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










Base de datos
Intervalo de año de publicación
1.
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
2.
Nat Rev Nephrol ; 18(9): 558-572, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35732833

RESUMEN

Cells are equipped with numerous sensors that recognize nucleic acids, which probably evolved for defence against viruses. Once triggered, these sensors stimulate the production of type I interferons and other cytokines that activate immune cells and promote an antiviral state. The evolutionary conserved enzyme cyclic GMP-AMP synthase (cGAS) is one of the most recently identified DNA sensors. Upon ligand engagement, cGAS dimerizes and synthesizes the dinucleotide second messenger 2',3'-cyclic GMP-AMP (cGAMP), which binds to the endoplasmic reticulum protein stimulator of interferon genes (STING) with high affinity, thereby unleashing an inflammatory response. cGAS-binding DNA is not restricted by sequence and must only be >45 nucleotides in length; therefore, cGAS can also be stimulated by self genomic or mitochondrial DNA. This broad specificity probably explains why the cGAS-STING pathway has been implicated in a number of autoinflammatory, autoimmune and neurodegenerative diseases; this pathway might also be activated during acute and chronic kidney injury. Therapeutic manipulation of the cGAS-STING pathway, using synthetic cyclic dinucleotides or inhibitors of cGAMP metabolism, promises to enhance immune responses in cancer or viral infections. By contrast, inhibitors of cGAS or STING might be useful in diseases in which this pro-inflammatory pathway is chronically activated.


Asunto(s)
Interferón Tipo I , Proteínas de la Membrana , ADN , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal
3.
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
5.
Sci Rep ; 10(1): 7908, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404939

RESUMEN

Most systemic lupus erythematosus (SLE) patients are photosensitive and ultraviolet B light (UVB) exposure worsens cutaneous disease and precipitates systemic flares of disease. The pathogenic link between skin disease and systemic exacerbations in SLE remains elusive. In an acute model of UVB-triggered inflammation, we observed that a single UV exposure triggered a striking IFN-I signature not only in the skin, but also in the blood and kidneys. The early IFN-I signature was significantly higher in female compared to male mice. The early IFN-I response in the skin was almost entirely, and in the blood partly, dependent on the presence of cGAS, as was skin inflammatory cell infiltration. Inhibition of cGAMP hydrolysis augmented the UVB-triggered IFN-I response. UVB skin exposure leads to cGAS-activation and both local and systemic IFN-I signature and could contribute to acute flares of disease in susceptible subjects such as patients with SLE.


Asunto(s)
Exposición a Riesgos Ambientales , Interferón Tipo I/biosíntesis , Nucleotidiltransferasas/metabolismo , Rayos Ultravioleta/efectos adversos , Animales , Citocinas/metabolismo , Dermatitis/etiología , Dermatitis/metabolismo , Dermatitis/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Noqueados , Nucleotidiltransferasas/genética , Factores Sexuales , Piel/metabolismo , Piel/patología , Piel/efectos de la radiación
6.
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
8.
Front Immunol ; 10: 1243, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31231380

RESUMEN

Signaling through Toll-like receptor 7 (TLR7) drives the production of type I IFN and promotes the activation of autoreactive B cells and is implicated in the pathogenesis of systemic lupus erythematosus (SLE). While TLR7 has been extensively studied in murine lupus, much less is known about its role in the pathogenesis of human SLE. Genetic studies support a link between the TLR7 rs3853839 C/G polymorphism, which affects TLR7 mRNA turnover, and SLE susceptibility; however, the effects of this polymorphism on B cells have not been studied. Here we determined how changes in TLR7 expression affect peripheral B cells and auto-Ab production in SLE patients. High TLR7 expression in SLE patients driven by TLR7 rs3853839 C/G polymorphism was associated with more active disease and upregulation of IFN-responsive genes. TLR7hi SLE patients showed an increase in peripheral B cells. Most notably, the percentage and numbers of CD19+CD24++CD38++ newly-formed transitional (TR) B cells were increased in TLR7hi SLE patients as compared to HCs and TLR7norm/lo SLE patients. Using auto-Ab arrays, we found an increase and enrichment of auto-Ab specificities in the TLR7hi SLE group, including the production of anti-RNA/RNP-Abs. Upon in vitro TLR7 ligand stimulation, TR B cells isolated from TLR7hi but not TLR7norm/lo SLE patients produced anti-nuclear auto-Abs (ANA). Exposure of TR B cells isolated from cord blood to IFNα induced the expression of TLR7 and enabled their activation in response to TLR7 ligation in vitro. Our study shows that overexpression of TLR7 in SLE patients drives the expansion of TR B cells. High TLR7 signaling in TR B cells promotes auto-Ab production, supporting a possible pathogenic role of TR B cells in human SLE.


Asunto(s)
Autoanticuerpos/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Expresión Génica , Lupus Eritematoso Sistémico/etiología , Lupus Eritematoso Sistémico/metabolismo , Receptor Toll-Like 7/genética , Autoinmunidad , Susceptibilidad a Enfermedades , Femenino , Humanos , Inmunofenotipificación , Interferón-alfa/metabolismo , Lupus Eritematoso Sistémico/patología , Masculino , Receptor Toll-Like 7/metabolismo
9.
Nat Rev Rheumatol ; 15(5): 254-255, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30914774
10.
Science ; 363(6423)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30630901

RESUMEN

Cytopenias are an important clinical problem associated with inflammatory disease and infection. We show that specialized phagocytes that internalize red blood cells develop in Toll-like receptor 7 (TLR7)-driven inflammation. TLR7 signaling caused the development of inflammatory hemophagocytes (iHPCs), which resemble splenic red pulp macrophages but are a distinct population derived from Ly6Chi monocytes. iHPCs were responsible for anemia and thrombocytopenia in TLR7-overexpressing mice, which have a macrophage activation syndrome (MAS)-like disease. Interferon regulatory factor 5 (IRF5), associated with MAS, participated in TLR7-driven iHPC differentiation. We also found iHPCs during experimental malarial anemia, in which they required endosomal TLR and MyD88 signaling for differentiation. Our findings uncover a mechanism by which TLR7 and TLR9 specify monocyte fate and identify a specialized population of phagocytes responsible for anemia and thrombocytopenia associated with inflammation and infection.


Asunto(s)
Anemia/fisiopatología , Síndrome de Activación Macrofágica/fisiopatología , Glicoproteínas de Membrana/fisiología , Fagocitos/citología , Transducción de Señal , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 9/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Inflamación/fisiopatología , Factores Reguladores del Interferón/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Factor 88 de Diferenciación Mieloide/fisiología , Plasmodium yoelii , Bazo/citología , Trombocitopenia/fisiopatología , Transcriptoma
11.
Arthritis Rheumatol ; 70(11): 1807-1819, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29781188

RESUMEN

OBJECTIVE: Type I interferon (IFN) is strongly implicated in the pathogenesis of systemic lupus erythematosus (SLE) as well as rare monogenic interferonopathies such as Aicardi-Goutières syndrome (AGS), a disease attributed to mutations in the DNA exonuclease TREX1. The DNA-activated type I IFN pathway cyclic GMP-AMP (cGAMP) synthase (cGAS) is linked to subsets of AGS and lupus. This study was undertaken to identify inhibitors of the DNA-cGAS interaction, and to test the lead candidate drug, X6, in a mouse model of AGS. METHODS: Trex1-/- mice were treated orally from birth with either X6 or hydroxychloroquine (HCQ) for 8 weeks. Expression of IFN-stimulated genes (ISGs) was quantified by quantitative polymerase chain reaction. Multiple reaction monitoring by ultra-performance liquid chromatography coupled with tandem mass spectrometry was used to quantify the production of cGAMP and X6 drug concentrations in the serum and heart tissue of Trex1-/- mice. RESULTS: On the basis of the efficacy-to-toxicity ratio established in vitro, drug X6 was selected as the lead candidate for treatment of Trex1-/- mice. X6 was significantly more effective than HCQ in attenuating ISG expression in mouse spleens (P < 0.01 for Isg15 and Isg20) and hearts (P < 0.05 for Isg15, Mx1, and Ifnb, and P < 0.01 for Cxcl10), and in reducing the production of cGAMP in mouse heart tissue (P < 0.05), thus demonstrating target engagement by the X6 compound. Of note, X6 was also more effective than HCQ in reducing ISG expression in vitro (P < 0.05 for IFI27 and MX1, and P < 0.01 for IFI44L and PKR) in human peripheral blood mononuclear cells from patients with SLE. CONCLUSION: This study demonstrates that X6 is superior to HCQ for the treatment of an experimental autoimmune myocarditis mediated in vivo by the cGAS/stimulator of IFN genes (cGAS/STING) pathway. The findings suggest that drug X6 could be developed as a novel treatment for AGS and/or lupus to inhibit activation of the cGAS/STING pathway.


Asunto(s)
Aminoacridinas/farmacología , Antimaláricos/farmacología , Exodesoxirribonucleasas/genética , Corazón/efectos de los fármacos , Interferón beta/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Nucleotidiltransferasas/efectos de los fármacos , Fosfoproteínas/genética , Animales , Quimiocina CXCL10/efectos de los fármacos , Quimiocina CXCL10/genética , Cromatografía Liquida , Citocinas/efectos de los fármacos , Citocinas/genética , Humanos , Hidroxicloroquina/farmacología , Técnicas In Vitro , Interferón beta/genética , Interferón beta/metabolismo , Leucocitos Mononucleares/metabolismo , Lupus Eritematoso Sistémico , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Proteínas de Resistencia a Mixovirus/efectos de los fármacos , Proteínas de Resistencia a Mixovirus/genética , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/genética , Tamaño de los Órganos , Reacción en Cadena de la Polimerasa , Bazo/efectos de los fármacos , Bazo/patología , Espectrometría de Masas en Tándem , Ubiquitinas/efectos de los fármacos , Ubiquitinas/genética
12.
Arthritis Rheumatol ; 70(6): 805-816, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29439290

RESUMEN

Cells of the innate immune system are rigged with sensors that detect nucleic acids derived from microbes, especially viruses. It has become clear that these same sensors that respond to nucleic acids derived from damaged cells or defective intracellular processing are implicated in triggering diseases such as lupus and arthritis. The ways in which cells die and the concomitant presence of proteins and peptides that allow nucleic acids to re-enter cells profoundly influence innate immune responses. In this review, we briefly discusses different types of programmed necrosis, such as pyroptosis, necroptosis, and NETosis, and explains how nucleic acids can engage intracellular receptors and stimulate inflammation. Host protective mechanisms that include compartmentalization of receptors and nucleases as well as the consequences of nuclease deficiencies are explored. In addition, proximal and distal targets in the nucleic acid stimulation of inflammation are discussed in terms of their potential amenability to therapy for the attenuation of innate immune activation and disease pathogenesis.


Asunto(s)
Muerte Celular/inmunología , Inmunidad Innata/genética , Inflamación/genética , Ácidos Nucleicos/inmunología , Humanos
13.
J Exp Med ; 214(7): 2103-2119, 2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28606989

RESUMEN

Neutrophils play a crucial role in host defense. However, neutrophil activation is also linked to autoimmune diseases such as systemic lupus erythematosus (SLE), where nucleic acid-containing immune complexes (IC) drive inflammation. The role of Toll-like receptor (TLR) signaling in processing of SLE ICs and downstream inflammatory neutrophil effector functions is not known. We observed that TLR7/8 activation leads to a furin-dependent proteolytic cleavage of the N-terminal part of FcgRIIA, shifting neutrophils away from phagocytosis of ICs toward the programmed form of necrosis, NETosis. TLR7/8-activated neutrophils promoted cleavage of FcgRIIA on plasmacytoid dendritic cells and monocytes, resulting in impaired overall clearance of ICs and increased complement C5a generation. Importantly, ex vivo derived activated neutrophils from SLE patients demonstrated a similar cleavage of FcgRIIA that was correlated with markers of disease activity, as well as complement activation. Therapeutic approaches aimed at blocking TLR7/8 activation would be predicted to increase phagocytosis of circulating ICs, while disarming their inflammatory potential.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Receptores de IgG/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología , Western Blotting , Células Cultivadas , Activación de Complemento/inmunología , Complemento C5a/inmunología , Complemento C5a/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Citometría de Flujo , Humanos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Neutrófilos/metabolismo , Receptores de IgG/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo
14.
Arthritis Rheumatol ; 69(1): 131-142, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27390188

RESUMEN

OBJECTIVE: Mutations in the ACP5 gene, which encodes tartrate-resistant acid phosphatase (TRAP), cause the immuno-osseous disorder spondyloenchondrodysplasia, which includes as disease features systemic lupus erythematosus (SLE) and a type I interferon (IFN) signature. Our aims were to identify TRAP substrates, determine the consequences of TRAP deficiency in immune cells, and assess whether ACP5 mutations are enriched in sporadic cases of SLE. METHODS: Interaction between TRAP and its binding partners was tested by a yeast 2-hybrid screening, confocal microscopy, and immunoprecipitation/Western blotting. TRAP knockdown was performed using small interfering RNA. Phosphorylation of osteopontin (OPN) was analyzed by mass spectrometry. Nucleotide sequence analysis of ACP5 was performed by Sanger sequencing or next-generation sequencing. RESULTS: TRAP and OPN colocalized and interacted in human macrophages and plasmacytoid dendritic cells (PDCs). TRAP dephosphorylated 3 serine residues on specific OPN peptides. TRAP knockdown resulted in increased OPN phosphorylation and increased nuclear translocation of IRF7 and P65, with resultant heightened expression of IFN-stimulated genes and IL6 and TNF following Toll-like receptor 9 stimulation. An excess of heterozygous ACP5 missense variants was observed in SLE compared to controls (P = 0.04), and transfection experiments revealed a significant reduction in TRAP activity in a number of variants. CONCLUSION: Our findings indicate that TRAP and OPN colocalize and that OPN is a substrate for TRAP in human immune cells. TRAP deficiency in PDCs leads to increased IFNα production, providing at least a partial explanation for how ACP5 mutations cause lupus in the context of spondyloenchondrodysplasia. Detection of ACP5 missense variants in a lupus cohort suggests that impaired TRAP functioning may increase susceptibility to sporadic lupus.


Asunto(s)
Lupus Eritematoso Sistémico/enzimología , Lupus Eritematoso Sistémico/etiología , Mutación , Fosfatasa Ácida Tartratorresistente/deficiencia , Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Humanos , Macrófagos , Osteocondrodisplasias/genética , Osteopontina/metabolismo , Fosforilación , Fosfatasa Ácida Tartratorresistente/genética
15.
Arthritis Rheumatol ; 69(4): 826-836, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27863141

RESUMEN

OBJECTIVE: Photosensitivity is common in patients with systemic lupus erythematosus, although the mechanisms linking ultraviolet (UV) light to flares are not well understood. We undertook this study to determine whether repetitive UVB exposure could induce type I interferon (IFN) production in normal mouse skin, and to investigate the roles of inflammatory monocytes and plasmacytoid dendritic cells (PDCs) in type I IFN production and development of UVB irradiation-induced inflammation. METHODS: Mice were irradiated with UVB at 100 mJ/cm2 for 5 days, and cutaneous manifestations were examined by messenger RNA expression of inflammatory and type I IFN response genes, histology, and flow cytometry. Inflammatory monocyte and PDC depletion experiments were performed in CCR2-diphtheria toxin receptor (DTR)-transgenic mice and blood dendritic cell antigen 2-DTR-transgenic mice. The roles of type I IFN and of the adaptor protein stimulator of IFN genes (STING) in UVB irradiation-induced inflammation were investigated using IFN-α/ß/ω receptor (IFNAR)-knockout mice and STING-knockout mice. RESULTS: Repeated UVB irradiation stimulated an inflammatory cell infiltrate and induction of type I IFN and proinflammatory cytokines. Interestingly, the type I IFN response was independent of PDCs but dependent on inflammatory monocytes, which were recruited following UVB irradiation. The adaptor protein STING was necessary for both type I IFN and proinflammatory cytokine expression in the skin. UVB-irradiated IFNAR-knockout mice showed increased levels of proinflammatory genes and more severe inflammation by histology, suggesting a protective role for type I IFN. CONCLUSION: In wild-type mice, repeated doses of UVB irradiation induce monocyte-dependent and PDC-independent expression of type I IFN together with expression of other proinflammatory cytokines. Induction is dependent on the adaptor protein STING. Surprisingly, studies using IFNAR-deficient mice revealed that type I IFN protects against UVB irradiation-induced skin inflammation, in part by attenuating proinflammatory cytokine expression and limiting tissue damage.


Asunto(s)
Interferón Tipo I/biosíntesis , Monocitos/efectos de la radiación , Piel/metabolismo , Piel/efectos de la radiación , Rayos Ultravioleta , Animales , Citocinas/biosíntesis , Células Dendríticas/fisiología , Células Dendríticas/efectos de la radiación , Inflamación/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monocitos/fisiología
16.
Arthritis Rheumatol ; 69(4): 800-807, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27863149

RESUMEN

OBJECTIVE: Type I interferon (IFN) is implicated in the pathogenesis of systemic lupus erythematosus (SLE) and interferonopathies such as Aicardi-Goutières syndrome. A recently discovered DNA-activated type I IFN pathway, cyclic GMP-AMP synthase (cGAS), has been linked to Aicardi-Goutières syndrome and mouse models of lupus. The aim of this study was to determine whether the cGAS pathway contributes to type I IFN production in patients with SLE. METHODS: SLE disease activity was measured by the Safety of Estrogens in Lupus Erythematosus National Assessment version of the Systemic Lupus Erythematosus Disease Activity Index. Expression of messenger RNA for cGAS and IFN-stimulated genes (ISGs) was determined by quantitative polymerase chain reaction analysis. Cyclic GMP-AMP (cGAMP) levels were examined by multiple reaction monitoring with ultra-performance liquid chromatography tandem mass spectrometry. RESULTS: Expression of cGAS in peripheral blood mononuclear cells (PBMCs) was significantly higher in SLE patients than in normal controls (n = 51 and n = 20 respectively; P < 0.01). There was a positive correlation between cGAS expression and the IFN score (P < 0.001). The expression of cGAS in PBMCs showed a dose response to type I IFN stimulation in vitro, consistent with it being an ISG. Targeted measurement of cGAMP by tandem mass spectrometry detected cGAMP in 15% of the SLE patients (7 of 48) but none of the normal (0 of 19) or rheumatoid arthritis (0 of 22) controls. Disease activity was higher in SLE patients with cGAMP versus those without cGAMP. CONCLUSION: Increased cGAS expression and cGAMP in a proportion of SLE patients indicates that the cGAS pathway should be considered as a contributor to type I IFN production. Whereas higher cGAS expression may be a consequence of exposure to type I IFN, detection of cGAMP in patients with increased disease activity indicates potential involvement of this pathway in disease expression.


Asunto(s)
Lupus Eritematoso Sistémico/enzimología , Nucleotidiltransferasas/biosíntesis , Adulto , Femenino , Humanos , Lupus Eritematoso Sistémico/sangre , Masculino , Nucleótidos Cíclicos/sangre , Transducción de Señal
17.
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
18.
Rambam Maimonides Med J ; 7(4)2016 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-27824546

RESUMEN

Anti-citrullinated protein antibodies (ACPAs) are highly specific serologic markers for rheumatoid arthritis (RA) and can pre-date clinical disease onset by up to 10 years, also predicting erosive disease. The process of citrullination, the post-translational conversion of arginine to citrulline residues, is mediated by peptidylarginine deiminase (PAD) enzymes present in polymorphonuclear cells (PMNs). Calcium ions (Ca2+) are required for PAD activation, but the intracellular Ca2+ concentration in normal cells is much lower than the optimal Ca2+ concentration needed for PAD activation. For this reason, it has been proposed that PAD activation, and thus citrullination, occurs only during PMN cell death when PAD enzymes leak out of the cells into the extracellular matrix, or extracellular Ca2+ enters the cells, with the high Ca2+ concentration activating PAD. Recently, using artificial in vitro systems to corroborate their hypothesis, Romero et al. demonstrated that "hypercitrullination," citrullination of multiple intracellular proteins, occurs within synovial fluid (SF) cells of RA patients, and that only modes of death leading to membranolysis such as perforin-granzyme pathway or complement membrane attack complex activation cause hypercitrullination. In order for Romero's hypothesis to hold, it is reasonable to surmise that PMN-directed lysis should occur in the rheumatoid joint or the circulation of RA patients. Research conducted thus far has shown that immunoglobulin G (IgG) targeting PMNs are present in RA SF and mediate PMN activation. However, the role of anti-PMN IgG in mediating complement activation and subsequent PMN lysis and hypercitrullination has not been fully evaluated.

19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...