Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.946
Filtrer
1.
Front Immunol ; 15: 1411979, 2024.
Article de Anglais | MEDLINE | ID: mdl-38989288

RÉSUMÉ

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.


Sujet(s)
Paroi cellulaire , Modèles animaux de maladie humaine , Fibrose , Lacticaseibacillus casei , Maladie de Kawasaki , Vascularite , Animaux , Souris , Paroi cellulaire/immunologie , Vascularite/immunologie , Vascularite/étiologie , Vascularite/anatomopathologie , Maladie de Kawasaki/immunologie , Maladie de Kawasaki/complications , Mâle , Myocardite/étiologie , Myocardite/anatomopathologie , Myocardite/immunologie , Inflammation/immunologie
2.
Front Immunol ; 15: 1423800, 2024.
Article de Anglais | MEDLINE | ID: mdl-39026684

RÉSUMÉ

Thymic epithelial tumors are rare malignancies with an incidence of 1.7 cases per million people per year. They pose significant management challenges due to their association with autoimmune disorders. In this case report, we present the 21-year history of a patient diagnosed with advanced B2/B3 thymoma and Good's syndrome. The patient achieved a complete and durable response after receiving only two cycles of the immune checkpoint inhibitor Nivolumab. However, this positive outcome was accompanied by the development of severe immune-related myocarditis complicated by reactivation of cytomegalovirus. Moreover, the patient developed a highly uncommon subdiaphragmatic pararectal dissemination of the thymic tumor, which is a condition rarely described in the literature. Despite the success in achieving complete and durable response with immune checkpoint inhibitors, the emergence of immune-related adverse events highlights the potential challenges associated with these treatments, emphasizing the need for careful monitoring and a comprehensive understanding of the intricate interplay between cancer, immune system dysregulations and immunotherapy.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Tumeurs du thymus , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Tumeurs du thymus/immunologie , Tumeurs du thymus/thérapie , Tumeurs du thymus/traitement médicamenteux , Thymome/immunologie , Thymome/thérapie , Nivolumab/usage thérapeutique , Nivolumab/effets indésirables , Mâle , Immunothérapie/méthodes , Immunothérapie/effets indésirables , Myocardite/étiologie , Myocardite/immunologie , Myocardite/thérapie , Myocardite/traitement médicamenteux , Résultat thérapeutique , Adulte d'âge moyen , Tumeurs épithéliales épidermoïdes et glandulaires
3.
Int J Mol Sci ; 25(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38891767

RÉSUMÉ

Myocarditis is characterized by an influx of inflammatory cells, predominantly of myeloid lineage. The progression of myocarditis to a dilated cardiomyopathy is markedly influenced by TGF-ß signalling. Here, we investigate the role of TGF-ß signalling in inflammatory cardiac macrophages in the development of myocarditis and post-inflammatory fibrosis. Experimental autoimmune myocarditis (EAM) was induced in the LysM-Cre × R26-stop-EYFP × Tgfbr2-fl/fl transgenic mice showing impaired TGF-ß signalling in the myeloid lineage and the LysM-Cre × R26-stop-EYFP control mice. In EAM, immunization led to acute myocarditis on day 21, followed by cardiac fibrosis on day 40. Both strains showed a similar severity of myocarditis and the extent of cardiac fibrosis. On day 21 of EAM, an increase in cardiac inflammatory macrophages was observed in both strains. These cells were sorted and analysed for differential gene expression using whole-genome transcriptomics. The analysis revealed activation and regulation of the inflammatory response, particularly the production of both pro-inflammatory and anti-inflammatory cytokines and cytokine receptors as TGF-ß-dependent processes. The analysis of selected cytokines produced by bone marrow-derived macrophages confirmed their suppressed secretion. In conclusion, our findings highlight the regulatory role of TGF-ß signalling in cytokine production within inflammatory cardiac macrophages during myocarditis.


Sujet(s)
Maladies auto-immunes , Cytokines , Macrophages , Souris transgéniques , Myocardite , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Myocardite/métabolisme , Myocardite/immunologie , Myocardite/anatomopathologie , Myocardite/étiologie , Facteur de croissance transformant bêta/métabolisme , Souris , Macrophages/métabolisme , Macrophages/immunologie , Maladies auto-immunes/métabolisme , Maladies auto-immunes/immunologie , Maladies auto-immunes/anatomopathologie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocarde/immunologie , Fibrose , Mâle
4.
Circ Res ; 134(12): 1703-1717, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843287

RÉSUMÉ

Fibroblasts are essential for building and maintaining the structural integrity of all organs. Moreover, fibroblasts can acquire an inflammatory phenotype to accommodate immune cells in specific niches and to provide migration, differentiation, and growth factors. In the heart, balancing of fibroblast activity is critical for cardiac homeostasis and optimal organ function during inflammation. Fibroblasts sustain cardiac homeostasis by generating local niche environments that support housekeeping functions and by actively engaging in intercellular cross talk. During inflammatory perturbations, cardiac fibroblasts rapidly switch to an inflammatory state and actively communicate with infiltrating immune cells to orchestrate immune cell migration and activity. Here, we summarize the current knowledge on the molecular landscape of cardiac fibroblasts, focusing on their dual role in promoting tissue homeostasis and modulating immune cell-cardiomyocyte interaction. In addition, we discuss potential future avenues for manipulating cardiac fibroblast activity during myocardial inflammation.


Sujet(s)
Fibroblastes , Homéostasie , Myocarde , Humains , Animaux , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibroblastes/immunologie , Myocarde/anatomopathologie , Myocarde/immunologie , Myocarde/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/immunologie , Myocardite/immunologie , Myocardite/anatomopathologie , Myocardite/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Communication cellulaire
5.
Circ Res ; 134(12): 1767-1790, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843292

RÉSUMÉ

Autoimmunity significantly contributes to the pathogenesis of myocarditis, underscored by its increased frequency in autoimmune diseases such as systemic lupus erythematosus and polymyositis. Even in cases of myocarditis caused by viral infections, dysregulated immune responses contribute to pathogenesis. However, whether triggered by existing autoimmune conditions or viral infections, the precise antigens and immunologic pathways driving myocarditis remain incompletely understood. The emergence of myocarditis associated with immune checkpoint inhibitor therapy, commonly used for treating cancer, has afforded an opportunity to understand autoimmune mechanisms in myocarditis, with autoreactive T cells specific for cardiac myosin playing a pivotal role. Despite their self-antigen recognition, cardiac myosin-specific T cells can be present in healthy individuals due to bypassing the thymic selection stage. In recent studies, novel modalities in suppressing the activity of pathogenic T cells including cardiac myosin-specific T cells have proven effective in treating autoimmune myocarditis. This review offers an overview of the current understanding of heart antigens, autoantibodies, and immune cells as the autoimmune mechanisms underlying various forms of myocarditis, along with the latest updates on clinical management and prospects for future research.


Sujet(s)
Maladies auto-immunes , Myocardite , Myocardite/immunologie , Myocardite/thérapie , Myocardite/étiologie , Humains , Maladies auto-immunes/immunologie , Maladies auto-immunes/thérapie , Maladies auto-immunes/traitement médicamenteux , Animaux , Autoanticorps/immunologie , Auto-immunité , Lymphocytes T/immunologie , Autoantigènes/immunologie , Myosines cardiaques/immunologie
6.
Circ Res ; 134(12): 1752-1766, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38843295

RÉSUMÉ

Heart failure (HF) is characterized by a progressive decline in cardiac function and represents one of the largest health burdens worldwide. Clinically, 2 major types of HF are distinguished based on the left ventricular ejection fraction (EF): HF with reduced EF and HF with preserved EF. While both types share several risk factors and features of adverse cardiac remodeling, unique hallmarks beyond ejection fraction that distinguish these etiologies also exist. These differences may explain the fact that approved therapies for HF with reduced EF are largely ineffective in patients suffering from HF with preserved EF. Improving our understanding of the distinct cellular and molecular mechanisms is crucial for the development of better treatment strategies. This article reviews the knowledge of the immunologic mechanisms underlying HF with reduced and preserved EF and discusses how the different immune profiles elicited may identify attractive therapeutic targets for these conditions. We review the literature on the reported mechanisms of adverse cardiac remodeling in HF with reduced and preserved EF, as well as the immune mechanisms involved. We discuss how the knowledge gained from preclinical models of the complex syndrome of HF as well as from clinical data obtained from patients may translate to a better understanding of HF and result in specific treatments for these conditions in humans.


Sujet(s)
Défaillance cardiaque , Débit systolique , Remodelage ventriculaire , Humains , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/immunologie , Animaux , Myocardite/physiopathologie , Myocardite/immunologie , Fonction ventriculaire gauche , Myocarde/anatomopathologie , Myocarde/métabolisme , Myocarde/immunologie
7.
J Clin Invest ; 134(13)2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38768074

RÉSUMÉ

Myocarditis is clinically characterized by chest pain, arrhythmias, and heart failure, and treatment is often supportive. Mutations in DSP, a gene encoding the desmosomal protein desmoplakin, have been increasingly implicated in myocarditis. To model DSP-associated myocarditis and assess the role of innate immunity, we generated engineered heart tissues (EHTs) using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from patients with heterozygous DSP truncating variants (DSPtvs) and a gene-edited homozygous deletion cell line (DSP-/-). At baseline, DSP-/- EHTs displayed a transcriptomic signature of innate immune activation, which was mirrored by cytokine release. Importantly, DSP-/- EHTs were hypersensitive to Toll-like receptor (TLR) stimulation, demonstrating more contractile dysfunction compared with isogenic controls. Relative to DSP-/- EHTs, heterozygous DSPtv EHTs had less functional impairment. DSPtv EHTs displayed heightened sensitivity to TLR stimulation, and when subjected to strain, DSPtv EHTs developed functional deficits, indicating reduced contractile reserve compared with healthy controls. Colchicine or NF-κB inhibitors improved strain-induced force deficits in DSPtv EHTs. Genomic correction of DSP p.R1951X using adenine base editing reduced inflammatory biomarker release from EHTs. Thus, EHTs replicate electrical and contractile phenotypes seen in human myocarditis, implicating cytokine release as a key part of the myogenic susceptibility to inflammation. The heightened innate immune activation and sensitivity are targets for clinical intervention.


Sujet(s)
Immunité innée , Cellules souches pluripotentes induites , Myocardite , Myocytes cardiaques , Humains , Myocardite/génétique , Myocardite/immunologie , Myocardite/anatomopathologie , Immunité innée/génétique , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/immunologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/immunologie , Myocytes cardiaques/anatomopathologie , Mâle , Prédisposition génétique à une maladie , Femelle
10.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38696536

RÉSUMÉ

Major 5'-terminally deleted (5'TD) RNA forms of group-B coxsackievirus (CVB-5'TD) has been associated with myocarditis in both mice and humans. Although it is known that interferon-ß (IFN-ß) signaling is critical for an efficient innate immune response against CVB-induced myocarditis, the link between CVB-5'TD RNA forms and type I IFN signaling in cardiomyocytes remains to be explored. In a mouse model of CVB3/28-induced myocarditis, major early-emerging forms of CVB-5'TD RNA have been characterized as replicative viral populations that impair IFN-ß production in the heart. Synthetic CVB3/28 RNA forms mimicking each of these major 5'TD virus populations were transfected in mice and have been shown to modulate innate immune responses in the heart and to induce myocarditis in mice. Remarkably, transfection of synthetic viral RNA with deletions in the secondary structures of the 5'-terminal CVB3 RNA domain I, modifying stem-loops "b", "c" or "d", were found to impair IFN-ß production in human cardiomyocytes. In addition, the activation of innate immune response by Poly(I:C), was found to restore IFN-ß production and to reduce the burden of CVB-5'TD RNA-forms in cardiac tissues, thereby reducing the mortality rate of infected mice. Overall, our results indicate that major early-emerging CVB3 populations deleted in the domain I of genomic RNA, in the 5' noncoding region, modulate the activation of the type I IFN pathway in cardiomyocytes and induce myocarditis in mice. These findings shed new light on the role of replicative CVB-5'TD RNA forms as key pathophysiological factors in CVB-induced human myocarditis.


Sujet(s)
Infections à virus coxsackie , Entérovirus humain B , Interféron de type I , Myocardite , Myocytes cardiaques , ARN viral , Myocardite/virologie , Myocardite/immunologie , Myocardite/génétique , Animaux , Myocytes cardiaques/virologie , Myocytes cardiaques/métabolisme , Souris , Entérovirus humain B/immunologie , Infections à virus coxsackie/immunologie , Infections à virus coxsackie/virologie , Infections à virus coxsackie/génétique , Interféron de type I/métabolisme , ARN viral/génétique , ARN viral/métabolisme , Humains , Immunité innée , Transduction du signal , Interféron bêta/métabolisme , Interféron bêta/génétique , Interféron bêta/immunologie , Mâle , Régions 5' non traduites
11.
Front Immunol ; 15: 1380697, 2024.
Article de Anglais | MEDLINE | ID: mdl-38715608

RÉSUMÉ

The Corona Virus Disease (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), has quickly spread worldwide and resulted in significant morbidity and mortality. Although most infections are mild, some patients can also develop severe and fatal myocarditis. In eukaryotic RNAs, 5-methylcytosine (m5C) is a common kind of post-transcriptional modification, which is involved in regulating various biological processes (such as RNA export, translation, and stability maintenance). With the rapid development of m5C modification detection technology, studies related to viral m5C modification are ever-increasing. These studies have revealed that m5C modification plays an important role in various stages of viral replication, including transcription and translation. According to recent studies, m5C methylation modification can regulate SARS-CoV-2 infection by modulating innate immune signaling pathways. However, the specific role of m5C modification in SARS-CoV-2-induced myocarditis remains unclear. Therefore, this review aims to provide insights into the molecular mechanisms of m5C methylation in SARS-CoV-2 infection. Moreover, the regulatory role of NSUN2 in viral infection and host innate immune response was also highlighted. This review may provide new directions for developing therapeutic strategies for SARS-CoV-2-associated myocarditis.


Sujet(s)
COVID-19 , Myocardite , SARS-CoV-2 , Myocardite/virologie , Myocardite/immunologie , Myocardite/thérapie , Myocardite/génétique , Humains , COVID-19/immunologie , COVID-19/génétique , COVID-19/thérapie , SARS-CoV-2/physiologie , Méthylation , 5-Méthyl-cytosine/métabolisme , Immunité innée , Traitements médicamenteux de la COVID-19 , Animaux , ARN viral/génétique , ARN viral/métabolisme , Maturation post-transcriptionnelle des ARN
12.
Nat Commun ; 15(1): 4153, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755212

RÉSUMÉ

Viral myocarditis, an inflammatory disease of the heart, causes significant morbidity and mortality. Type I interferon (IFN)-mediated antiviral responses protect against myocarditis, but the mechanisms are poorly understood. We previously identified A Disintegrin And Metalloproteinase domain 9 (ADAM9) as an important factor in viral pathogenesis. ADAM9 is implicated in a range of human diseases, including inflammatory diseases; however, its role in viral infection is unknown. Here, we demonstrate that mice lacking ADAM9 are more susceptible to encephalomyocarditis virus (EMCV)-induced death and fail to mount a characteristic type I IFN response. This defect in type I IFN induction is specific to positive-sense, single-stranded RNA (+ ssRNA) viruses and involves melanoma differentiation-associated protein 5 (MDA5)-a key receptor for +ssRNA viruses. Mechanistically, ADAM9 binds to MDA5 and promotes its oligomerization and thereby downstream mitochondrial antiviral-signaling protein (MAVS) activation in response to EMCV RNA stimulation. Our findings identify a role for ADAM9 in the innate antiviral response, specifically MDA5-mediated IFN production, which protects against virus-induced cardiac damage, and provide a potential therapeutic target for treatment of viral myocarditis.


Sujet(s)
Protéines ADAM , Infections à cardiovirus , Virus de l'encéphalomyocardite , Immunité innée , Interféron de type I , Hélicase IFIH1 inductrice de l'interféron , Protéines membranaires , Souris knockout , Myocardite , Animaux , Virus de l'encéphalomyocardite/immunologie , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Hélicase IFIH1 inductrice de l'interféron/génétique , Hélicase IFIH1 inductrice de l'interféron/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie , Infections à cardiovirus/immunologie , Infections à cardiovirus/virologie , Protéines ADAM/métabolisme , Protéines ADAM/génétique , Protéines ADAM/immunologie , Souris , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines membranaires/immunologie , Myocardite/immunologie , Myocardite/virologie , Humains , Souris de lignée C57BL , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/immunologie , Transduction du signal/immunologie , Mâle , Cellules HEK293
13.
Free Radic Biol Med ; 218: 149-165, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38570171

RÉSUMÉ

Proper protein degradation is required for cellular protein homeostasis and organ function. Particularly, in post-mitotic cells, such as cardiomyocytes, unbalanced proteolysis due to inflammatory stimuli and oxidative stress contributes to organ dysfunction. To ensure appropriate protein turnover, eukaryotic cells exert two main degradation systems, the ubiquitin-proteasome-system and the autophagy-lysosome-pathway. It has been shown that proteasome activity affects the development of cardiac dysfunction differently, depending on the type of heart failure. Studies analyzing the inducible subtype of the proteasome, the immunoproteasome (i20S), demonstrated that the i20S plays a double role in diseased hearts. While i20S subunits are increased in cardiac hypertrophy, atrial fibrillation and partly in myocarditis, the opposite applies to diabetic cardiomyopathy and ischemia/reperfusion injury. In addition, the i20S appears to play a role in autophagy modulation depending on heart failure phenotype. This review summarizes the current literature on the i20S in different heart failure phenotypes, emphasizing the two faces of i20S in injured hearts. A selection of established i20S inhibitors is introduced and signaling pathways linking the i20S to autophagy are highlighted. Mapping the interplay of the i20S and autophagy in different types of heart failure offers potential approaches for developing treatment strategies against heart failure.


Sujet(s)
Autophagie , Défaillance cardiaque , Proteasome endopeptidase complex , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/génétique , Défaillance cardiaque/immunologie , Humains , Proteasome endopeptidase complex/métabolisme , Animaux , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Phénotype , Transduction du signal , Protéolyse , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/génétique , Myocardite/anatomopathologie , Myocardite/métabolisme , Myocardite/immunologie , Myocardite/génétique , Cardiomégalie/anatomopathologie , Cardiomégalie/métabolisme , Cardiomégalie/génétique
14.
Immunology ; 172(2): 235-251, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38425094

RÉSUMÉ

Myocarditis has emerged as a rare but lethal immune checkpoint inhibitor (ICI)-associated toxicity. However, the exact mechanism and the specific therapeutic targets remain underexplored. In this study, we aim to characterise the transcriptomic profiles based on single-cell RNA sequencing from ICI-related myocarditis. Peripheral blood mononuclear cell (PBMC) samples were collected from four groups for single-cell RNA sequencing: (1) patients with newly diagnosed lung squamous cell carcinoma before treatment (Control Group); (2) patients with lung squamous cell carcinoma with PD-1 inhibitor therapy who did not develop myocarditis (PD-1 Group); (3) patients during fulminant ICI-related myocarditis onset (Myocarditis Group); and (4) Patients with fulminant ICI-related myocarditis during disease remission (Recovery Group). Subcluster determination, functional analysis, single-cell trajectory and cell-cell interaction analysis were performed after scRNA-seq. Bulk-RNA sequencing was performed for further validation. Our results revealed the diversity of cellular populations in ICI-related myocarditis, marked by their distinct transcriptional profiles and biological functions. Monocytes, NKs as well as B cells contribute to the regulation of innate immunity and inflammation in ICI-related myocarditis. With integrated analysis of scRNA-seq and bulk sequencing, we identified S100A protein family as a potential serum marker for ICI-related myocarditis. Our study has created a cell atlas of PBMC during ICI-related myocarditis, which would shed light on the pathophysiological mechanism and potential therapeutic targets of ICI-related myocarditis in continuous exploration.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Immunité innée , Tumeurs du poumon , Myocardite , Analyse sur cellule unique , Humains , Myocardite/immunologie , Myocardite/induit chimiquement , Myocardite/génétique , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/génétique , Transcriptome , Analyse de séquence d'ARN , Agranulocytes/immunologie , Agranulocytes/métabolisme , Sujet âgé , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/génétique , Analyse de profil d'expression de gènes
16.
Immunol Cell Biol ; 101(8): 735-745, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37253434

RÉSUMÉ

Coxsackievirus B3 (CVB3)-induced viral myocarditis (VMC) is characterized by immune cell infiltration and myocardial damage. High mobility group box 1 (HMGB1) is a highly conserved nuclear DNA-binding protein that participates in DNA replication, transcriptional regulation, repair response and inflammatory response in different disease models. To investigate the exact function of HMGB1 in CVB3-induced VMC, we crossed Hmgb1-floxed (Hmgb1f/f ) mice with mice carrying a suitable Cre recombinase transgenic strain to achieve conditional inactivation of the Hmgb1 gene in a cardiomyocyte-specific manner and to establish myocarditis. In this study, we found that cardiomyocyte-specific Hmgb1-deficient (Hmgb1f/f TgCre/+ ) mice exhibited exacerbated myocardial injury. Hmgb1-deficient cardiomyocytes may promote early apoptosis via the p53-mediated Bax mitochondrial pathway, as evidenced by the higher localization of p53 protein in the cytosol of Hmgb1-deficient cardiomyocytes upon CVB3 infection. Moreover, cardiomyocyte Hmgb1-deficient mice are more susceptible to cardiac dysfunction after infection. This study provides new insights into HMGB1 in VMC pathogenesis and a strategy for appropriate blocking of HMGB1 in the clinical treatment of VMC.


Sujet(s)
Infections à virus coxsackie , Entérovirus humain B , Protéine HMGB1 , Myocardite , Animaux , Souris , Apoptose/génétique , Protéine HMGB1/métabolisme , Souris de lignée BALB C , Myocardite/immunologie , Myocardite/anatomopathologie , Myocardite/virologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Protéine p53 suppresseur de tumeur/métabolisme , Infections à virus coxsackie/immunologie
17.
Nat Commun ; 14(1): 1299, 2023 03 09.
Article de Anglais | MEDLINE | ID: mdl-36894554

RÉSUMÉ

mRNA-based vaccines dramatically reduce the occurrence and severity of COVID-19, but are associated with rare vaccine-related adverse effects. These toxicities, coupled with observations that SARS-CoV-2 infection is associated with autoantibody development, raise questions whether COVID-19 vaccines may also promote the development of autoantibodies, particularly in autoimmune patients. Here we used Rapid Extracellular Antigen Profiling to characterize self- and viral-directed humoral responses after SARS-CoV-2 mRNA vaccination in 145 healthy individuals, 38 patients with autoimmune diseases, and 8 patients with mRNA vaccine-associated myocarditis. We confirm that most individuals generated robust virus-specific antibody responses post vaccination, but that the quality of this response is impaired in autoimmune patients on certain modes of immunosuppression. Autoantibody dynamics are remarkably stable in all vaccinated patients compared to COVID-19 patients that exhibit an increased prevalence of new autoantibody reactivities. Patients with vaccine-associated myocarditis do not have increased autoantibody reactivities relative to controls. In summary, our findings indicate that mRNA vaccines decouple SARS-CoV-2 immunity from autoantibody responses observed during acute COVID-19.


Sujet(s)
Maladies auto-immunes , Vaccins contre la COVID-19 , COVID-19 , Immunité humorale , Vaccins synthétiques , Vaccins à ARNm , Humains , Anticorps antiviraux/immunologie , Autoanticorps/immunologie , Maladies auto-immunes/immunologie , Auto-immunité/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , Vaccins contre la COVID-19/effets indésirables , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/usage thérapeutique , Effets secondaires indésirables des médicaments/immunologie , Immunité humorale/immunologie , Myocardite/immunologie , ARN messager , SARS-CoV-2 , Vaccination , Vaccins synthétiques/effets indésirables , Vaccins synthétiques/immunologie , Vaccins synthétiques/usage thérapeutique , Vaccins à ARNm/effets indésirables , Vaccins à ARNm/immunologie , Vaccins à ARNm/usage thérapeutique
20.
Int J Cardiol ; 359: 108-112, 2022 07 15.
Article de Anglais | MEDLINE | ID: mdl-35395284

RÉSUMÉ

BACKGROUND: Early detection and initiation of treatment in cardiac sarcoidosis (CS) is believed to be crucial to reduce morbidity and mortality. The diagnosis of CS is challenging, especially in isolated CS (ICS). Certain human leukocyte antigen (HLA-DRB1) alleles associate with different phenotypes of sarcoidosis. Phenotypic and genotypic characterization of patients with CS may improve our ability to identify patients being at risk for developing CS. METHODS: 87 patients with CS, identified at two Swedish university hospitals were included. Phenotypic characteristics were extracted from the medical records and the patients were HLA-DRB1 typed. RESULTS: Median age at diagnosis was 55 years, 37% were women. HLA-DRB1 distribution was similar to a general sarcoidosis population. A majority of patients (51/87) had CS as the first sarcoidosis presentation. They were younger (p = 0.04), more often presenting with ventricular tachycardia (VT) or atrioventricular block (AVB) grade II or III (p < 0.001), had lower left ventricular ejection fraction (LVEF) (p = 0.002), lower serum angiotensin converting enzyme (s-ACE) (p = 0.025), and fewer extra cardiac manifestations (ECM) (p = 0.02) than those presenting with CS later. CONCLUSIONS: Of Swedish CS patients, 59% presented with cardiac involvement as first manifestation. They had more severe cardiac symptoms than patients presenting with CS later. This phenotype disclosed less ECM and lower s-ACE thus diagnosis can be missed or delayed. We did not observe significant differences in HLA-DRB1 allele frequency between patients with CS compared to sarcoidosis in general. Awareness of CS as a primary manifestation can enable early detection and adequate intervention.


Sujet(s)
Chaines HLA-DRB1 , Myocardite , Sarcoïdose , Allèles , Femelle , Chaines HLA-DRB1/génétique , Chaines HLA-DRB1/immunologie , Humains , Mâle , Myocardite/génétique , Myocardite/immunologie , Phénotype , Sarcoïdose/génétique , Sarcoïdose/immunologie , Débit systolique , Suède , Fonction ventriculaire gauche
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE