Together, the results demonstrate a combined requirement for FcgammaRIII and FcgammaRIV in autoimmune injury, and identify the linker for activation of T cells adaptor as an integral component of linked FcgammaR and C5a anaphylatoxin receptor activation to generate inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Autoinmunidad/inmunología , Proteínas de la Membrana/inmunología , Fosfoproteínas/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de IgG/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anemia Hemolítica Autoinmune/genética , Anemia Hemolítica Autoinmune/inmunología , Anemia Hemolítica Autoinmune/metabolismo , Animales , Femenino , Citometría de Flujo , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos , Ratones Noqueados , Microscopía Confocal , Fagocitosis/inmunología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neumonía/genética , Neumonía/inmunología , Neumonía/metabolismo , Unión Proteica , Receptor de Anafilatoxina C5a/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo
11.
J Clin Invest ; 116(2): 512-20, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16453025

RESUMEN

Complement C5a, a potent anaphylatoxin, is a candidate target molecule for the treatment of inflammatory diseases, such as myocardial ischemia/reperfusion injury, RA, and the antiphospholipid syndrome. In contrast, up until now, no specific contribution of C5a and its receptor, C5aR, was recognized in diseases of antibody-dependent type II autoimmunity. Here we identify C5a as a novel key mediator of autoimmune hemolytic anemia (AIHA) and show that mice lacking C5aR are partially resistant to this IgG autoantibody-induced disease model. Upon administration of anti-erythrocyte antibodies, upregulation of activating Fcgamma receptors (FcgammaRs) on Kupffer cells, as observed in WT mice, was absent in C5aR-deficient mice, and FcgammaR-mediated in vivo erythrophagocytosis was impaired. Surprisingly, in mice deficient in FcgammaRI and FcgammaRIII, anti-erythrocyte antibody-induced C5 and C5a production was abolished, demonstrating the existence of a previously unidentified FcgammaR-mediated C5a-generating pathway. These results show that the development of a full-blown antibody-dependent autoimmune disease requires C5a--produced by and acting on FcgammaR--and may suggest therapeutic benefits of C5 and/or C5a/C5aR blockade in AIHA and other diseases closely related to type II autoimmune injury.


Asunto(s)
Anemia Hemolítica Autoinmune/inmunología , Autoinmunidad/fisiología , Complemento C5a/inmunología , Inmunoglobulina G/inmunología , Receptor de Anafilatoxina C5a/inmunología , Anemia Hemolítica Autoinmune/genética , Animales , Anticuerpos Monoclonales/inmunología , Complemento C5a/genética , Eritrocitos/metabolismo , Macrófagos del Hígado/citología , Macrófagos del Hígado/metabolismo , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Fagocitosis/fisiología , Receptor de Anafilatoxina C5a/genética , Receptores de IgG , Tasa de Supervivencia
12.
J Clin Invest ; 110(12): 1823-30, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12488432

RESUMEN

IgG Fc receptors (FcgammaRs, especially FcgammaRIII) and complement (in particular, C5a anaphylatoxin) are critical effectors of the acute inflammatory response to immune complexes (ICs). However, it is unknown whether and how these two key components can interact with each other in vivo. We use here a mouse model of the acute pulmonary IC hypersensitivity reaction to analyze their potential interaction. FcgammaRIII and C5aR are coexpressed on alveolar macrophages (AMs), and both FcgammaRIII and C5aR mutant mice display impaired immune responses. We find that recombinant human C5a (rhC5a) can control inverse expression of various FcgammaRs, and costimulation of ICs with rhC5a results in strong enhancement of FcgammaRIII-triggered cellular activation in vitro and in vivo. Moreover, we show here that early IC-induced bioactive C5a, and its interaction with C5aR, causes induction of activating FcgammaRIII and suppression of inhibitory FcgammaRII on AMs that appears crucial for efficient cytokine production and neutrophil recruitment in lung pathology. Therefore, C5a, which is a potent chemoattractant, has a broader critical function in regulating the inhibitory/activating FcgammaRII/III receptor pair to connect complement and FcgammaR effector pathways in immune inflammation.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Complemento C5a/inmunología , Enfermedades del Complejo Inmune/inmunología , Enfermedades Pulmonares/inmunología , Receptores de IgG/inmunología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Separación Celular , Células Cultivadas , Quimiotaxis/fisiología , Complemento C5a/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Enfermedades del Complejo Inmune/metabolismo , Enfermedades Pulmonares/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/metabolismo , Receptor de Anafilatoxina C5a , Receptores de Complemento/metabolismo , Receptores de IgG/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo
13.
Immunol Lett ; 100(1): 56-67, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16125792

RESUMEN

Genetic studies in mice indicate a crucial role for Fc receptors (FcR) in antibody-mediated autoimmune diseases. Like other immune regulatory receptor pairs, the FcR system is constituted by activating and inhibitory receptors that bind the same ligand, the Fc portion of Ig. Analyses of animal models have shown that the inhibitory Fc receptor, FcgammaRIIB can suppress antibody-mediated autoimmunity, whereas activating-type FcR, such as FcgammaRIII promote disease development. This review summarizes recent advances of FcR, as obtained from gene deletion studies in mice, and highlights the importance of factors that interact with FcR in autoimmunity. There is emerging evidence for an indispensable role of the complement component C5a in the regulation of FcR and the sensing of FcR-dependent effector cell responses. On the other hand, FcR might be alternatives to serum complement in the generation of C5a at sites of inflammation. Thus, FcR and complement interact with each other at the level of C5a by linking regulatory events with effector cell activities in autoimmunity. This connecting pathway is now proposed to be a promising new therapeutic target for the treatment of inflammation and autoimmune disease in both mice and humans.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos CD/inmunología , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Activación de Complemento/inmunología , Complemento C5a/inmunología , Receptores de IgG/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Antígenos CD/genética , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/terapia , Autoinmunidad/genética , Activación de Complemento/genética , Complemento C5a/genética , Eliminación de Gen , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/terapia , Ratones , Receptores de IgG/genética
14.
J Exp Med ; 212(8): 1255-66, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26169941

RESUMEN

Spontaneous organ hemorrhage is the major complication in thrombocytopenia with a potential fatal outcome. However, the exact mechanisms regulating vascular integrity are still unknown. Here, we demonstrate that neutrophils recruited to inflammatory sites are the cellular culprits inducing thrombocytopenic tissue hemorrhage. Exposure of thrombocytopenic mice to UVB light provokes cutaneous petechial bleeding. This phenomenon is also observed in immune-thrombocytopenic patients when tested for UVB tolerance. Mechanistically, we show, analyzing several inflammatory models, that it is neutrophil diapedesis through the endothelial barrier that is responsible for the bleeding defect. First, bleeding is triggered by neutrophil-mediated mechanisms, which act downstream of capturing, adhesion, and crawling on the blood vessel wall and require Gαi signaling in neutrophils. Second, mutating Y731 in the cytoplasmic tail of VE-cadherin, known to selectively affect leukocyte diapedesis, but not the induction of vascular permeability, attenuates bleeding. Third, and in line with this, simply destabilizing endothelial junctions by histamine did not trigger bleeding. We conclude that specifically targeting neutrophil diapedesis through the endothelial barrier may represent a new therapeutic avenue to prevent fatal bleeding in immune-thrombocytopenic patients.


Asunto(s)
Antígenos CD/genética , Cadherinas/genética , Dermatitis por Contacto/patología , Hemorragia/fisiopatología , Neutrófilos/fisiología , Trombocitopenia/fisiopatología , Migración Transendotelial y Transepitelial/fisiología , Vasculitis/patología , Análisis de Varianza , Animales , Dermatitis por Contacto/etiología , Dermatitis por Contacto/inmunología , Citometría de Flujo , Hemorragia/etiología , Hemorragia/prevención & control , Técnicas Histológicas , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación Missense/genética , Migración Transendotelial y Transepitelial/efectos de los fármacos , Rayos Ultravioleta , Vasculitis/etiología , Vasculitis/inmunología
15.
Arch Immunol Ther Exp (Warsz) ; 51(6): 399-406, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14692661

RESUMEN

Fc-receptors for IgG (FcyRs) are critically involved at multiple stages of an immune response, ranging from antigen presentation and regulation of antibody production to the end-stage effector mechanisms of inflammation. IgG autoantibodies that are detectable in the majority of autoimmune diseases are ligands for FcgammaRs. The three classes, FcgammaRI, FcgammaRII, and FcgammaRIII, vary in their antibody affinity, cellular expression and in vivo function. We review the current knowledge on the regulation and diverse functions of the distinct FcgammaRs and describe the evidence of their important immunoregulatory roles in autoimmunity based on recent work in animal models.


Asunto(s)
Autoinmunidad/fisiología , Isoformas de Proteínas , Receptores de IgG , Animales , Presentación de Antígeno , Proteínas del Sistema Complemento/inmunología , Eliminación de Gen , Humanos , Sistema Inmunológico/fisiología , Inflamación/inmunología , Polimorfismo Genético , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología
16.
Immunobiology ; 218(9): 1131-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23642836

RESUMEN

Tissue loss after myocardial ischemia with reperfusion (MI/R) is in part conveyed by neutrophil recruitment to post-ischemic myocardium. Strategies to prevent reperfusion injury would help to limit myocardial damage. The receptor for activated complement factor 5 (C5aR) plays a prominent role in inflammation. We examine the effects of C5aR-deficiency on reperfusion injury after MI/R. C5aR(-/-)-mice and their C57BL/6- (WT) littermates underwent transient myocardial ischemia followed by different time points of reperfusion. Infarct size and leukocyte infiltration were determined. Expression of C5aR, inflammatory cytokines and adhesion molecules were analyzed by real-time RT-PCR. Leukocyte-endothelial interactions were assessed by low-shear adhesion- and transmigration-assays in vitro. Myocardial C5aR mRNA expression was 2.8-fold increased by ischemia. Infarct size per area-at-risk and leukocyte recruitment into infarctions were reduced in C5aR(-/-)-compared to WT-mice as well as in WT mice treated with the C5aR-antagonist JPE1375. IL-6, IL-1ß, ICAM-1 and VCAM-1 expression were not different, while TNFα expression was reduced in C5aR(-/-)-mice after MI/R. In vitro, C5aR on leukocytes is required for effective transendothelial migration but not adhesion. Expression of MMP9 and JAM-A, molecules that are involved in leukocyte transmigration, were reduced in C5aR(-/-) mice in vivo. Genetic C5aR deficiency blunts the inflammatory response in murine MI/R resulting in reduced inflammatory cell recruitment, which is due to a C5aR-dependent effect on leukocyte transmigration across inflamed endothelium into the ischemic myocardium. This effect could be related to MMP9- and JAM-A expression in response to ischemia and reperfusion.


Asunto(s)
Infarto del Miocardio/inmunología , Daño por Reperfusión Miocárdica/complicaciones , Miocardio/metabolismo , Neutrófilos/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/genética , Células Cultivadas , Regulación de la Expresión Génica/genética , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/complicaciones , Daño por Reperfusión Miocárdica/etiología , Miocardio/inmunología , Miocardio/patología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Migración Transendotelial y Transepitelial/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Vaccine ; 28(3): 707-13, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19887136

RESUMEN

Vaccination against Pseudomonas aeruginosa is a desirable, yet challenging strategy for prevention of airway infection in patients with cystic fibrosis. We compared the formation of antibodies in lower airways induced by systemic and mucosal vaccination strategies. We immunised 48 volunteers in six vaccination groups with either a systemic, a nasal, or four newly constructed oral live vaccines based on attenuated live Salmonella (strains CVD908 and Ty21a), followed by a systemic booster vaccination. All vaccines were based on a recombinant fusion protein of the highly conserved P. aeruginosa outer membrane proteins OprF and OprI as antigen. While systemic and mucosal vaccines induced a comparable rise of serum antibody titers, a significant rise of IgA and IgG antibodies in the lower airways was noted only after nasal and oral vaccinations. We conclude that nasal and oral OprF-OprI vaccines are promising candidates for development of antipseudomonal immunisation through inducing a specific antibody response in the lung.


Asunto(s)
Infecciones por Pseudomonas/prevención & control , Vacunas contra la Infección por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Administración Intranasal , Administración Oral , Adulto , Anticuerpos Antibacterianos/análisis , Anticuerpos Antibacterianos/sangre , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Femenino , Vectores Genéticos , Experimentación Humana , Humanos , Inmunización Secundaria/métodos , Inmunoglobulina A/análisis , Inmunoglobulina G/análisis , Masculino , Persona de Mediana Edad , Vacunas contra la Infección por Pseudomonas/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Sistema Respiratorio/inmunología , Salmonella/genética , Vacunación/métodos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Adulto Joven
18.
J Innate Immun ; 2(1): 87-92, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20375626

RESUMEN

The complement system is a key component of the innate immune response. Here, we have examined the role of complement anaphylatoxin C5a in an experimental model of Staphylococcus aureus bacteremia. Our data provide compelling evidence for a protective role of C5a during staphylococcal bloodstream infection.


Asunto(s)
Bacteriemia/inmunología , Complemento C5a/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Complemento C5a/genética , Femenino , Inmunidad Innata/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA
19.
Inflamm Bowel Dis ; 15(12): 1812-23, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19714742

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is a critical public health issue; more and more people are affected, but treatment options remain limited. Complement activation and the anaphylatoxin C5a have been shown to play a role in IBD. In this study, mouse models of acute and chronic dextran sulfate-induced colitis were used to further elucidate the impact of C5a and its receptor (C5aR) on disease development. METHODS: In C57BL/6J wildtype and C5aR(-/-) mice the extent of complement activation, changes in weight, and water/food consumption were determined. Disease severity was evaluated via a clinical score, histology, cytokine- and myeloperoxidase-determination as well as real-time reverse-transcriptase polymerase chain reaction (RT-PCR) for expression of anaphylatoxin receptors and inflammatory mediators. RESULTS: C5aR(-/-) mice showed milder disease symptoms, less histological damage, and a lower expression of inflammatory mediators in acute colitis, a setting where complement was activated. In chronic colitis the knockout mice exhibited aggravated weight loss, a higher degree of histological damage and granulocyte infiltration. Intriguingly, increases in C3a-receptor and C5L2 mRNA were dependent on C5aR. Compared to wildtype mice, C5aR(-/-) animals displayed smaller lymph nodes in acute colitis, but extensive swelling and diminished IL-4 and IFN-γ responses in the chronic disease, demonstrating that C5aR modifies T-helper cell polarization. CONCLUSIONS: C5aR exerts detrimental functions in acute colitis, strongly supporting the idea that a C5aR-antagonist might be useful for IBD treatment. However, since the absence of C5aR was no longer protective and in some regards disadvantageous in chronic IBD, future studies should address the efficacy and the possible side effects of a sustained antagonist treatment.


Asunto(s)
Enfermedades Inflamatorias del Intestino/inmunología , Receptor de Anafilatoxina C5a/inmunología , Enfermedad Aguda , Animales , Polaridad Celular/inmunología , Activación de Complemento/inmunología , Sulfato de Dextran/inmunología , Sulfato de Dextran/toxicidad , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/patología , Interferón gamma/inmunología , Interleucina-4/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/genética , Índice de Severidad de la Enfermedad , Linfocitos T Colaboradores-Inductores/inmunología
20.
J Biol Chem ; 283(48): 33296-303, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18786920

RESUMEN

Fcgamma receptors (FcgammaR) and the C5a receptor (C5aR) are key effectors of the acute inflammatory response to IgG immune complexes (IC). Their coordinated activation is critical in IC-induced diseases, although the significance of combined signaling by these two different receptor classes in tissue injury is unclear. Here we used the mouse model of the passive reverse lung Arthus reaction to define their requirements for distinct phosphoinositide 3-kinase (PI3K) activities in vivo. We show that genetic deletion of class IB PI3Kgamma abrogates C5aR signaling that is crucial for FcgammaR-mediated activation of lung macrophages. Thus, in PI3Kgamma(-/-) mice, IgG IC-induced FcgammaR regulation, cytokine release, and neutrophil recruitment were blunted. Notably, however, C5a production occurred normally in PI3Kgamma(-/-) mice but was impaired in PI3Kdelta(-/-) mice. Consequently, class IA PI3Kdelta deficiency caused resistance to acute IC lung injury. These results demonstrate that PI3Kgamma and PI3Kdelta coordinate the inflammatory effects of C5aR and FcgammaR and define PI3Kdelta as a novel and essential element of FcgammaR signaling in the generation of C5a in IC disease.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Reacción de Arthus/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Receptores de IgG/metabolismo , Animales , Complejo Antígeno-Anticuerpo/genética , Complejo Antígeno-Anticuerpo/inmunología , Reacción de Arthus/genética , Reacción de Arthus/inmunología , Fosfatidilinositol 3-Quinasa Clase I , Fosfatidilinositol 3-Quinasa Clase Ib , Modelos Animales de Enfermedad , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Isoenzimas/genética , Isoenzimas/inmunología , Isoenzimas/metabolismo , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA