Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
Arthritis Rheumatol ; 74(5): 902-907, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34725956

RESUMEN

OBJECTIVE: Defibrotide is a heterogenous mixture of polyanionic oligonucleotides currently approved for treatment of transplant-associated venoocclusive disease. While defibrotide has a known role in limiting endothelial cell activation, some studies have also demonstrated anti-leukocyte properties. In a recent study, we found that neutrophil extracellular traps (NETs) play a role in the thrombotic complications of antiphospholipid syndrome (APS). In the present study, we investigated the hypothesis that defibrotide might act to mitigate APS-relevant NET formation in vitro and in mouse models. METHODS: We used in vitro assays and a mouse model to determine the mechanisms by which defibrotide inhibits NET formation and venous thrombosis in APS. RESULTS: At doses ranging from 1 to 10 µg/ml, defibrotide significantly suppressed NET formation from control neutrophils stimulated with IgG isolated from patients with APS. Defibrotide increased levels of intracellular cyclic AMP in neutrophils, and its suppressive effects on NET formation were mitigated by blocking adenosine A2A receptor or by inhibiting the cyclic AMP-dependent kinase protein kinase A. Defibrotide at doses ranging from 15 to 150 mg/kg/day inhibited NET formation and venous thrombosis in a model of antiphospholipid antibody-accelerated thrombosis-an effect that was reduced in adenosine A2A receptor-knockout mice. CONCLUSION: This study is the first to demonstrate mechanisms by which defibrotide counteracts neutrophil-mediated thrombotic inflammation inherent to APS.


Asunto(s)
Síndrome Antifosfolípido , Trampas Extracelulares , Trombosis , Trombosis de la Vena , Animales , Anticuerpos Antifosfolípidos , Síndrome Antifosfolípido/tratamiento farmacológico , Síndrome Antifosfolípido/metabolismo , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Humanos , Ratones , Neutrófilos/metabolismo , Polidesoxirribonucleótidos , Receptor de Adenosina A2A , Trombosis/etiología , Trombosis/prevención & control
2.
Viruses ; 13(11)2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34835015

RESUMEN

Men are disproportionately affected by the coronavirus disease-2019 (COVID-19), and face higher odds of severe illness and death compared to women. The vascular effects of androgen signaling and inflammatory cytokines in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-mediated endothelial injury are not defined. We determined the effects of SARS-CoV-2 spike protein-mediated endothelial injury under conditions of exposure to androgen dihydrotestosterone (DHT) and tumor necrosis factor-a (TNF-α) and tested potentially therapeutic effects of mineralocorticoid receptor antagonism by spironolactone. Circulating endothelial injury markers VCAM-1 and E-selectin were measured in men and women diagnosed with COVID-19. Exposure of endothelial cells (ECs) in vitro to DHT exacerbated spike protein S1-mediated endothelial injury transcripts for the cell adhesion molecules E-selectin, VCAM-1 and ICAM-1 and anti-fibrinolytic PAI-1 (p < 0.05), and increased THP-1 monocyte adhesion to ECs (p = 0.032). Spironolactone dramatically reduced DHT+S1-induced endothelial activation. TNF-α exacerbated S1-induced EC activation, which was abrogated by pretreatment with spironolactone. Analysis from patients hospitalized with COVID-19 showed concordant higher circulating VCAM-1 and E-Selectin levels in men, compared to women. A beneficial effect of the FDA-approved drug spironolactone was observed on endothelial cells in vitro, supporting a rationale for further evaluation of mineralocorticoid antagonism as an adjunct treatment in COVID-19.


Asunto(s)
COVID-19/patología , Dihidrotestosterona/farmacología , Endotelio Vascular/patología , Inflamación , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/fisiología , Espironolactona/farmacología , Antagonistas de Receptores de Angiotensina/farmacología , COVID-19/fisiopatología , COVID-19/virología , Moléculas de Adhesión Celular/sangre , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Humanos , Masculino , Caracteres Sexuales , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/fisiología , Valsartán/farmacología
3.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34264868

RESUMEN

Neutrophil-mediated activation and injury of the endothelium play roles in the pathogenesis of diverse disease states ranging from autoimmunity to cancer to COVID-19. Neutralization of cationic proteins (such as neutrophil extracellular trap-derived [NET-derived] histones) with polyanionic compounds has been suggested as a potential strategy for protecting the endothelium from such insults. Here, we report that the US Food and Drug Administration-approved polyanionic agent defibrotide (a pleiotropic mixture of oligonucleotides) directly engages histones and thereby blocks their pathological effects on endothelium. In vitro, defibrotide counteracted endothelial cell activation and pyroptosis-mediated cell death, whether triggered by purified NETs or recombinant histone H4. In vivo, defibrotide stabilized the endothelium and protected against histone-accelerated inferior vena cava thrombosis in mice. Mechanistically, defibrotide demonstrated direct and tight binding to histone H4 as detected by both electrophoretic mobility shift assay and surface plasmon resonance. Taken together, these data provide insights into the potential role of polyanionic compounds in protecting the endothelium from thromboinflammation with potential implications for myriad NET- and histone-accelerated disease states.


Asunto(s)
Fibrinolíticos/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Polidesoxirribonucleótidos/farmacología , Trombosis/tratamiento farmacológico , Animales , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Fibrinolíticos/uso terapéutico , Histonas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Polidesoxirribonucleótidos/uso terapéutico , Piroptosis
4.
medRxiv ; 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33655266

RESUMEN

Neutrophil-mediated activation and injury of the endothelium play a role in the pathogenesis of diverse disease states ranging from autoimmunity to cancer to COVID-19. Neutralization of cationic proteins (such as neutrophil extracellular trap/NET-derived histones) with polyanionic compounds has been suggested as a potential strategy for protecting the endothelium from such insults. Here, we report that the FDA-approved polyanionic agent defibrotide (a pleiotropic mixture of oligonucleotides) directly engages histones and thereby blocks their pathological effects on endothelium. In vitro , defibrotide counteracted endothelial cell activation and pyroptosis-mediated cell death, whether triggered by purified NETs or recombinant histone H4. In vivo , defibrotide stabilized the endothelium and protected against histone-accelerated inferior vena cava thrombosis in mice. Mechanistically, defibrotide demonstrated direct and tight binding to histone H4 as detected by both electrophoretic mobility shift assay and surface plasmon resonance. Taken together, these data provide insights into the potential role of polyanionic compounds in protecting the endothelium from thromboinflammation with potential implications for myriad NET- and histone-accelerated disease states.

5.
J Leukoc Biol ; 109(1): 67-72, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32869342

RESUMEN

Severe cases of coronavirus disease 2019 (COVID-19) are regularly complicated by respiratory failure. Although it has been suggested that elevated levels of blood neutrophils associate with worsening oxygenation in COVID-19, it is unknown whether neutrophils are drivers of the thrombo-inflammatory storm or simple bystanders. To better understand the potential role of neutrophils in COVID-19, we measured levels of the neutrophil activation marker S100A8/A9 (calprotectin) in hospitalized patients and determined its relationship to severity of illness and respiratory status. Patients with COVID-19 (n = 172) had markedly elevated levels of calprotectin in their blood. Calprotectin tracked with other acute phase reactants including C-reactive protein, ferritin, lactate dehydrogenase, and absolute neutrophil count, but was superior in identifying patients requiring mechanical ventilation. In longitudinal samples, calprotectin rose as oxygenation worsened. When tested on day 1 or 2 of hospitalization (n = 94 patients), calprotectin levels were significantly higher in patients who progressed to severe COVID-19 requiring mechanical ventilation (8039 ± 7031 ng/ml, n = 32) as compared to those who remained free of intubation (3365 ± 3146, P < 0.0001). In summary, serum calprotectin levels track closely with current and future COVID-19 severity, implicating neutrophils as potential perpetuators of inflammation and respiratory compromise in COVID-19.


Asunto(s)
COVID-19 , Calgranulina A , Calgranulina B , Activación Neutrófila , Neutrófilos , SARS-CoV-2 , COVID-19/sangre , COVID-19/inmunología , COVID-19/patología , COVID-19/terapia , Calgranulina A/sangre , Calgranulina A/inmunología , Calgranulina B/sangre , Calgranulina B/inmunología , Femenino , Hospitalización , Humanos , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo , Índice de Severidad de la Enfermedad , Factores de Tiempo
6.
Nat Commun ; 10(1): 1916, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015489

RESUMEN

Potentiation of neutrophil extracellular trap (NET) release is one mechanism by which antiphospholipid antibodies (aPL Abs) effect thrombotic events in patients with antiphospholipid syndrome (APS). Surface adenosine receptors trigger cyclic AMP (cAMP) formation in neutrophils, and this mechanism has been proposed to regulate NETosis in some contexts. Here we report that selective agonism of the adenosine A2A receptor (CGS21680) suppresses aPL Ab-mediated NETosis in protein kinase A-dependent fashion. CGS21680 also reduces thrombosis in the inferior vena cavae of both control mice and mice administered aPL Abs. The antithrombotic medication dipyridamole is known to potentiate adenosine signaling by increasing extracellular concentrations of adenosine and interfering with the breakdown of cAMP. Like CGS21680, dipyridamole suppresses aPL Ab-mediated NETosis via the adenosine A2A receptor and mitigates venous thrombosis in mice. In summary, these data suggest an anti-inflammatory therapeutic paradigm in APS, which may extend to thrombotic disease in the general population.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Adenosina/análogos & derivados , Síndrome Antifosfolípido/tratamiento farmacológico , Trampas Extracelulares/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Fenetilaminas/farmacología , Trombosis de la Vena/tratamiento farmacológico , Adenosina/inmunología , Adenosina/metabolismo , Adenosina/farmacología , Animales , Anticuerpos Antifosfolípidos/sangre , Síndrome Antifosfolípido/genética , Síndrome Antifosfolípido/inmunología , Síndrome Antifosfolípido/patología , AMP Cíclico/inmunología , AMP Cíclico/metabolismo , Dipiridamol/farmacología , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Fibrinolíticos/farmacología , Regulación de la Expresión Génica , Humanos , Inmunoglobulina G/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/inmunología , Transducción de Señal , Vena Cava Inferior/efectos de los fármacos , Vena Cava Inferior/inmunología , Vena Cava Inferior/metabolismo , Trombosis de la Vena/genética , Trombosis de la Vena/inmunología , Trombosis de la Vena/patología
7.
J Clin Invest ; 129(7): 2872-2877, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30990798

RESUMEN

Deep vein thrombosis (DVT), caused by alterations in venous homeostasis is the third most common cause of cardiovascular mortality; however, key molecular determinants in venous thrombosis have not been fully elucidated. Several lines of evidence indicate that DVT occurs at the intersection of dysregulated inflammation and coagulation. The enzyme ectonucleoside tri(di)phosphohydrolase (ENTPD1, also known as CD39) is a vascular ecto-apyrase on the surface of leukocytes and the endothelium that inhibits intravascular inflammation and thrombosis by hydrolysis of phosphodiester bonds from nucleotides released by activated cells. Here, we evaluated the contribution of CD39 to venous thrombosis in a restricted-flow model of murine inferior vena cava stenosis. CD39-deficiency conferred a >2-fold increase in venous thrombogenesis, characterized by increased leukocyte engagement, neutrophil extracellular trap formation, fibrin, and local activation of tissue factor in the thrombotic milieu. This was orchestrated by increased phosphorylation of the p65 subunit of NFκB, activation of the NLRP3 inflammasome, and interleukin-1ß (IL-1ß) release in CD39-deficient mice. Substantiating these findings, an IL-1ß-neutralizing antibody attenuated the thrombosis risk in CD39-deficient mice. These data demonstrate that IL-1ß is a key accelerant of venous thrombo-inflammation, which can be suppressed by CD39. CD39 inhibits in vivo crosstalk between inflammation and coagulation pathways, and is a critical vascular checkpoint in venous thrombosis.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Trombosis de la Vena/metabolismo , Animales , Antígenos CD/genética , Apirasa/genética , Modelos Animales de Enfermedad , Trampas Extracelulares/genética , Trampas Extracelulares/metabolismo , Humanos , Inflamasomas/genética , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/genética , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neutrófilos/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Trombosis de la Vena/genética , Trombosis de la Vena/patología
8.
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
9.
Front Immunol ; 9: 1322, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29942314

RESUMEN

OBJECTIVES: CD39 and CD73 are surface enzymes that jut into the extracellular space where they mediate the step-wise phosphohydrolysis of the autocrine and paracrine danger signals ATP and ADP into anti-inflammatory adenosine. Given the role of vascular and immune cells' "purinergic halo" in maintaining homeostasis, we hypothesized that the ectonucleotidases CD39 and CD73 might play a protective role in lupus. METHODS: Lupus was modeled by intraperitoneal administration of pristane to three groups of mice: wild-type (WT), CD39-/-, and CD73-/-. After 36 weeks, autoantibodies, endothelial function, kidney disease, splenocyte activation/polarization, and neutrophil activation were characterized. RESULTS: As compared with WT mice, CD39-/- mice developed exaggerated splenomegaly in response to pristane, while both groups of ectonucleotidase-deficient mice demonstrated heightened anti-ribonucleoprotein production. The administration of pristane to WT mice triggered only subtle dysfunction of the arterial endothelium; however, both CD39-/- and CD73-/- mice demonstrated striking endothelial dysfunction following induction of lupus, which could be reversed by superoxide dismutase. Activated B cells and plasma cells were expanded in CD73-/- mice, while deficiency of either ectonucleotidase led to expansion of TH17 cells. CD39-/- and CD73-/- mice demonstrated exaggerated neutrophil extracellular trap release, while CD73-/- mice additionally had higher levels of plasma cell-free DNA. CONCLUSION: These data are the first to link ectonucleotidases with lupus autoimmunity and vascular disease. New therapeutic strategies may harness purinergic nucleotide dissipation or signaling to limit the damage inflicted upon organs and blood vessels by lupus.

10.
Nat Commun ; 8: 14252, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165452

RESUMEN

Novel therapeutics are required for improving the management of chronic inflammatory diseases. Aptamers are single-stranded RNA or DNA molecules that have recently shown utility in a clinical setting, as they can specifically neutralize biomedically relevant proteins, particularly cell surface and extracellular proteins. The nuclear chromatin protein DEK is a secreted chemoattractant that is abundant in the synovia of patients with juvenile idiopathic arthritis (JIA). Here, we show that DEK is crucial to the development of arthritis in mouse models, thus making it an appropriate target for aptamer-based therapy. Genetic depletion of DEK or treatment with DEK-targeted aptamers significantly reduces joint inflammation in vivo and greatly impairs the ability of neutrophils to form neutrophil extracellular traps (NETs). DEK is detected in spontaneously forming NETs from JIA patient synovial neutrophils, and DEK-targeted aptamers reduce NET formation. DEK is thus key to joint inflammation, and anti-DEK aptamers hold promise for the treatment of JIA and other types of arthritis.


Asunto(s)
Aptámeros de Nucleótidos/uso terapéutico , Artritis Juvenil/terapia , Factores Quimiotácticos/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Trampas Extracelulares/inmunología , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/genética , Adulto , Animales , Artritis Juvenil/inmunología , Factores Quimiotácticos/genética , Factores Quimiotácticos/inmunología , Factores Quimiotácticos/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/inmunología , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Proteínas Oncogénicas/inmunología , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/inmunología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Cultivo Primario de Células , Líquido Sinovial/química , Líquido Sinovial/citología , Líquido Sinovial/inmunología , Zimosan/inmunología
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.
J Clin Invest ; 123(7): 2981-93, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23722903

RESUMEN

Recent evidence suggests that enhanced neutrophil extracellular trap (NET) formation activates plasmacytoid dendritic cells and serves as a source of autoantigens in SLE. We propose that aberrant NET formation is also linked to organ damage and to the premature vascular disease characteristic of human SLE. Here, we demonstrate enhanced NET formation in the New Zealand mixed 2328 (NZM) model of murine lupus. NZM mice also developed autoantibodies to NETs as well as the ortholog of human cathelicidin/LL37 (CRAMP), a molecule externalized in the NETs. NZM mice were treated with Cl-amidine, an inhibitor of peptidylarginine deiminases (PAD), to block NET formation and were evaluated for lupus-like disease activity, endothelial function, and prothrombotic phenotype. Cl-amidine treatment inhibited NZM NET formation in vivo and significantly altered circulating autoantibody profiles and complement levels while reducing glomerular IgG deposition. Further, Cl-amidine increased the differentiation capacity of bone marrow endothelial progenitor cells, improved endothelium-dependent vasorelaxation, and markedly delayed time to arterial thrombosis induced by photochemical injury. Overall, these findings suggest that PAD inhibition can modulate phenotypes crucial for lupus pathogenesis and disease activity and may represent an important strategy for mitigating cardiovascular risk in lupus patients.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Hidrolasas/antagonistas & inhibidores , Ornitina/análogos & derivados , Animales , Autoanticuerpos/sangre , Células de la Médula Ósea/fisiología , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/inmunología , Trombosis de las Arterias Carótidas/prevención & control , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio/patología , Femenino , Humanos , Inyecciones Subcutáneas , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/enzimología , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Ornitina/administración & dosificación , Fenotipo , Desiminasas de la Arginina Proteica , Células Madre/fisiología , Vasodilatación/efectos de los fármacos
13.
Sci Transl Med ; 5(178): 178ra40, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23536012

RESUMEN

The early events leading to the development of rheumatoid arthritis (RA) remain unclear, but formation of autoantibodies to citrullinated protein antigens (ACPAs) is considered a key pathogenic event. Neutrophils isolated from patients with various autoimmune diseases display enhanced neutrophil extracellular trap (NET) formation, a phenomenon that exposes autoantigens in the context of immunostimulatory molecules. We investigated whether aberrant NETosis occurs in RA, determined its triggers, and examined its deleterious inflammatory consequences. Enhanced NETosis was observed in circulating and RA synovial fluid neutrophils compared to neutrophils from healthy controls and from patients with osteoarthritis (OA). Further, netting neutrophils infiltrated RA synovial tissue, rheumatoid nodules, and skin. NETosis correlated with ACPA presence and levels and with systemic inflammatory markers. RA sera and immunoglobulin fractions from RA patients with high levels of ACPA and/or rheumatoid factor significantly enhanced NETosis, and the NETs induced by these autoantibodies displayed distinct protein content. Indeed, during NETosis, neutrophils externalized the citrullinated autoantigens implicated in RA pathogenesis, and anti-citrullinated vimentin antibodies potently induced NET formation. Moreover, the inflammatory cytokines interleukin-17A (IL-17A) and tumor necrosis factor-α (TNF-α) induced NETosis in RA neutrophils. In turn, NETs significantly augmented inflammatory responses in RA and OA synovial fibroblasts, including induction of IL-6, IL-8, chemokines, and adhesion molecules. These observations implicate accelerated NETosis in RA pathogenesis, through externalization of citrullinated autoantigens and immunostimulatory molecules that may promote aberrant adaptive and innate immune responses in the joint and in the periphery, and perpetuate pathogenic mechanisms in this disease.


Asunto(s)
Artritis Reumatoide/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Citrulina/metabolismo , Humanos , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Microscopía Fluorescente , Neutrófilos/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA