Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 143(12): 1193-1197, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38237140

RESUMEN

ABSTRACT: Many patients with antiphospholipid syndrome had decreased ectonucleotidase activity on neutrophils and platelets, which enabled extracellular nucleotides to trigger neutrophil-platelet aggregates. This phenotype was replicated by treating healthy neutrophils and platelets with patient-derived antiphospholipid antibodies or ectonucleotidase inhibitors.


Asunto(s)
Síndrome Antifosfolípido , Humanos , Neutrófilos , Anticuerpos Antifosfolípidos , Plaquetas
2.
Clin Immunol ; 261: 109926, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38355030

RESUMEN

Our study aimed to evaluate the presence, clinical associations, and potential mechanistic roles of non-criteria antiphospholipid antibodies (aPL) and circulating calprotectin, a highly stable marker of neutrophil extracellular trap release (NETosis), in pediatric APS patients. We found that 79% of pediatric APS patients had at least one non-criteria aPL at moderate-to-high titer. Univariate logistic regression demonstrated that positive anti-beta-2 glycoprotein I domain 1 (anti-D1) IgG (p = 0.008), anti-phosphatidylserine/prothrombin (aPS/PT) IgG (p < 0.001), and aPS/PT IgM (p < 0.001) were significantly associated with venous thrombosis. Positive anti-D1 IgG (p < 0.001), aPS/PT IgG (p < 0.001), and aPS/PT IgM (p = 0.001) were also associated with non-thrombotic manifestations of APS, such as thrombocytopenia. Increased levels of calprotectin were detected in children with APS. Calprotectin correlated positively with absolute neutrophil count (r = 0.63, p = 0.008) and negatively with platelet count (r = -0.59, p = 0.015). Mechanistically, plasma from pediatric APS patients with high calprotectin levels impaired platelet viability in a dose-dependent manner.


Asunto(s)
Anticuerpos Antifosfolípidos , Síndrome Antifosfolípido , Humanos , Niño , Biomarcadores , beta 2 Glicoproteína I , Inmunoglobulina G , Inmunoglobulina M , Protrombina , Complejo de Antígeno L1 de Leucocito
3.
Clin Immunol ; 255: 109714, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37527733

RESUMEN

Beta-2 glycoprotein I (ß2GPI) is a phospholipid-binding plasma protein and prominent autoantigen in antiphospholipid syndrome (APS). Here, we tested the hypothesis that ß2GPI might bind to not only phospholipids, but also cell-free DNA and neutrophil extracellular traps (NETs). We report that ß2GPI interacts with cell-free DNA from different species, as well as NETs, in a dose-dependent manner, retarding their migration in an agarose-gel electrophoretic mobility shift assay. We confirm the direct binding interaction of ß2GPI with DNA and NETs by ELISA. We also demonstrate that ß2GPI colocalizes with NET strands by immunofluorescence microscopy. Finally, we provide evidence that ß2GPI-DNA complexes can be detected in the plasma of APS patients, where they positively correlate with an established biomarker of NET remnants. Taken together, our findings indicate that ß2GPI interacts with DNA and NETs, and suggest that this interaction may play a role as a perpetuator and/or instigator of autoimmunity in APS.

4.
Int J Mol Sci ; 24(4)2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36834634

RESUMEN

This study analyzed the nuclease- and serum-driven degradation of millimeter-scale, circular DNA-histone mesostructures (DHMs). DHMs are bioengineered chromatin meshes of defined DNA and histone compositions designed as minimal mimetics of physiological extracellular chromatin structures, such as neutrophil extracellular traps (NETs). Taking advantage of the defined circular shape of the DHMs, an automated time-lapse imaging and image analysis method was developed and used to track DHM degradation and shape changes over time. DHMs were degraded well by 10 U/mL concentrations of deoxyribonuclease I (DNase I) but not by the same level of micrococcal nuclease (MNase), whereas NETs were degraded well by both nucleases. These comparative observations suggest that DHMs have a less accessible chromatin structure compared to NETs. DHMs were degraded by normal human serum, although at a slower rate than NETs. Interestingly, time-lapse images of DHMs revealed qualitative differences in the serum-mediated degradation process compared to that mediated by DNase I. Importantly, despite their reduced susceptibility to degradation and compositional simplicity, the DHMs mimicked NETs in being degraded to a greater extent by normal donor serum compared to serum from a lupus patient with high disease activity. These methods and insights are envisioned to guide the future development and expanded use of DHMs, beyond the previously reported antibacterial and immunostimulatory analyses, to extracellular chromatin-related pathophysiological and diagnostic studies.


Asunto(s)
Cromatina , Trampas Extracelulares , Humanos , Cromatina/metabolismo , Histonas/metabolismo , Neutrófilos/metabolismo , Trampas Extracelulares/metabolismo , ADN/metabolismo , Desoxirribonucleasa I/metabolismo
5.
Rheumatology (Oxford) ; 61(12): 4962-4974, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-35325077

RESUMEN

OBJECTIVE: Leucocyte immunoglobulin-like receptor A3 (LILRA3) belongs to a family of leucocyte receptors. Our previous study reported LILRA3 transcripts were markedly upregulated in neutrophils from patients with APS. We undertook this study to investigate clinical implications of LILRA3 in APS and its potential role in APS-associated thrombosis. METHODS: Two independent cohorts were studied. The first consisted of 294 APS patients, 48 asymptomatic aPL carriers and 150 healthy controls (HCs) from Peking University People's Hospital. The second included 99 APS patients, 25 aPL carriers and 40 HCs from United States APS centres. Serum or plasma concentrations of LILRA3 and MPO-DNA complexes were measured. Additionally, 35 patients with thrombotic APS (tAPS) were evaluated to determine potential effects of immunosuppressive therapy on serum concentrations of LILRA3 and MPO-DNA complexes. RESULTS: Both positivity and serum concentration of LILRA3 were significantly increased in APS patients, especially in those with tAPS. LILRA3-positive tAPS patients displayed more severe thrombotic manifestations. Serum LILRA3 was positively correlated with MPO-DNA complexes in LILRA3-positive tAPS. After immunosuppressive treatment, LILRA3 and MPO-DNA complexes were consistently decreased in tAPS patients. Key findings from the Peking cohort were confirmed in the United States cohort. CONCLUSION: Our study provides first evidence that LILRA3 is aberrantly expressed in APS, especially in patients with tAPS. Serum LILRA3 correlated with MPO-DNA complexes, and the two indices were consistently decreased in tAPS patients after treatment. LILRA3 may play a role in thrombosis of APS and may serve as a biomarker and/or therapeutic target in tAPS.


Asunto(s)
Síndrome Antifosfolípido , Trombosis , Humanos , Síndrome Antifosfolípido/complicaciones , Anticuerpos Antifosfolípidos , Trombosis/etiología , Trombosis/tratamiento farmacológico , Biomarcadores , Estudios de Cohortes , Receptores Inmunológicos
6.
J Thromb Thrombolysis ; 51(2): 446-453, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33151461

RESUMEN

Studies of patients with COVID-19 have demonstrated markedly dysregulated coagulation and a high risk of morbid arterial and venous thrombotic events. Elevated levels of blood neutrophils and neutrophil extracellular traps (NETs) have recently been described in patients with COVID-19. However, their potential role in COVID-19-associated thrombosis remains incompletely understood. In order to elucidate the potential role of hyperactive neutrophils and NET release in COVID-19-associated thrombosis, we conducted a case-control study of patients hospitalized with COVID-19 who developed thrombosis, as compared with gender- and age-matched COVID-19 patients without clinical thrombosis. We found that remnants of NETs (cell-free DNA, myeloperoxidase-DNA complexes, and citrullinated histone H3) and neutrophil-derived S100A8/A9 (calprotectin) in patient sera were associated with higher risk of morbid thrombotic events in spite of prophylactic anticoagulation. These observations underscore the need for urgent investigation into the potential relationship between NETs and unrelenting thrombosis in COVID-19, as well as novel approaches for thrombosis prevention.


Asunto(s)
COVID-19/sangre , Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , SARS-CoV-2/metabolismo , Trombosis/sangre , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/complicaciones , Estudios de Casos y Controles , Femenino , Histonas/sangre , Humanos , Complejo de Antígeno L1 de Leucocito/sangre , Masculino , Persona de Mediana Edad , Trombosis/etiología
7.
Clin Immunol ; 196: 110-116, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30471352

RESUMEN

Antiphospholipid syndrome (APS) is a systemic autoimmune disease characterized by thromboembolic events and pregnancy loss. We sought to characterize the DNA methylation profile of primary APS in comparison to healthy controls and individuals with SLE. In primary APS neutrophils compared to controls, 17 hypomethylated and 25 hypermethylated CpG sites were identified. Notable hypomethylated genes included ETS1, a genetic risk locus for SLE, and PTPN2, a genetic risk locus for other autoimmune diseases. Gene ontology analysis of hypomethylated genes revealed enrichment of genes involved in pregnancy. None of the differentially methylated sites in primary APS were differentially methylated in SLE neutrophils, and there was no demethylation of interferon signature genes in primary APS as is seen in SLE. Hypomethylation within a single probe in the IFI44L promoter (cg06872964) was able to distinguish SLE from primary APS with a sensitivity of 93.3% and specificity of 80.0% at a methylation fraction of 0.329.


Asunto(s)
Síndrome Antifosfolípido/genética , Metilación de ADN , Neutrófilos/metabolismo , Adulto , Anciano , Síndrome Antifosfolípido/inmunología , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Genoma , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Proto-Oncogénica c-ets-1/genética , Proteínas Supresoras de Tumor/genética
8.
Ann Rheum Dis ; 76(2): 450-457, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27432357

RESUMEN

OBJECTIVES: Patients with antiphospholipid syndrome (APS) are at risk for subclinical endothelial injury, as well as accelerated atherosclerosis. In the related disease systemic lupus erythematosus, there is a well-established defect in circulating endothelial progenitors, which leads to an accrual of endothelial damage over time. This defect has been at least partially attributed to exaggerated expression of type I interferons (IFNs). We sought to determine whether these pathways are important in APS. METHODS: We studied 68 patients with primary APS. Endothelial progenitors were assessed by flow cytometry and functional assay. Type I IFN activity was determined by a well-accepted bioassay, while peripheral blood mononuclear cells were scored for expression of IFN-responsive genes. RESULTS: Endothelial progenitors from patients with APS demonstrated a marked defect in the ability to differentiate into endothelial cells, a phenotype which could be mimicked by treating control progenitors with APS sera. Elevated type I IFN activity was detected in the circulation of patients with APS (a finding that was then replicated in an independent cohort). While IgG depletion from APS sera did not rescue endothelial progenitor function, the dysfunction was successfully reversed by a type I IFN receptor-neutralising antibody. CONCLUSIONS: We describe, for the first time to our knowledge, an IFN signature in primary APS and show that this promotes impaired endothelial progenitor function. This work opens the door to novel approaches that may mitigate vascular damage in APS, such as anti-IFN drugs.


Asunto(s)
Síndrome Antifosfolípido/inmunología , Diferenciación Celular/fisiología , Células Progenitoras Endoteliales/fisiología , Interferón-alfa/inmunología , Leucocitos Mononucleares/inmunología , Adulto , Anciano , Anticuerpos Neutralizantes/farmacología , Síndrome Antifosfolípido/fisiopatología , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Células Progenitoras Endoteliales/efectos de los fármacos , Femenino , Citometría de Flujo , Humanos , Interferón Tipo I/inmunología , Masculino , Persona de Mediana Edad , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Adulto Joven
9.
Rheumatology (Oxford) ; 55(11): 1987-1992, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27498356

RESUMEN

OBJECTIVE: Alterations in phenotype and function of endothelial progenitor cells (EPCs) have been associated with poor vascular outcomes and impaired vascular repair in various conditions. Our hypothesis was that patients with PM and DM have dysregulation of EPCs driven by type I IFN and IL-18 similar to other autoimmune diseases. METHODS: Quantification of circulating EPCs was performed by flow cytometry in patients with PM/DM and matched healthy controls. The ability of EPCs to differentiate into mature endothelial cells was investigated by light and fluorescence microscopy quantification in the presence or absence of PM/DM or control serum, neutralizing antibodies to type I IFN receptor or IL-18. Serum type I IFN activity was quantified by induction of type I IFN-inducible genes in HeLa cells. Circulating IL-18 concentrations were assessed by ELISA. RESULTS: Circulating EPCs were significantly lower in PM/DM patients compared with controls. PM/DM EPCs displayed a decreased capacity to differentiate into mature endothelial cells and PM/DM serum significantly inhibited differentiation of control EPCs. This effect was reversed in the majority of samples with neutralizing antibodies to IL-18 or to type I IFN receptor or by a combination of these antibodies. Patients with associated impairments in EPC function had higher type I IFN serum activity. CONCLUSION: PM/DM is associated with dysregulation of EPC phenotype and function that may be attributed, at least in part, to aberrant IL-18 and type I IFN pathways. The implication of these vasculopathic findings for disease prognosis and complications remains to be determined.


Asunto(s)
Dermatomiositis/patología , Células Progenitoras Endoteliales/fisiología , Interferón Tipo I/metabolismo , Polimiositis/patología , Adulto , Anciano , Diferenciación Celular/fisiología , Dermatomiositis/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interleucina-18/metabolismo , Masculino , Persona de Mediana Edad , Polimiositis/metabolismo , Estudios Prospectivos , Receptor de Interferón alfa y beta/metabolismo
10.
Circ Res ; 114(6): 947-56, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24425713

RESUMEN

RATIONALE: Neutrophil extracellular trap (NET) formation promotes vascular damage, thrombosis, and activation of interferon-α-producing plasmacytoid dendritic cells in diseased arteries. Peptidylarginine deiminase inhibition is a strategy that can decrease in vivo NET formation. OBJECTIVE: To test whether peptidylarginine deiminase inhibition, a novel approach to targeting arterial disease, can reduce vascular damage and inhibit innate immune responses in murine models of atherosclerosis. METHODS AND RESULTS: Apolipoprotein-E (Apoe)(-/-) mice demonstrated enhanced NET formation, developed autoantibodies to NETs, and expressed high levels of interferon-α in diseased arteries. Apoe(-/-) mice were treated for 11 weeks with daily injections of Cl-amidine, a peptidylarginine deiminase inhibitor. Peptidylarginine deiminase inhibition blocked NET formation, reduced atherosclerotic lesion area, and delayed time to carotid artery thrombosis in a photochemical injury model. Decreases in atherosclerosis burden were accompanied by reduced recruitment of netting neutrophils and macrophages to arteries, as well as by reduced arterial interferon-α expression. CONCLUSIONS: Pharmacological interventions that block NET formation can reduce atherosclerosis burden and arterial thrombosis in murine systems. These results support a role for aberrant NET formation in the pathogenesis of atherosclerosis through modulation of innate immune responses.


Asunto(s)
Aterosclerosis/prevención & control , Inhibidores Enzimáticos/uso terapéutico , Hidrolasas/antagonistas & inhibidores , Inmunidad Innata/efectos de los fármacos , Ornitina/análogos & derivados , Animales , Enfermedades de la Aorta/tratamiento farmacológico , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/enzimología , Aterosclerosis/etiología , Aterosclerosis/inmunología , Aterosclerosis/patología , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Citrulina/análisis , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Espacio Extracelular , Histonas/metabolismo , Hidrolasas/fisiología , Interferón-alfa/biosíntesis , Interferón-alfa/genética , Selectina L/análisis , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutropenia/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/ultraestructura , Ornitina/farmacología , Ornitina/uso terapéutico , Procesos Fotoquímicos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Arginina Deiminasa Proteína-Tipo 4 , Receptor de Interferón alfa y beta/deficiencia , Seno Aórtico/patología , Túnica Íntima/patología
11.
J Immunol ; 192(3): 906-18, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24379124

RESUMEN

Inappropriate activation of type I IFN plays a key role in the pathogenesis of autoimmune disease, including systemic lupus erythematosus (SLE). In this study, we report the presence of IFN activation in SLE bone marrow (BM), as measured by an IFN gene signature, increased IFN regulated chemokines, and direct production of IFN by BM-resident cells, associated with profound changes in B cell development. The majority of SLE patients had an IFN signature in the BM that was more pronounced than the paired peripheral blood and correlated with both higher autoantibodies and disease activity. Pronounced alterations in B cell development were noted in SLE in the presence of an IFN signature with a reduction in the fraction of pro/pre-B cells, suggesting an inhibition in early B cell development and an expansion of B cells at the transitional stage. These B cell changes strongly correlated with an increase in BAFF and APRIL expression in the IFN-high BM. Furthermore, we found that BM neutrophils in SLE were prime producers of IFN-α and B cell factors. In NZM lupus-prone mice, similar changes in B cell development were observed and mediated by IFN, given abrogation in NZM mice lacking type-I IFNR. BM neutrophils were abundant, responsive to, and producers of IFN, in close proximity to B cells. These results indicate that the BM is an important but previously unrecognized target organ in SLE with neutrophil-mediated IFN activation and alterations in B cell ontogeny and selection.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Médula Ósea/inmunología , Interferón Tipo I/fisiología , Lupus Eritematoso Sistémico/inmunología , Linfopoyesis/inmunología , Neutrófilos/inmunología , Adulto , Animales , Factor Activador de Células B/biosíntesis , Factor Activador de Células B/genética , Subgrupos de Linfocitos B/patología , Médula Ósea/metabolismo , Quimiocinas/biosíntesis , Quimiocinas/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Humanos , Interferón Tipo I/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos , Persona de Mediana Edad , Neutrófilos/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Regulación hacia Arriba/inmunología
12.
Ann Rheum Dis ; 74(7): 1417-24, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24570026

RESUMEN

RATIONALE: The structural and functional integrity of the endothelium is crucial in maintaining vascular homeostasis and preventing atherosclerosis. Patients with systemic lupus erythematosus (SLE) have an increased risk of developing endothelial dysfunction and premature cardiovascular disease. Neutrophil extracellular trap (NET) formation is increased in SLE and has been proposed to contribute to endothelial damage, but the mechanism remains unclear. OBJECTIVE: To determine the mechanism by which enhanced NET formation by low-density granulocytes (LDGs) in SLE contributes to endothelial damage and disrupts the endothelium. RESULTS: The putative role of NET-externalised matrix metalloproteinases (MMPs) in altering the functional integrity of the endothelium was examined. MMP-9 externalised by lupus LDGs during NET formation specifically impaired murine aortic endothelium-dependent vasorelaxation and induced endothelial cell apoptosis. Endothelial dysfunction correlated with the activation of endothelial MMP-2 by MMP-9 present in NETs, while inhibition of MMP-2 activation restored endothelium-dependent function and decreased NET-induced vascular cytotoxicity. Moreover, immunogenic complexes composed of MMP-9 and anti-MMP-9 were identified in SLE sera. These complexes, as well as anti-MMP-9 autoantibodies, induced NETosis and enhanced MMP-9 activity. CONCLUSIONS: These observations implicate activation of endothelial MMP-2 by MMP-9 contained in NETs as an important player in endothelial dysfunction, and MMP-9 as a novel self-antigen in SLE. These results further support that aberrant NET formation plays pathogenic roles in SLE.


Asunto(s)
Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Trampas Extracelulares/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/fisiopatología , Metaloproteinasa 2 de la Matriz/metabolismo , Animales , Aorta/patología , Artritis Reumatoide , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Células Cultivadas , Endotelio Vascular/patología , Femenino , Granulocitos/metabolismo , Granulocitos/patología , Humanos , Lupus Eritematoso Sistémico/patología , Metaloproteinasa 9 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Venas Umbilicales/patología , Regulación hacia Arriba
13.
Ann Rheum Dis ; 74(12): 2199-206, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25104775

RESUMEN

OBJECTIVES: An imbalance between neutrophil extracellular trap (NET) formation and degradation has been described in systemic lupus erythematosus (SLE), potentially contributing to autoantigen externalisation, type I interferon synthesis and endothelial damage. We have demonstrated that peptidylarginine deiminase (PAD) inhibition reduces NET formation and protects against lupus-related vascular damage in the New Zealand Mixed model of lupus. However, another strategy for inhibiting NETs--knockout of NOX2--accelerates lupus in a different murine model, MRL/lpr. Here, we test the effects of PAD inhibition on MRL/lpr mice in order to clarify whether some NET inhibitory pathways may be consistently therapeutic across models of SLE. METHODS: NET formation and autoantibodies to NETs were characterised in lupus-prone MRL/lpr mice. MRL/lpr mice were also treated with two different PAD inhibitors, Cl-amidine and the newly described BB-Cl-amidine. NET formation, endothelial function, interferon signature, nephritis and skin disease were examined in treated mice. RESULTS: Neutrophils from MRL/lpr mice demonstrate accelerated NET formation compared with controls. MRL/lpr mice also form autoantibodies to NETs and have evidence of endothelial dysfunction. PAD inhibition markedly improves endothelial function, while downregulating the expression of type I interferon-regulated genes. PAD inhibition also reduces proteinuria and immune complex deposition in the kidneys, while protecting against skin disease. CONCLUSIONS: PAD inhibition reduces NET formation, while protecting against lupus-related damage to the vasculature, kidneys and skin in various lupus models. The strategy by which NETs are inhibited will have to be carefully considered if human studies are to be undertaken.


Asunto(s)
Amidinas/farmacología , Trampas Extracelulares/metabolismo , Hidrolasas/antagonistas & inhibidores , Enfermedades Renales/prevención & control , Lupus Eritematoso Sistémico/tratamiento farmacológico , Enfermedades Vasculares/prevención & control , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Enfermedades Renales/etiología , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos MRL lpr , Desiminasas de la Arginina Proteica , Enfermedades Vasculares/etiología
14.
J Autoimmun ; 58: 59-66, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25638528

RESUMEN

Recent evidence suggests that neutrophils play an important role in the pathogenesis of lupus. The goal of this study was to characterize the epigenetic architecture, by studying the DNA methylome, of neutrophils and low density granulocytes (LDGs) in lupus patients. We studied 15 lupus patients and 15 healthy age, sex, and ethnicity matched controls. Genome-wide DNA methylation was assessed using the Illumina HumanMethylation 450 BeadChip array, which includes over 485,000 methylation sites across the entire genome. Bisulfite DNA sequencing was used to validate the array results. Statistical and bioinformatic analysis was performed to identify and characterize differentially methylated loci and genes. We identified 293 differentially methylated CG sites in neutrophils between lupus patients and controls. The majority (68%) of differentially methylated CG sites were hypomethylated in lupus neutrophils compared to controls, suggesting overall hypomethylation. We found a robust and consistent demethylation of interferon signature genes in lupus neutrophils, and similar demethylation in the same genes in autologous LDGs. Indeed, the DNA methylome in lupus neutrophils and LDGs was almost identical, suggesting similar chromatin architecture in the two granulocyte subsets. A notable exception was the hypomethylation of a CG site in the promoter region of the cytoskeleton-regulating gene RAC1 in LDGs. Our findings demonstrate a pattern of robust demethylation of interferon signature genes in lupus patients supporting a pathogenic role for neutrophils in lupus. We suggest a model whereby DNA from lupus neutrophils and LDGs externalized by NETosis enhance type-I IFN production via TLR-9 stimulation by hypomethylated DNA.


Asunto(s)
Epigénesis Genética , Interferón Tipo I/genética , Lupus Eritematoso Cutáneo/genética , Neutrófilos/inmunología , Adulto , Cromatina/metabolismo , Metilación de ADN , Epigenómica , Femenino , Perfilación de la Expresión Génica , Humanos , Análisis por Micromatrices , Persona de Mediana Edad , Regulación hacia Arriba , Adulto Joven
15.
J Clin Invest ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38869951

RESUMEN

Neutrophil hyperactivity and neutrophil extracellular trap release (NETosis) appear to play important roles in the pathogenesis of the thromboinflammatory autoimmune disease known as antiphospholipid syndrome (APS). The understanding of neutrophil metabolism has advanced tremendously in the past decade, and accumulating evidence suggests that a variety of metabolic pathways guide neutrophil activities in health and disease. Our previous work characterizing the transcriptome of APS neutrophils revealed that genes related to glycolysis, glycogenolysis, and the pentose phosphate pathway (PPP) were significantly upregulated. Here, we found that APS patient neutrophils used glycolysis more avidly than healthy control neutrophils, especially when the neutrophils were from APS patients with a history of microvascular disease. In vitro, inhibiting either glycolysis or the PPP tempered phorbol myristate acetate- and APS IgG-induced NETosis, but not NETosis triggered by a calcium ionophore. In mice, inhibiting either glycolysis or the PPP reduced neutrophil reactive oxygen species production and suppressed APS IgG-induced NETosis ex vivo. When APS-associated thrombosis was evaluated in mice, inhibiting either glycolysis or the PPP markedly suppressed thrombosis and circulating NET remnants. In summary, these data identify a potential role for restraining neutrophil glucose flux in the treatment of APS.

16.
Arthritis Rheumatol ; 76(6): 928-935, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38225923

RESUMEN

OBJECTIVE: While thrombosis and pregnancy loss are the best-known clinical features of antiphospholipid syndrome (APS), many patients also exhibit "extra-criteria" manifestations, such as thrombocytopenia. The mechanisms that drive APS thrombocytopenia are not completely understood, and no clinical biomarkers are available for predicting antiphospholipid antibody (aPL)-mediated thrombocytopenia. Calprotectin is a heterodimer of S100A8 and S100A9 that is abundant in the neutrophil cytoplasm and released upon proinflammatory neutrophil activation. Here, we sought to evaluate the presence, clinical associations, and potential mechanistic roles of circulating calprotectin in a cohort of primary APS and aPL-positive patients. METHODS: Levels of circulating calprotectin were determined in plasma by the QUANTA Flash chemiluminescent assay. A viability dye-based platelet assay was used to assess the potential impact of calprotectin on aPL-mediated thrombocytopenia. RESULTS: Circulating calprotectin was measured in 112 patients with primary APS and 30 aPL-positive (without APS criteria manifestations or lupus) patients as compared to patients with lupus (without APS), patients with unprovoked venous thrombosis (without aPL), and healthy controls. Levels of calprotectin were higher in patients with primary APS and aPL-positive patients compared to healthy controls. After adjustment for age and sex, calprotectin level correlated positively with absolute neutrophil count (r = 0.41, P < 0.001), positively with C-reactive protein level (r = 0.34, P = 0.002), and negatively with platelet count (r = -0.24, P = 0.004). Mechanistically, we found that calprotectin provoked aPL-mediated thrombocytopenia by engaging platelet surface toll-like receptor 4 and activating the NLRP3-inflammasome, thereby reducing platelet viability in a caspase-1-dependent manner. CONCLUSION: These data suggest that calprotectin has the potential to be a functional biomarker and a new therapeutic target for APS thrombocytopenia.


Asunto(s)
Síndrome Antifosfolípido , Plaquetas , Complejo de Antígeno L1 de Leucocito , Trombocitopenia , Humanos , Síndrome Antifosfolípido/sangre , Femenino , Complejo de Antígeno L1 de Leucocito/sangre , Masculino , Persona de Mediana Edad , Adulto , Trombocitopenia/sangre , Plaquetas/metabolismo , Biomarcadores/sangre , Receptor Toll-Like 4/sangre , Anticuerpos Antifosfolípidos/sangre
17.
J Immunol ; 187(1): 490-500, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21606249

RESUMEN

IL-17 and IL-23 are known to be absolutely central to psoriasis pathogenesis because drugs targeting either cytokine are highly effective treatments for this disease. The efficacy of these drugs has been attributed to blocking the function of IL-17-producing T cells and their IL-23-induced expansion. However, we demonstrate that mast cells and neutrophils, not T cells, are the predominant cell types that contain IL-17 in human skin. IL-17(+) mast cells and neutrophils are found at higher densities than IL-17(+) T cells in psoriasis lesions and frequently release IL-17 in the process of forming specialized structures called extracellular traps. Furthermore, we find that IL-23 and IL-1ß can induce mast cell extracellular trap formation and degranulation of human mast cells. Release of IL-17 from innate immune cells may be central to the pathogenesis of psoriasis, representing a fundamental mechanism by which the IL-23-IL-17 axis mediates host defense and autoimmunity.


Asunto(s)
Espacio Extracelular/metabolismo , Interleucina-17/metabolismo , Mastocitos/metabolismo , Neutrófilos/metabolismo , Psoriasis/metabolismo , Psoriasis/patología , Quimasas/biosíntesis , Humanos , Inmunidad Innata , Recuento de Leucocitos , Mastocitos/enzimología , Mastocitos/patología , Neutrófilos/enzimología , Neutrófilos/patología , Piel/inmunología , Piel/metabolismo , Piel/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Triptasas/biosíntesis
18.
J Immunol ; 187(1): 538-52, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21613614

RESUMEN

An abnormal neutrophil subset has been identified in the PBMC fractions from lupus patients. We have proposed that these low-density granulocytes (LDGs) play an important role in lupus pathogenesis by damaging endothelial cells and synthesizing increased levels of proinflammatory cytokines and type I IFNs. To directly establish LDGs as a distinct neutrophil subset, their gene array profiles were compared with those of autologous normal-density neutrophils and control neutrophils. LDGs significantly overexpress mRNA of various immunostimulatory bactericidal proteins and alarmins, relative to lupus and control neutrophils. In contrast, gene profiles of lupus normal-density neutrophils do not differ from those of controls. LDGs have heightened capacity to synthesize neutrophils extracellular traps (NETs), which display increased externalization of bactericidal, immunostimulatory proteins, and autoantigens, including LL-37, IL-17, and dsDNA. Through NETosis, LDGs have increased capacity to kill endothelial cells and to stimulate IFN-α synthesis by plasmacytoid dendritic cells. Affected skin and kidneys from lupus patients are infiltrated by netting neutrophils, which expose LL-37 and dsDNA. Tissue NETosis is associated with increased anti-dsDNA in sera. These results expand the potential pathogenic roles of aberrant lupus neutrophils and suggest that dysregulation of NET formation and its subsequent responses may play a prominent deleterious role.


Asunto(s)
Adyuvantes Inmunológicos/toxicidad , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Infiltración Neutrófila/inmunología , Autoantígenos/inmunología , Autoantígenos/toxicidad , Línea Celular , Pruebas Inmunológicas de Citotoxicidad , Humanos , Recuento de Leucocitos , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Análisis de Secuencia por Matrices de Oligonucleótidos
19.
Inhal Toxicol ; 25(10): 587-92, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23919441

RESUMEN

CONTEXT: Fine particulate matter (PM) air pollution has been associated with alterations in circulating endothelial progenitor cell (EPC) levels, which may be one mechanism whereby exposures promote cardiovascular diseases. However, the impact of coarse PM on EPCs is unknown. OBJECTIVE: We aimed to determine the effect of acute exposure to coarse concentrated ambient particles (CAP) on circulating EPC levels. METHODS: Thirty-two adults (25.9 ± 6.6 years) were exposed to coarse CAP (76.2 ± 51.5 µg m(-3)) in a rural location and filtered air (FA) for 2 h in a randomized double-blind crossover study. Peripheral venous blood was collected 2 and 20 h post-exposures for circulating EPC (n = 21), white blood cell (n = 24) and vascular endothelial growth factor (VEGF) (n = 16-19) levels. The changes between exposures were compared by matched Wilcoxon signed-rank tests. RESULTS: Circulating EPC levels were elevated 2 [108.29 (6.24-249.71) EPC mL(-1); median (25th-75th percentiles), p = 0.052] and 20 h [106.86 (52.91-278.35) EPC mL(-1), p = 0.008] post-CAP exposure compared to the same time points following FA [38.47 (0.00-84.83) and 50.16 (0.00-104.79) EPC mL(-1)]. VEGF and white blood cell (WBC) levels did not differ between exposures. CONCLUSIONS: Brief inhalation of coarse PM from a rural location elicited an increase in EPCs that persisted for at least 20 h. The underlying mechanism responsible may reflect a systemic reaction to an acute "endothelial injury" and/or a circulating EPC response to sympathetic nervous system activation.


Asunto(s)
Células Endoteliales , Tamaño de la Partícula , Material Particulado/toxicidad , Células Madre , Adulto , Estudios Cruzados , Femenino , Humanos , Masculino , Material Particulado/química , Población Rural , Adulto Joven
20.
Sci Adv ; 9(43): eadj1019, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37878711

RESUMEN

While neutrophil extracellular traps (NETs) have previously been linked to some diabetes-associated complications, such as dysfunctional wound healing, their potential role in diabetic vascular dysfunction has not been studied. Diabetic Akita mice were crossed with either Elane-/- or Pad4-/- mice to generate NET-deficient diabetic mice. By 24 weeks of age, Akita aortae showed markedly impaired relaxation in response to acetylcholine, indicative of vascular dysfunction. Both Akita-Elane-/- mice and Akita-Pad4-/- mice had reduced levels of circulating NETs and improved acetylcholine-mediated aortic relaxation. Compared with wild-type aortae, the thromboxane metabolite TXB2 was roughly 10-fold higher in both intact and endothelium-denuded aortae of Akita mice. In contrast, Akita-Elane-/- and Akita-Pad4-/- aortae had TXB2 levels similar to wild type. In summary, inhibition of NETosis by two independent strategies prevented the development of vascular dysfunction in diabetic Akita mice. Thromboxane was up-regulated in the vessel walls of NETosis-competent diabetic mice, suggesting a role for neutrophils in driving the production of this vasoconstrictive and atherogenic prostanoid.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Trampas Extracelulares , Ratones , Animales , Trampas Extracelulares/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/metabolismo , Acetilcolina , Neutrófilos/metabolismo , Tromboxanos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA