Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 482
Filtrar
1.
Turk J Pediatr ; 66(4): 439-447, 2024 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-39387427

RESUMEN

BACKGROUND: Viral myocarditis (VMC) is common in children. Previous studies have reported the clinical value of nuclear paraspeckle assembly transcript 1 (NEAT1) and microRNA-425-3p (miR-425-3p) in certain diseases, but not in VMC. This article was designed to investigate the expression of long noncoding RNA (lncRNA) NEAT1 and miR-425-3p in the serum of patients with VMC and their clinical significance. METHODS: We assessed VMC and healthy patients and analyzed differences in the expression levels of NEAT1 and miR-425-3p. The correlation and targeting relationship between the two were reported by Spearman correlation analysis and luciferase reporter assay. ROC curves were plotted to reflect the diagnostic effect of both. In addition, according to the 12-month prognostic effect grouping, patients with VMC were separated into a group with good vs. poor prognosis, and the difference in the expression levels of NEAT1 and miR-425-3p between the two groups were analyzed. The ability of the two markers in the prognosis of VMC was further analyzed by multiple logistic regression. RESULTS: NEAT1 expression was up-regulated in VMC and miR-425-3p expression was down-regulated, and there was a negative correlation and targeting link between the two. The diagnostic efficacy of both NEAT1 and miR-425-3p was higher than that of a single indicator. High expression of NEAT1 and low expression of miR-425-3p were found in VMC patients with poor prognosis. Both were independent influencers of VMC prognosis. CONCLUSION: NEAT1 and miR-425-3p expressions were affected by VMC and had important clinical implications for VMC, indicating for the first time the clinical function of NEAT1 and miR-425-3p in VMC.


Asunto(s)
MicroARNs , Miocarditis , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Miocarditis/genética , Miocarditis/metabolismo , Miocarditis/diagnóstico , Miocarditis/virología , MicroARNs/genética , MicroARNs/metabolismo , Masculino , Femenino , Pronóstico , Preescolar , Niño , Biomarcadores/sangre , Biomarcadores/metabolismo , Lactante , Estudios de Casos y Controles , Relevancia Clínica
2.
Int J Mol Sci ; 25(17)2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39273613

RESUMEN

Myocarditis is an inflammatory disease that may lead to dilated cardiomyopathy. Viral infection of the myocardium triggers immune responses, which involve, among others, macrophage infiltration, oxidative stress, expression of pro-inflammatory cytokines, and microRNAs (miRNAs). The cardioprotective role of estrogen in myocarditis is well documented; however, sex differences in the miRNA expression in chronic myocarditis are still poorly understood, and studying them further was the aim of the present study. Male and female ABY/SnJ mice were infected with CVB3. Twenty-eight days later, cardiac tissue from both infected and control mice was used for real-time PCR and Western blot analysis. NFκB, IL-6, iNOS, TNF-α, IL-1ß, MCP-1, c-fos, and osteopontin (OPN) were used to examine the inflammatory state in the heart. Furthermore, the expression of several inflammation- and remodeling-related miRNAs was analyzed. NFκB, IL-6, TNF-α, IL-1ß, iNOS, and MCP-1 were significantly upregulated in male mice with CVB3-induced chronic myocarditis, whereas OPN mRNA expression was increased only in females. Further analysis revealed downregulation of some anti-inflammatory miRNA in male hearts (let7a), with upregulation in female hearts (let7b). In addition, dysregulation of remodeling-related miRNAs (miR27b and mir199a) in a sex-dependent manner was observed. Taken together, the results of the present study suggest a sex-specific expression of pro-inflammatory markers as well as inflammation- and remodeling-related miRNAs, with a higher pro-inflammatory response in male CVB3 myocarditis mice.


Asunto(s)
Infecciones por Coxsackievirus , Modelos Animales de Enfermedad , MicroARNs , Miocarditis , Animales , Miocarditis/metabolismo , Miocarditis/virología , Miocarditis/genética , MicroARNs/genética , MicroARNs/metabolismo , Femenino , Masculino , Ratones , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/virología , Enterovirus Humano B , Biomarcadores/metabolismo , Caracteres Sexuales , Citocinas/metabolismo , Citocinas/genética , Miocardio/metabolismo , Miocardio/patología , Inflamación/genética , Inflamación/metabolismo , Factores Sexuales , Regulación de la Expresión Génica
3.
Genes (Basel) ; 15(9)2024 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-39336825

RESUMEN

Myocarditis is an inflammatory condition of cardiac tissue presenting significant variability in clinical manifestations and outcomes. Its etiology is diverse, encompassing infectious agents (primarily viruses, but also bacteria, protozoa, and helminths) and non-infectious factors (autoimmune responses, toxins, and drugs), though often the specific cause remains unidentified. Recent research has highlighted the potential role of genetic susceptibility in the development of myocarditis (and in some cases the development of inflammatory dilated cardiomyopathy, i.e., the condition in which there is chronic inflammation (>3 months) and left ventricular dysfunction\dilatation), with several studies indicating a correlation between myocarditis and genetic backgrounds. Notably, pathogenic genetic variants linked to dilated or arrhythmic cardiomyopathy are found in 8-16% of myocarditis patients. Genetic predispositions can lead to recurrent myocarditis and a higher incidence of ventricular arrhythmias and heart failure. Moreover, the presence of DSP mutations has been associated with distinct pathological patterns and clinical outcomes in arrhythmogenic cardiomyopathy (hot phases). The interplay between genetic factors and environmental triggers, such as viral infections and physical stress, is crucial in understanding the pathogenesis of myocarditis. Identifying these genetic markers can improve the diagnosis, risk stratification, and management of patients with myocarditis, potentially guiding tailored therapeutic interventions. This review aims to synthesize current knowledge on the genetic underpinnings of myocarditis, with an emphasis on desmoplakin-related arrhythmogenic cardiomyopathy, to enhance clinical understanding and inform future research directions.


Asunto(s)
Desmoplaquinas , Miocarditis , Humanos , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Desmoplaquinas/genética , Predisposición Genética a la Enfermedad , Inflamación/genética , Inflamación/patología , Mutación , Miocarditis/genética , Miocarditis/patología , Fenotipo
4.
Int J Mol Sci ; 25(16)2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39201692

RESUMEN

Acute pancreatitis (AP) is an inflammatory disease initiated by the death of exocrine acinar cells, but its pathogenesis remains unclear. Signal transducer and activator of transcription 3 (STAT3) is a multifunctional factor that regulates immunity and the inflammatory response. The protective role of STAT3 is reported in Coxsackievirus B3 (CVB3)-induced cardiac fibrosis, yet the exact role of STAT3 in modulating viral-induced STAT1 activation and type I interferon (IFN)-stimulated gene (ISG) transcription in the pancreas remains unclarified. In this study, we tested whether STAT3 regulated viral-induced STAT1 translocation. We found that CVB3, particularly capsid VP1 protein, markedly upregulated the phosphorylation and nuclear import of STAT3 (p-STAT3) while it significantly impeded the nuclear translocation of p-STAT1 in the pancreases and hearts of mice on day 3 postinfection (p.i.). Immunoblotting and an immunofluorescent assay demonstrated the increased expression and nuclear translocation of p-STAT3 but a blunted p-STAT1 nuclear translocation in CVB3-infected acinar 266-6 cells. STAT3 shRNA knockdown or STAT3 inhibitors reduced viral replication via the rescue of STAT1 nuclear translocation and increasing the ISRE activity and ISG transcription in vitro. The knockdown of STAT1 blocked the antiviral effect of the STAT3 inhibitor. STAT3 inhibits STAT1 activation by virally inducing a potent inhibitor of IFN signaling, the suppressor of cytokine signaling-3 ((SOCS)-3). Sustained pSTAT1 and the elevated expression of ISGs were induced in SOCS3 knockdown cells. The in vivo administration of HJC0152, a pharmaceutical STAT3 inhibitor, mitigated the viral-induced AP and myocarditis pathology via increasing the IFNß as well as ISG expression on day 3 p.i. and reducing the viral load in multi-organs. These findings define STAT3 as a negative regulator of the type I IFN response via impeding the nuclear STAT1 translocation that otherwise triggers ISG induction in infected pancreases and hearts. Our findings identify STAT3 as an antagonizing factor of the IFN-STAT1 signaling pathway and provide a potential therapeutic target for viral-induced AP and myocarditis.


Asunto(s)
Enterovirus Humano B , Miocarditis , Pancreatitis , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Replicación Viral , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Miocarditis/virología , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Animales , Pancreatitis/metabolismo , Pancreatitis/virología , Pancreatitis/patología , Pancreatitis/genética , Enterovirus Humano B/fisiología , Ratones , Humanos , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/virología , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/genética , Núcleo Celular/metabolismo , Masculino , Transporte Activo de Núcleo Celular , Regulación de la Expresión Génica , Enfermedad Aguda , Línea Celular , Transducción de Señal
5.
PLoS One ; 19(8): e0307779, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39150929

RESUMEN

OBJECTIVE: The main pathological change of myocarditis is an inflammatory injury of cardiomyocytes. Long noncoding RNAs (lncRNAs) are closely related to inflammation, and our previous study showed that differential expression of lncRNAs is associated with myocarditis. This study aimed to investigate the impact of lncRNAs on the onset of myocarditis. METHODS: RNA expression was measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR). Lipopolysaccharide (LPS) was used to induce inflammation in human cardiomyocytes (HCMs). The expression of inflammatory cytokines and myocardial injury markers was detected by enzyme-linked immunosorbent assay (ELISA) and RT-qPCR. Cell viability and apoptosis were measured by the cell counting kit-8 assay and flow cytometry. The binding force between lncRNA NONHSAT122636.2 and microRNA miRNA-2110 was detected using the dual-luciferase assay. RESULTS: NONHSAT122636.2 was dynamically expressed in patients with myocarditis and negatively correlated with inflammation severity. The overexpression of NONHSAT122636.2 improved inflammatory injury in LPS-stimulated HCMs. The study observed that there was a weak binding force between NONHSAT122636.2 and miR-2110. CONCLUSION: NONHSAT122636.2 attenuates myocardial inflammation and apoptosis in myocarditis. Additionally, its expression decreases in the peripheral blood of children suffering from myocarditis and in patients who are diagnosed for the first time showing higher diagnostic sensitivity and specificity. This decrease is negatively correlated with the degree of inflammation. Overall, the study suggests that NONHSAT122636.2 can be exploited as a potential diagnostic biomarker for pediatric myocarditis.


Asunto(s)
Apoptosis , MicroARNs , Miocarditis , Miocitos Cardíacos , ARN Largo no Codificante , Miocarditis/genética , Miocarditis/patología , Miocarditis/metabolismo , ARN Largo no Codificante/genética , Humanos , Apoptosis/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Masculino , Femenino , Lipopolisacáridos/farmacología , Niño , Inflamación/genética , Inflamación/patología , Preescolar , Citocinas/metabolismo , Citocinas/genética
6.
Int J Biol Macromol ; 278(Pt 1): 134193, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39069042

RESUMEN

Long non-coding RNAs (lncRNAs) have been implicated in dilated cardiomyopathy (DCM). However, the biological functions and regulatory mechanisms of lncRNAs in DCM remain elusive. Using a mouse model of experimental autoimmune myocarditis (EAM) to mimic DCM, we successfully constructed a dynamic lncRNA expression library for EAM by lncRNA microarray and found that the expression of a macrophage-enriched lncRNA, MAAMT, was significantly increased in the myocardial tissue of mice at the acute stage of EAM. Functionally, MAAMT knockdown alleviated the recruitment and proinflammatory activation of macrophages in the heart, spleen, and peripheral blood of mice at the acute stage of EAM, reduced myocardial inflammation and injury, and eventually reversed ventricular remodelling and improved cardiac function in mice at the chronic stage of EAM. Mechanistically, we identified serine/arginine-rich splicing factor 1 (SRSF1) as an MAAMT-interacting protein in macrophages using RNA pull-down assays coupled with mass spectrometry. MAAMT knockdown attenuated the ubiquitination-mediated degradation of SRSF1, increased the protein expression of SRSF1, and restrained the activation of the NF-κB pathway in macrophages, thereby inhibiting the proinflammatory activation of macrophages. Collectively, our results demonstrate that MAAMT is a key proinflammatory regulator of myocarditis that promotes macrophage activation through the SRSF1-NF-κB axis, providing a new insight into early effective treatment strategies for DCM.


Asunto(s)
Enfermedades Autoinmunes , Macrófagos , Miocarditis , FN-kappa B , ARN Largo no Codificante , Factores de Empalme Serina-Arginina , Transducción de Señal , Animales , Miocarditis/metabolismo , Miocarditis/inmunología , Miocarditis/genética , Miocarditis/patología , ARN Largo no Codificante/genética , Factores de Empalme Serina-Arginina/metabolismo , Factores de Empalme Serina-Arginina/genética , Ratones , FN-kappa B/metabolismo , Macrófagos/metabolismo , Macrófagos/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/inmunología , Masculino , Modelos Animales de Enfermedad , Activación de Macrófagos , Inflamación/genética , Inflamación/metabolismo
7.
Cancer Immunol Res ; 12(8): 954-955, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38967235

RESUMEN

Immune checkpoint therapies can drive antitumor responses and benefit patients but can also induce life-threatening immune-related adverse events such as myocarditis and myositis. These immune-related adverse events are rare but carry substantial morbidity and mortality. In this issue, Siddiqui and colleagues use single-cell RNA and T-cell receptor sequencing to identify novel cellular subsets and propose various mechanisms that could contribute to the pathogenesis of immune checkpoint inhibitor-associated myocarditis and myositis. These new insights should help move the field toward the development of improved treatment and prevention options, ultimately improving patient outcomes. See related article by Siddiqui et al., p. 964 (1).


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Miocarditis , Miositis , Miocarditis/genética , Miocarditis/etiología , Miocarditis/metabolismo , Humanos , Miositis/genética , Miositis/inmunología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Animales
8.
BMC Cardiovasc Disord ; 24(1): 375, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39026189

RESUMEN

BACKGROUND: Acute myocardial injury, cytokine storms, hypoxemia and pathogen-mediated damage were the major causes responsible for mortality induced by coronavirus disease 2019 (COVID-19)-related myocarditis. These need ECMO treatment. We investigated differentially expressed genes (DEGs) in patients with COVID-19-related myocarditis and ECMO prognosis. METHODS: GSE150392 and GSE93101 were analyzed to identify DEGs. A Venn diagram was used to obtain the same transcripts between myocarditis-related and ECMO-related DEGs. Enrichment pathway analysis was performed and hub genes were identified. Pivotal miRNAs, transcription factors, and chemicals with the screened gene interactions were identified. The GSE167028 dataset and single-cell sequencing data were used to validate the screened genes. RESULTS: Using a Venn diagram, 229 overlapping DEGs were identified between myocarditis-related and ECMO-related DEGs, which were mainly involved in T cell activation, contractile actin filament bundle, actomyosin, cyclic nucleotide phosphodiesterase activity, and cytokine-cytokine receptor interaction. 15 hub genes and 15 neighboring DEGs were screened, which were mainly involved in the positive regulation of T cell activation, integrin complex, integrin binding, the PI3K-Akt signaling pathway, and the TNF signaling pathway. Data in GSE167028 and single-cell sequencing data were used to validate the screened genes, and this demonstrated that the screened genes CCL2, APOE, ITGB8, LAMC2, COL6A3 and TNC were mainly expressed in fibroblast cells; IL6, ITGA1, PTK2, ITGB5, IL15, LAMA4, CAV1, SNCA, BDNF, ACTA2, CD70, MYL9, DPP4, ENO2 and VEGFC were expressed in cardiomyocytes; IL6, PTK2, ITGB5, IL15, APOE, JUN, SNCA, CD83, DPP4 and ENO2 were expressed in macrophages; and IL6, ITGA1, PTK2, ITGB5, IL15, VCAM1, LAMA4, CAV1, ACTA2, MYL9, CD83, DPP4, ENO2, VEGFC and IL32 were expressed in vascular endothelial cells. CONCLUSION: The screened hub genes, IL6, ITGA1, PTK2, ITGB3, ITGB5, CCL2, IL15, VCAM1, GZMB, APOE, ITGB8, LAMA4, LAMC2, COL6A3 and TNFRSF9, were validated using GEO dataset and single-cell sequencing data, which may be therapeutic targets patients with myocarditis to prevent MI progression and adverse cardiovascular events.


Asunto(s)
COVID-19 , Oxigenación por Membrana Extracorpórea , Miocarditis , Humanos , COVID-19/genética , COVID-19/terapia , COVID-19/complicaciones , Miocarditis/genética , Miocarditis/terapia , Miocarditis/virología , Pronóstico , Perfilación de la Expresión Génica , Bases de Datos Genéticas , SARS-CoV-2 , Redes Reguladoras de Genes , Transcriptoma
9.
BMC Genom Data ; 25(1): 51, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844841

RESUMEN

Vaccine-related myocarditis associated with the BNT162b2 vaccine is a rare complication, with a higher risk observed in male adolescents. However, the contribution of genetic factors to this condition remains uncertain. In this study, we conducted a comprehensive genetic association analysis in a cohort of 43 Hong Kong Chinese adolescents who were diagnosed with myocarditis shortly after receiving the BNT162b2 mRNA COVID-19 vaccine. A comparison of whole-genome sequencing data was performed between the confirmed myocarditis cases and a control group of 481 healthy individuals. To narrow down potential genomic regions of interest, we employed a novel clustering approach called ClusterAnalyzer, which prioritised 2,182 genomic regions overlapping with 1,499 genes for further investigation. Our pathway analysis revealed significant enrichment of these genes in functions related to cardiac conduction, ion channel activity, plasma membrane adhesion, and axonogenesis. These findings suggest a potential genetic predisposition in these specific functional areas that may contribute to the observed side effect of the vaccine. Nevertheless, further validation through larger-scale studies is imperative to confirm these findings. Given the increasing prominence of mRNA vaccines as a promising strategy for disease prevention and treatment, understanding the genetic factors associated with vaccine-related myocarditis assumes paramount importance. Our study provides valuable insights that significantly advance our understanding in this regard and serve as a valuable foundation for future research endeavours in this field.


Asunto(s)
Vacuna BNT162 , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Miocarditis , Humanos , Vacuna BNT162/efectos adversos , Miocarditis/genética , Miocarditis/epidemiología , Miocarditis/etiología , Miocarditis/inducido químicamente , Masculino , Adolescente , Hong Kong/epidemiología , Femenino , Vacunas contra la COVID-19/efectos adversos , COVID-19/prevención & control , COVID-19/genética , COVID-19/epidemiología , Secuenciación Completa del Genoma , SARS-CoV-2/genética , SARS-CoV-2/inmunología
11.
Circ Genom Precis Med ; 17(4): e004487, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38910558

RESUMEN

BACKGROUND: Inflammatory heart disease can be triggered by a variety of causes, both infectious and noninfectious in nature. We hypothesized that inflammatory cardiomyopathy is potentially related to microbial infection. METHODS: In this retrospective study, we used deep RNA sequencing on formalin-fixed paraffin-embedded heart tissue specimens to detect pathogenic agents. We first investigated 4 single-sample cases to test the feasibility of this diagnostic protocol and further 3 control-sample paired cases to improve the protocol with differential metatranscriptomics next-generation sequencing (mtNGS) analysis. RESULTS: We demonstrate that differential mtNGS allows identification of various microbials as potentially pathogenic, for example, Cutibacterium acnes, Corynebacterium aurimucosum, and Pseudomonas denitrificans, which are usually commensal in healthy individuals. Differential mtNGS also allows characterization of human host response in each individual by profiling alterations of gene expression, networked pathways, and inferred immune cell compositions, information of which is beneficial for us to understand different etiologies and immunity roles in each case. Additionally, differential mtNGS allows the identification of genetic variants in patients that may contribute to their susceptibility to particular microbial infections. CONCLUSIONS: The demonstrated power of differential mtNGS in simultaneous capture of both the infectious microbial(s) and the status of human host immune response could help us better understand the pathogenesis of complex inflammatory cardiomyopathy, if conducted on a larger scale of the population. The developed differential mtNGS method could also shed light on its translation and adoption of such a laboratory test in clinic practice, allowing for a more effective diagnosis to guide therapeutic treatment of the disease.


Asunto(s)
Cardiomiopatías , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estudios Retrospectivos , Cardiomiopatías/genética , Cardiomiopatías/microbiología , Cardiomiopatías/diagnóstico , Masculino , Femenino , Análisis de Secuencia de ARN , Persona de Mediana Edad , Miocarditis/microbiología , Miocarditis/diagnóstico , Miocarditis/genética , Adulto , Anciano , Inflamación/microbiología , Inflamación/genética , Inflamación/diagnóstico
12.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38696536

RESUMEN

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.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus Humano B , Interferón Tipo I , Miocarditis , Miocitos Cardíacos , ARN Viral , Miocarditis/virología , Miocarditis/inmunología , Miocarditis/genética , Animales , Miocitos Cardíacos/virología , Miocitos Cardíacos/metabolismo , Ratones , Enterovirus Humano B/inmunología , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/virología , Infecciones por Coxsackievirus/genética , Interferón Tipo I/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Humanos , Inmunidad Innata , Transducción de Señal , Interferón beta/metabolismo , Interferón beta/genética , Interferón beta/inmunología , Masculino , Regiones no Traducidas 5'
13.
J Clin Invest ; 134(13)2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38768074

RESUMEN

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.


Asunto(s)
Inmunidad Innata , Células Madre Pluripotentes Inducidas , Miocarditis , Miocitos Cardíacos , Humanos , Miocarditis/genética , Miocarditis/inmunología , Miocarditis/patología , Inmunidad Innata/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Masculino , Predisposición Genética a la Enfermedad , Femenino
14.
BMC Cardiovasc Disord ; 24(1): 282, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811883

RESUMEN

Sudden cardiac death (SCD) is a major public health issue worldwide. In the young (< 40 years of age), genetic cardiomyopathies and viral myocarditis, sometimes in combination, are the most frequent, but underestimated, causes of SCD. Molecular autopsy is essential for prevention. Several studies have shown an association between genetic cardiomyopathies and viral myocarditis, which is probably underestimated due to insufficient post-mortem investigations. We report on four autopsy cases illustrating the pathogenesis of these combined pathologies. In two cases, a genetic hypertrophic cardiomyopathy was diagnosed in combination with Herpes Virus Type 6 (HHV6) and/or Parvovirus-B19 (PVB19) in the heart. In the third case, autopsy revealed a dilated cardiomyopathy and virological analyses revealed acute myocarditis caused by three viruses: PVB19, HHV6 and Epstein-Barr virus. Genetic analyses revealed a mutation in the gene coding for desmin. The fourth case illustrated a channelopathy and a PVB19/HHV6 coinfection. Our four cases illustrate the highly probable deleterious role of cardiotropic viruses in the occurrence of SCD in subjects with genetic cardiomyopathies. We discuss the pathogenetic link between viral myocarditis and genetic cardiomyopathy. Molecular autopsy is essential in prevention of these SCD, and a close collaboration between cardiologists, pathologists, microbiologists and geneticians is mandatory.


Asunto(s)
Autopsia , Muerte Súbita Cardíaca , Herpesvirus Humano 6 , Miocarditis , Parvovirus B19 Humano , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/virología , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Causas de Muerte , Coinfección , Muerte Súbita Cardíaca/etiología , Muerte Súbita Cardíaca/patología , Muerte Súbita Cardíaca/prevención & control , Infecciones por Virus de Epstein-Barr/complicaciones , Resultado Fatal , Predisposición Genética a la Enfermedad , Herpesvirus Humano 4/genética , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/aislamiento & purificación , Mutación , Miocarditis/virología , Miocarditis/patología , Miocarditis/genética , Infecciones por Parvoviridae/complicaciones , Parvovirus B19 Humano/genética , Infecciones por Roseolovirus/complicaciones , Infecciones por Roseolovirus/virología , Infecciones por Roseolovirus/diagnóstico , Infecciones por Roseolovirus/patología
15.
Circ Heart Fail ; 17(6): e011204, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38813684

RESUMEN

BACKGROUND: Acute myocarditis has been genetically linked to dilated cardiomyopathy (DCM), but the clinical significance remains uncertain. We investigated the prevalence and long-term prognosis of DCM and heart failure (HF) among unselected patients hospitalized with acute myocarditis and their first-degree relatives compared with an age- and sex-matched cohort. METHODS: This was an observational study utilizing the Danish nationwide registries, where all patients with a first-time myocarditis diagnosis from 1995 to 2018 were identified and matched (on birth year and sex) with 10 controls from the general population. RESULTS: Totally 3176 patients with acute myocarditis and 31 760 controls were included (median age, 49.8 [Q1-Q3, 32.5-70.2] years; 35.6% female). At baseline, patients with myocarditis had a higher prevalence of DCM (7 [0.2%] versus 8 [0.0%]) and HF (336 [10.6%] versus 695 [2.2%]) than controls; P<0.0001 for both. Patients with myocarditis more often had siblings with DCM (12 [0.4%] versus 17 [0.05%]) or HF (36 [1.1%] versus 89 [0.3%]); P<0.0001, odds ratios 7.09 (3.38-14.85) and 2.92 (1.25-6.80), respectively, whereas parental DCM and HF did not differ among patients with myocarditis and controls. Patients with myocarditis had greater 20-year incidence of DCM, HF, and all-cause mortality (0.5% [0.3%-0.9%], 15% [13%-17%], and 47% [44%-50%]) compared with controls (0.06% [0.03%-0.11%], 6.8% [6.4%-7.3%], and 34% [33%-35%]; P<0.0001). Having a first-degree relative with DCM or HF was associated with increased long-term mortality among the patients with myocarditis (hazard ratio, 1.40 [1.11-1.77]) but not among the controls (hazard ratio, 0.90 [0.81-1.01]; Pdifference=0.0008). CONCLUSIONS: Acute myocarditis aggregates with DCM within families, where it carries a worsened prognosis. A differential association between parents and siblings (with sibling preponderance) could suggest that additional environmental factors are important for myocarditis development even in predisposed individuals.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Miocarditis , Sistema de Registros , Humanos , Miocarditis/epidemiología , Miocarditis/genética , Miocarditis/mortalidad , Masculino , Femenino , Persona de Mediana Edad , Adulto , Prevalencia , Pronóstico , Dinamarca/epidemiología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/epidemiología , Cardiomiopatía Dilatada/mortalidad , Anciano , Insuficiencia Cardíaca/epidemiología , Insuficiencia Cardíaca/genética , Enfermedad Aguda , Factores de Riesgo , Predisposición Genética a la Enfermedad
16.
Front Immunol ; 15: 1380697, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38715608

RESUMEN

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.


Asunto(s)
COVID-19 , Miocarditis , SARS-CoV-2 , Miocarditis/virología , Miocarditis/inmunología , Miocarditis/terapia , Miocarditis/genética , Humanos , COVID-19/inmunología , COVID-19/genética , COVID-19/terapia , SARS-CoV-2/fisiología , Metilación , 5-Metilcitosina/metabolismo , Inmunidad Innata , Tratamiento Farmacológico de COVID-19 , Animales , ARN Viral/genética , ARN Viral/metabolismo , Procesamiento Postranscripcional del ARN
18.
Int J Cardiovasc Imaging ; 40(6): 1397-1398, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38652395

RESUMEN

We present a case of a young patient with chest pain. Labs and cardiac imaging were suspicious for acute myocarditis. Genetic testing revealed a diagnosis of desmoplakin cardiomyopathy. Desmoplakin cardiomyopathy may be considered in patients with recurrent acute myocarditis or a family history of cardiac disease to avoid the potential for misdiagnosis.


Asunto(s)
Cardiomiopatías , Miocarditis , Valor Predictivo de las Pruebas , Humanos , Enfermedad Aguda , Biopsia , Cardiomiopatías/genética , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatía Dilatada , Desmoplaquinas/genética , Diagnóstico Diferencial , Electrocardiografía , Predisposición Genética a la Enfermedad , Enfermedades del Cabello , Queratodermia Palmoplantar , Mutación , Miocarditis/diagnóstico por imagen , Miocarditis/genética , Fenotipo , Femenino , Adolescente
19.
JACC Heart Fail ; 12(6): 1101-1111, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38573261

RESUMEN

BACKGROUND: Acute myocarditis is an inflammatory condition that may precede the development of dilated or arrhythmogenic cardiomyopathy. OBJECTIVES: The aim of this study was to investigate the reported prevalence of pathogenic or likely pathogenic (P/LP) variants in cardiomyopathy-associated genes in patients with acute myocarditis. METHODS: For this systematic review and meta-analysis, the PubMed and Embase databases were searched on March 4, 2023. Observational studies evaluating the prevalence of P/LP variants in cardiomyopathy-associated genes in patients with acute myocarditis were included. Studies were stratified into adult and pediatric age groups and for the following scenarios: 1) complicated myocarditis (ie, presenting with acute heart failure, reduced left ventricular ejection fraction, or life-threatening ventricular arrhythmias); and 2) uncomplicated myocarditis. The study was registered with the International Prospective Register of Systematic Reviews (CRD42023408668) and followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS: Of 732 studies identified, 8 met the inclusion criteria, providing data for 586 patients with acute myocarditis. A total of 89 P/LP variants in cardiomyopathy-associated genes were reported in 85 patients. For uncomplicated myocarditis, the pooled prevalence was 4.2% (95% CI: 1.8%-7.4%; I2 = 1.4%), whereas for complicated myocarditis, the pooled prevalence was 21.9% (95% CI: 14.3%-30.5%; I2 = 38.8%) and 44.5% (95% CI: 22.7%-67.4%; I2 = 52.8%) in adults and children, respectively. P/LP variants in desmosomal genes were predominant in uncomplicated myocarditis (64%), whereas sarcomeric gene variants were more prevalent in complicated myocarditis (58% in adults and 71% in children). CONCLUSIONS: Genetic variants are present in a large proportion of patients with acute myocarditis. The prevalence of genetic variants and the genes involved vary according to age and clinical presentation.


Asunto(s)
Miocarditis , Humanos , Miocarditis/genética , Miocarditis/epidemiología , Enfermedad Aguda , Prevalencia , Cardiomiopatías/genética , Cardiomiopatías/epidemiología
20.
Free Radic Biol Med ; 218: 149-165, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38570171

RESUMEN

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.


Asunto(s)
Autofagia , Insuficiencia Cardíaca , Complejo de la Endopetidasa Proteasomal , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/inmunología , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Fenotipo , Transducción de Señal , Proteolisis , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/genética , Miocarditis/patología , Miocarditis/metabolismo , Miocarditis/inmunología , Miocarditis/genética , Cardiomegalia/patología , Cardiomegalia/metabolismo , Cardiomegalia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA