Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 166(1): 88-101, 2016 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-27293190

RESUMEN

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.


Asunto(s)
Autoanticuerpos/inmunología , Micropartículas Derivadas de Células/química , Cromatina/inmunología , ADN/inmunología , Endodesoxirribonucleasas/genética , Lupus Eritematoso Sistémico/inmunología , Animales , Micropartículas Derivadas de Células/metabolismo , Modelos Animales de Enfermedad , Endodesoxirribonucleasas/deficiencia , Endodesoxirribonucleasas/metabolismo , Humanos , Células Jurkat , Lupus Eritematoso Sistémico/enzimología , Lupus Eritematoso Sistémico/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33397815

RESUMEN

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.


Asunto(s)
Inflamación/sangre , Riñón/metabolismo , Neutrófilos/efectos de la radiación , Piel/efectos de la radiación , Animales , Calgranulina B/genética , Movimiento Celular/efectos de la radiación , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Inflamación/etiología , Inflamación/patología , Interleucina-17/genética , Riñón/lesiones , Riñón/patología , Riñón/efectos de la radiación , Lipocalina 2/genética , Ratones , Neutrófilos/metabolismo , Neutrófilos/patología , Piel/lesiones , Rayos Ultravioleta/efectos adversos , Molécula 1 de Adhesión Celular Vascular/genética
3.
J Immunol ; 204(10): 2627-2640, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32238460

RESUMEN

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.


Asunto(s)
Enfermedades por Deficiencia de Complemento Hereditario/genética , Riñón/patología , Nefritis Lúpica/inmunología , Animales , Anticuerpos Antinucleares/sangre , Antígenos de Superficie/genética , Activación de Complemento , Complemento C1q/genética , Complemento C3/genética , Modelos Animales de Enfermedad , Glomerulonefritis , Enfermedades por Deficiencia de Complemento Hereditario/inmunología , Humanos , Nefritis Lúpica/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Leche/genética , Herencia Multifactorial
4.
Immunity ; 36(1): 120-31, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22284419

RESUMEN

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.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Autoinmunidad/inmunología , Interferones , Linfocitos/inmunología , Animales , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes del Sistema Nervioso/fisiopatología , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Interferones/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Malformaciones del Sistema Nervioso/fisiopatología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transducción de Señal
5.
Annu Rev Med ; 68: 317-330, 2017 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-27813878

RESUMEN

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.


Asunto(s)
Acridinas/farmacología , Antimaláricos/farmacología , Enfermedades Autoinmunes/tratamiento farmacológico , Inmunidad Innata/efectos de los fármacos , Inmunomodulación/efectos de los fármacos , Quinolinas/farmacología , Acridinas/química , Animales , Antimaláricos/química , Artemisininas/farmacología , Artritis Reumatoide/tratamiento farmacológico , Autofagia/efectos de los fármacos , Endosomas/efectos de los fármacos , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Quinolinas/química , Transducción de Señal , Receptores Toll-Like/metabolismo
6.
Annu Rev Med ; 67: 323-36, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26526766

RESUMEN

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.


Asunto(s)
Autoinmunidad/inmunología , Inmunidad Innata , Inflamación/inmunología , Ácidos Nucleicos/inmunología , Receptores Toll-Like/agonistas , Receptores Toll-Like/antagonistas & inhibidores , Adyuvantes Inmunológicos/uso terapéutico , Aminoquinolinas/uso terapéutico , Animales , Citosol/inmunología , ADN Viral/inmunología , Endosomas/inmunología , Humanos , Imiquimod , Inflamación/metabolismo , Lupus Eritematoso Sistémico/inmunología , Psoriasis/inmunología , Transducción de Señal , Receptores Toll-Like/inmunología , Virosis/inmunología
7.
J Immunol ; 197(7): 2854-63, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27534558

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Interferones/inmunología , Lupus Eritematoso Sistémico/genética , ARN/sangre , Adulto , Reacciones Antígeno-Anticuerpo , Autoanticuerpos/inmunología , Estudios de Casos y Controles , Estudios Transversales , Femenino , Humanos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Masculino , Persona de Mediana Edad , ARN/inmunología , ARN Citoplasmático Pequeño/sangre , ARN Nuclear Pequeño/sangre
8.
J Immunol ; 194(9): 4089-93, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25821216

RESUMEN

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.


Asunto(s)
Antimaláricos/farmacología , ADN/metabolismo , Interferón beta/biosíntesis , Nucleotidiltransferasas/metabolismo , Línea Celular , ADN/química , Humanos , Modelos Moleculares , Nucleotidiltransferasas/química , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad
9.
J Immunol ; 195(11): 5309-17, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26500348

RESUMEN

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.


Asunto(s)
Complemento C3/inmunología , Inmunoglobulina M/inmunología , Antígeno de Macrófago-1/inmunología , Microglía/inmunología , Fagocitosis/inmunología , Animales , Anticuerpos Bloqueadores/farmacología , Antígeno CD11b/inmunología , Antígenos CD18/genética , Antígenos CD18/inmunología , Línea Celular , Complemento C3/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Fc/biosíntesis , Receptores Fc/genética , Receptores Fc/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología
10.
Clin Immunol ; 163: 84-90, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26769276

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Complemento C1q/efectos de los fármacos , Complemento C1s/antagonistas & inhibidores , Citocinas/efectos de los fármacos , Citofagocitosis/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Apoptosis/inmunología , Activación de Complemento/inmunología , Complemento C1q/inmunología , Complemento C1q/metabolismo , Complemento C1s/metabolismo , Complemento C3b/efectos de los fármacos , Complemento C3b/inmunología , Complemento C3b/metabolismo , Citocinas/inmunología , Citofagocitosis/inmunología , Humanos , Técnicas In Vitro , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Células Jurkat , Macrófagos/inmunología , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
11.
J Immunol ; 190(3): 886-91, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23303674

RESUMEN

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.


Asunto(s)
Interferón Tipo I/fisiología , Glicoproteínas de Membrana/fisiología , Mielopoyesis/fisiología , Receptor Toll-Like 7/fisiología , Animales , Antígenos Ly/análisis , Médula Ósea/patología , División Celular , Linaje de la Célula , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/inmunología , Granulocitos/patología , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Macrófagos/patología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/análisis , Ratones , Ratones Transgénicos , Modelos Inmunológicos , Células Mieloides/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Quimera por Radiación , Receptor de Interferón alfa y beta/deficiencia , Transducción de Señal/fisiología , Bazo/patología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/biosíntesis , Receptor Toll-Like 7/genética
12.
J Immunol ; 190(6): 2536-43, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23382559

RESUMEN

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.


Asunto(s)
Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ribonucleasa Pancreática/genética , Receptor Toll-Like 7/biosíntesis , Receptor Toll-Like 7/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Animales , Bovinos , Células Cultivadas , Células Madre Embrionarias , Hepatitis/enzimología , Hepatitis/inmunología , Hepatitis/patología , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/prevención & control , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/mortalidad , Lupus Eritematoso Sistémico/prevención & control , Masculino , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Ribonucleasa Pancreática/sangre , Ribonucleasa Pancreática/fisiología , Bazo/enzimología , Bazo/inmunología , Bazo/patología , Análisis de Supervivencia , Receptor Toll-Like 7/fisiología
13.
Curr Opin Rheumatol ; 26(5): 459-66, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25036095

RESUMEN

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.


Asunto(s)
Apoptosis/inmunología , Muerte Celular/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Apoptosis/genética , Autofagia/genética , Autofagia/inmunología , Muerte Celular/genética , Humanos , Lupus Eritematoso Sistémico/genética , MicroARNs/genética , Modelos Biológicos , Necrosis , Proteínas Opsoninas/sangre , Receptores de Superficie Celular/inmunología
14.
Arthritis Rheum ; 65(10): 2713-23, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23840006

RESUMEN

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.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Células Dendríticas/metabolismo , Inmunoglobulina G/farmacología , Interferón-alfa/antagonistas & inhibidores , Interferón-alfa/efectos de los fármacos , Receptores Toll-Like/metabolismo , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Dinoprostona/metabolismo , Humanos , Inmunoglobulina G/administración & dosificación , Inyecciones Intravenosas , Interferón-alfa/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Receptores de IgG/metabolismo , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/agonistas
15.
J Immunol ; 188(2): 902-15, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22147767

RESUMEN

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.


Asunto(s)
Complejo Antígeno-Anticuerpo/fisiología , Complemento C1q/fisiología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Células Dendríticas/patología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interferón-alfa/biosíntesis , Receptores de Lipopolisacáridos/biosíntesis , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Células U937
16.
Front Immunol ; 14: 1050528, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36923413

RESUMEN

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.


Asunto(s)
Enfermedades Autoinmunes , Lupus Eritematoso Sistémico , Animales , Ratones , Autoanticuerpos , Enfermedades Autoinmunes/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Interleucina-4/metabolismo , Monocitos/metabolismo , Neutrófilos , Receptor Toll-Like 7/metabolismo
17.
Curr Opin Rheumatol ; 24(5): 499-505, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22832823

RESUMEN

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.


Asunto(s)
Interferón Tipo I/metabolismo , Lupus Eritematoso Sistémico/etiología , Lupus Eritematoso Sistémico/inmunología , Fosfatasa Ácida/deficiencia , Complejo Antígeno-Anticuerpo/metabolismo , Biomarcadores/metabolismo , Plaquetas/inmunología , Complemento C1q/metabolismo , Exodesoxirribonucleasas/deficiencia , Humanos , Inmunidad Innata , Isoenzimas/deficiencia , Células Asesinas Naturales/inmunología , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/terapia , Neutrófilos/inmunología , Fosfoproteínas/deficiencia , Transducción de Señal , Fosfatasa Ácida Tartratorresistente , Receptores Toll-Like/metabolismo
18.
Arthritis Rheum ; 63(1): 118-26, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20862680

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Animales , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Ratones , Ratones Endogámicos DBA , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Factor de Crecimiento Transformador beta1/inmunología , Factor de Crecimiento Transformador beta1/metabolismo
19.
J Immunol ; 185(8): 4738-49, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20844193

RESUMEN

Almost all humans with homozygous deficiency of C1q develop systemic lupus erythematosus (SLE). The precise cellular mechanism(s) by which C1q prevents the development of SLE remains unclear. In this study, we tested the role of C1q in the regulation of IFN-α induced by immune complexes (ICs) in vitro, as well as the consequences of lack of C1q in vivo. Our experiments revealed that C1q preferentially promotes the binding of SLE ICs to monocytes rather than plasmacytoid dendritic cells, but this inhibition was not due to the induction of inhibitory soluble factors. The presence of C1q also altered the trafficking of ICs within monocytes such that ICs persisted in early endosomes. In patients with C1q deficiency, serum and cerebrospinal fluid levels of IFN-α and IFN-γ-inducible protein-10 levels were elevated and strongly correlated with Ro autoantibodies, demonstrating the clinical significance of these observations. These studies therefore associate C1q deficiency with defective regulation of IFN-α and provide a better understanding of the cellular mechanisms by which C1q prevents the development of IC-stimulated autoimmunity.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Complemento C1q/deficiencia , Células Dendríticas/metabolismo , Interferón-alfa/biosíntesis , Lupus Eritematoso Sistémico/inmunología , Nucleoproteínas/inmunología , Adolescente , Complejo Antígeno-Anticuerpo/metabolismo , Autoanticuerpos/sangre , Autoanticuerpos/líquido cefalorraquídeo , Autoantígenos/inmunología , Separación Celular , Complemento C1q/inmunología , Citocinas/sangre , Citocinas/líquido cefalorraquídeo , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Interferón-alfa/inmunología , Receptores de Lipopolisacáridos/inmunología , Receptores de Lipopolisacáridos/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/fisiopatología , Masculino , Monocitos/inmunología , Monocitos/metabolismo , Nucleoproteínas/metabolismo , Linaje , Adulto Joven
20.
Adv Exp Med Biol ; 750: 14-26, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22903663

RESUMEN

Subsets of IgM naturally occurring autoantibodies (NAbs) bind to the cell surface membranes of dying cells. The antibodies predominantly have specificities against lipid antigens or oxidized lipids. Chief among these lipid antigens are phosphorylcholine (PC) and malondialdehyde (MDA). Antibodies to negatively charged phospholipids such as phosphatidylserine (PS) have been described and there is controversy as to whether these antibodies are related to anticardiolipin antibodies observed in disease states. IgM NAbs that bind to apoptotic cells recruit classical complement pathway components and facilitate phagocytosis by both macrophages and dendritic cells, and may block inflammatory pathways. Under these circumstances, pathologic immune responses to self (autoimmunity) are avoided, whereas mice lacking serum IgM develop a lupus-like disease with associated IgG autoantibody responses. Based on these observations, IgM anti-PC NAbs were found to attenuate inflammation in mouse models of arthritis. IgMNAbs antibodies therefore appear to play pivotal roles in the dampening inflammation and maintenance of tolerance.


Asunto(s)
Autoanticuerpos/inmunología , Inmunidad Innata , Inmunoglobulina M/inmunología , Malondialdehído/inmunología , Fosforilcolina/inmunología , Animales , Apoptosis , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Humanos , Tolerancia Inmunológica , Inmunoglobulina G/inmunología , Ratones , Oxidación-Reducción , Fagocitosis/inmunología , Fosforilcolina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA