Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 407
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
BMC Nephrol ; 23(1): 25, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35016642

RESUMO

BACKGROUND: The addition of sialic acid alters IgG from a pro-inflammatory state to an anti-inflammatory state. However, there is a lack of research on the changes of IgG sialylation in IgA nephropathy (IgAN). METHODS: This study included a total of 184 IgAN patients. The sialylated IgG (SA-IgG), IgG-galactose-deficient IgA1 complex (IgG-Gd-IgA1-IC), IL-6, TNF-α, and TGF-ß were detected using commercial ELISA kits. SA-IgG, non-sialylated IgG (NSA-IgG), sialylated IgG-IgA1 complex (SA-IgG-IgA1), and non-sialylated IgG-IgA1 complex (NSA-IgG-IgA1) were purified from IgAN patients and healthy controls (HCs). RESULTS: The mean SA-IgG levels in plasma and B lymphocytes in IgAN patients were significantly higher than those of healthy controls. A positive correlation was found between SA-IgG levels in plasma and B lymphocytes. In vitro, the results showed that the release of IgG-Gd-IgA1-IC was significantly decreased in peripheral blood mononuclear cells (PBMCs) cultured with SA-IgG from both IgAN patients and healthy controls. The proliferation ability and the release of IL-6, TNF-α, and TGF-ß in human mesangial cells (HMCs) were measured after stimulating with SA-IgG-IgA1-IC and NSA-IgG-IgA1-IC. The mesangial cell proliferation levels induced by NSA-IgG-IgA1-IC derived from IgAN patients were significantly higher than those caused by SA-IgG-IgA1-IC derived from IgAN patients and healthy controls. Compared with NSA-IgG-IgA1 from healthy controls, IgAN-NSA-IgG-IgA1 could significantly upregulate the expression of IL-6 and TNF-α in mesangial cells. The data showed that there weren't any significant differences in the levels of IL-6, TNF-α, and TGF-ß when treated with IgAN-SA-IgG-IgA1 and HC-NSA-IgG-IgA1. CONCLUSIONS: The present study demonstrated that the sialylation of IgG increased in patients with IgA nephropathy. It exerted an inhibitory effect on the formation of Gd-IgA1-containing immune complexes in PBMCs and the proliferation and inflammation activation in mesangial cells.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Glomerulonefrite por IGA/imunologia , Glomerulonefrite por IGA/metabolismo , Imunoglobulina A/imunologia , Imunoglobulina G/metabolismo , Células Mesangiais , Ácido N-Acetilneuramínico/metabolismo , Feminino , Galactose , Humanos , Adulto Jovem
2.
Eur J Clin Invest ; 48 Suppl 2: e12948, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29734519

RESUMO

Neutrophils are amongst the first cells to be recruited to sites of infection or trauma. Neutrophil functional responsiveness is tightly regulated by many agents including immune complexes. These immune cells can generate reactive oxygen species and degranulate to release abundant cytotoxic products, making them efficient at killing invading microorganisms. If neutrophil function is dysregulated, however, these cells have the potential to cause unwanted host tissue damage as exemplified by pathological and chronic inflammatory conditions. In physiological inflammation, once the initial insult has efficiently been dealt with, neutrophils are thought to leave the tissues or undergo programmed cells death, especially apoptosis. Apoptotic neutrophils are then rapidly removed by other phagocytes, primarily macrophages, by mechanisms that do not elicit a pro-inflammatory response. In this review, we discuss the interesting observations and consequences that immune complexes have on neutrophil cell death processes such as apoptosis.


Assuntos
Morte Celular/fisiologia , Neutrófilos/fisiologia , Complexo Antígeno-Anticorpo/fisiologia , Apoptose/fisiologia , Morte Celular/imunologia , Humanos , Imunoglobulinas/fisiologia , Neutrófilos/imunologia , Transdução de Sinais/fisiologia
3.
Mediators Inflamm ; 2015: 267590, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26300590

RESUMO

Microparticles (MPs) are induced during apoptosis, cell activation, and even "spontaneous" release. Initially MPs were considered to be inert cellular products with no biological function. However, an extensive research and functional characterization have shown that the molecular composition and the effects of MPs depend upon the cellular background and the mechanism inducing them. They possess a wide spectrum of biological effects on intercellular communication by transferring different molecules able to modulate other cells. MPs interact with their target cells through different mechanisms: membrane fusion, macropinocytosis, and receptor-mediated endocytosis. However, when MPs remain in the extracellular milieu, they undergo modifications such as citrullination, glycosylation, and partial proteolysis, among others, becoming a source of neoantigens. In rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), reports indicated elevated levels of MPs with different composition, content, and effects compared with those isolated from healthy individuals. MPs can also form immune complexes amplifying the proinflammatory response and tissue damage. Their early detection and characterization could facilitate an appropriate diagnosis optimizing the pharmacological strategies, in different diseases including cancer, infection, and autoimmunity. This review focuses on the current knowledge about MPs and their involvement in the immunopathogenesis of SLE and RA.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Artrite Reumatoide/imunologia , Micropartículas Derivadas de Células/fisiologia , Lúpus Eritematoso Sistêmico/imunologia , Plaquetas/fisiologia , Humanos , Fator de Necrose Tumoral alfa/biossíntese
4.
J Am Soc Nephrol ; 25(5): 918-25, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24357670

RESUMO

The neonatal Fc receptor (FcRn) is a major regulator of IgG and albumin homeostasis systemically and in the kidneys. We investigated the role of FcRn in the development of immune complex-mediated glomerular disease in mice. C57Bl/6 mice immunized with the noncollagenous domain of the α3 chain of type IV collagen (α3NC1) developed albuminuria associated with granular capillary loop deposition of exogenous antigen, mouse IgG, C3 and C5b-9, and podocyte injury. High-resolution imaging showed abundant IgG deposition in the expanded glomerular basement membrane, especially in regions corresponding to subepithelial electron dense deposits. FcRn-null and -humanized mice immunized with α3NC1 developed no albuminuria and had lower levels of serum IgG anti-α3NC1 antibodies and reduced glomerular deposition of IgG, antigen, and complement. Our results show that FcRn promotes the formation of subepithelial immune complexes and subsequent glomerular pathology leading to proteinuria, potentially by maintaining higher serum levels of pathogenic IgG antibodies. Therefore, reducing pathogenic IgG levels by pharmacologic inhibition of FcRn may provide a novel approach for the treatment of immune complex-mediated glomerular diseases. As proof of concept, we showed that a peptide inhibiting the interaction between human FcRn and human IgG accelerated the degradation of human IgG anti-α3NC1 autoantibodies injected into FCRN-humanized mice as effectively as genetic ablation of FcRn, thus preventing the glomerular deposition of immune complexes containing human IgG.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Glomerulonefrite/etiologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Receptores Fc/fisiologia , Albuminúria/etiologia , Albuminúria/metabolismo , Animais , Doença Antimembrana Basal Glomerular/etiologia , Doença Antimembrana Basal Glomerular/imunologia , Doença Antimembrana Basal Glomerular/metabolismo , Complexo Antígeno-Anticorpo/efeitos adversos , Autoantígenos/fisiologia , Colágeno Tipo IV/fisiologia , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Células HEK293 , Humanos , Imunoglobulina G/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Arthritis Rheum ; 65(10): 2583-93, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23860661

RESUMO

OBJECTIVE: Previous studies have demonstrated a protective role of Toll-like receptor 2 (TLR-2) and a proinflammatory function of TLR-4 during chronic T cell-driven arthritis. The involvement of TLRs in T cell-independent arthritic processes, however, remains unclear. This study was undertaken to determine the functional significance of TLR-2 and TLR-4 in T cell-independent immune complex-driven arthritis. METHODS: Serum-transfer arthritis was induced in wild-type and TLR-deficient mice by intraperitoneal injections of arthritogenic K/BxN mouse serum. Arthritis was assessed macroscopically and by histologic analysis. The influence of TLR-2 on macrophage cytokine profile, Fcγ receptor (FcγR) expression, and response to immune complexes was determined. RESULTS: While TLR-4, unexpectedly, did not play any significant role, TLR-2 deficiency accelerated the onset and markedly increased the severity of acute immune complex-driven arthritis in mice. TLR-2 deficiency resulted in a substantial increase in joint inflammation, bone erosion, and cartilage pathology, indicating a protective function of TLR-2 in passive FcγR-driven disease. Ex vivo study of the macrophage inflammatory phenotype revealed increased production of tumor necrosis factor α (TNFα) and interleukin-6 (IL-6) despite similar levels of IL-10, along with a significant increase in FcγR-specific response, in TLR-2-/- mouse macrophages early in the disease. Although distinct FcγR messenger RNA expression was not affected, cell surface protein expression of the inhibitory FcγRIIB in TLR-2-/- naive primary macrophages was specifically diminished, resulting in a higher proinflammatory response. Accordingly, TLR-2 stimulation specifically up-regulated FcγRIIB, but not the activating FcγR, on macrophages. CONCLUSION: TLR-2 regulates acute immune complex-driven arthritis by controlling macrophage FcγR response. Our findings indicate that the protective role of TLR-2 is extended beyond its previously described role in promoting Treg cells during T cell-mediated arthritis.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Artrite Experimental/fisiopatologia , Receptores de IgG/fisiologia , Transdução de Sinais/fisiologia , Receptor 2 Toll-Like/fisiologia , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Técnicas In Vitro , Interleucina-6/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Soro/imunologia , Índice de Gravidade de Doença , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
6.
J Immunol ; 188(2): 902-15, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22147767

RESUMO

Immune complexes (ICs) play a pivotal role in causing inflammation in systemic lupus erythematosus (SLE). Yet, it remains unclear what the dominant blood cell type(s) and inflammation-related gene programs stimulated by lupus ICs are. To address these questions, we exposed normal human PBMCs or CD14(+) isolated monocytes to SLE ICs in the presence or absence of C1q and performed microarray analysis and other tests for cell activation. By microarray analysis, we identified genes and pathways regulated by SLE ICs that are both type I IFN dependent and independent. We also found that C1q-containing ICs markedly reduced expression of the majority of IFN-response genes and also influenced the expression of multiple other genes induced by SLE ICs. Surprisingly, IC activation of isolated CD14(+) monocytes did not upregulate CD40 and CD86 and only modestly stimulated inflammatory gene expression. However, when monocyte subsets were purified and analyzed separately, the low-abundance CD14(dim) ("patrolling") subpopulation was more responsive to ICs. These observations demonstrate the importance of plasmacytoid dendritic cells, CD14(dim) monocytes, and C1q as key regulators of inflammatory properties of ICs and identify many pathways through which they act.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Complemento C1q/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Células Dendríticas/patologia , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interferon-alfa/biossíntese , Receptores de Lipopolissacarídeos/biossíntese , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Células U937
7.
J Immunol ; 189(5): 2645-55, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22865916

RESUMO

Targeting of Ags directly to dendritic cells (DCs) through anti-DC receptor Ab fused to Ag proteins is a promising approach to vaccine development. However, not all Ags can be expressed as a rAb directly fused to a protein Ag. In this study, we show that noncovalent assembly of Ab-Ag complexes, mediated by interaction between dockerin and cohesin domains from cellulose-degrading bacteria, can greatly expand the range of Ags for this DC-targeting vaccine technology. rAbs with a dockerin domain fused to the rAb H chain C terminus are efficiently secreted by mammalian cells, and many Ags not secreted as rAb fusion proteins are readily expressed as cohesin directly fused to Ag either via secretion from mammalian cells or as soluble cytoplasmic Escherichia coli products. These form very stable and homogeneous complexes with rAb fused to dockerin. In vitro, these complexes can efficiently bind to human DC receptors followed by presentation to Ag-specific CD4⁺ and CD8⁺ T cells. Low doses of the HA1 subunit of influenza hemagglutinin conjugated through this means to anti-Langerin rAbs elicited Flu HA1-specific Ab and T cell responses in mice. Thus, the noncovalent assembly of rAb and Ag through dockerin and cohesin interaction provides a useful modular strategy for development and testing of prototype vaccines for elicitation of Ag-specific T and B cell responses, particularly when direct rAb fusions to Ag cannot be expressed.


Assuntos
Anticorpos/administração & dosagem , Anticorpos/fisiologia , Antígenos/administração & dosagem , Antígenos/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Animais , Complexo Antígeno-Anticorpo/administração & dosagem , Complexo Antígeno-Anticorpo/biossíntese , Complexo Antígeno-Anticorpo/fisiologia , Antígenos/isolamento & purificação , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Terapia de Alvo Molecular , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia
8.
J Am Soc Nephrol ; 24(10): 1529-36, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23833258

RESUMO

Regulatory T cells (Tregs) can suppress immunologic damage in renal ischemia-reperfusion injury (IRI), but the isolation and ex vivo expansion of these cells for clinical application remains challenging. Here, we investigated whether the IL-2/anti-IL-2 complex (IL-2C), a mediator of Treg expansion, can attenuate renal IRI in mice. IL-2C administered before bilateral renal IRI induced Treg expansion in both spleen and kidney, improved renal function, and attenuated histologic renal injury and apoptosis after IRI. Furthermore, IL-2C administration reduced the expression of inflammatory cytokines and attenuated the infiltration of neutrophils and macrophages in renal tissue. Depletion of Tregs with anti-CD25 antibodies abrogated the beneficial effects of IL-2C. However, IL-2C-mediated renal protection was not dependent on either IL-10 or TGF-ß. Notably, IL-2C administered after IRI also enhanced Treg expansion in spleen and kidney, increased tubular cell proliferation, improved renal function, and reduced renal fibrosis. In conclusion, these results indicate that IL-2C-induced Treg expansion attenuates acute renal damage and improves renal recovery in vivo, suggesting that IL-2C may be a therapeutic strategy for renal IRI.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complexo Antígeno-Anticorpo/fisiologia , Diferenciação Celular/imunologia , Interleucina-2/fisiologia , Insuficiência Renal/prevenção & controle , Traumatismo por Reperfusão/prevenção & controle , Linfócitos T Reguladores/imunologia , Animais , Avaliação Pré-Clínica de Medicamentos , Fibrose , Interleucina-2/imunologia , Rim/imunologia , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência Renal/imunologia , Insuficiência Renal/patologia , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/patologia
9.
J Immunol ; 186(5): 2699-704, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21325219

RESUMO

IgG FcRs are important mediators of immunity and play a key role during Ab-based immunotherapy. Within the leukocyte IgG receptor family, only FcγRI is capable of IgG binding with high affinity. FcγRI exists as a complex of a ligand binding α-chain and an FcR γ-chain. The receptors' α-chain can, furthermore, elicit several functions independent of the ITAM-bearing FcR γ-chain. Functional implications of high-affinity IgG binding and mechanisms underlying FcR γ-chain-independent signaling remain unclear to this day. In this paper, we provide an overview of past literature on FcγRI and address the implications of recently described interactions between cytosolic proteins and the FcγRI α-chain, as well as cytokine-enhanced FcγRI immune complex binding. Furthermore, an analysis of potential polymorphisms within the FCGR1A gene is provided.


Assuntos
Sítios de Ligação de Anticorpos , Receptores de IgG/fisiologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/genética , Complexo Antígeno-Anticorpo/fisiologia , Sítios de Ligação de Anticorpos/genética , Modelos Animais de Doenças , Humanos , Dados de Sequência Molecular , Ligação Proteica/genética , Ligação Proteica/imunologia , Estrutura Terciária de Proteína/genética , Receptores de IgG/química , Receptores de IgG/genética
10.
J Immunol ; 186(3): 1432-41, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21191075

RESUMO

Immune complex-induced acute lung injury (IC-ALI) has been implicated in various pulmonary disease states. However, the role of NKT cells in IC-ALI remains unknown. Therefore, we explored NKT cell functions in IC-ALI using chicken egg albumin and anti-chicken egg albumin IgG. The bronchoalveolar lavage fluid of CD1d(-/-) and Jα18(-/-) mice contained few Ly6G(+)CD11b(+) granulocytes, whereas levels in B6 mice were greater and were increased further by α-galactosyl ceramide. IFN-γ and MIP-1α production in the lungs was greater in B6 than CD1d(-/-) mice. Adoptive transfer of wild type (WT) but not IFN-γ-, MIP-1α-, or FcγR-deficient NKT cells into CD1d(-/-) mice caused recruitment of inflammatory cells to the lungs. Moreover, adoptive transfer of IFN-γR-deficient NKT cells enhanced MIP-1α production and cell recruitment in the lungs of CD1d(-/-) or CD1d(-/-)IFN-γ(-/-) mice, but to a lesser extent than WT NKT cells. This suggests that IFN-γ-producing NKT cells enhance MIP-1α production in both an autocrine and a paracrine manner. IFN-γ-deficient NKT cells induced less IL-1ß and TNF-α production by alveolar macrophages and dendritic cells in CD1d(-/-) mice than did WT NKT cells. Taken together, these data suggest that CD1d-restricted IFN-γ-producing NKT cells promote IC-ALI by producing MIP-1α and enhancing proinflammatory cytokine production by alveolar macrophages and dendritic cells.


Assuntos
Lesão Pulmonar Aguda/imunologia , Complexo Antígeno-Anticorpo/fisiologia , Antígenos CD1d/administração & dosagem , Quimiocina CCL3/biossíntese , Células Dendríticas/imunologia , Interferon gama/metabolismo , Macrófagos Alveolares/imunologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Transferência Adotiva , Animais , Antígenos CD1d/genética , Antígenos CD1d/metabolismo , Quimiocina CCL3/deficiência , Quimiocina CCL3/genética , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Imunoglobulina G/administração & dosagem , Interferon gama/deficiência , Interferon gama/genética , Ativação de Macrófagos/imunologia , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/transplante , Ovalbumina/administração & dosagem , Ovalbumina/imunologia
11.
J Immunol ; 186(6): 3484-96, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21307287

RESUMO

IgE-mediated allergic inflammation occurs when allergens cross-link IgE on the surface of immune cells, thereby triggering the release of inflammatory mediators as well as enhancing Ag presentations. IgE is frequently present in airway secretions, and its level can be enhanced in human patients with allergic rhinitis and bronchial asthma. However, it remains completely unknown how IgE appears in the airway secretions. In this study, we show that CD23 (FcεRII) is constitutively expressed in established or primary human airway epithelial cells, and its expression is significantly upregulated when airway epithelial cells were subjected to IL-4 stimulation. In a transcytosis assay, human IgE or IgE-derived immune complex (IC) was transported across a polarized Calu-3 monolayer. Exposure of the Calu-3 monolayer to IL-4 stimulation also enhanced the transcytosis of either human IgE or the IC. A CD23-specific Ab or soluble CD23 significantly reduced the efficiency of IgE or IC transcytosis, suggesting a specific receptor-mediated transport by CD23. Transcytosis of both IgE and the IC was further verified in primary human airway epithelial cell monolayers. Furthermore, the transcytosed Ag-IgE complexes were competent in inducing degranulation of the cultured human mast cells. Because airway epithelial cells are the first cell layer to come into contact with inhaled allergens, our study implies CD23-mediated IgE transcytosis in human airway epithelial cells may play a critical role in initiating and contributing to the perpetuation of airway allergic inflammation.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Polaridade Celular/imunologia , Imunoglobulina E/metabolismo , Receptores de IgE/fisiologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Transcitose/imunologia , Alérgenos/administração & dosagem , Animais , Complexo Antígeno-Anticorpo/fisiologia , Brônquios/imunologia , Brônquios/metabolismo , Brônquios/patologia , Células CHO , Linhagem Celular , Linhagem Celular Tumoral , Polaridade Celular/genética , Cricetinae , Cricetulus , Células HEK293 , Células HT29 , Humanos , Imunoglobulina E/fisiologia , Mediadores da Inflamação/administração & dosagem , Mastócitos/imunologia , Mastócitos/metabolismo , Mastócitos/patologia , Receptores de IgE/biossíntese , Receptores de IgE/genética , Mucosa Respiratória/patologia , Transcitose/genética , Células U937
12.
Clin Exp Immunol ; 167(2): 235-45, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22235999

RESUMO

In systemic lupus erythematosus (SLE), the autoantibodies that form immune complexes (ICs) trigger activation of the complement system. This results in the formation of membrane attack complex (MAC) on cell membrane and the soluble terminal complement complex (TCC). Hyperactive T cell responses are hallmark of SLE pathogenesis. How complement activation influences the T cell responses in SLE is not fully understood. We observed that aggregated human γ-globulin (AHG) bound to a subset of CD4(+) T cells in peripheral blood mononuclear cells and this population increased in the SLE patients. Human naive CD4(+) T cells, when treated with purified ICs and TCC, triggered recruitment of the FcRγ chain with the membrane receptor and co-localized with phosphorylated Syk. These events were also associated with aggregation of membrane rafts. Thus, results presented suggest a role for ICs and complement in the activation of Syk in CD4(+) T cells. Thus, we propose that the shift in signalling from ζ-chain-ZAP70 to FcRγ chain-Syk observed in T cells of SLE patients is triggered by ICs and complement. These results demonstrate a link among ICs, complement activation and phosphorylation of Syk in CD4(+) T cells.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Autoanticorpos/fisiologia , Linfócitos T CD4-Positivos/enzimologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lúpus Eritematoso Sistêmico/enzimologia , Processamento de Proteína Pós-Traducional/imunologia , Proteínas Tirosina Quinases/metabolismo , Adolescente , Adulto , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas/enzimologia , Células Cultivadas/imunologia , Ativação Enzimática/imunologia , Feminino , Humanos , Células Jurkat , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Microdomínios da Membrana , Pessoa de Meia-Idade , Fosforilação , Receptores de IgG/biossíntese , Receptores de IgG/genética , Receptores de IgG/imunologia , Transdução de Sinais/imunologia , Quinase Syk , Adulto Jovem , Proteína-Tirosina Quinase ZAP-70/fisiologia , gama-Globulinas/imunologia
13.
J Immunol ; 185(3): 1577-83, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20585032

RESUMO

Anti-CD40L immunotherapy in systemic lupus erythematosus patients was associated with thromboembolism of unknown cause. We previously showed that monoclonal anti-CD40L immune complexes (ICs) activated platelets in vitro via the IgG receptor (FcgammaRIIa). In this study, we examined the prothrombotic effects of anti-CD40L ICs in vivo. Because mouse platelets lack FcgammaRIIa, we used FCGR2A transgenic mice. FCGR2A mice were injected i.v. with preformed ICs consisting of either anti-human CD40L mAb (M90) plus human CD40L, or a chimerized anti-mouse CD40L mAb (hMR1) plus mouse CD40L. ICs containing an aglycosylated form of hMR1, which does not bind FcgammaRIIa, were also injected. M90 IC caused shock and thrombocytopenia in FCGR2A but not in wild-type mice. Animals injected with hMR1 IC also experienced these effects, whereas those injected with aglycosylated-hMR1 IC did not, demonstrating that anti-CD40L IC-induced platelet activation in vivo is FcgammaRIIa-dependent. Sequential injections of individual IC components caused similar effects, suggesting that ICs were able to assemble in circulation. Analysis of IC-injected mice revealed pulmonary thrombi consisting of platelet aggregates and fibrin. Mice pretreated with a thrombin inhibitor became moderately thrombocytopenic in response to anti-CD40L ICs and had pulmonary platelet-thrombi devoid of fibrin. In conclusion, we have shown for the first time that anti-CD40L IC-induced thrombosis can be replicated in mice transgenic for FcgammaRIIa. This molecular mechanism may be important for understanding thrombosis associated with CD40L immunotherapy. The FCGR2A mouse model may also be useful for assessing the hemostatic safety of other therapeutic Abs.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Autoanticorpos/toxicidade , Ligante de CD40/imunologia , Ativação Plaquetária/imunologia , Receptores de IgG/genética , Trombose/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/toxicidade , Complexo Antígeno-Anticorpo/administração & dosagem , Complexo Antígeno-Anticorpo/toxicidade , Autoanticorpos/administração & dosagem , Autoanticorpos/uso terapêutico , Humanos , Hibridomas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ativação Plaquetária/genética , Receptores de IgG/deficiência , Receptores de IgG/fisiologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/fisiologia , Proteínas Recombinantes de Fusão/toxicidade , Trombose/sangue
14.
J Immunol ; 184(4): 1977-89, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20083655

RESUMO

The FcgammaRIIB is a potent inhibitory coreceptor that blocks BCR signaling in response to immune complexes and, as such, plays a decisive role in regulating Ab responses. The recent application of high-resolution live cell imaging to B cell studies is providing new molecular details of the earliest events in the initiation BCR signaling that follow within seconds of Ag binding. In this study, we report that when colligated to the BCR through immune complexes, the FcgammaRIIB colocalizes with the BCR in microscopic clusters and blocks the earliest events that initiate BCR signaling, including the oligomerization of the BCR within these clusters, the active recruitment of BCRs to these clusters, and the resulting spreading and contraction response. Fluorescence resonance energy transfer analyses indicate that blocking these early events may not require molecular proximity of the cytoplasmic domains of the BCR and FcgammaRIIB, but relies on the rapid and sustained association of FcgammaRIIB with raft lipids in the membrane. These results may provide novel early targets for therapies aimed at regulating the FcgammaRIIB to control Ab responses in autoimmune disease.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Antígenos/fisiologia , Receptores de Antígenos de Linfócitos B/antagonistas & inibidores , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de IgG/fisiologia , Sequência de Aminoácidos , Animais , Complexo Antígeno-Anticorpo/metabolismo , Subpopulações de Linfócitos B/enzimologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/imunologia , Células Cultivadas , Humanos , Inositol Polifosfato 5-Fosfatases , Ligantes , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Monoéster Fosfórico Hidrolases/fisiologia , Fosforilação/imunologia , Transporte Proteico/imunologia , Agregação de Receptores/imunologia , Receptores de Antígenos de Linfócitos B/fisiologia , Receptores de IgG/metabolismo , Transdução de Sinais/imunologia , Quinases da Família src/metabolismo , Quinases da Família src/fisiologia
15.
J Infect Dis ; 203(10): 1405-14, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21502081

RESUMO

It has been reported that levels of viremia reflect the severity of illness in dengue virus infection. We assessed the levels of viremia in patients with primary and secondary infections, using 2 cell lines: FcγR-expressing BHK cells and FcγR-negative cells. In primary infection, virus titers were at similar levels between FcγR-expressing and FcγR-negative cells. In secondary infection, however, virus titers were ∼10 times higher in FcγR-expressing cells on days 1-6 when compared with FcγR-negative cells, indicating discrepancy in viremia titers between FcγR-expressing and FcγR-negative cells. The results suggest that dengue virus-antibody complexes with infectious capacity exist in patients with secondary infection, and these immune complexes can be detected by using FcγR-expressing cells. As it has been reported that principal target cells of dengue virus infection are FcγR-positive, monocyte/macrophage lineage cells, virus titers determined using FcγR-expressing cells may better reflect the actual viremic conditions in vivo.


Assuntos
Complexo Antígeno-Anticorpo/análise , Vírus da Dengue/imunologia , Dengue/imunologia , Receptores de IgG/metabolismo , Viremia/imunologia , Adolescente , Adulto , Animais , Complexo Antígeno-Anticorpo/fisiologia , Linhagem Celular , Cricetinae , Dengue/sangue , Dengue/virologia , Vírus da Dengue/classificação , Vírus da Dengue/genética , Vírus da Dengue/crescimento & desenvolvimento , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Viral/análise , Receptores de IgG/genética , Recidiva , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaio de Placa Viral , Viremia/sangue , Viremia/virologia , Adulto Jovem
16.
J Immunol ; 183(4): 2330-6, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19620297

RESUMO

Sphingosine 1-phosphate (S1P) is a bioactive phospholipid that is released by platelets and endothelial cells and has been implicated in diverse biological functions. We hypothesized that S1P may influence immune complex-mediated polymorphonuclear neutrophil activation. Using flow cytometry and fluorescence spectrometry, we found that exogenous addition of S1P led to an enhanced polymorphonuclear neutrophil Fcgamma receptor-mediated rise in intracellular Ca(2+) and reactive oxygen species generation in a pertussis toxin-independent manner, while having only a small effect by itself. Thus, S1P amplifies a positive feedback loop where Fcgamma receptor-mediated rises in Ca(2+) and reactive oxygen species are interdependent, with reactive oxygen species acting to increase tyrosine phosphorylation and activity of upstream signaling intermediates. S1P augmentation of Fcgamma receptor signaling translates to downstream functional consequences, including shape change and recruitment to endothelial surfaces coated with suboptimal levels of immune complexes. Taken together, S1P from activated platelets or endothelial cells may serve to amplify leukocyte recruitment and tissue injury at sites of immune complex deposition in vasculitis.


Assuntos
Adjuvantes Imunológicos/sangue , Quimiotaxia de Leucócito/imunologia , Lisofosfolipídeos/sangue , Ativação de Neutrófilo/imunologia , Infiltração de Neutrófilos/imunologia , Receptores de IgG/sangue , Esfingosina/análogos & derivados , Regulação para Cima/imunologia , Adjuvantes Imunológicos/metabolismo , Complexo Antígeno-Anticorpo/sangue , Complexo Antígeno-Anticorpo/fisiologia , Sinalização do Cálcio/imunologia , Adesão Celular/imunologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Células HL-60 , Humanos , Mediadores da Inflamação/sangue , Mediadores da Inflamação/fisiologia , Lisofosfolipídeos/metabolismo , Microcirculação/imunologia , Neutrófilos/enzimologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores de IgG/fisiologia , Esfingosina/sangue , Esfingosina/metabolismo
17.
J Immunol ; 183(5): 3109-17, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19648272

RESUMO

Although TLR9 was originally thought to specifically recognize microbial DNA, it is now evident that mammalian DNA can be an effective TLR9 ligand. However, the DNA sequence required for TLR9 activation is controversial, as studies have shown conflicting results depending on the nature of the DNA backbone, the route of DNA uptake, and the cell type being studied. In systemic lupus erythematosus, a major route whereby DNA gains access to intracellular TLR9, and thereby activates dendritic cells (DCs), is through uptake as a DNA-containing immune complex. In this report, we used defined dsDNA fragments with a natural (phosphodiester) backbone and show that unmethylated CpG dinucleotides within dsDNA are required for murine DC TLR9 activation induced by a DNA-containing immune complex. The strongest activation is seen with dsDNA fragments containing optimal CpG motifs (purine-purine-CpG-pyrimidine-pyrimidine) that are common in microbial DNA but rare in mammalian DNA. Importantly, however, activation can also be induced by CpG-rich DNA fragments that lack these optimal CpG motifs and that we show are plentiful in CpG islands within mammalian DNA. No activation is induced by DNA fragments lacking CpG dinucleotides, although this CpG-free DNA can induce DC activation if internalized by liposomal transfection instead of as an immune complex. Overall, the data suggest that the release of CpG-rich DNA from mammalian DNA may contribute to the pathogenesis of autoimmune diseases such as systemic lupus erythematosus and psoriasis in which activation of TLR9 in DCs by self DNA has been implicated in disease pathogenesis.


Assuntos
Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/genética , Ilhas de CpG/imunologia , DNA/química , DNA/fisiologia , Células Dendríticas/imunologia , Oligonucleotídeos Fosforotioatos/fisiologia , Receptor Toll-Like 9/fisiologia , Adjuvantes Imunológicos/genética , Adjuvantes Imunológicos/metabolismo , Adjuvantes Imunológicos/fisiologia , Animais , Complexo Antígeno-Anticorpo/fisiologia , Células Cultivadas , Ilhas de CpG/genética , DNA/metabolismo , Fragmentação do DNA , Metilação de DNA/imunologia , Células Dendríticas/química , Células Dendríticas/metabolismo , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Oligonucleotídeos Fosforotioatos/química , Oligonucleotídeos Fosforotioatos/genética , Psoríase/genética , Psoríase/imunologia , Psoríase/metabolismo , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/metabolismo
18.
J Immunol ; 183(7): 4509-20, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19734236

RESUMO

Rheumatoid arthritis (RA) is a common autoimmune disease leading to profound disability and premature death. Although a role for FcgammaRs and TLRs is accepted, their precise involvement remains to be elucidated. FcgammaRIIb is an inhibitory FcR important in the maintenance of tolerance. We hypothesized that the inhibitory FcgammaRIIb inhibits TLR responses on monocyte-derived dendritic cells (DC) and serves as a counterregulatory mechanism to dampen inflammation, and we surmised that this mechanism might be defective in RA. The expression of the inhibitory FcgammaRIIb was found to be significantly higher on DCs from RA patients having low RA disease activity in the absence of treatment with antirheumatic drugs. The expression of activating FcgammaRs was similarly distributed among all RA patients and healthy controls. Intriguingly, only DCs with a high expression of FcgammaRIIb were able to inhibit TLR4-mediated secretion of proinflammatory cytokines when stimulated with immune complexes. In addition, when these DCs were coincubated with the combination of a TLR4 agonist and immune complexes, a markedly inhibited T cell proliferation was apparent, regulatory T cell development was promoted, and T cells were primed to produce high levels of IL-13 compared with stimulation of the DCs with the TLR4 agonist alone. Blocking FcgammaRIIb with specific Abs fully abrogated these effects demonstrating the full dependence on the inhibitory FcgammaRIIb in the induction of these phenomena. This TLR4-FcgammaRIIb interaction was shown to dependent on the PI3K and Akt pathway.


Assuntos
Artrite Reumatoide/imunologia , Células Dendríticas/imunologia , Regulação para Baixo/imunologia , Inibidores do Crescimento/fisiologia , Receptores de IgG/fisiologia , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/fisiologia , Regulação para Cima/imunologia , Idoso , Complexo Antígeno-Anticorpo/fisiologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Células Cultivadas , Estudos de Coortes , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Regulação para Baixo/genética , Inibidores do Crescimento/biossíntese , Inibidores do Crescimento/genética , Humanos , Mediadores da Inflamação/metabolismo , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Estudos Prospectivos , Receptores de IgG/biossíntese , Receptores de IgG/genética , Regulação para Cima/genética
19.
J Immunol ; 182(1): 554-62, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19109188

RESUMO

Excessive activation of TLR may induce endotoxin shock and inflammatory diseases, so the negative regulation of TLR-triggered inflammatory response attracts much attention. Nonpathogenic immune complex (IC) and Ig (IC/Ig) have been shown to play important roles in the regulation of immune responses and to be therapeutic in some kinds of autoimmune diseases. However, the role of IC/Ig in the regulation of TLR-triggered inflammatory responses and the underlying mechanisms remain to be fully understood. In this study we demonstrate that IC/Ig can significantly inhibit LPS-induced secretion of TNF-alpha and IL-6 from macrophages by preferentially inducing PGE(2). Pretreatment of mice with IC can protect wild-type mice, but not Fc gammaRIIb(-/-) mice, from lethal endotoxin shock, and significantly reduce the levels of serum TNF-alpha and IL-6 in wild-type mice but not in Fc gammaR IIb(-/-) mice. Furthermore, blockade of PGE(2) by celecoxib restores LPS-induced production of TNF-alpha and IL-6 in the presence of IC both in vitro and in vivo. Accordingly, blockade of PGE(2) production in vivo results in the increased sensitivity of IC-pretreated mice to lethal endotoxin shock. Therefore, IC/Ig can negatively regulate TLR4-triggered inflammatory response in macrophages through Fc gammaRIIb-dependent PGE(2). In addition, our results suggest that down-regulation of NF-kappaB activation and TLR4 expression but activation of protein kinase A pathway in macrophages by IC/Ig contribute to the negative regulatory process. Thus we provide new manner for the immune regulation and mechanistic explanation for nonpathogenic IC/Ig in the treatment of inflammatory or autoimmune diseases.


Assuntos
Complexo Antígeno-Anticorpo/fisiologia , Dinoprostona/biossíntese , Regulação para Baixo/imunologia , Mediadores da Inflamação/fisiologia , Macrófagos/imunologia , Macrófagos/patologia , Receptores de IgG/fisiologia , Receptor 4 Toll-Like/fisiologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Linhagem Celular , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Citocinas/fisiologia , Dinoprostona/fisiologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Líquido Intracelular/enzimologia , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/deficiência , Receptores de IgG/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/antagonistas & inibidores
20.
Infect Immun ; 78(7): 3129-35, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20439480

RESUMO

Complement-containing immune complexes can be presented to phagocytes by human erythrocytes bearing complement receptor 1 (CR1). Although this has long been assumed to be a mechanism by which humans are able to protect themselves from "extracellular" bacteria such as pneumococci, there is little direct evidence. In these studies we have investigated this question by comparing results for erythrocytes from transgenic mice expressing human CR1 on their erythrocytes to the results for wild-type mouse erythrocytes that do not express CR1. We demonstrate that human CR1 expression on murine erythrocytes allows immune adherence to beads opsonized with either mouse or human serum as a source of complement. The role of CR1 in immune adherence was supported by studies showing that it was blocked by the addition of antibody to human CR1. Furthermore, human CR1 expression enhances the immune adherence of opsonized pneumococci to erythrocytes in vitro, and the pneumococci attached to erythrocytes via CR1 can be transferred in vitro to live macrophages. Even more importantly, we observed that if complement-opsonized pneumococci are injected intravenously with CR1(+) mouse erythrocytes into wild-type mice (after a short in vitro incubation), they are cleared faster than opsonized pneumococci similarly injected with wild-type mouse erythrocytes. Finally, we have shown that the intravenous (i.v.) injection of pneumococci into CR1(+) mice also results in more rapid blood clearance than in wild-type mice. These data support that immune adherence via CR1 on erythrocytes likely plays an important role in the clearance of opsonized bacteria from human blood.


Assuntos
Membrana Eritrocítica/fisiologia , Infecções Pneumocócicas/imunologia , Receptores de Complemento/biossíntese , Animais , Complexo Antígeno-Anticorpo/imunologia , Complexo Antígeno-Anticorpo/fisiologia , Linhagem Celular , Membrana Eritrocítica/imunologia , Expressão Gênica/imunologia , Expressão Gênica/fisiologia , Humanos , Macrófagos/imunologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagocitose/imunologia , Fagocitose/fisiologia , Receptores de Complemento/fisiologia , Streptococcus pneumoniae/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA