Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.953
Filtrar
1.
Front Immunol ; 15: 1411979, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38989288

RESUMEN

Background: Kawasaki disease (KD), an acute febrile illness and systemic vasculitis, is the leading cause of acquired heart disease in children in industrialized countries. KD leads to the development of coronary artery aneurysms (CAA) in affected children, which may persist for months and even years after the acute phase of the disease. There is an unmet need to characterize the immune and pathological mechanisms of the long-term complications of KD. Methods: We examined cardiovascular complications in the Lactobacillus casei cell wall extract (LCWE) mouse model of KD-like vasculitis over 4 months. The long-term immune, pathological, and functional changes occurring in cardiovascular lesions were characterized by histological examination, flow cytometric analysis, immunofluorescent staining of cardiovascular tissues, and transthoracic echocardiogram. Results: CAA and abdominal aorta dilations were detected up to 16 weeks following LCWE injection and initiation of acute vasculitis. We observed alterations in the composition of circulating immune cell profiles, such as increased monocyte frequencies in the acute phase of the disease and higher counts of neutrophils. We determined a positive correlation between circulating neutrophil and inflammatory monocyte counts and the severity of cardiovascular lesions early after LCWE injection. LCWE-induced KD-like vasculitis was associated with myocarditis and myocardial dysfunction, characterized by diminished ejection fraction and left ventricular remodeling, which worsened over time. We observed extensive fibrosis within the inflamed cardiac tissue early in the disease and myocardial fibrosis in later stages. Conclusion: Our findings indicate that increased circulating neutrophil counts in the acute phase are a reliable predictor of cardiovascular inflammation severity in LCWE-injected mice. Furthermore, long-term cardiac complications stemming from inflammatory cell infiltrations in the aortic root and coronary arteries, myocardial dysfunction, and myocardial fibrosis persist over long periods and are still detected up to 16 weeks after LCWE injection.


Asunto(s)
Pared Celular , Modelos Animales de Enfermedad , Fibrosis , Lacticaseibacillus casei , Síndrome Mucocutáneo Linfonodular , Vasculitis , Animales , Ratones , Pared Celular/inmunología , Vasculitis/inmunología , Vasculitis/etiología , Vasculitis/patología , Síndrome Mucocutáneo Linfonodular/inmunología , Síndrome Mucocutáneo Linfonodular/complicaciones , Masculino , Miocarditis/etiología , Miocarditis/patología , Miocarditis/inmunología , Inflamación/inmunología
2.
Kaohsiung J Med Sci ; 40(7): 612-620, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38828518

RESUMEN

Palpable purpura, gastrointestinal symptoms, joint involvement, and renal disease characterize immunoglobulin A vasculitis (IgAV). Renal involvement ranging from mild proteinuria to severe nephritic or nephrotic syndrome highlights the importance of monitoring kidney function in patients with IgAV. Recognizing these key features is crucial for early diagnosis and appropriate management to prevent long-term complications related to kidney disease. However, the pathogenesis of IgAV remains unclear. Disease mechanisms involve various factors, including the interplay of aberrantly glycosylated IgA, anti-endothelial cell antibodies, and neutrophils following infection triggers, which are the main pathogenic mechanisms of IgAV. Insights from cases of IgAV related to Coronavirus disease 2019 have offered additional understanding of the connection between infection and IgAV pathogenesis. This review provides a valuable resource for healthcare professionals and rheumatology researchers seeking a better understanding of the clinical features and pathophysiology of IgAV.


Asunto(s)
COVID-19 , Inmunoglobulina A , Humanos , Inmunoglobulina A/inmunología , COVID-19/inmunología , COVID-19/fisiopatología , COVID-19/virología , COVID-19/complicaciones , Vasculitis/inmunología , Vasculitis/fisiopatología , SARS-CoV-2/inmunología , Vasculitis por IgA/inmunología , Vasculitis por IgA/fisiopatología , Vasculitis por IgA/diagnóstico , Autoanticuerpos/inmunología , Neutrófilos/inmunología
4.
Front Immunol ; 15: 1354349, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38707895

RESUMEN

Antiphospholipid antibodies (aPL) are both laboratory evidence and causative factors for a broad spectrum of clinical manifestations of antiphospholipid syndrome (APS), with thrombotic and obstetric events being the most prevalent. Despite the aPL-triggered vasculopathy nature of APS, vasculitic-like manifestations rarely exist in APS and mainly appear associated with other concurrent connective tissue diseases like systemic lupus erythematous. Several studies have characterized pulmonary capillaritis related to pathogenic aPL, suggesting vasculitis as a potential associated non-thrombotic manifestation. Here, we describe a 15-year-old girl who develops hepatic infarction in the presence of highly positive aPL, temporally related to prior non-severe COVID-19 infection. aPL-related hepatic vasculitis, which has not been reported before, contributes to liver ischemic necrosis. Immunosuppression therapy brings about favorable outcomes. Our case together with retrieved literature provides supportive evidence for aPL-related vasculitis, extending the spectrum of vascular changes raised by pathogenic aPL. Differentiation between thrombotic and vasculitic forms of vascular lesions is essential for appropriate therapeutic decision to include additional immunosuppression therapy. We also perform a systematic review to characterize the prevalence and clinical features of new-onset APS and APS relapses after COVID-19 for the first time, indicating the pathogenicity of aPL in a subset of COVID-19 patients.


Asunto(s)
Anticuerpos Antifosfolípidos , Síndrome Antifosfolípido , COVID-19 , SARS-CoV-2 , Vasculitis , Humanos , COVID-19/complicaciones , COVID-19/inmunología , Femenino , Adolescente , Anticuerpos Antifosfolípidos/sangre , Anticuerpos Antifosfolípidos/inmunología , Vasculitis/inmunología , Vasculitis/etiología , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/inmunología , SARS-CoV-2/inmunología , Hígado/patología
5.
Z Rheumatol ; 83(4): 283-296, 2024 May.
Artículo en Alemán | MEDLINE | ID: mdl-38587633

RESUMEN

The diagnosis of systemic vasculitis (SV) is a major clinical challenge due to the very different forms of presentation and requires an interdisciplinary approach. Targeted laboratory diagnostics support making the diagnosis, differential diagnosis and classification and are also a key component in the detection of active organ manifestations and treatment complications. The basic laboratory tests include the erythrocyte sedimentation rate (ESR), C­reactive protein (CRP), blood count, serum creatinine, urinalysis, specific autoantibodies, complement, immunoglobulins, cryoglobulins and hepatitis B and C serology. Antineutrophil cytoplasmic autoantibodies (ANCA), antiglomerular basement membrane antibodies (anti-GBM antibodies) and anti-C1q antibodies are valuable laboratory markers for the diagnosis of the various forms of small vessel vasculitis. There are no specific laboratory tests for the diagnosis of medium and large vessel vasculitis. Despite advances in our understanding of the pathogenesis of vasculitis, no biomarkers have yet been identified that can be reliably used to guide treatment or that are useful in distinguishing vasculitis from other inflammatory diseases such as infections or treatment complications.


Asunto(s)
Anticuerpos Anticitoplasma de Neutrófilos , Biomarcadores , Vasculitis , Humanos , Biomarcadores/sangre , Vasculitis/diagnóstico , Vasculitis/sangre , Vasculitis/inmunología , Anticuerpos Anticitoplasma de Neutrófilos/sangre , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Autoanticuerpos/sangre , Técnicas de Laboratorio Clínico/métodos , Diagnóstico Diferencial
6.
Semin Arthritis Rheum ; 66: 152440, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38579593

RESUMEN

BACKGROUND/PURPOSE: Vasculitis as an immune-related adverse event (irAE) from checkpoint inhibitor therapy (ICI) to treat cancer is a rare clinical event, and little is known regarding its nosology, clinical manifestations, or response to treatment and outcomes. METHODS: To address these gaps, we used the Preferred Reporting Items for Systemic Reviews and Meta-Analyses Extension for Scoping Reviews (PRISMA-ScR) framework to further define this complication. Two independent PUBMED searches in September and November of 2022 revealed 127 publications with 37 excluded from title by relevance, 43 excluded by article type, and 23 excluded due to lack of biopsy results, or biopsy negative for vasculitis. Twenty-nine documented cases from 24 publications were included for final analysis. Basic demographics, ICI details, timing of onset of vasculitis symptoms, irAE treatment and outcomes were collected. The vasculitides were classified using 2022 ACR/EULAR Vasculitis Classification Criteria as well as 2012 Revised Chapel-Hill Nomenclature. Adaptations from Naidoo et al. 2023 [1] consensus definitions for irAEs were used and efforts were made to classify steroid-responsive versus unresponsive irAEs. RESULTS: Of the 29 cases reviewed, the average age of patients was 62.1 ± 11.0, composed of 58.6 % (n = 17) male and 41.3 % (n = 12) female. Prominent cancer types were lung cancer (41.4 %; n = 12), melanoma (41.4 %; n = 12), and renal cancer (10.3 %; n = 3), with majority being stage 4 (75.9 %, n = 22) and stage 3 (10.3 %, n = 3). Only 8 cases met the ACR/EULAR criteria, and by Chapel-Hill Nomenclature, approximately a third were small-vessel vasculitis (31.0 %; n = 9) with n = 4 positive for ANCA. Most biopsies were taken from the skin (37.9 %, n = 11) and kidney (24.1 %, n = 7). Patients were either treated with single (65.5 %, n = 19), dual (17.2 %; n = 5), or sequential (17.2 %; n = 5) ICI regimen which included anti-PD-1 therapy in all but one case, with mean of 8.7 ± 10.5 cycles received. Mean time to onset of symptoms from start of ICI was 7.2 ± 7.8 months, with 55.2 % occurring >3 months since the initial immunotherapy. Vasculitis treatment included glucocorticoids in 96 % of cases and immunotherapy was often discontinued (44.8 %; n = 13). Clinical improvement of irAE was documented in 86.2 % (n = 25). Data were missing in terms of fate of ICI (34.5 %; n = 10) and tumor outcomes (41.4 %; n = 12). Cancer progressed in 20.7 % (n = 6), stable in 34.5 % (n = 10) cases, and 6 patients died of all-causes. CONCLUSION: Vasculitis as an irAE appears clinically heterogeneous and rare. Among reported cases with adequate documentation, vasculitis is of delayed onset following the initiation of immunotherapy. Outcomes of ICI-vasculitis were generally favorable, responding to glucocorticoids and immunotherapy withdrawal. There is an urgent need for more standardized reporting of rare irAEs such as vasculitis to clarify clinical risks, classification, relationship to immunotherapy and outcomes.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Vasculitis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Vasculitis/inducido químicamente , Vasculitis/inmunología , Anciano
8.
Hypertension ; 81(6): 1218-1232, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38511317

RESUMEN

Inflammatory responses in small vessels play an important role in the development of cardiovascular diseases, including hypertension, stroke, and small vessel disease. This involves various complex molecular processes including oxidative stress, inflammasome activation, immune-mediated responses, and protein misfolding, which together contribute to microvascular damage. In addition, epigenetic factors, including DNA methylation, histone modifications, and microRNAs influence vascular inflammation and injury. These phenomena may be acquired during the aging process or due to environmental factors. Activation of proinflammatory signaling pathways and molecular events induce low-grade and chronic inflammation with consequent cardiovascular damage. Identifying mechanism-specific targets might provide opportunities in the development of novel therapeutic approaches. Monoclonal antibodies targeting inflammatory cytokines and epigenetic drugs, show promise in reducing microvascular inflammation and associated cardiovascular diseases. In this article, we provide a comprehensive discussion of the complex mechanisms underlying microvascular inflammation and offer insights into innovative therapeutic strategies that may ameliorate vascular injury in cardiovascular disease.


Asunto(s)
Inflamación , Animales , Humanos , Arterias/metabolismo , Enfermedades Cardiovasculares/metabolismo , Epigénesis Genética , Inflamación/metabolismo , Inflamación/inmunología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Vasculitis/metabolismo , Vasculitis/inmunología
11.
Int J Mol Sci ; 24(14)2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37511357

RESUMEN

Prolonged B cells stimulation due to the Hepatitis C virus (HCV) can result in autoimmunity, stigmatized by rising levels of cryoglobulins (CGs), the rheumatoid factor (RF), and free light chains (FLC) of immunoglobulins (Ig) associated with a range of symptoms, from their absence to severe cryoglobulinemic vasculitis and lymphoma. Here, we aimed to identify an immunological signature for the earliest stages of vasculitis when cryoprecipitate is still not detectable. We firstly analyzed the IgG subclasses, FLC, and RF in 120 HCV-RNA-positive patients divided into four groups according to the type of cryoprecipitate and symptoms: 30 asymptomatic without cryoprecipitate (No Cryo), 30 with vasculitis symptoms but without CGs that we supposed were circulating but still not detectable (Circulating), 30 type II and 30 type III mixed cryoglobulinemia (Cryo II and Cryo III, respectively). Our results revealed that patients with supposed circulating CGs displayed a pattern of serological parameters that closely resembled Cryo II and Cryo III, with a stronger similarity to Cryo II. Accordingly, we analyzed the groups of Circulating and Cryo II for their immunoglobulin heavy chain (IgH) and T-cell receptor (TCR) gene rearrangements, finding a similar mixed distribution of monoclonal, oligoclonal, and polyclonal responses compared to a control group of ten HCV-RNA-negative patients recovered from infection, who displayed a 100% polyclonal response. Our results strengthened the hypothesis that circulating CGs are the origin of symptoms in HCV-RNA-positive patients without cryoprecipitate and demonstrated that an analysis of clonal IGH and TCR rearrangements is the best option for the early diagnosis of extrahepatic complications.


Asunto(s)
Crioglobulinemia , Crioglobulinas , Hepatitis C Crónica , Vasculitis , Vasculitis/diagnóstico , Vasculitis/inmunología , Vasculitis/virología , Humanos , Masculino , Femenino , Crioglobulinemia/diagnóstico , Crioglobulinemia/virología , Crioglobulinas/análisis , Factor Reumatoide/sangre , Inmunoglobulinas/sangre , Hepatitis C Crónica/sangre , Hepatitis C Crónica/complicaciones
12.
BMC Nephrol ; 23(1): 67, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35172758

RESUMEN

BACKGROUND: Hypocomplementemic urticarial vasculitis (HUV) is a rare systemic vasculitis. We aimed to describe the kidney involvement of HUV in a multicenter national cohort with an extended follow-up. METHODS: All patients with HUV (international Schwartz criteria) with a biopsy-proven kidney involvement, identified through a survey of the French Vasculitis Study Group (FVSG), were included. A systematic literature review on kidney involvement of HUV was performed. RESULTS: Twelve patients were included, among whom 8 had positive anti-C1q antibodies. All presented with proteinuria, from mild to nephrotic, and 8 displayed acute kidney injury (AKI), requiring temporary haemodialysis in 2. Kidney biopsy showed membrano-proliferative glomerulonephritis (MPGN) in 8 patients, pauci-immune crescentic GN or necrotizing vasculitis in 3 patients (with a mild to severe interstitial inflammation), and an isolated interstitial nephritis in 1 patient. C1q deposits were observed in the glomeruli (n = 6), tubules (n = 4) or renal arterioles (n = 3) of 8 patients. All patients received corticosteroids, and 9 were also treated with immunosuppressants or apheresis. After a mean follow-up of 8.9 years, 6 patients had a preserved renal function, but 2 patients had developed stage 3-4 chronic kidney disease (CKD) and 4 patients had reached end-stage kidney disease (ESKD), among whom 1 had received a kidney transplant. CONCLUSION: Renal involvement of HUV can be responsible for severe AKI, CKD and ESRD. It is not always associated with circulating anti-C1q antibodies. Kidney biopsy shows mostly MPGN or crescentic GN, with frequent C1q deposits in the glomeruli, tubules or arterioles.


Asunto(s)
Glomerulonefritis Membranoproliferativa/complicaciones , Urticaria/complicaciones , Vasculitis/complicaciones , Corticoesteroides/uso terapéutico , Adulto , Anciano , Biopsia , Eliminación de Componentes Sanguíneos , Niño , Preescolar , Complemento C1q/metabolismo , Ciclofosfamida/uso terapéutico , Femenino , Estudios de Seguimiento , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Glomerulonefritis Membranoproliferativa/inmunología , Glomerulonefritis Membranoproliferativa/patología , Humanos , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Rituximab/uso terapéutico , Síndrome , Urticaria/inmunología , Vasculitis/inmunología
13.
Front Immunol ; 13: 823853, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154143

RESUMEN

The correlation of infections with vascular autoinflammatory diseases such as vasculitis and atherosclerosis has been long recognized, and progressive inflammation with the formation of tertiary lymphoid organs in arterial adventitia intensively studied, the immunological basis of the nondiseased vasculatures that predispose to subsequent vascular autoimmunity during inflammation, however, is not well characterized. Here, we investigated the vascular immunity in situ of steady-state C57BL/6 mice and found that healthy vascular tissues contained a comprehensive set of immune cells with relatively higher proportion of innate components than lymphoid organs. Notably, a complete set of dendritic cell (DC) subsets was observed with monocyte-derived DCs (moDCs) constituting a major proportion; this is in contrast to moDCs being considered rare in the steady state. Interestingly, these vascular DCs constitutively expressed more suppressive factors with cDC1 for PD-L1 and moDCs for IL-10; this is concordant with the inhibitive phenotype of T cells in normal vascular tissues. The immunotolerant state of the vascular tissues, however, was readily eroded by systemic inflammation, demonstrated by the upregulation of proinflammatory cytokines and enhanced antigen presentation by vascular DCs to activate both cellular and humoral immunity in situ, which ultimately led to vascular destruction. Different vascular DC subsets elicited selective effects: moDCs were potent cytokine producers and B-cell activators, whereas cDCs, particularly, cDC1, were efficient at presenting antigens to stimulate T cells. Together, we unveil regional immunological features of vascular tissues to explain their dual facets under physiological versus pathological conditions for the better understanding and treatment of cardiovascular autoinflammation.


Asunto(s)
Células Dendríticas/inmunología , Inflamación/inmunología , Autotolerancia/fisiología , Linfocitos T/inmunología , Vasculitis/inmunología , Animales , Presentación de Antígeno/inmunología , Autoinmunidad , Diferenciación Celular/inmunología , Citocinas/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología
14.
J Mol Cell Cardiol ; 164: 69-82, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34838588

RESUMEN

The global propagation of SARS-CoV-2 leads to an unprecedented public health emergency. Despite that the lungs are the primary organ targeted by COVID-19, systemic endothelial inflammation and dysfunction is observed particularly in patients with severe COVID-19, manifested by elevated endothelial injury markers, endotheliitis, and coagulopathy. Here, we review the clinical characteristics of COVID-19 associated endothelial dysfunction; and the likely pathological mechanisms underlying the disease including direct cell entry or indirect immune overreactions after SARS-CoV-2 infection. In addition, we discuss potential biomarkers that might indicate the disease severity, particularly related to the abnormal development of thrombosis that is a fatal vascular complication of severe COVID-19. Furthermore, we summarize clinical trials targeting the direct and indirect pathological pathways after SARS-CoV-2 infection to prevent or inhibit the virus induced endothelial disorders.


Asunto(s)
COVID-19/patología , Endotelio Vascular/patología , SARS-CoV-2 , Adolescente , Adulto , Anciano , Enzima Convertidora de Angiotensina 2/fisiología , Animales , COVID-19/sangre , COVID-19/complicaciones , COVID-19/fisiopatología , COVID-19/terapia , Ensayos Clínicos como Asunto , Células Endoteliales/patología , Células Endoteliales/virología , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Proteína HMGB1/fisiología , Humanos , Macaca mulatta , Ratones , Neuropilina-1/fisiología , Estrés Oxidativo , Especies Reactivas de Oxígeno , Receptores Virales/fisiología , Receptores Depuradores de Clase B/fisiología , Índice de Severidad de la Enfermedad , Transducción de Señal , Síndrome de Respuesta Inflamatoria Sistémica/patología , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Trombofilia/etiología , Trombofilia/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Vasculitis/etiología , Vasculitis/inmunología , Vasculitis/fisiopatología , Adulto Joven
15.
Cardiovasc Res ; 118(2): 372-385, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33483732

RESUMEN

Nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) is an intracellular innate immune receptor that recognizes a diverse range of stimuli derived from pathogens, damaged or dead cells, and irritants. NLRP3 activation causes the assembly of a large multiprotein complex termed the NLRP3 inflammasome, and leads to the secretion of bioactive interleukin (IL)-1ß and IL-18 as well as the induction of inflammatory cell death termed pyroptosis. Accumulating evidence indicates that NLRP3 inflammasome plays a key role in the pathogenesis of sterile inflammatory diseases, including atherosclerosis and other vascular diseases. Indeed, the results of the Canakinumab Anti-inflammatory Thrombosis Outcome Study trial demonstrated that IL-1ß-mediated inflammation plays an important role in atherothrombotic events and suggested that NLRP3 inflammasome is a key driver of atherosclerosis. In this review, we will summarize the current state of knowledge regarding the role of NLRP3 inflammasome in vascular diseases, in particular in atherosclerosis, vascular injury, aortic aneurysm, and Kawasaki disease vasculitis, and discuss NLRP3 inflammasome as a therapeutic target for these disorders.


Asunto(s)
Arterias/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Vasculares/metabolismo , Vasculitis/metabolismo , Animales , Antiinflamatorios/farmacología , Arterias/efectos de los fármacos , Arterias/inmunología , Arterias/patología , Humanos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Terapia Molecular Dirigida , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Transducción de Señal , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/patología , Vasculitis/tratamiento farmacológico , Vasculitis/inmunología , Vasculitis/patología
16.
Sci Rep ; 11(1): 20019, 2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34625602

RESUMEN

Dysfunction of immunoinhibitory signals and persistent T cell activation reportedly play important roles in the development of vasculitis. The skin is one of the most accessible organs, and it is suitable for the characterization of immune cell signatures. However, the inhibitory checkpoint molecules in the skin and their relevance to vasculitis have not been studied. Here, we investigated the profile of immune checkpoint molecules in the skin and peripheral blood of patients with vasculitis and healthy donors. We found that some of the inhibitory checkpoint molecules, including programmed cell death 1 receptor (PD-1), were elevated in T-cells in the blood of patients with systemic and cutaneous vasculitis. In addition, programmed death-ligand 1 (PD-L1) expression was elevated in the skin of patients with cutaneous vasculitis. Histologically, PD-L1 was highly expressed in the vessels in the skin along with CD4+ and CD8+ T-cell infiltration in patients with cutaneous vasculitis. Notably, plasma soluble PD-L1 levels were increased, and these correlated with C-reactive protein in patients with systemic vasculitis. Our findings suggest that inhibitory checkpoint molecules might be differentially modulated in the skin and peripheral blood of patients with vasculitis, and that the alteration of the PD-L1/PD-1 axis may be associated with the regulation of T-cell activation in vasculitis.


Asunto(s)
Antígeno B7-H1 , Biomarcadores , Activación de Linfocitos/inmunología , Receptor de Muerte Celular Programada 1 , Vasculitis/inmunología , Adulto , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Piel/metabolismo
17.
Nat Rev Rheumatol ; 17(11): 651-664, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34599320

RESUMEN

Fatigue is a complex phenomenon and an important health concern for many people with chronic inflammatory rheumatic diseases, such as rheumatoid arthritis, psoriatic arthritis, primary Sjögren syndrome and systemic lupus erythematosus. Although some clinical trials have shown the benefits of cognitive behavioural therapy in fatigue management, the effect of this approach is relatively modest, and no curative treatment has been identified. The pathogenesis of fatigue remains unclear. Despite many challenges and limitations, a growing body of research points to roles for the immune system, the central and autonomic nervous systems and the neuroendocrine system in the induction and maintenance of fatigue in chronic diseases. New insights indicate that sleep, genetic susceptibility, metabolic disturbances and other biological and physiological mechanisms contribute to fatigue. Furthermore, understanding of the relationships between psychosocial factors and fatigue is increasing. However, the interrelationships between these diverse mechanisms and fatigue remain poorly defined. In this Review, we outline various biological, physiological and psychosocial determinants of fatigue in inflammatory rheumatic diseases, and propose mechanistic and conceptual models of fatigue to summarize current understanding, stimulate debate and develop further research ideas.


Asunto(s)
Artritis , Enfermedades del Tejido Conjuntivo , Fatiga , Enfermedades Reumáticas , Vasculitis , Artritis/inmunología , Investigación Biomédica , Enfermedad Crónica , Enfermedades del Tejido Conjuntivo/inmunología , Fatiga/diagnóstico , Fatiga/etiología , Fatiga/fisiopatología , Fatiga/terapia , Predicción , Humanos , Inflamación/inmunología , Enfermedades Reumáticas/complicaciones , Enfermedades Reumáticas/inmunología , Vasculitis/inmunología
19.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298979

RESUMEN

Platelet extravasation during inflammation is under-appreciated. In wild-type (WT) mice, a central corneal epithelial abrasion initiates neutrophil (PMN) and platelet extravasation from peripheral limbal venules. The same injury in mice expressing low levels of the ß2-integrin, CD18 (CD18hypo mice) shows reduced platelet extravasation with PMN extravasation apparently unaffected. To better define the role of CD18 on platelet extravasation, we focused on two relevant cell types expressing CD18: PMNs and mast cells. Following corneal abrasion in WT mice, we observed not only extravasated PMNs and platelets but also extravasated erythrocytes (RBCs). Ultrastructural observations of engorged limbal venules showed platelets and RBCs passing through endothelial pores. In contrast, injured CD18hypo mice showed significantly less venule engorgement and markedly reduced platelet and RBC extravasation; mast cell degranulation was also reduced compared to WT mice. Corneal abrasion in mast cell-deficient (KitW-sh/W-sh) mice showed less venule engorgement, delayed PMN extravasation, reduced platelet and RBC extravasation and delayed wound healing compared to WT mice. Finally, antibody-induced depletion of circulating PMNs prior to corneal abrasion reduced mast cell degranulation, venule engorgement, and extravasation of PMNs, platelets, and RBCs. In summary, in the injured cornea, platelet and RBC extravasation depends on CD18, PMNs, and mast cell degranulation.


Asunto(s)
Plaquetas/fisiología , Antígenos CD18/fisiología , Degranulación de la Célula , Córnea/irrigación sanguínea , Eritrocitos/fisiología , Hiperemia/fisiopatología , Mastocitos/fisiología , Neutrófilos/fisiología , Migración Transendotelial y Transepitelial/fisiología , Vasculitis/inmunología , Vénulas/metabolismo , Animales , Antígenos CD18/deficiencia , Movimiento Celular , Quimiotaxis de Leucocito , Lesiones de la Cornea/metabolismo , Lesiones de la Cornea/patología , Epitelio Corneal/fisiología , Femenino , Hiperemia/sangre , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Microscopía Electrónica , Modelos Animales , Fagocitosis , Regeneración/fisiología , Vasculitis/sangre , Vénulas/patología , Cicatrización de Heridas/fisiología
20.
J Mol Cell Cardiol ; 160: 121-127, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34303670

RESUMEN

Immune checkpoint inhibitors (ICI) have changed the landscape of cancer therapy, but their use carries a high risk of cardiac immune related adverse events (iRAEs). With the expanding utilization of ICI therapy, there is a growing need to understand the underlying mechanisms behind their anti-tumor activity as well as their immune-mediated toxicities. In this review, we will focus on clinical characteristics and immune pathways of ICI cardiotoxicity, with an emphasis on single-cell technologies used to gain insights in this field. We will focus on three key areas of ICI-mediated immune pathways, including the anti-tumor immune response, the augmentation of the immune response by ICIs, and the pathologic "autoimmune" response in some individuals leading to immune-mediated toxicity, as well as local factors in the myocardial immune environment predisposing to autoimmunity. Discerning the underlying mechanisms of these immune pathways is necessary to inform the development of targeted therapies for ICI cardiotoxicities and reduce treatment related morbidity and mortality.


Asunto(s)
Antineoplásicos/efectos adversos , Arritmias Cardíacas/inducido químicamente , Aterosclerosis/inducido químicamente , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunoterapia/métodos , Miocarditis/inducido químicamente , Pericarditis/inducido químicamente , Vasculitis/inducido químicamente , Animales , Arritmias Cardíacas/inmunología , Aterosclerosis/inmunología , Autoinmunidad/efectos de los fármacos , Cardiotoxicidad/inmunología , Humanos , Ratones , Miocarditis/inmunología , Pericarditis/inmunología , Placa Aterosclerótica/inducido químicamente , Placa Aterosclerótica/inmunología , Factores de Riesgo , Resultado del Tratamiento , Vasculitis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA