Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 166(1): 88-101, 2016 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-27293190

RESUMO

Antibodies to DNA and chromatin drive autoimmunity in systemic lupus erythematosus (SLE). Null mutations and hypomorphic variants of the secreted deoxyribonuclease DNASE1L3 are linked to familial and sporadic SLE, respectively. We report that DNASE1L3-deficient mice rapidly develop autoantibodies to DNA and chromatin, followed by an SLE-like disease. Circulating DNASE1L3 is produced by dendritic cells and macrophages, and its levels inversely correlate with anti-DNA antibody response. DNASE1L3 is uniquely capable of digesting chromatin in microparticles released from apoptotic cells. Accordingly, DNASE1L3-deficient mice and human patients have elevated DNA levels in plasma, particularly in circulating microparticles. Murine and human autoantibody clones and serum antibodies from human SLE patients bind to DNASE1L3-sensitive chromatin on the surface of microparticles. Thus, extracellular microparticle-associated chromatin is a potential self-antigen normally digested by circulating DNASE1L3. The loss of this tolerance mechanism can contribute to SLE, and its restoration may represent a therapeutic opportunity in the disease.


Assuntos
Autoanticorpos/imunologia , Micropartículas Derivadas de Células/química , Cromatina/imunologia , DNA/imunologia , Endodesoxirribonucleases/genética , Lúpus Eritematoso Sistêmico/imunologia , Animais , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Endodesoxirribonucleases/deficiência , Endodesoxirribonucleases/metabolismo , Humanos , Células Jurkat , Lúpus Eritematoso Sistêmico/enzimologia , Lúpus Eritematoso Sistêmico/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33397815

RESUMO

Photosensitivity to ultraviolet (UV) light affects up to ∼80% of lupus patients. Sunlight exposure can exacerbate local as well as systemic manifestations of lupus, including nephritis, by mechanisms that are poorly understood. Here, we report that acute skin exposure to UV light triggers a neutrophil-dependent injury response in the kidney characterized by upregulated expression of endothelial adhesion molecules as well as inflammatory and injury markers associated with transient proteinuria. We showed that UV light stimulates neutrophil migration not only to the skin but also to the kidney in an IL-17A-dependent manner. Using a photoactivatable lineage tracing approach, we observed that a subset of neutrophils found in the kidney had transited through UV light-exposed skin, suggesting reverse transmigration. Besides being required for the renal induction of genes encoding mediators of inflammation (vcam-1, s100A9, and Il-1b) and injury (lipocalin-2 and kim-1), neutrophils significantly contributed to the kidney type I interferon signature triggered by UV light. Together, these findings demonstrate that neutrophils mediate subclinical renal inflammation and injury following skin exposure to UV light. Of interest, patients with lupus have subpopulations of blood neutrophils and low-density granulocytes with similar phenotypes to reverse transmigrating neutrophils observed in the mice post-UV exposure, suggesting that these cells could have transmigrated from inflamed tissue, such as the skin.


Assuntos
Inflamação/sangue , Rim/metabolismo , Neutrófilos/efeitos da radiação , Pele/efeitos da radiação , Animais , Calgranulina B/genética , Movimento Celular/efeitos da radiação , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Inflamação/etiologia , Inflamação/patologia , Interleucina-17/genética , Rim/lesões , Rim/patologia , Rim/efeitos da radiação , Lipocalina-2/genética , Camundongos , Neutrófilos/metabolismo , Neutrófilos/patologia , Pele/lesões , Raios Ultravioleta/efeitos adversos , Molécula 1 de Adesão de Célula Vascular/genética
3.
Ann Rheum Dis ; 81(6): 845-853, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35236659

RESUMO

OBJECTIVE: Systemic lupus erythematosus (SLE) is the prototypical systemic autoimmune disease. While the long-term prognosis has greatly improved, better long-term survival is still necessary. The type I interferon (IFN) signature, a prominent feature of SLE, is not an ideal therapeutic target or outcome predictor. To explore immunological pathways in SLE more precisely, we performed transcriptomic, epigenomic and genomic analyses using 19 immune cell subsets from peripheral blood. METHODS: We sorted 19 immune cell subsets and identified the mRNA expression profiles and genetic polymorphisms in 107 patients with SLE and 92 healthy controls. Combined differentially expressed genes and expression quantitative trait loci analysis was conducted to find key driver genes in SLE pathogenesis. RESULTS: We found transcriptomic, epigenetic and genetic importance of oxidative phosphorylation (OXPHOS)/mitochondrial dysfunction in SLE memory B cells. Particularly, we identified an OXPHOS-regulating gene, PRDX6 (peroxiredoxin 6), as a key driver in SLE B cells. Prdx6-deficient B cells showed upregulated mitochondrial respiration as well as antibody production. We revealed OXPHOS signature was associated with type I IFN signalling-related genes (ISRGs) signature in SLE memory B cells. Furthermore, the gene sets related to innate immune signalling among ISRGs presented correlation with OXPHOS and these two signatures showed associations with SLE organ damage as well as specific clinical phenotypes. CONCLUSION: This work elucidated the potential prognostic marker for SLE. Since OXPHOS consists of the electron transport chain, a functional unit in mitochondria, these findings suggest the importance of mitochondrial dysfunction as a key immunological pathway involved in SLE.


Assuntos
Interferon Tipo I , Lúpus Eritematoso Sistêmico , Linfócitos B/metabolismo , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Fosforilação Oxidativa , Transcriptoma
4.
J Immunol ; 204(10): 2627-2640, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32238460

RESUMO

Lupus nephritis (LN) is a major contributor to morbidity and mortality in lupus patients, but the mechanisms of kidney damage remain unclear. In this study, we introduce, to our knowledge, novel models of LN designed to resemble the polygenic nature of human lupus by embodying three key genetic alterations: the Sle1 interval leading to anti-chromatin autoantibodies; Mfge8-/- , leading to defective clearance of apoptotic cells; and either C1q-/- or C3-/- , leading to low complement levels. We report that proliferative glomerulonephritis arose only in the presence of all three abnormalities (i.e., in Sle1.Mfge8 -/- C1q -/- and Sle1.Mfge8 -/- C3 -/- triple-mutant [TM] strains [C1q -/-TM and C3-/- TM, respectively]), with structural kidney changes resembling those in LN patients. Unexpectedly, both TM strains had significant increases in autoantibody titers, Ag spread, and IgG deposition in the kidneys. Despite the early complement component deficiencies, we observed assembly of the pathogenic terminal complement membrane attack complex in both TM strains. In C1q-/- TM mice, colocalization of MASP-2 and C3 in both the glomeruli and tubules indicated that the lectin pathway likely contributed to complement activation and tissue injury in this strain. Interestingly, enhanced thrombin activation in C3-/- TM mice and reduction of kidney injury following attenuation of thrombin generation by argatroban in a serum-transfer nephrotoxic model identified thrombin as a surrogate pathway for complement activation in C3-deficient mice. These novel mouse models of human lupus inform the requirements for nephritis and provide targets for intervention.


Assuntos
Doenças da Deficiência Hereditária de Complemento/genética , Rim/patologia , Nefrite Lúpica/imunologia , Animais , Anticorpos Antinucleares/sangue , Antígenos de Superfície/genética , Ativação do Complemento , Complemento C1q/genética , Complemento C3/genética , Modelos Animais de Doenças , Glomerulonefrite , Doenças da Deficiência Hereditária de Complemento/imunologia , Humanos , Nefrite Lúpica/genética , Serina Proteases Associadas a Proteína de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Leite/genética , Herança Multifatorial
5.
Immunity ; 36(1): 120-31, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22284419

RESUMO

The type I interferon (IFN) response initiated by detection of nucleic acids is important for antiviral defense but is also associated with specific autoimmune diseases. Mutations in the human 3' repair exonuclease 1 (Trex1) gene cause Aicardi-Goutières syndrome (AGS), an IFN-associated autoimmune disease. However, the source of the type I IFN response and the precise mechanisms of disease in AGS remain unknown. Here, we demonstrate that Trex1 is an essential negative regulator of the STING-dependent antiviral response. We used an in vivo reporter of IFN activity in Trex1-deficient mice to localize the initiation of disease to nonhematopoietic cells. These IFNs drove T cell-mediated inflammation and an autoantibody response that targeted abundant, tissue-restricted autoantigens. However, B cells contributed to mortality independently of T cell-mediated tissue damage. These findings reveal a stepwise progression of autoimmune disease in Trex1-deficient mice, with implications for the treatment of AGS and related disorders.


Assuntos
Doenças Autoimunes/fisiopatologia , Autoimunidade/imunologia , Interferons , Linfócitos/imunologia , Animais , Doenças Autoimunes/enzimologia , Doenças Autoimunes do Sistema Nervoso/fisiopatologia , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Interferons/fisiologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Malformações do Sistema Nervoso/fisiopatologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transdução de Sinais
6.
Annu Rev Med ; 68: 317-330, 2017 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-27813878

RESUMO

The best known of the naturally occurring antimalarial compounds are quinine, extracted from cinchona bark, and artemisinin (qinghao), extracted from Artemisia annua in China. These and other derivatives are now chemically synthesized and remain the mainstay of therapy to treat malaria. The beneficial effects of several of the antimalarial drugs (AMDs) on clinical features of autoimmune disorders were discovered by chance during World War II. In this review, we discuss the chemistry of AMDs and their mechanisms of action, emphasizing how they may impact multiple pathways of innate immunity. These pathways include Toll-like receptors and the recently described cGAS-STING pathway. Finally, we discuss the current and future impact of AMDs on systemic lupus erythematosus, rheumatoid arthritis, and devastating monogenic disorders (interferonopathies) characterized by expression of type I interferon in the brain.


Assuntos
Acridinas/farmacologia , Antimaláricos/farmacologia , Doenças Autoimunes/tratamento farmacológico , Imunidade Inata/efeitos dos fármacos , Imunomodulação/efeitos dos fármacos , Quinolinas/farmacologia , Acridinas/química , Animais , Antimaláricos/química , Artemisininas/farmacologia , Artrite Reumatoide/tratamento farmacológico , Autofagia/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Quinolinas/química , Transdução de Sinais , Receptores Toll-Like/metabolismo
7.
Annu Rev Med ; 67: 323-36, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26526766

RESUMO

An important concept in immunology is the classification of immune responses as either innate or adaptive, based on whether the antigen receptors are encoded in the germline or generated somatically by gene rearrangement. The innate immune system is an ancient mode of immunity, and by being a first layer in our defense against infectious agents, it is essential for our ability to develop rapid and sustained responses to pathogens. We discuss the importance of nucleic acid recognition by the innate immune system to mounting an appropriate immune response to pathogens and also how inflammation driven by uncontrolled recognition of self-nucleic acids can lead to autoimmune diseases. We also summarize current efforts to either harness the immune system using agonists of nucleic acid-specific innate sensors or, on the contrary, by using inhibitors in autoimmune situations.


Assuntos
Autoimunidade/imunologia , Imunidade Inata , Inflamação/imunologia , Ácidos Nucleicos/imunologia , Receptores Toll-Like/agonistas , Receptores Toll-Like/antagonistas & inibidores , Adjuvantes Imunológicos/uso terapêutico , Aminoquinolinas/uso terapêutico , Animais , Citosol/imunologia , DNA Viral/imunologia , Endossomos/imunologia , Humanos , Imiquimode , Inflamação/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Psoríase/imunologia , Transdução de Sinais , Receptores Toll-Like/imunologia , Viroses/imunologia
8.
J Immunol ; 197(7): 2854-63, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27534558

RESUMO

The loss of tolerance and the presence of circulating autoantibodies directed against nuclear Ags is the hallmark of systemic lupus erythematosus (SLE). Many of these Ags are complexed with short, noncoding RNAs, such as U1 and Y1. The amount of U1 and Y1 RNA complexed with SLE patient Abs and immune complexes was measured in a cross-section of 228 SLE patients to evaluate the role of these RNA molecules within the known biochemical framework of SLE. The study revealed that SLE patients had significantly elevated levels of circulating U1 and/or Y1 RNA compared with healthy volunteers. In addition, the blood-borne RNA molecules were correlated with SLE disease activity and increased expression of IFN-inducible genes. To our knowledge, this study provides the first systematic examination of the role of circulating RNA in a large group of SLE patients and provides an important link with IFN dysregulation.


Assuntos
Regulação da Expressão Gênica , Interferons/imunologia , Lúpus Eritematoso Sistêmico/genética , RNA/sangue , Adulto , Reações Antígeno-Anticorpo , Autoanticorpos/imunologia , Estudos de Casos e Controles , Estudos Transversais , Feminino , Humanos , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Pessoa de Meia-Idade , RNA/imunologia , RNA Citoplasmático Pequeno/sangue , RNA Nuclear Pequeno/sangue
9.
J Immunol ; 194(9): 4089-93, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25821216

RESUMO

Type I IFN is strongly implicated in the pathogenesis of systemic autoimmune diseases, such as lupus, and rare monogenic IFNopathies, including Aicardi-Goutières syndrome. Recently, a new DNA-activated pathway involving the enzyme cyclic GMP-AMP synthase (cGAS) was described and potentially linked to Aicardi-Goutières syndrome. To identify drugs that could potentially inhibit cGAS activity, we performed in silico screening of drug libraries. By computational analysis, we identified several antimalarial drugs (AMDs) that were predicted to interact with the cGAS/dsDNA complex. Our studies validated that several AMDs were effective inhibitors of IFN-ß production and that they functioned by inhibiting dsDNA stimulation of cGAS. Because AMDs have been widely used in human diseases and have an excellent safety profile, our findings suggest new therapeutic strategies for the treatment of severe debilitating diseases associated with type I IFNs due to cGAS activation.


Assuntos
Antimaláricos/farmacologia , DNA/metabolismo , Interferon beta/biossíntese , Nucleotidiltransferases/metabolismo , Linhagem Celular , DNA/química , Humanos , Modelos Moleculares , Nucleotidiltransferases/química , Ligação Proteica/efeitos dos fármacos , Relação Estrutura-Atividade
10.
J Immunol ; 195(11): 5309-17, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26500348

RESUMO

Microglia play an important role in receptor-mediated phagocytosis in the CNS. In brain abscess and other CNS infections, invading bacteria undergo opsonization with Igs or complement. Microglia recognize these opsonized pathogens by Fc or complement receptors triggering phagocytosis. In this study, we investigated the role of Fcα/µR, the less-studied receptor for IgM and IgA, in microglial phagocytosis. We showed that primary microglia, as well as N9 microglial cells, express Fcα/µR. We also showed that anti-Staphylococcus aureus IgM markedly increased the rate of microglial S. aureus phagocytosis. To unequivocally test the role of Fcα/µR in IgM-mediated phagocytosis, we performed experiments in microglia from Fcα/µR(-/-) mice. Surprisingly, we found that IgM-dependent phagocytosis of S. aureus was similar in microglia derived from wild-type or Fcα/µR(-/-) mice. We hypothesized that IgM-dependent activation of complement receptors might contribute to the IgM-mediated increase in phagocytosis. To test this, we used immunologic and genetic inactivation of complement receptor 3 components (CD11b and CD18) as well as C3. IgM-, but not IgG-mediated phagocytosis of S. aureus was reduced in wild-type microglia and macrophages following preincubation with an anti-CD11b blocking Ab. IgM-dependent phagocytosis of S. aureus was also reduced in microglia derived from CD18(-/-) and C3(-/-) mice. Taken together, our findings implicate complement receptor 3 and C3, but not Fcα/µR, in IgM-mediated phagocytosis of S. aureus by microglia.


Assuntos
Complemento C3/imunologia , Imunoglobulina M/imunologia , Antígeno de Macrófago 1/imunologia , Microglia/imunologia , Fagocitose/imunologia , Animais , Anticorpos Bloqueadores/farmacologia , Antígeno CD11b/imunologia , Antígenos CD18/genética , Antígenos CD18/imunologia , Linhagem Celular , Complemento C3/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Fc/biossíntese , Receptores Fc/genética , Receptores Fc/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia
11.
Clin Immunol ; 163: 84-90, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26769276

RESUMO

Complement activation contributes to inflammation in many diseases, yet it also supports physiologic apoptotic cells (AC) clearance and its downstream immunosuppressive effects. The roles of individual complement components in AC phagocytosis have been difficult to dissect with artificially depleted sera. Using human in vitro systems and the novel antibody complement C1s inhibitor TNT003, we uncoupled the role of the enzymatic activation of the classical pathway from the opsonizing role of C1q in mediating a) the phagocytosis of early and late AC, and b) the immunosuppressive capacity of early AC. We found that C1s inhibition had a small impact on the physiologic clearance of early AC, leaving their immunosuppressive properties entirely unaffected, while mainly inhibiting the phagocytosis of late apoptotic/secondary necrotic cells. Our data suggest that C1s inhibition may represent a valuable therapeutic strategy to control classical pathway activation without causing significant AC accumulation in diseases without defects in AC phagocytosis.


Assuntos
Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Complemento C1q/efeitos dos fármacos , Complemento C1s/antagonistas & inibidores , Citocinas/efeitos dos fármacos , Citofagocitose/efeitos dos fármacos , Tolerância Imunológica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Apoptose/imunologia , Ativação do Complemento/imunologia , Complemento C1q/imunologia , Complemento C1q/metabolismo , Complemento C1s/metabolismo , Complemento C3b/efeitos dos fármacos , Complemento C3b/imunologia , Complemento C3b/metabolismo , Citocinas/imunologia , Citofagocitose/imunologia , Humanos , Técnicas In Vitro , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Células Jurkat , Macrófagos/imunologia , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/imunologia
12.
J Immunol ; 190(3): 886-91, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23303674

RESUMO

Mice overexpressing TLR7 (TLR7.1 mice) are a model of systemic lupus erythematosus pathogenesis and exhibit peripheral myeloid expansion. We show that TLR7.1 mice have a dramatic expansion of splenic cells that derive from granulocyte/macrophage progenitors (GMP) compared with wild-type mice. In the bone marrow, TLR7.1 mice exhibited hallmarks of emergency myelopoiesis and contained a discrete population of Sca-1(+) GMP, termed emergency GMP, which are more proliferative and superior myeloid precursors than classical Sca-1(-) GMP. The emergency myelopoiesis and peripheral myeloid expansion in TLR7.1 mice was dependent on type I IFN signaling. TLR7 agonist administration to nontransgenic mice also drove type I IFN-dependent emergency myelopoiesis. TLR7.1 plasmacytoid dendritic cells were cell-intrinsically activated by TLR7 overexpression and constitutively produced type I IFN mRNA. This study shows that type I IFN can act upon myeloid progenitors to promote the development of emergency GMP, which leads to an expansion of their progeny in the periphery.


Assuntos
Interferon Tipo I/fisiologia , Glicoproteínas de Membrana/fisiologia , Mielopoese/fisiologia , Receptor 7 Toll-Like/fisiologia , Animais , Antígenos Ly/análise , Medula Óssea/patologia , Divisão Celular , Linhagem da Célula , Células Dendríticas/imunologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/imunologia , Granulócitos/patologia , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Macrófagos/patologia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Proteínas de Membrana/análise , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Células Mieloides/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Quimera por Radiação , Receptor de Interferon alfa e beta/deficiência , Transdução de Sinais/fisiologia , Baço/patologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/biossíntese , Receptor 7 Toll-Like/genética
13.
J Immunol ; 190(6): 2536-43, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23382559

RESUMO

TLR7 activation is implicated in the pathogenesis of systemic lupus erythematosus. Mice that overexpress TLR7 develop a lupus-like disease with autoantibodies and glomerulonephritis and early death. To determine whether degradation of the TLR7 ligand RNA would alter the course of disease, we created RNase A transgenic (Tg) mice. We then crossed the RNase Tg to TLR7 Tg mice to create TLR7 × RNase double Tg (DTg) mice. DTg mice had a significantly increased survival associated with reduced activation of T and B lymphocytes and reduced kidney deposition of IgG and C3. We observed massive hepatic inflammation and cell death in TLR7 Tg mice. In contrast, hepatic inflammation and necrosis were strikingly reduced in DTg mice. These findings indicate that high concentrations of serum RNase protect against immune activation and inflammation associated with TLR7 stimulation and that RNase may be a useful therapeutic strategy in the prevention or treatment of inflammation in systemic lupus erythematosus and, possibly, liver diseases.


Assuntos
Regulação para Baixo/genética , Regulação para Baixo/imunologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Ribonuclease Pancreático/genética , Receptor 7 Toll-Like/biossíntese , Receptor 7 Toll-Like/genética , Regulação para Cima/genética , Regulação para Cima/imunologia , Animais , Bovinos , Células Cultivadas , Células-Tronco Embrionárias , Hepatite/enzimologia , Hepatite/imunologia , Hepatite/patologia , Humanos , Inflamação/enzimologia , Inflamação/imunologia , Inflamação/prevenção & controle , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/mortalidade , Lúpus Eritematoso Sistêmico/prevenção & controle , Masculino , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ribonuclease Pancreático/sangue , Ribonuclease Pancreático/fisiologia , Baço/enzimologia , Baço/imunologia , Baço/patologia , Análise de Sobrevida , Receptor 7 Toll-Like/fisiologia
14.
Curr Opin Rheumatol ; 26(5): 459-66, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25036095

RESUMO

PURPOSE OF REVIEW: Systemic lupus erythematosus (SLE) is characterized by autoantibodies directed against nuclear autoantigens normally concealed from immune recognition in healthy individuals. Here, we summarize recently identified mechanisms of abnormal cell death leading to exposure and aberrant processing of nucleoprotein self antigens, and discuss their role in the SLE pathogenesis. RECENT FINDINGS: During the past few years, the unveiling of several new forms of cell death has expanded our understanding beyond the simple view of 'apoptotic' versus 'necrotic' cell death. SLE patients show abnormalities in cell death at several levels, including increased rates of apoptosis, necrosis, and autophagy, as well as reduced clearance of dying cells. These abnormalities lead to an increased autoantigen burden and antigen modifications, such as nucleic acid oxidation that increases the inflammatory properties of self antigens. Recent investigations have highlighted the role of opsonins in determining the immunogenic versus tolerogenic characteristics of self antigens. SUMMARY: Dysregulation of different forms of programmed cell death contributes to increased exposure, availability, and immunogenic characteristics of intracellular self antigens, which all participate in development of lupus autoimmunity. As our understanding of abnormalities of cell death in SLE advances, potential therapeutic opportunities await human implementation.


Assuntos
Apoptose/imunologia , Morte Celular/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Apoptose/genética , Autofagia/genética , Autofagia/imunologia , Morte Celular/genética , Humanos , Lúpus Eritematoso Sistêmico/genética , MicroRNAs/genética , Modelos Biológicos , Necrose , Proteínas Opsonizantes/sangue , Receptores de Superfície Celular/imunologia
15.
Arthritis Rheum ; 65(10): 2713-23, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23840006

RESUMO

OBJECTIVE: Plasmacytoid dendritic cells (PDCs) produce high concentrations of interferon-α (IFNα) following exposure to immune complexes containing nucleic acids. We previously reported that serum from healthy donors inhibits IFNα production by PDCs in response to systemic lupus erythematosus (SLE) immune complexes, and that inhibition is mediated, in part, by IgG. IgG is the major component of intravenous immunoglobulin and is well known to exert antiinflammatory properties. Although suppression of inflammation by the sialylated subfraction of IgG has been implicated in some models, the mechanism of IFNα inhibition by IgG and the importance of sialylation have not been studied. METHODS: SLE immune complexes or synthetic Toll-like receptor (TLR) agonists were used to stimulate total or individual cell-depleted human mononuclear cell cultures in the presence or absence of IgG, Fc fragments, F(ab')2 fragments, and their sialylated or unsialylated subfractions. Cytokines were quantified by enzyme-linked immunosorbent assay. RESULTS: We identified 2 distinct mechanisms by which IgG inhibits IFNα production. First, IgG Fc fragments inhibited SLE immune complex-stimulated IFNα production via a sialic acid-independent mechanism, by inhibiting immune complex binding to Fcγ receptor IIa on PDCs. In contrast, the F(ab')2 fragment of the sialylation-enriched fraction of IgG inhibited TLR-7 or TLR-9 agonist-induced IFNα production but did not require the sialic acid residue itself. The inhibitory activity of IgG on TLR agonist-induced IFNα required monocyte production of prostaglandin E2, a potent suppressor of IFNα production by PDCs. CONCLUSION: IgG attenuates IFNα production by PDCs by both cell surface receptor and intracellular pathways, depending on the nature of the inducing stimulus.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Células Dendríticas/metabolismo , Imunoglobulina G/farmacologia , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/efeitos dos fármacos , Receptores Toll-Like/metabolismo , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/patologia , Dinoprostona/metabolismo , Humanos , Imunoglobulina G/administração & dosagem , Injeções Intravenosas , Interferon-alfa/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Receptores de IgG/metabolismo , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/agonistas
16.
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
17.
Sci Rep ; 14(1): 15511, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38969707

RESUMO

Anti-citrullinated protein autoantibodies (ACPA) are diagnostic for rheumatoid arthritis (RA). The antigens recognized by these autoantibodies are produced by protein arginine deiminases (PADs), particularly PAD4. However, it remains unknown why and how PAD4 causes this aberrant citrullination in RA. Here, we report that poly-perforin pores are present on freshly isolated neutrophils from RA patients, but not on healthy donor neutrophils. Neutrophils with perforin pores also contained intracellular citrullinated proteins in the region adjacent to the pores. This response was replicated in vitro by treating neutrophils with purified perforin, which generated intense dots of anti-perforin immunofluorescence, calcium influx, and intracellular citrullination. Extensive neutrophil killing in Felty's syndrome, an aggressive form of RA, correlated with particularly high ACPA, and PAD4 autoantibodies. In contrast, other forms of death, including NETosis, apoptosis, and pyroptosis, produced minimal citrullination. We conclude that neutrophil targeting by perforin leading to intracellular citrullination takes place in patients with RA.


Assuntos
Anticorpos Antiproteína Citrulinada , Artrite Reumatoide , Citrulinação , Neutrófilos , Perforina , Proteína-Arginina Desiminase do Tipo 4 , Humanos , Neutrófilos/metabolismo , Neutrófilos/imunologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Artrite Reumatoide/imunologia , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Anticorpos Antiproteína Citrulinada/metabolismo , Anticorpos Antiproteína Citrulinada/imunologia , Perforina/metabolismo , Feminino , Masculino , Pessoa de Meia-Idade , Autoanticorpos/imunologia , Desiminases de Arginina em Proteínas/metabolismo , Adulto , Síndrome de Felty/metabolismo , Síndrome de Felty/patologia , Armadilhas Extracelulares/metabolismo , Citrulina/metabolismo , Idoso
18.
Front Immunol ; 14: 1050528, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36923413

RESUMO

Purpose and methods: B cell-activating factor (BAFF) contributes to the pathogenesis of autoimmune diseases including systemic lupus erythematosus (SLE). Although several anti-BAFF Abs and derivatives have been developed for the treatment of SLE, the specific sources of BAFF that sustain autoantibody (auto-Ab) producing cells have not been definitively identified. Using BAFF-RFP reporter mice, we identified major changes in BAFF-producing cells in two mouse spontaneous lupus models (Tlr7 Tg mice and Sle1), and in a pristane-induced lupus (PIL) model. Results: First, we confirmed that similar to their wildtype Tlr7 Tg and Sle1 mice counterparts, BAFF-RFP Tlr7 Tg mice and BAFF-RFP Sle1 mice had increased BAFF serum levels, which correlated with increases in plasma cells and auto-Ab production. Next, using the RFP reporter, we defined which cells had dysregulated BAFF production. BAFF-producing neutrophils (Nphs), monocytes (MOs), cDCs, T cells and B cells were all expanded in the spleens of BAFF-RFP Tlr7 Tg mice and BAFF-RFP Sle1 mice compared to controls. Furthermore, Ly6Chi inflammatory MOs and T cells had significantly increased BAFF expression per cell in both spontaneous lupus models, while CD8- DCs up-regulated BAFF expression only in the Tlr7 Tg mice. Similarly, pristane injection of BAFF-RFP mice induced increases in serum BAFF levels, auto-Abs, and the expansion of BAFF-producing Nphs, MOs, and DCs in both the spleen and peritoneal cavity. BAFF expression in MOs and DCs, in contrast to BAFF from Nphs, was required to maintain homeostatic and pristane-induced systemic BAFF levels and to sustain mature B cell pools in spleens and BMs. Although acting through different mechanisms, Nph, MO and DC sources of BAFF were each required for the development of auto-Abs in PIL mice. Conclusions: Our findings underscore the importance of considering the relative roles of specific myeloid BAFF sources and B cell niches when developing treatments for SLE and other BAFF-associated autoimmune diseases.


Assuntos
Doenças Autoimunes , Lúpus Eritematoso Sistêmico , Animais , Camundongos , Autoanticorpos , Doenças Autoimunes/metabolismo , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Interleucina-4/metabolismo , Monócitos/metabolismo , Neutrófilos , Receptor 7 Toll-Like/metabolismo
19.
Curr Opin Rheumatol ; 24(5): 499-505, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22832823

RESUMO

PURPOSE OF REVIEW: Type I interferon (IFN-I) is strongly implicated in the pathogenesis of systemic lupus erythematosus (SLE). Here, we focus on new developments in pathways of IFN-I stimulation, the role of IFN-I in syndromes associated with lupus-like diseases, the utility of IFN-I signatures as biomarkers, and the progress of therapeutic agents targeting IFN-I pathways in SLE. RECENT FINDINGS: Immune complexesimmune complex are a dominant driver of IFN-I production by activating toll-like receptors (TLRs) in plasmacytoid dendritic cells (pDC) in SLE. The level of IFN-I production is attenuated by C1q in immune complexes and enhanced by natural killer (NK) cells as well as by activated platelets that express CD40L. In addition to immune complexs, cell-intrinsic activation pathways utilize recently described non-TLR RNA and DNA sensors. Some modules or clusters of IFN-I stimulated genes or proteins correlate with disease activity, whereas IFN-I biomarkers of disease flare or specific clinical manifestations need further study. IFN-I blocking studies have reached phase II clinical trials. SUMMARY: Significant progress has been made in defining both TLR as well as non-TLR mediated stimulation of IFN-I. This has helped elucidate the mechanisms of several mutations and common genetic variations in predisposing to SLE. Challenges remain in the establishing the utility of biomarkers and the role of IFN-I blockade in the clinical management of patients with this disease.


Assuntos
Interferon Tipo I/metabolismo , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/imunologia , Fosfatase Ácida/deficiência , Complexo Antígeno-Anticorpo/metabolismo , Biomarcadores/metabolismo , Plaquetas/imunologia , Complemento C1q/metabolismo , Exodesoxirribonucleases/deficiência , Humanos , Imunidade Inata , Isoenzimas/deficiência , Células Matadoras Naturais/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/terapia , Neutrófilos/imunologia , Fosfoproteínas/deficiência , Transdução de Sinais , Fosfatase Ácida Resistente a Tartarato , Receptores Toll-Like/metabolismo
20.
Arthritis Rheum ; 63(1): 118-26, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20862680

RESUMO

OBJECTIVE: To investigate the molecular mechanism for biased interleukin-17 (IL-17) production by DBA/1 CD4+ T cells upon T cell receptor (TCR) and transforming growth factor ß (TGFß) stimulation. METHODS: Purified naive CD4+ T cells were stimulated with anti-CD3/CD28 under Th1, Th2, Th17, and induced T regulatory (iTreg) cell conditions. Cytokine production was assayed by intracellular staining and enzyme-linked immunosorbent assay. Expression of transcription factors was determined by reverse transcription-polymerase chain reaction, flow cytometry, and immunoblotting techniques. RESULTS: Naive CD4+ T cells from DBA/1 mice produced more IL-17 under Th17 cell polarizing conditions as compared with those from C57BL/6 or BALB/c mice. Further investigation revealed no difference among the strains in terms of CD4+ T cell survival, upstream TCR signaling, or CD69 expression or in the phosphorylation of STAT-3 and expression of suppressor of cytokine signaling 3 that positively or negatively regulate IL-17 cell production. However, DBA/1 CD4+ T cells expressed increased levels of retinoic acid-related orphan receptor γt (RORγt). Furthermore, under iTreg cell polarizing conditions, DBA/1 CD4+ T cells showed a strikingly reduced level of FoxP3 expression. When interferon-γ and IL-4 were blocked, FoxP3 expression increased but remained lower in DBA/1 CD4+ T cells following exposure to TGFß as compared with C57BL/6 CD4+ T cells. Moreover, DBA/1 CD4+ T cells showed reduced phosphorylation of Smad2 and Smad3 under both Th17 and iTreg cell polarizing conditions. CONCLUSION: These results indicate that naive CD4+ T cells from DBA/1 mice have a dichotomous response to TGFß: enhanced RORγt, yet reduced FoxP3, up-regulation. This observation may help to elucidate the branch point of TGFß signaling that leads to skewed Th17, but reduced iTreg, cell differentiation.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Camundongos , Camundongos Endogâmicos DBA , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Fator de Crescimento Transformador beta1/imunologia , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA