Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 942
Filter
1.
Sci Rep ; 14(1): 12653, 2024 06 02.
Article in English | MEDLINE | ID: mdl-38825590

ABSTRACT

Nonischaemic myocardial fibrosis is associated with cardiac dysfunction, malignant arrhythmias and sudden cardiac death. In the absence of a specific aetiology, its finding as late gadolinium enhancement (LGE) on cardiac magnetic resonance imaging is often attributed to preceding viral myocarditis. Athletes presenting with ventricular arrhythmias often have nonischaemic LGE. Previous studies have demonstrated an adverse effect of exercise on the course of acute viral myocarditis. In this study, we have investigated, for the first time, the impact of endurance training on longer-term outcomes such as myocardial fibrosis and arrhythmogenicity in a murine coxsackievirus B3 (CVB)-induced myocarditis model. Male C57BL/6J mice (n = 72) were randomly assigned to 8 weeks of forced treadmill running (EEX) or no exercise (SED). Myocarditis was induced 2 weeks later by a single intraperitoneal injection with CVB, versus vehicle in the controls (PBS). In a separate study, mice (n = 30) were subjected to pretraining for 13 weeks (preEEX), without continuation of exercise during myocarditis. Overall, continuation of exercise resulted in a milder clinical course of viral disease, with less weight loss and better preserved running capacity. CVB-EEX and preEEX-CVB mice tended to have a lower mortality rate. At sacrifice (i.e. 6 weeks after inoculation), the majority of virus was cleared from the heart. Histological assessment demonstrated prominent myocardial inflammatory infiltration and cardiomyocyte loss in both CVB groups. Inflammatory lesions in the CVB-EEX group contained higher numbers of pro-inflammatory cells (iNOS-reactive macrophages and CD8+ T lymphocytes) compared to these in CVB-SED. Treadmill running during myocarditis increased interstitial fibrosis [82.4% (CVB-EEX) vs. 56.3% (CVB-SED); P = 0.049]. Additionally, perivascular and/or interstitial fibrosis with extensive distribution was more likely to occur with exercise [64.7% and 64.7% (CVB-EEX) vs. 50% and 31.3% (CVB-SED); P = 0.048]. There was a numerical, but not significant, increase in the number of scars per cross-section (1.9 vs. 1.2; P = 0.195), with similar scar distribution and histological appearance in CVB-EEX and CVB-SED. In vivo electrophysiology studies did not induce sustained monomorphic ventricular tachycardia, only nonsustained (usually polymorphic) runs. Their cumulative beat count and duration paralleled the increased fibrosis between CVB-EEX and CVB-SED, but the difference was not significant (P = 0.084 for each). Interestingly, in mice that were subjected to pretraining only without continuation of exercise during myocarditis, no differences between pretrained and sedentary mice were observed at sacrifice (i.e. 6 weeks after inoculation and training cessation) with regard to myocardial inflammation, fibrosis, and ventricular arrhythmogenicity. In conclusion, endurance exercise during viral myocarditis modulates the inflammatory process with more pro-inflammatory cells and enhances perivascular and interstitial fibrosis development. The impact on ventricular arrhythmogenesis requires further exploration.


Subject(s)
Arrhythmias, Cardiac , Coxsackievirus Infections , Disease Models, Animal , Enterovirus B, Human , Fibrosis , Mice, Inbred C57BL , Myocarditis , Physical Conditioning, Animal , Animals , Myocarditis/virology , Myocarditis/pathology , Male , Mice , Arrhythmias, Cardiac/etiology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/complications , Myocardium/pathology , Endurance Training
2.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38836810

ABSTRACT

Coxsackievirus A10 (CV-A10) infection, a prominent cause of childhood hand-foot-and-mouth disease (HFMD), frequently manifests with the intriguing phenomenon of onychomadesis, characterized by nail shedding. However, the underlying mechanism is elusive. Here, we found that CV-A10 infection in mice could suppress Wnt/ß-catenin signaling by restraining LDL receptor-related protein 6 (LRP6) phosphorylation and ß-catenin accumulation and lead to onychomadesis. Mechanistically, CV-A10 mimics Dickkopf-related protein 1 (DKK1) to interact with Kringle-containing transmembrane protein 1 (KRM1), the CV-A10 cellular receptor. We further found that Wnt agonist (GSK3ß inhibitor) CHIR99021 can restore nail stem cell differentiation and protect against nail shedding. These findings provide novel insights into the pathogenesis of CV-A10 and related viruses in onychomadesis and guide prognosis assessment and clinical treatment of the disease.


Subject(s)
Intercellular Signaling Peptides and Proteins , Low Density Lipoprotein Receptor-Related Protein-6 , Wnt Signaling Pathway , Animals , Wnt Signaling Pathway/drug effects , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Mice , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Humans , beta Catenin/metabolism , Nail Diseases/metabolism , Nail Diseases/virology , Nail Diseases/pathology , Nails/metabolism , Nails/pathology , Cell Differentiation/drug effects , Mice, Inbred C57BL , Hand, Foot and Mouth Disease/virology , Hand, Foot and Mouth Disease/metabolism , Hand, Foot and Mouth Disease/pathology , Hand, Foot and Mouth Disease/complications , Phosphorylation/drug effects , Coxsackievirus Infections/complications , Coxsackievirus Infections/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Pyridines/pharmacology , Pyrimidines
3.
Medicine (Baltimore) ; 103(10): e37248, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38457543

ABSTRACT

INTRODUCTION: In rare occasions, coxsackievirus infections can cause serious illness, such as encephalitis and myocarditis. The immunotherapies of cancer could increase the risk of myocarditis, especially when applying immune checkpoint inhibitors. Herein, we report a rare case of Coxsackie B virus-induced myocarditis in a patient with a history of lymphoma. CASE PRESENTATION: A 32-year-old woman was admitted to the hospital with recurrent fever for more than 20 days, and she had a history of lymphoma. Before admission, the positron emission tomography/computed tomography result indicated that the patient had no tumor progression, and she was not considered the cancer-related fever upon arriving at our hospital. Patient's red blood cell, platelet count, and blood pressure were decreased. In addition, she had sinus bradycardia and 3 branch blocks, which was consistent with acute high lateral and anterior wall myocardial infarction. During hospitalization, the patient had recurrent arrhythmia, repeated sweating, poor mentation, dyspnea, and Coxsackie B virus were detected in patient's blood samples by pathogen-targeted next-generation sequencing. The creatine kinase, creatine kinase MB, and N-terminal pro-brain natriuretic peptide were persistently elevated. Consequently, the patient was diagnosed with viral myocarditis induced by Coxsackie B virus, and treated with acyclovir, gamma globulin combined with methylprednisolone shock therapy, trimetazidine, levosimendan, sildenan, continuous pump pressors with m-hydroxylamine, entecavir, adefovir, glutathione, pantoprazole, and low-molecular-weight heparin. Her symptoms worsened and died. CONCLUSION: We reported a case with a history of lymphoma presented with fever, myocardial injury, who was ultimately diagnosed with Coxsackie B virus-induced myocarditis. Moreover, pathogen-targeted next-generation sequencing indeed exhibited higher sensitivity compared to mNGS in detecting Coxsackie B virus.


Subject(s)
Coxsackievirus Infections , Lymphoma , Myocarditis , Virus Diseases , Humans , Female , Adult , Myocarditis/diagnosis , Myocarditis/etiology , Enterovirus B, Human , Coxsackievirus Infections/complications , Coxsackievirus Infections/diagnosis , Fever
4.
Int J Mol Sci ; 25(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38339075

ABSTRACT

Recent research suggests that T-cell receptor (TCR) sequences expanded during human immunodeficiency virus and SARS-CoV-2 infections unexpectedly mimic these viruses. The hypothesis tested here is that TCR sequences expanded in patients with type 1 diabetes mellitus (T1DM) and autoimmune myocarditis (AM) mimic the infectious triggers of these diseases. Indeed, TCR sequences mimicking coxsackieviruses, which are implicated as triggers of both diseases, are statistically significantly increased in both T1DM and AM patients. However, TCRs mimicking Clostridia antigens are significantly expanded in T1DM, whereas TCRs mimicking Streptococcal antigens are expanded in AM. Notably, Clostridia antigens mimic T1DM autoantigens, such as insulin and glutamic acid decarboxylase, whereas Streptococcal antigens mimic cardiac autoantigens, such as myosin and laminins. Thus, T1DM may be triggered by combined infections of coxsackieviruses with Clostridia bacteria, while AM may be triggered by coxsackieviruses with Streptococci. These TCR results are consistent with both epidemiological and clinical data and recent experimental studies of cross-reactivities of coxsackievirus, Clostridial, and Streptococcal antibodies with T1DM and AM antigens. These data provide the basis for developing novel animal models of AM and T1DM and may provide a generalizable method for revealing the etiologies of other autoimmune diseases. Theories to explain these results are explored.


Subject(s)
Autoimmune Diseases , Coxsackievirus Infections , Diabetes Mellitus, Type 1 , Enterovirus , Myocarditis , Streptococcal Infections , Animals , Humans , Autoimmune Diseases/complications , Coxsackievirus Infections/complications , Autoantigens , Streptococcus , Streptococcal Infections/complications , Antigens, Bacterial , Receptors, Antigen, T-Cell
5.
JAMA Ophthalmol ; 142(1): e234721, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38236264

ABSTRACT

This case report describes a diagnosis of unilateral retinopathy secondary to coxsackie B virus in a male patient aged 41 years who presented with a central scotoma and blistering rash of the hands, feet, and mouth for 4 days.


Subject(s)
Coxsackievirus Infections , Herpesviridae Infections , Retinal Diseases , Humans , Retinal Diseases/diagnosis , Retinal Diseases/etiology , Male , Adult , Scotoma/etiology , Coxsackievirus Infections/complications , Hand, Foot and Mouth Disease
6.
Int Immunopharmacol ; 127: 111304, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38091826

ABSTRACT

Acute viral myocarditis can progress to chronic myocarditis leading to dilated cardiomyopathy (DCM). Persistent CD4+ T-cell-mediated autoimmunity triggered by infection plays a critical role in this progression. Increasing evidence demonstrates that effector memory CD4+T (CD4+TEM) cells, a subset of memory CD4+ T cells, are crucial pathogenic mediators of many autoimmune diseases. However, the role of CD4+TEM cells during the progression from acute viral myocarditis to DCM remains unknown. In this study, we observed an increase in CD4+TEM cells both in the periphery and the heart, and memory CD4+ T cells were the predominant sources of IL-17A and IFN-γ among inflamed heart-infiltrating CD4+ T cells during the progression from acute myocarditis to chronic myocarditis and DCM in CVB3-induced BALB/c mice. Moreover, splenic CD4+TEM cells sorted from DCM mice induced by CVB3 were found to respond to cardiac self-antigens ex vivo. Additionally, adoptive transfer experiments substantiated their pathogenic impact, inducing sustained myocardial inflammation, tissue fibrosis, cardiac injury, and impairment of cardiac systolic function in vivo. Our findings illustrate that long-lived CD4+TEM cells are important contributors to the progression from acute viral myocarditis into DCM.


Subject(s)
Autoimmune Diseases , Cardiomyopathy, Dilated , Coxsackievirus Infections , Myocarditis , Mice , Animals , Cardiomyopathy, Dilated/pathology , T-Lymphocytes/pathology , Mice, Inbred BALB C , Myocardium/pathology , Coxsackievirus Infections/complications , Enterovirus B, Human
7.
Virus Res ; 339: 199250, 2024 01 02.
Article in English | MEDLINE | ID: mdl-37865350

ABSTRACT

Evidence is emerging on the roles of long noncoding RNAs (lncRNAs) as regulatory factors in a variety of viral infection processes, but the mechanisms underlying their functions in coxsackievirus group B type3 (CVB3)-induced acute viral myocarditis have not been explicitly delineated. We previously demonstrated that CVB3 infection decreases miRNA-21 expression; however, lncRNAs that regulate the miRNA-21-dependent CVB3 disease process have yet to be identified. To evaluate lncRNAs upstream of miRNA-21, differentially expressed lncRNAs in CVB3-infected mouse hearts were identified by microarray analysis and lncRNA/miRNA-21 interactions were predicted bioinformatically. MEG3 was identified as a candidate miRNA-21-interacting lncRNA upregulated in CVB3-infected mouse hearts. MEG3 expression was verified to be upregulated in HeLa cells 48 h post CVB3 infection and to act as a competitive endogenous RNA of miRNA-21. MEG3 knockdown resulted in the upregulation of miRNA-21, which inhibited CVB3 replication by attenuating P38-MAPK signaling in vitro and in vivo. Knockdown of MEG3 expression before CVB3 infection inhibited viral replication in mouse hearts and alleviated cardiac injury, which improved survival. Furthermore, the knockdown of CREB5, which was predicted bioinformatically to function upstream of MEG3, was demonstrated to decrease MEG3 expression and CVB3 viral replication. This study identifies the function of the lncRNA MEG3/miRNA-21/P38 MAPK axis in the process of CVB3 replication, for which CREB5 could serve as an upstream modulator.


Subject(s)
Coxsackievirus Infections , Enterovirus , MicroRNAs , Myocarditis , RNA, Long Noncoding , Virus Diseases , Animals , Humans , Mice , Coxsackievirus Infections/complications , Coxsackievirus Infections/genetics , Enterovirus/genetics , Enterovirus B, Human/genetics , Enterovirus B, Human/metabolism , HeLa Cells/virology , MicroRNAs/genetics , MicroRNAs/metabolism , Myocarditis/genetics , Myocarditis/metabolism , Myocarditis/virology , RNA, Long Noncoding/genetics , Virus Replication
9.
Endocr Rev ; 44(4): 737-751, 2023 07 11.
Article in English | MEDLINE | ID: mdl-36884282

ABSTRACT

The evidence for an association between coxsackievirus B (CVB) infection, pancreatic islet autoimmunity, and clinical type 1 diabetes is increasing. Results from prospective cohorts and pancreas histopathology studies have provided a compelling case. However, the demonstration of a causal relationship is missing, and is likely to remain elusive until tested in humans by avoiding exposure to this candidate viral trigger. To this end, CVB vaccines have been developed and are entering clinical trials. However, the progress made in understanding the biology of the virus and in providing tools to address the long-standing question of causality contrasts with the scarcity of information about the antiviral immune responses triggered by infection. Beta-cell death may be primarily induced by CVB itself, possibly in the context of poor immune protection, or secondarily provoked by T-cell responses against CVB-infected beta cells. The possible involvement of epitope mimicry mechanisms skewing the physiological antiviral response toward autoimmunity has also been suggested. We here review the available evidence for each of these 3 non-mutually exclusive scenarios. Understanding which ones are at play is critical to maximize the odds of success of CVB vaccination, and to develop suitable tools to monitor the efficacy of immunization and its intermingling with autoimmune onset or prevention.


Subject(s)
Coxsackievirus Infections , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Humans , Diabetes Mellitus, Type 1/prevention & control , Prospective Studies , Enterovirus B, Human/physiology , Coxsackievirus Infections/prevention & control , Coxsackievirus Infections/complications
10.
Pediatr Infect Dis J ; 42(5): e177-e179, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36795579

ABSTRACT

During July-September 2022, 14 children suffering from meningoencephalitis tested positive for Coxsackievirus B2 (8 cerebrospinal fluid, 9 stool samples). Mean age 22 months (range 0-60 months); 8 were males. Seven of the children presented with ataxia and 2 had imaging features of rhombencephalitis, not previously described in association with Coxsackievirus B2.


Subject(s)
Coxsackievirus Infections , Meningoencephalitis , Male , Child , Humans , Infant, Newborn , Infant , Child, Preschool , Female , Coxsackievirus Infections/epidemiology , Coxsackievirus Infections/complications , Israel/epidemiology , Enterovirus B, Human , Meningoencephalitis/epidemiology , Disease Outbreaks
11.
J Virol ; 97(1): e0142622, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36475766

ABSTRACT

Group B enteroviruses, including coxsackievirus B3 (CVB3), can persistently infect cardiac tissue and cause dilated cardiomyopathy. Persistence is linked to 5' terminal deletions of viral genomic RNAs that have been detected together with minor populations of full-length genomes in human infections. In this study, we explored the functions and interactions of the different viral RNA forms found in persistently infected patients and their putative role(s) in pathogenesis. Since enterovirus cardiac pathogenesis is linked to the viral proteinase 2A, we investigated the effect of different terminal genomic RNA deletions on 2A activity. We discovered that 5' terminal deletions in CVB3 genomic RNAs decreased the levels of 2A proteinase activity but could not abrogate it. Using newly generated viral reporters encoding nano-luciferase, we found that 5' terminal deletions resulted in decreased levels of viral protein and RNA synthesis in singly transfected cardiomyocyte cultures. Unexpectedly, when full-length and terminally deleted forms were cotransfected into cardiomyocytes, a cooperative interaction was observed, leading to increased viral RNA and protein production. However, when viral infections were carried out in cells harboring 5' terminally deleted CVB3 RNAs, a decrease in infectious particle production was observed. Our results provide a possible explanation for the necessity of full-length viral genomes during persistent infection, as they would stimulate efficient viral replication compared to that of the deleted genomes alone. To avoid high levels of viral particle production that would trigger cellular immune activation and host cell death, the terminally deleted RNA forms act to limit the production of viral particles, possibly as trans-dominant inhibitors. IMPORTANCE Enteroviruses like coxsackievirus B3 are able to initiate acute infections of cardiac tissue and, in some cases, to establish a long-term persistent infection that can lead to serious disease sequelae, including dilated cardiomyopathy. Previous studies have demonstrated the presence of 5' terminally deleted forms of enterovirus RNAs in heart tissues derived from patients with dilated cardiomyopathy. These deleted RNAs are found in association with very low levels of full-length enterovirus genomic RNAs, an interaction that may facilitate continued persistence while limiting virus particle production. Even in the absence of detectable infectious virus particle production, these deleted viral RNA forms express viral proteinases at levels capable of causing viral pathology. Our studies provide mechanistic insights into how full-length and deleted forms of enterovirus RNA cooperate to stimulate viral protein and RNA synthesis without stimulating infectious viral particle production. They also highlight the importance of targeting enteroviral proteinases to inhibit viral replication while at the same time limiting the long-term pathologies they trigger.


Subject(s)
Cardiomyopathy, Dilated , Coxsackievirus Infections , Enterovirus B, Human , Humans , Antigens, Viral , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/virology , Coxsackievirus Infections/complications , Enterovirus B, Human/metabolism , Genomics , Myocytes, Cardiac/virology , Peptide Hydrolases , Persistent Infection , RNA, Viral/genetics , Viral Proteins/metabolism , Virus Replication
12.
Birth Defects Res ; 115(2): 171-178, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36094067

ABSTRACT

BACKGROUND: Different serotypes of coxsackievirus B (CVB), which is the most common cause of viral myocarditis, target cardiomyocytes through Coxsackie and Adenovirus Receptor and Decay-Accelerating Factor. Both receptors are expressed in the fetal heart. We hypothesized that infection with different serotypes of CVB during early pregnancy plays a role in pathogenesis of congenital heart defect (CHD). METHODS: In this study, we use a murine model to infect with CVB1, CVB4, and combination of CVB3 + CVB4 during a critical period in gestation. We examined offspring of pregnant mice for fetal death and heart defects following viral infection. RESULT: Fetuses from uninfected control dams showed normal heart development, while maternal CVB infection precipitates CHD: majorly ventricular septal defects (VSD) and non-compaction of ventricular myocardium (NC), with some infrequent cases of double outlet right ventricle, left ventricle wall rupture, right ventricle hypertrophy, and thickened/dysplastic semilunar valves. Infection of pregnant dams with CVB1 leads to 44% VSD and 41.2% NC cases, while with CVB4 leads to 31.7% VSD and 13.3% NC cases. Co-infection with CVB3 + CVB4 increases fetal pathology to 51.3% VSD and 41% NC cases. Infection can also result in fetal death, with higher incidences with CVB3 + CVB4 with 46.2% cases, compared to 33.3% by CVB1 and 21.7% by CVB4. Male fetuses were more susceptible to all phenotypes. CONCLUSION: Our report shows that prenatal CVB infections can lead to pathogenesis of certain heart defects in mouse model, particularly exacerbated with co-infections. This data confirms a link between prenatal CVB infection and CHD development and highlights it is not unique to just one serotype of CVB.


Subject(s)
Coxsackievirus Infections , Heart Defects, Congenital , Myocarditis , Pregnancy Complications, Infectious , Animals , Female , Male , Mice , Pregnancy , Enterovirus B, Human/genetics , Heart Defects, Congenital/etiology , Heart Defects, Congenital/pathology , Myocarditis/etiology , Myocarditis/pathology , Myocardium/pathology , Myocytes, Cardiac , Coxsackievirus Infections/complications , Coxsackievirus Infections/pathology
13.
Virologie (Montrouge) ; 26(6): 415-430, 2022.
Article in French | MEDLINE | ID: mdl-36565260

ABSTRACT

Epidemiological and experimental studies suggest that enteroviruses (EV) and particularly coxsackieviruses B (CVB) are likely to trigger or accelerate the onset of islet autoimmunity and the development of type 1 diabetes (T1D) in genetically susceptible individuals. Several mutually non-exclusive mechanisms have been proposed to explain the involvement of CVB in the pathogenesis of T1D. CVB can infect and persist in the intestine, thymic cells, monocytes/macrophages, ductal cells and pancreatic ß-cells, which leads to structural or functional alterations of these cells. A chronic inflammatory response and disruption of tolerance towards ß-cells due to CVB infections are able to promote the recruitment and activation of pre-existing autoreactive T-cells and the destruction of ß-cells. Vaccine or therapeutic strategies to control EV infections have been developed and open perspectives for the prevention or treatment of T1D.


Subject(s)
Coxsackievirus Infections , Diabetes Mellitus, Type 1 , Enterovirus Infections , Enterovirus , Humans , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Coxsackievirus Infections/complications , Enterovirus B, Human/physiology , Enterovirus Infections/complications , Enterovirus Infections/epidemiology
16.
Am J Case Rep ; 23: e935974, 2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35799414

ABSTRACT

BACKGROUND Myocarditis is an inflammatory process that can present as acute or chronic with either focal or diffuse involvement of the myocardium. Its incidence is approximately 1.5 million cases per year worldwide. In the United States, viral infection is the most common cause of myocarditis. Most of the reported cases are singular and self-limiting in nature. We present the case of severe recurrent myocarditis in a young adult who was transferred to the Intensive Care Unit. CASE REPORT An 18-year-old man presented with chest pressure and troponin I 33 ng/mL. He had presented to another hospital with similar symptoms 3 months prior and was diagnosed with myocarditis that had resolved with colchicine. As part of his workup during this admission, coronary angiogram was normal and biopsy obtained without evidence of an inflammatory process; however, cardiac magnetic resonance imaging (MRI) was consistent with myocarditis and Coxsackie B titers indicated prior infection, leading to a diagnosis of clinically suspected recurrent viral myocarditis. He was treated with intravenous immunoglobulin (IV Ig) and a steroid taper, with rapid improvement in symptoms over the ensuing weeks without evidence of further recurrence or sequelae. CONCLUSIONS We present a case of recurrent Coxsackie B myocarditis based on presentation and imaging. Myocarditis is an important diagnosis to consider when a young, healthy individual presents with chest pain mimicking acute coronary syndrome, especially during the COVID pandemic. If there is evidence of myocarditis on MRI or endomyocardial biopsy, immunosuppressive therapy should be considered in patients with recurrent and severe presentations.


Subject(s)
COVID-19 , Coxsackievirus Infections , Myocarditis , Adolescent , Coxsackievirus Infections/complications , Humans , Immunoglobulins, Intravenous/therapeutic use , Male , Myocarditis/diagnosis , Myocarditis/drug therapy , Myocarditis/etiology , Myocardium/pathology , Steroids
17.
Int J Biochem Cell Biol ; 146: 106208, 2022 05.
Article in English | MEDLINE | ID: mdl-35381374

ABSTRACT

Viral myocarditis (VMC) is the main cause of sudden acute heart failure and cardiac death in adolescents; however, treatment for VMC is limited. Trehalose is a natural non-reductive disaccharide that protects against cardiovascular diseases by inducing autophagy. The protective effect of trehalose on VMC and the specific mechanism remains unclear. In this study, we established a VMC mouse model, treated with trehalose in vivo, and cultured B cells from VMC mice with trehalose in vitro to elucidate the effect of trehalose on B cells in acute VMC. Trehalose alleviated myocardial injury in VMC mice and increased the number of autophagosomes, LC3II/LC3I ratio, and expression level of LAMP2, whereas it decreased the expression of p62 in VMC-B cells. Bafilomycin A1 suppressed VMC-B cell autophagy induced by trehalose. At the mechanistic level, trehalose treatment significantly upregulated the phosphorylation of AMPK and ULK1 in VMC-B cells. Dorsomorphin and SBI-0206965 abolished the increased phosphorylation level and altered the expression levels of autophagy-related proteins. In conclusion, trehalose alleviates myocardial inflammatory damage of VMC by inducing B cell autophagy, mediated by the AMPK/ULK1 signalling pathway. Thus, trehalose may be a potentially useful molecule for alleviating myocardial injury in VMC.


Subject(s)
Coxsackievirus Infections , Myocarditis , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy , Autophagy-Related Protein-1 Homolog , Coxsackievirus Infections/complications , Coxsackievirus Infections/drug therapy , Coxsackievirus Infections/metabolism , Mice , Mice, Inbred BALB C , Myocarditis/drug therapy , Trehalose/pharmacology , Trehalose/therapeutic use
18.
Int J Exp Pathol ; 103(4): 149-155, 2022 08.
Article in English | MEDLINE | ID: mdl-35363404

ABSTRACT

Atrial dysfunction is a relatively common complication of acute myocarditis, although its pathophysiology is unclear. There is limited information on myocarditis-associated histological changes in the atria and how they develop in time. The aim of this study therefore was to investigate inflammation, fibrosis and viral genome in the atria in time after mild CVB3-induced viral myocarditis (VM) in mice. C3H mice (n = 68) were infected with 105 PFU of Coxsackievirus B3 (CVB3) and were compared with uninfected mice (n = 10). Atrial tissue was obtained at days 4, 7, 10, 21, 35 or 49 post-infection. Cellular infiltration of CD45+ lymphocytes, MAC3+ macrophages, Ly6G+ neutrophils and mast cells was quantified by (immuno)histochemical staining. The CVB3 RNA was determined by in situ hybridization, and fibrosis was evaluated by elastic van Gieson (EvG) staining. In the atria of VM mice, the numbers of lymphocytes on days 4 and 7 (p < .05) and days 10 (p < .01); macrophages on days 7 (p < .01) and 10 (p < .05); neutrophils on days 4 (p < .05); and mast cells on days 4 and 7 (p < .05) increased significantly compared with control mice and decreased thereafter to basal levels. No cardiomyocyte death was observed, and the CVB3 genome was detected in only one infected mouse on Day 4 post-infection. No significant changes in the amount of atrial fibrosis were found between VM and control mice. A temporary increase in inflammation is induced in the atria in the acute phase of CVB3-induced mild VM, which may facilitate the development of atrial arrhythmia and contractile dysfunction.


Subject(s)
Coxsackievirus Infections , Myocarditis , Animals , Coxsackievirus Infections/complications , Coxsackievirus Infections/pathology , Disease Models, Animal , Enterovirus B, Human/genetics , Fibrosis , Inflammation/pathology , Mice , Mice, Inbred C3H , Myocarditis/pathology , Myocardium/pathology
20.
Pediatr Cardiol ; 43(5): 960-968, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35022808

ABSTRACT

Gestational viral infection has been associated with congenital heart disease (CHD). Few studies, however, have studied the potential role of gestational Coxsackievirus B (CVB) exposure in the pathogenesis of CHD. We prospectively enrolled women with pregnancies affected by CHD to explore possible associations with in utero CVB exposure. Serum samples were obtained from 122 women referred for fetal echocardiography between 2006 and 2018. We quantified CVB IgG and IgM levels, with titers ≥ 15.0 U/mL considered positive and measured neutralizing antibodies for three CVB serotypes: CVB1, CVB3, and CVB4. Using data from the national enterovirus surveillance system, we compared the annual exposure rates for each serotype in our cohort to infections reported across the United States. 98 pregnancies with no genetic defects were included. Overall, 29.6% (29/98) had positive IgG and 4.1% (4/98) of women had positive CVB IgM titers. To explore first-trimester CVB exposure, we focused exclusively on the 26 women with positive IgG and negative IgM titers. 61.5% (16/26) had neutralizing antibodies against a single serotype and 38.5% (10/26) against multiple CVB serotypes. CVB4 neutralizing antibodies were the most common (65.4%, 17/26), followed by CVB3 (53.9%, 14/26) and CVB1 (30.8%, 8/26). Among these, 30.8% of babies presented pulmonary valve anomalies: 19.2% (5/26) pulmonary atresia, and 11.5% (3/26) pulmonary stenosis. 23.1% (6/26) of babies had coronary sinusoids. CVB exposure in our cohort mirrored that of reported infections in the United States. Our results suggest a possible association between gestational CVB exposure and specific CHD, particularly pulmonary valve anomalies and coronary sinusoids.


Subject(s)
Coxsackievirus Infections , Heart Defects, Congenital , Pulmonary Atresia , Antibodies, Neutralizing , Antibodies, Viral , Coxsackievirus Infections/complications , Coxsackievirus Infections/pathology , Enterovirus B, Human/genetics , Female , Heart Defects, Congenital/complications , Heart Defects, Congenital/epidemiology , Humans , Immunoglobulin G , Immunoglobulin M , Pulmonary Atresia/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...