Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 694
Filtrar
1.
Circ Res ; 134(10): 1330-1347, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38557119

RESUMEN

BACKGROUND: Tetraspanin CD151 is highly expressed in endothelia and reinforces cell adhesion, but its role in vascular inflammation remains largely unknown. METHODS: In vitro molecular and cellular biological analyses on genetically modified endothelial cells, in vivo vascular biological analyses on genetically engineered mouse models, and in silico systems biology and bioinformatics analyses on CD151-related events. RESULTS: Endothelial ablation of Cd151 leads to pulmonary and cardiac inflammation, severe sepsis, and perilous COVID-19, and endothelial CD151 becomes downregulated in inflammation. Mechanistically, CD151 restrains endothelial release of proinflammatory molecules for less leukocyte infiltration. At the subcellular level, CD151 determines the integrity of multivesicular bodies/lysosomes and confines the production of exosomes that carry cytokines such as ANGPT2 (angiopoietin-2) and proteases such as cathepsin-D. At the molecular level, CD151 docks VCP (valosin-containing protein)/p97, which controls protein quality via mediating deubiquitination for proteolytic degradation, onto endolysosomes to facilitate VCP/p97 function. At the endolysosome membrane, CD151 links VCP/p97 to (1) IFITM3 (interferon-induced transmembrane protein 3), which regulates multivesicular body functions, to restrain IFITM3-mediated exosomal sorting, and (2) V-ATPase, which dictates endolysosome pH, to support functional assembly of V-ATPase. CONCLUSIONS: Distinct from its canonical function in strengthening cell adhesion at cell surface, CD151 maintains endolysosome function by sustaining VCP/p97-mediated protein unfolding and turnover. By supporting protein quality control and protein degradation, CD151 prevents proteins from (1) buildup in endolysosomes and (2) discharge through exosomes, to limit vascular inflammation. Also, our study conceptualizes that balance between degradation and discharge of proteins in endothelial cells determines vascular information. Thus, the IFITM3/V-ATPase-tetraspanin-VCP/p97 complexes on endolysosome, as a protein quality control and inflammation-inhibitory machinery, could be beneficial for therapeutic intervention against vascular inflammation.


Asunto(s)
COVID-19 , Endosomas , Lisosomas , Tetraspanina 24 , Animales , Lisosomas/metabolismo , Tetraspanina 24/metabolismo , Tetraspanina 24/genética , Humanos , Ratones , COVID-19/metabolismo , COVID-19/inmunología , COVID-19/patología , Endosomas/metabolismo , Ratones Noqueados , Vasculitis/metabolismo , Ratones Endogámicos C57BL , SARS-CoV-2 , Inflamación/metabolismo , Inflamación/patología , Sepsis/metabolismo
2.
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 , Humanos , Inflamación/metabolismo , Inflamación/inmunología , Enfermedades Cardiovasculares/metabolismo , Estrés Oxidativo/fisiología , Epigénesis Genética , Arterias/metabolismo , Transducción de Señal/fisiología , Vasculitis/metabolismo , Vasculitis/inmunología , Animales
3.
Int J Biol Macromol ; 261(Pt 2): 129658, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38266857

RESUMEN

A category of very uncommon systemic inflammatory blood vessel illnesses known as vasculitides. The pathogenesis and etiology of vasculitis are still poorly known. Despite all of the progress made in understanding the genetics and causes behind vasculitis, there is still more to learn. Epigenetic dysregulation is a significant contributor to immune-mediated illnesses, and epigenetic aberrancies in vasculitis are becoming more widely acknowledged. Less than 2 % of the genome contains protein-encoding DNA. Studies have shown that a variety of RNAs originating from the non-coding genome exist. Long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) have attracted the most attention in recent years as they are becoming more and more important regulators of different biological processes, such as diseases of the veins. Extracellular vehicles (EVs) such as exosomes, are membrane-bound vesicular structures that break free either during programmed cell death, such as apoptosis, pyroptosis, and necroptosis or during cell activation. Exosomes may be involved in harmful ways in inflammation, procoagulation, autoimmune reactions, endothelial dysfunction/damage, intimal hyperplasia and angiogenesis, all of which may be significant in vasculitis. Herein, we summarized various non-coding RNAs that are involved in vasculitides pathogenesis. Moreover, we highlighted the role of exosomes in vasculitides.


Asunto(s)
Exosomas , MicroARNs , ARN Largo no Codificante , Vasculitis , Humanos , MicroARNs/genética , Vasculitis/genética , Vasculitis/metabolismo , Exosomas/genética , Exosomas/metabolismo , ARN Largo no Codificante/genética , ARN Circular/metabolismo
4.
Cytokine ; 169: 156304, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37487381

RESUMEN

OBJECTIVES: By using GWAS(genome-wide association studies) and linkage disequilibrium analysis to investigate the susceptibility genes of KD(Kawasaki disease), previous studies have identified that the CaN(calcineurin)-NFAT(the nuclear factor of activated T cell) signal pathway were significantly associated with susceptibility to KD. However, little is known about the molecular basis of the CaN/NFAT pathway involved in KD. Therefore, in our study we investigate the role of Ca2+/CaN/NFAT signaling pathway in macrophages in vitro and in vivo on coronary artery lesions induced by LCWE (Lactobacillus casei cell wall extract). METHODS AND RESULTS: We observed that LCWE could increase the expression of NFAT1 and NFAT2 in macrophages in vitro, and also enhance the transcriptional activity of NFAT by promoting the nucleus translocation. Similarly, in LCWE-induced mice model, the expression of NFAT1 and NFAT2 and associated proinflammatory factors were increased significantly. In addition, by knocking down or overexpressing NFAT1 or NFAT2 in macrophages, the results indicated that NFAT signaling pathway mediated LCWE-induced immune responses in macrophages and regulated the synthesis of IL(interleukin)-6, IL-1ß and TNF(tumor necrosis factor)-α in LCWE-induced macrophage activation. As well, we found that this process could be suppressed by CaN inhibitor CsA(cyclosporinA). CONCLUSIONS: Therefore, the CaN/NFAT signaling pathway mediated LCWE-induced immune responses in macrophages, and also participated in the LCWE-induced CALs(coronary artery lesions). And also the inhibitory effect of CsA in LCWE-induced cell model towards a strategy to modulate the CaN/NFAT pathway during the acute course of KD might be helpful in alleviate KD-induced CALs.


Asunto(s)
Lacticaseibacillus casei , Síndrome Mucocutáneo Linfonodular , Vasculitis , Animales , Ratones , Síndrome Mucocutáneo Linfonodular/genética , Extractos Celulares/efectos adversos , Estudio de Asociación del Genoma Completo , Vasculitis/complicaciones , Vasculitis/metabolismo , Macrófagos/metabolismo , Transducción de Señal , Interleucina-6/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Pared Celular/metabolismo , Pared Celular/patología , Factores de Transcripción NFATC/metabolismo
5.
Biochim Biophys Acta Mol Basis Dis ; 1869(5): 166707, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37001702

RESUMEN

INTRODUCTION: The COVID-19 pandemic provide the opportunities to explore the numerous similarities in clinical symptoms with Kawasaki disease (KD), including severe vasculitis. Despite this, the underlying mechanisms of vascular injury in both KD and COVID-19 remain elusive. To identify these mechanisms, this study employs single-cell RNA sequencing to explore the molecular mechanisms of immune responses in vasculitis, and validate the results through in vitro experiments. METHOD: The single-cell RNA sequencing (scRNA-seq) analysis of peripheral blood mononuclear cells (PBMCs) was carried out to investigate the molecular mechanisms of immune responses in vasculitis in KD and COVID-19. The analysis was performed on PBMCs from six children diagnosed with complete KD, three age-matched KD healthy controls (KHC), six COVID-19 patients (COV), three influenza patients (FLU), and four healthy controls (CHC). The results from the scRNA-seq analysis were validated through flow cytometry and immunofluorescence experiments on additional human samples. Subsequently, monocyte adhesion assays, immunofluorescence, and quantitative polymerase chain reaction (qPCR) were used to analyze the damages to endothelial cells post-interaction with monocytes in HUVEC and THP1 cultures. RESULTS: The scRNA-seq analysis revealed the potential cellular types involved and the alterations in genetic transcriptions in the inflammatory responses. The findings indicated that while the immune cell compositions had been altered in KD and COV patients, and the ratio of CD14+ monocytes were both elevated in KD and COV. While the CD14+ monocytes share a large scale of same differentiated expressed geens between KD and COV. The differential activation of CD14 and CD16 monocytes was found to respond to both endothelial and epithelial dysfunctions. Furthermore, SELL+/CCR1+/XAF1+ CD14 monocytes were seen to enhance the adhesion and damage to endothelial cells. The results also showed that different types of B cells were involved in both KD and COV, while only the activation of T cells was recorded in KD. CONCLUSION: In conclusion, our study demonstrated the role of the innate immune response in the regulation of endothelial dysfunction in both KD and COVID-19. Additionally, our findings indicate that the adaptive immunity activation differs between KD and COVID-19. Our results demonstrate that monocytes in COVID-19 exhibit adhesion to both endothelial cells and alveolar epithelial cells, thus providing insight into the mechanisms and shared phenotypes between KD and COVID-19.


Asunto(s)
COVID-19 , Síndrome Mucocutáneo Linfonodular , Vasculitis , Niño , Humanos , Monocitos/metabolismo , Síndrome Mucocutáneo Linfonodular/genética , Síndrome Mucocutáneo Linfonodular/metabolismo , Leucocitos Mononucleares/metabolismo , Células Endoteliales/metabolismo , Pandemias , RNA-Seq , Receptores de Lipopolisacáridos/metabolismo , COVID-19/metabolismo , Vasculitis/genética , Vasculitis/metabolismo , Receptores CCR1
6.
Cardiovasc Res ; 119(5): 1265-1278, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36534975

RESUMEN

AIMS: The nuclear factor-κB (NF-κB) signalling pathway plays a critical role in the pathogenesis of multiple vascular diseases. However, in endothelial cells (ECs), the molecular mechanisms responsible for the negative regulation of the NF-κB pathway are poorly understood. In this study, we investigated a novel role for protein tyrosine phosphatase type IVA1 (PTP4A1) in NF-κB signalling in ECs. METHODS AND RESULTS: In human tissues, human umbilical artery ECs, and mouse models for loss of function and gain of function of PTP4A1, we conducted histological analysis, immunostaining, laser-captured microdissection assay, lentiviral infection, small interfering RNA transfection, quantitative real-time PCR and reverse transcription-PCR, as well as luciferase reporter gene and chromatin immunoprecipitation assays. Short hairpin RNA-mediated knockdown of PTP4A1 and overexpression of PTP4A1 in ECs indicated that PTP4A1 is critical for inhibiting the expression of cell adhesion molecules (CAMs). PTP4A1 increased the transcriptional activity of upstream stimulatory factor 1 (USF1) by dephosphorylating its S309 residue and subsequently inducing the transcription of tumour necrosis factor-alpha-induced protein 3 (TNFAIP3/A20) and the inhibition of NF-κB activity. Studies on Ptp4a1 knockout or transgenic mice demonstrated that PTP4A1 potently regulates the interleukin 1ß-induced expression of CAMs in vivo. In addition, we verified that PTP4A1 deficiency in apolipoprotein E knockout mice exacerbated high-fat high-cholesterol diet-induced atherogenesis with upregulated expression of CAMs. CONCLUSION: Our data indicate that PTP4A1 is a novel negative regulator of vascular inflammation by inducing USF1/A20 axis-mediated NF-κB inactivation. Therefore, the expression and/or activation of PTP4A1 in ECs might be useful for the treatment of vascular inflammatory diseases.


Asunto(s)
Células Endoteliales , FN-kappa B , Vasculitis , Animales , Humanos , Ratones , Proteínas de Ciclo Celular/metabolismo , Células Endoteliales/metabolismo , Inflamación/genética , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal , Factores Estimuladores hacia 5'/metabolismo , Vasculitis/genética , Vasculitis/metabolismo
7.
Cell Prolif ; 55(6): e13251, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35582751

RESUMEN

OBJECTIVES: Melatonin has been reported to be an appropriate candidate for mitigating various cardiovascular injuries, owing to its versatility. This study aimed to explore the role of melatonin in Kawasaki disease (KD)-associated vasculitis and its underlying mechanisms. MATERIAL AND METHODS: The role of melatonin was evaluated in human coronary artery endothelial cells (HCAECs), peripheral blood mononuclear cells from KD patients, human THP1 cell line in vitro, and a Candida albicans water-soluble fraction (CAWS)-induced KD mouse model in vivo. Cell proliferation assay, cell apoptosis assay, cell co-culture, RNA extraction, RNA sequencing, reverse transcription quantitative PCR, enzyme-linked immunosorbent assay (ELISA), transwell assay, western blot, dual-luciferase reporter assay, and autophagic flux assay were performed to investigate the function and regulatory mechanisms of melatonin in vitro, while haematoxylin and eosin staining, Verhoeff's van Gieson staining, ELISA, and immunohistochemical analysis were performed to detect the effect of melatonin in vivo. RESULTS: Melatonin suppressed cell apoptosis directly reduced the expression of endothelial cell damage markers in HCAECs, and alleviated vasculitis in the CAWS-induced KD mouse model. Mechanistically, melatonin promoted autophagy by activating the melatonin/ melatonin receptor (MT)/cAMP-response element binding protein (CREB) pathway and upregulating the expression of autophagy-related gene-3, thereby suppressing cell apoptosis in an autophagy-dependent manner. Additionally, melatonin decreased the production of pro-inflammatory cytokines in macrophages and indirectly reduced the immunopathological damage of HCAECs. CONCLUSIONS: This study revealed that melatonin protects vascular endothelial cells in KD, by suppressing cell apoptosis in an autophagy-dependent manner and reducing the immunopathological damage mediated by macrophages.


Asunto(s)
Melatonina , Síndrome Mucocutáneo Linfonodular , Vasculitis , Animales , Apoptosis , Autofagia , Células Endoteliales/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Melatonina/farmacología , Melatonina/uso terapéutico , Ratones , Síndrome Mucocutáneo Linfonodular/tratamiento farmacológico , Síndrome Mucocutáneo Linfonodular/genética , Síndrome Mucocutáneo Linfonodular/metabolismo , Vasculitis/metabolismo
8.
Semin Immunopathol ; 44(3): 325-345, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35254509

RESUMEN

The discovery of anti-neutrophil cytoplasmic antibodies (ANCA) and their antigenic targets, myeloperoxidase (MPO) and proteinase 3 (PR3), has led to further understanding as to the pathophysiologic processes that underlie vascular and tissue damage in ANCA vasculitis. ANCA trigger neutrophil activation leading to vascular damage in ANCA vasculitis. However, decades of study have determined that neutrophil activation alone is not sufficient to cause disease. Inflammatory stimuli are drivers of ANCA autoantigen expression and ANCA production. Certain infections or bacterial peptides may be crucial players in the initial steps of ANCA immunopathogenesis. Genetic and epigenetic alterations of gene encoding for MPO and PR3 provide additional disturbances to the immune homeostasis which provide a substrate for pathogenic ANCA formation from an adaptive immune system predisposed to autoreactivity. Promoted by inflammatory cytokines, ANCA binding leads to neutrophil activation, a process characterized by conformational changes, production and release of cytotoxic substances, and alternative complement pathway activation, thus creating an intense inflammatory milieu. This cascade of events perpetuates a vicious cycle of further inflammatory cell recruitment and activation, culminating in tissue necrosis. Our understanding of the pathogenic process in ANCA vasculitis paves the way for the development of therapies targeting crucial steps in this process. The greater appreciation of the role for complement, monocytes, and the adaptive immune system has already led to novel complement blockers and is poised to lead to further innovations which will allow for tailored antigen- or cell-specific immunotherapy targeting the autoimmune process without exposure to undue risks or toxicities.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos , Vasculitis , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/etiología , Anticuerpos Anticitoplasma de Neutrófilos , Autoantígenos , Humanos , Monocitos/metabolismo , Mieloblastina/genética , Vasculitis/metabolismo
9.
J Pathol ; 257(3): 300-313, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35239186

RESUMEN

P2RX7, an ionotropic receptor for extracellular adenosine triphosphate (ATP), is expressed on immune cells, including macrophages, monocytes, and dendritic cells and is upregulated on nonimmune cells following injury. P2RX7 plays a role in many biological processes, including production of proinflammatory cytokines such as interleukin (IL)-1ß via the canonical inflammasome pathway. P2RX7 has been shown to be important in inflammation and fibrosis and may also play a role in autoimmunity. We have developed and phenotyped a novel P2RX7 knockout (KO) inbred rat strain and, taking advantage of the human-resembling unique histopathological features of rat models of glomerulonephritis, we induced three models of disease: nephrotoxic nephritis, experimental autoimmune glomerulonephritis, and experimental autoimmune vasculitis. We found that deletion of P2RX7 does not protect rats from models of experimental glomerulonephritis or the development of autoimmunity. Notably, treatment with A-438079, a P2RX7 antagonist, was equally protective in WKY WT and P2RX7 KO rats, revealing its 'off-target' properties. We identified a novel ATP/P2RX7/K+ efflux-independent and caspase-1/8-dependent pathway for the production of IL-1ß in rat dendritic cells, which was absent in macrophages. Taken together, these results comprehensively establish that inflammation and autoimmunity in glomerulonephritis is independent of P2RX7 and reveals the off-target properties of drugs previously known as selective P2RX7 antagonists. Rat mononuclear phagocytes may be able to utilise an 'alternative inflammasome' pathway to produce IL-1ß independently of P2RX7, which may account for the susceptibility of P2RX7 KO rats to inflammation and autoimmunity in glomerulonephritis. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Glomerulonefritis , Receptores Purinérgicos P2X7 , Vasculitis , Adenosina Trifosfato/metabolismo , Animales , Caspasa 1/metabolismo , Caspasas , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas , Ratas Endogámicas WKY , Receptores Purinérgicos P2X7/metabolismo , Vasculitis/metabolismo , Vasculitis/patología
10.
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
11.
Cells ; 10(8)2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34440897

RESUMEN

Perinuclear anti-neutrophilic cytoplasmic antibodies (P-ANCA) recognize heterogeneous antigens, including myeloperoxidase (MPO), lactoferrin, elastase, cathepsin-G and bactericidal/permeability-increasing protein. Although P-ANCA have diagnostic utility in vasculitides, they may also be found in patients with various other systemic autoimmune rheumatic diseases (SARDs). Nevertheless, the clinical significance and the targets recognized by P-ANCA in such patients remain unclear. For this purpose, herein we investigated the occurrence of ANCA-related antigenic specificities in 82 P-ANCA-positive sera by multiplex ELISA, as well as their association with other autoantibodies. The P-ANCA-positive sera corresponded to patients with vasculitides (n = 24), systemic lupus erythematosus (n = 28), antiphospholipid syndrome (n = 5), Sjögren's syndrome (n = 7), rheumatoid arthritis (n = 3), systemic scleroderma (n = 1), sarcoidosis (n = 1) and Hashimoto's thyroiditis (n = 13). In most P-ANCA-positive patients studied (51/82, 62.3%), these autoantibodies occurred in high titers (>1:160). The analysis of P-ANCA-positive sera revealed reactivity to MPO in only 50% of patients with vasculitides, whereas it was infrequent in the other disease groups studied. Reactivity to other P-ANCA-related autoantigens was also rarely detected. Our findings support that high P-ANCA titers occur in SARD. The P-ANCA-positive staining pattern is associated with MPO specificity in vasculitides, while in other autoimmune diseases, it mostly involves unknown autoantigens.


Asunto(s)
Anticuerpos Anticitoplasma de Neutrófilos/metabolismo , Enfermedades Autoinmunes/metabolismo , Vasculitis/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Elastasa Pancreática/metabolismo , Adulto Joven
12.
Int J Mol Sci ; 22(14)2021 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-34299162

RESUMEN

IgA, previously called Henoch-Schönlein vasculitis, is an essential immune component that drives the host immune response to the external environment. As IgA has the unique characteristic of a flexible response to broad types of microorganisms, it sometimes causes an autoreactive response in the host human body. IgA vasculitis and related organ dysfunction are representative IgA-mediated autoimmune diseases; bacterial and viral infections often trigger IgA vasculitis. Recent drug developments and the presence of COVID-19 have revealed that these agents can also trigger IgA vasculitis. These findings provide a novel understanding of the pathogenesis of IgA vasculitis. In this review, we focus on the characteristics of IgA and symptoms of IgA vasculitis and other organ dysfunction. We also mention the therapeutic approach, biomarkers, novel triggers for IgA vasculitis, and epigenetic modifications in patients with IgA vasculitis.


Asunto(s)
Biomarcadores/análisis , Epigénesis Genética , Inmunoglobulina A/metabolismo , Vasculitis/terapia , Animales , Humanos , Vasculitis/diagnóstico , Vasculitis/etiología , Vasculitis/metabolismo
13.
Front Immunol ; 12: 640315, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34079539

RESUMEN

Innate immune activity plays an essential role in the development of Kawasaki disease (KD) vasculitis. Extracellular release of high mobility group box-1 (HMGB-1), an endogenous damage-associated molecular pattern protein that can activate the innate immune system and drive host inflammatory responses, may contribute to the development of coronary artery abnormalities in KD. Prednisolone (PSL) added to intravenous immunoglobulin treatment for acute KD may reduce such abnormalities. Here, we evaluate the dynamics of HMGB-1 and therapeutic effects of PSL on HMGB-1-mediated inflammatory pathways on KD vasculitis in vitro. Serum samples were collected prior to initial treatment from patients with KD, systemic juvenile idiopathic arthritis (sJIA), and from healthy controls (VH), then incubated with human coronary artery endothelial cells (HCAECs). Following treatment of KD serum-activated HCAECs with PSL or PBS as a control, effects on the HMGB-1 signaling pathway were evaluated. Compared to that from VH and sJIA, KD serum activation induced HCAEC cytotoxicity and triggered extracellular release of HMGB-1. KD serum-activated HCAECs up-regulated extracellular signal-regulated kinase (ERK)1/2, c-Jun N-terminal kinase (JNK) and, p38 phosphorylation in the cytoplasm and nuclear factor kappa B (NF-κB) phosphorylation in the nucleus and increased interleukin (IL)-1ß and tumor necrosis factor (TNF)-α production. PSL treatment of KD serum-activated HCAECs inhibited extracellular release of HMGB-1, down-regulated ERK1/2, JNK, p38, and NF-κB signaling pathways, and decreased IL-1ß and TNF-α production. Our findings suggest that extracellular HMGB-1 plays an important role in mediating KD pathogenesis and that PSL treatment during the acute phase of KD may ameliorate HMGB-1-mediated inflammatory responses in KD vasculitis.


Asunto(s)
Antiinflamatorios/farmacología , Proteína HMGB1/metabolismo , Síndrome Mucocutáneo Linfonodular/metabolismo , Prednisolona/farmacología , Células Cultivadas , Vasos Coronarios , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteína HMGB1/efectos de los fármacos , Humanos , Inflamación/metabolismo , Síndrome Mucocutáneo Linfonodular/complicaciones , Síndrome Mucocutáneo Linfonodular/tratamiento farmacológico , Vasculitis/etiología , Vasculitis/metabolismo
14.
Front Immunol ; 12: 663412, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34079548

RESUMEN

Endothelial inflammation and damage are the main drivers of cardiovascular risk/disease. Endothelial repair is mediated in part by recruitment of bone marrow endothelial progenitor/endothelial colony forming cells (EPC/ECFC). People with HIV (PWH) have increased cardiovascular risk and the impact of infection in endothelial repair is not well defined. The low frequencies and challenges to in vitro isolation and differentiation of EPC/ECFC from PBMCs had made it difficult to study their role in this context. We hypothesized that HIV driven inflammation induces phenotypic changes that reflects the impact of infection. To test this hypothesis, we evaluated expression of markers of trafficking, endothelial differentiation, and angiogenesis, and study their association with biomarkers of inflammation in a cohort of PWH. In addition, we investigated the relationship of circulating endothelial progenitors and angiogenic T cells, a T cell subset with angiogenic function. Using a flow cytometry approach, we identified two subsets of circulating progenitors LIN4-CD45-CD34+ and LIN4-CD45dimCD34+ in PWH. We found that the phenotype but not frequencies were associated with biomarkers of inflammation. In addition, the percentage of LIN4-CD45dimCD34+ was associated with serum levels of lipids. This data may provide a new tool to better address the impact of HIV infection in endothelial inflammation and repair.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Vasculitis/etiología , Vasculitis/metabolismo , Anciano , Biomarcadores , Relación CD4-CD8 , Enfermedad Crónica , Células Progenitoras Endoteliales/patología , Femenino , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Humanos , Inmunofenotipificación , Mediadores de Inflamación , Metabolismo de los Lípidos , Lípidos/sangre , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Vasculitis/patología
15.
Int J Mol Sci ; 22(8)2021 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-33917744

RESUMEN

Coronary artery disease (CAD) and its complications are the leading cause of death worldwide. Inflammatory activation and dysfunction of the endothelium are key events in the development and pathophysiology of atherosclerosis and are associated with an elevated risk of cardiovascular events. There is great interest to further understand the pathophysiologic mechanisms underlying endothelial dysfunction and atherosclerosis progression, and to identify novel biomarkers and therapeutic strategies to prevent endothelial dysfunction, atherosclerosis and to reduce the risk of developing CAD and its complications. The use of liquid biopsies and new molecular biology techniques have allowed the identification of a growing list of molecular and cellular markers of endothelial dysfunction, which have provided insight on the molecular basis of atherosclerosis and are potential biomarkers and therapeutic targets for the prevention and or treatment of atherosclerosis and CAD. This review describes recent information on normal vascular endothelium function, as well as traditional and novel potential biomarkers of endothelial dysfunction and inflammation, and pharmacological and non-pharmacological therapeutic strategies aimed to protect the endothelium or reverse endothelial damage, as a preventive treatment for CAD and related complications.


Asunto(s)
Biomarcadores , Enfermedad de la Arteria Coronaria/etiología , Enfermedad de la Arteria Coronaria/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Vasculitis/etiología , Vasculitis/metabolismo , Animales , Permeabilidad Capilar , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/fisiopatología , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Hemostasis , Humanos , Terapia Molecular Dirigida/métodos , Vasculitis/tratamiento farmacológico , Vasculitis/fisiopatología
17.
Mol Med Rep ; 23(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495831

RESUMEN

Chronic vascular inflammatory response is an important pathological basis of cardiovascular disease. Genistein (GEN), a natural compound, exhibits anti­inflammatory effects. The aim of the present study was to investigate the effects of GEN on lipopolysaccharide (LPS)­induced chronic vascular inflammatory response in mice and explore the underlying anti­inflammatory mechanisms. C57BL/6 mice were fed with a high­fat diet combined with intraperitoneal injection of LPS to induce chronic vascular inflammation. The expression levels of TNF­α, IL­6 and microRNA (miR)­21 in the vasculature were detected via reverse transcription­quantitative (RT­q)PCR. The protein levels of inducible nitric oxide synthase (iNOS) and NF­κB p65 were detected via western blotting. NF­κB p65 was also analyzed via immunohistochemistry and immunofluorescence (IF). In addition, after transfection with miR­21 mimic or inhibitor for 24 h, vascular endothelial cells (VECs) were treated with GEN and LPS. RT­qPCR and western blot analyses were performed to detect the expression of TNF­α, IL­6, miR­21 and iNOS, and the protein levels of iNOS and NF­κB p65, respectively. IF was used to measure NF­κB p65 nuclear translocation. The results revealed that GEN significantly decreased the expression of inflammation­associated vascular factors in LPS­treated C57BL/6 mice, including TNF­α, IL­6, iNOS, NF­κB p65 and miR­21. Furthermore, miR­21 antagomir enhanced the anti­inflammatory effects of GEN. In LPS­induced VECs, miR­21 mimic increased inflammation­associated factor expression and attenuated the anti­inflammatory effects of GEN, whereas miR­21 inhibitor induced opposing effects. Therefore, the results of the present study suggested that GEN inhibited chronic vascular inflammatory response in mice, which may be associated with the inhibition of VEC inflammatory injury via the miR­21/NF­κB p65 pathway.


Asunto(s)
Células Endoteliales/metabolismo , Genisteína/farmacología , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , MicroARNs/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Vasculitis/metabolismo , Animales , Enfermedad Crónica , Células Endoteliales/patología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/genética , Masculino , Ratones , MicroARNs/genética , Factor de Transcripción ReIA/genética , Vasculitis/inducido químicamente , Vasculitis/tratamiento farmacológico , Vasculitis/genética
18.
J Thorac Cardiovasc Surg ; 161(5): e377-e393, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32007256

RESUMEN

OBJECTIVE: It is unclear whether nucleophosmin (NPM) participates in cardiovascular disease. The present study aimed to investigate the role and underlying mechanisms of NPM in atherosclerosis. METHODS: Levels and location of NPM in human carotid atherosclerotic plaques and healthy controls were detected by real-time polymerase chain reaction, immunoblots, and immunofluorescence. Atherosclerotic prone ApoE-/- mice were fed with a Western diet for 16 weeks as an in vivo model. Human primary umbilical vein endothelial cells (HUVECs) were cultured as an in vitro model. RESULTS: Compared with controls, we found that NPM levels in human carotid atherosclerotic plaques were more than twice as high as in normal arteries, which mainly localized in endothelial cells. In vivo, adenovirus-containing NPM small hairpin RNA attenuated atherosclerotic lesion and promoted plaque stabilization in ApoE-/- mice fed a Western diet by reducing vascular inflammation, maintaining endothelial function, and decreasing macrophage infiltration. Furthermore, NPM knockdown decreased nuclear factor-κB (NF-κB) p65 phosphorylation. In cultured HUVECs, palmitic acid increased the protein levels of NPM and induced the expression of inflammatory cytokines and monocyte adhesion, whereas NPM knockdown attenuated this effect. In HUVECs, NPM protein physically interacted with NF-κB p65 subunit and promoted its nuclear transposition. NPM also increased the transcriptional activity of NF-κB p65 promoter and enhance its binding to target genes, including interleukin-1ß, interleukin-6, intercellular adhesion molecule-1, and E-selectin. CONCLUSIONS: These data provide novel evidence that NPM promotes atherosclerosis by inducing vascular inflammation and endothelial dysfunction through the NF-κB signaling pathway and suggest that NPM may be a promising target for atherosclerosis prevention and treatment.


Asunto(s)
Aterosclerosis/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Endotelio Vascular/metabolismo , Proteínas Nucleares/metabolismo , Placa Aterosclerótica/metabolismo , Vasculitis/metabolismo , Animales , Aterosclerosis/etiología , Enfermedades de las Arterias Carótidas/etiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/etiología , Inflamación/metabolismo , Ratones , FN-kappa B/metabolismo , Proteínas Nucleares/efectos adversos , Proteínas Nucleares/análisis , Nucleofosmina , Transducción de Señal , Vasculitis/etiología
19.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-32990682

RESUMEN

Dysfunction of immune and vascular systems has been implicated in aging and Alzheimer disease; however, their interrelatedness remains poorly understood. The complement pathway is a well-established regulator of innate immunity in the brain. Here, we report robust age-dependent increases in vascular inflammation, peripheral lymphocyte infiltration, and blood-brain barrier (BBB) permeability. These phenotypes were subdued by global inactivation and by endothelial cell-specific ablation of C3ar1. Using an in vitro model of the BBB, we identified intracellular Ca2+ as a downstream effector of C3a/C3aR signaling and a functional mediator of vascular endothelial cadherin junction and barrier integrity. Endothelial C3ar1 inactivation also dampened microglia reactivity and improved hippocampal and cortical volumes in the aging brain, demonstrating a crosstalk between brain vasculature dysfunction and immune cell activation and neurodegeneration. Further, prominent C3aR-dependent vascular inflammation was also observed in a tau-transgenic mouse model. Our studies suggest that heightened C3a/C3aR signaling through endothelial cells promotes vascular inflammation and BBB dysfunction and contributes to overall neuroinflammation in aging and neurodegenerative disease.


Asunto(s)
Envejecimiento/metabolismo , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Endotelio Vascular/metabolismo , Receptores de Complemento/metabolismo , Vasculitis/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Barrera Hematoencefálica/patología , Complemento C3a/genética , Complemento C3a/metabolismo , Endotelio Vascular/patología , Ratones , Ratones Noqueados , Receptores de Complemento/genética , Vasculitis/genética , Vasculitis/patología
20.
J Mol Neurosci ; 71(2): 225-233, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32748330

RESUMEN

The proliferation and migration of vascular smooth muscle cells (VSMCs) are involved in the pathogenesis of intracranial aneurysm (IA) formation and rupture. Interleukin enhancer binding factor 2 (ILF2) is known as the nuclear factor of activated T cells and regulates cell growth. This study was aimed to explore the effects of ILF2 on IA progression. Human brain VSMCs (hBVSMCs) were transfected with pCDNA3.1(+), pCDNA3.1(+)-ILF2, siRNA-negative control, and siRNA-ILF2. The transfection efficiency was then evaluated by determining ILF2 expression. The cell viability and apoptosis were determined using Cell Counting Kit-8 and Annexin V-FITC cell apoptosis assay kit, respectively. Real-time quantification PCR (RT-qPCR) was applied to measure the expression levels of apoptosis-related and inflammation-related genes. Finally, western blot was used to detect the expression level of Fas cell surface death receptor 95 (CD95) and Caspase 8. Overexpression of ILF2 could significantly increase cell viability and decrease cell apoptosis (P < 0.05), while knock-down of ILF2 showed opposite trends for hBVSMCs on cell viability and apoptosis (P < 0.05). RT-qPCR results showed that ILF2 knock-down downregulated the expression levels of BCL2 apoptosis regulator (BCL2), transcriptional regulator Myc-like (c-Myc), and caspase 1 (ICE) whereas upregulated the expression levels of CD95, p21, p53, and interleukin-13 (IL-13). Additionally, the protein expression levels of CD95 and Caspase 8 were significantly decreased after ILF2 overexpression while were significantly increased after ILF2 knock-down (P < 0.05). ILF2 knock-down may inhibit cell viability and promote cell apoptosis of hBVSMCs by regulating the expression levels of apoptosis-related genes and suppressing inflammatory response.


Asunto(s)
Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Proteína del Factor Nuclear 45/fisiología , Apoptosis/efectos de los fármacos , Encéfalo/irrigación sanguínea , Caspasa 8/biosíntesis , Caspasa 8/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Citocinas/genética , Técnicas de Silenciamiento del Gen , Humanos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/metabolismo , Transfección , Vasculitis/metabolismo , Receptor fas/biosíntesis , Receptor fas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...