Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.742
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.
Sci Rep ; 14(1): 10289, 2024 05 04.
Article in English | MEDLINE | ID: mdl-38704437

ABSTRACT

Myocarditis is considered a fatal form of foot-and-mouth disease (FMD) in suckling calves. In the present study, a total of 17 calves under 4 months of age and suspected clinically for FMD were examined for clinical lesions, respiratory rate, heart rate, and heart rhythm. Lesion samples, saliva, nasal swabs, and whole blood were collected from suspected calves and subjected to Sandwich ELISA and reverse transcription multiplex polymerase chain reaction (RT-mPCR) for detection and serotyping of FMD virus (FMDV). The samples were found to be positive for FMDV serotype "O". Myocarditis was suspected in 6 calves based on tachypnoea, tachycardia, and gallop rhythm. Serum aspartate aminotransferase (AST), creatinine kinase myocardial band (CK-MB) and lactate dehydrogenase (LDH), and cardiac troponins (cTnI) were measured. Mean serum AST, cTn-I and LDH were significantly higher (P < 0.001) in < 2 months old FMD-infected calves showing clinical signs suggestive of myocarditis (264.833 ± 4.16; 11.650 ± 0.34 and 1213.33 ± 29.06) than those without myocarditis (< 2 months old: 110.00 ± 0.00, 0.06 ± 0.00, 1050.00 ± 0.00; > 2 months < 4 months: 83.00 ± 3.00, 0.05 ± 0.02, 1159.00 ± 27.63) and healthy control groups (< 2 months old: 67.50 ± 3.10, 0.047 ± 0.01, 1120.00 ± 31.62; > 2 months < 4 months: 72.83 ± 2.09, 0.47 ± 0.00, 1160.00 ± 18.44). However, mean serum CK-MB did not differ significantly amongst the groups. Four calves under 2 months old died and a necropsy revealed the presence of a pathognomic gross lesion of the myocardial form of FMD known as "tigroid heart". Histopathology confirmed myocarditis. This study also reports the relevance of clinical and histopathological findings and biochemical markers in diagnosing FMD-related myocarditis in suckling calves.


Subject(s)
Foot-and-Mouth Disease , Myocarditis , Animals , Cattle , Myocarditis/veterinary , Myocarditis/virology , Myocarditis/pathology , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease/pathology , Cattle Diseases/virology , Cattle Diseases/blood , Cattle Diseases/pathology , Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease Virus/isolation & purification , Animals, Suckling , Age Factors , Aspartate Aminotransferases/blood , Male , L-Lactate Dehydrogenase/blood
4.
Nat Commun ; 15(1): 4153, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755212

ABSTRACT

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.


Subject(s)
ADAM Proteins , Cardiovirus Infections , Encephalomyocarditis virus , Immunity, Innate , Interferon Type I , Interferon-Induced Helicase, IFIH1 , Membrane Proteins , Mice, Knockout , Myocarditis , Animals , Encephalomyocarditis virus/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interferon Type I/metabolism , Interferon Type I/immunology , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , ADAM Proteins/metabolism , ADAM Proteins/genetics , ADAM Proteins/immunology , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Myocarditis/immunology , Myocarditis/virology , Humans , Mice, Inbred C57BL , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Signal Transduction/immunology , Male , HEK293 Cells
5.
Sci Rep ; 14(1): 11124, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750107

ABSTRACT

Influenza is a significant public health and economic threat around the world. Epidemiological studies have demonstrated a close association between influenza pandemics and cardiovascular mortality. Moreover, it has been shown that there is a decrease in cardiovascular mortality in high-risk patients following vaccination with the influenza vaccine. Here, we have investigated the role of anti-viral STAT1 signaling in influenza-induced myocarditis. Wild-type mice (C57BL/6) were infected with either influenza A/PR/8/34 or control, and cellular response and gene expression analysis from the heart samples were assessed 7 days later. The expression of interferon response genes STAT1, STAT2, Mx1, OASL2, ISG15, chemokines CCL2, CCL3, CXCL9 and CXCL10, and the frequency of neutrophils (CD45+CD11b+Ly6G+) and CD4+ T cells (CD45+CD4+) were all significantly increased in influenza-infected mice when compared to vehicle controls. These data suggest that influenza infection induces interferons, inflammatory chemokines, and cellular recruitment during influenza infection. We further investigated the role of STAT1 in influenza-induced myocarditis. The frequency of neutrophils and the levels of lipocalin 2 were significantly increased in STAT1-/- mice when compared to WT controls. Finally, we investigated the role of Lcn2 in viral-induced myocarditis. We found that in the absence of Lcn2, there was preserved cardiac function in Lcn2-/- mice when compared to WT controls. These data suggest that the absence of Lcn2 is cardioprotective during viral-induced myocarditis.


Subject(s)
Lipocalin-2 , Mice, Inbred C57BL , Myocarditis , Orthomyxoviridae Infections , STAT1 Transcription Factor , Animals , Myocarditis/virology , Myocarditis/metabolism , Myocarditis/etiology , Lipocalin-2/metabolism , Lipocalin-2/genetics , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Mice , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Neutrophils/metabolism , Neutrophils/immunology , Male , Mice, Knockout
6.
Front Immunol ; 15: 1380697, 2024.
Article in English | MEDLINE | ID: mdl-38715608

ABSTRACT

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.


Subject(s)
COVID-19 , Myocarditis , SARS-CoV-2 , Myocarditis/virology , Myocarditis/immunology , Myocarditis/therapy , Myocarditis/genetics , Humans , COVID-19/immunology , COVID-19/genetics , COVID-19/therapy , SARS-CoV-2/physiology , Methylation , 5-Methylcytosine/metabolism , Immunity, Innate , COVID-19 Drug Treatment , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , RNA Processing, Post-Transcriptional
7.
J Zhejiang Univ Sci B ; 25(5): 422-437, 2024 May 15.
Article in English, Chinese | MEDLINE | ID: mdl-38725341

ABSTRACT

Viral myocarditis (VMC) is one of the most common acquired heart diseases in children and teenagers. However, its pathogenesis is still unclear, and effective treatments are lacking. This study aimed to investigate the regulatory pathway by which exosomes alleviate ferroptosis in cardiomyocytes (CMCs) induced by coxsackievirus B3 (CVB3). CVB3 was utilized for inducing the VMC mouse model and cellular model. Cardiac echocardiography, left ventricular ejection fraction (LVEF), and left ventricular fractional shortening (LVFS) were implemented to assess the cardiac function. In CVB3-induced VMC mice, cardiac insufficiency was observed, as well as the altered levels of ferroptosis-related indicators (glutathione peroxidase 4 (GPX4), glutathione (GSH), and malondialdehyde (MDA)). However, exosomes derived from human umbilical cord mesenchymal stem cells (hucMSCs-exo) could restore the changes caused by CVB3 stimulation. Let-7a-5p was enriched in hucMSCs-exo, and the inhibitory effect of hucMSCs-exolet-7a-5p mimic on CVB3-induced ferroptosis was higher than that of hucMSCs-exomimic NC (NC: negative control). Mothers against decapentaplegic homolog 2 (SMAD2) increased in the VMC group, while the expression of zinc-finger protein 36 (ZFP36) decreased. Let-7a-5p was confirmed to interact with SMAD2 messenger RNA (mRNA), and the SMAD2 protein interacted directly with the ZFP36 protein. Silencing SMAD2 and overexpressing ZFP36 inhibited the expression of ferroptosis-related indicators. Meanwhile, the levels of GPX4, solute carrier family 7, member 11 (SLC7A11), and GSH were lower in the SMAD2 overexpression plasmid (oe-SMAD2)+let-7a-5p mimic group than in the oe-NC+let-7a-5p mimic group, while those of MDA, reactive oxygen species (ROS), and Fe2+ increased. In conclusion, these data showed that ferroptosis could be regulated by mediating SMAD2 expression. Exo-let-7a-5p derived from hucMSCs could mediate SMAD2 to promote the expression of ZFP36, which further inhibited the ferroptosis of CMCs to alleviate CVB3-induced VMC.


Subject(s)
Enterovirus B, Human , Exosomes , Ferroptosis , Mesenchymal Stem Cells , MicroRNAs , Myocytes, Cardiac , Signal Transduction , Smad2 Protein , Umbilical Cord , Mesenchymal Stem Cells/metabolism , Exosomes/metabolism , Animals , Humans , Mice , Smad2 Protein/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Enterovirus B, Human/physiology , Myocytes, Cardiac/metabolism , Umbilical Cord/cytology , Coxsackievirus Infections/metabolism , Male , Myocarditis/metabolism , Myocarditis/virology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
8.
BMC Cardiovasc Disord ; 24(1): 282, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811883

ABSTRACT

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.


Subject(s)
Autopsy , Death, Sudden, Cardiac , Herpesvirus 6, Human , Myocarditis , Parvovirus B19, Human , Humans , Myocarditis/virology , Myocarditis/pathology , Myocarditis/genetics , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , Death, Sudden, Cardiac/prevention & control , Male , Adult , Female , Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/isolation & purification , Parvovirus B19, Human/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/virology , Cardiomyopathy, Dilated/pathology , Roseolovirus Infections/complications , Roseolovirus Infections/virology , Roseolovirus Infections/diagnosis , Roseolovirus Infections/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Parvoviridae Infections/complications , Young Adult , Genetic Predisposition to Disease , Fatal Outcome , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Coinfection , Cause of Death , Mutation , Middle Aged
9.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Article in English | MEDLINE | ID: mdl-38696536

ABSTRACT

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.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , Interferon Type I , Myocarditis , Myocytes, Cardiac , RNA, Viral , Myocarditis/virology , Myocarditis/immunology , Myocarditis/genetics , Animals , Myocytes, Cardiac/virology , Myocytes, Cardiac/metabolism , Mice , Enterovirus B, Human/immunology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Coxsackievirus Infections/genetics , Interferon Type I/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , Immunity, Innate , Signal Transduction , Interferon-beta/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Male , 5' Untranslated Regions
11.
Viruses ; 16(5)2024 04 25.
Article in English | MEDLINE | ID: mdl-38793559

ABSTRACT

Coxsackievirus B3 (CVB3) is a positive single-strand RNA genome virus which belongs to the enterovirus genus in the picornavirus family, like poliovirus. It is one of the most prevalent pathogens that cause myocarditis and pancreatitis in humans. However, a suitable therapeutic medication and vaccination have yet to be discovered. Caboxamycin, a benzoxazole antibiotic isolated from the culture broth of the marine strain Streptomyces sp., SC0774, showed an antiviral effect in CVB3-infected HeLa cells and a CVB3-induced myocarditis mouse model. Caboxamycin substantially decreased CVB3 VP1 production and cleavage of translation factor eIF4G1 from CVB3 infection. Virus-positive and -negative strand RNA was dramatically reduced by caboxamycin treatment. In addition, the cleavage of the pro-apoptotic molecules BAD, BAX, and caspase3 was significantly inhibited by caboxamycin treatment. In animal experiments, the survival rate of mice was improved following caboxamycin treatment. Moreover, caboxamycin treatment significantly decreased myocardial damage and inflammatory cell infiltration. Our study showed that caboxamycin dramatically suppressed cardiac inflammation and mouse death. This result suggests that caboxamycin may be suitable as a potential antiviral drug for CVB3.


Subject(s)
Antiviral Agents , Coxsackievirus Infections , Disease Models, Animal , Enterovirus B, Human , Myocarditis , Animals , Myocarditis/drug therapy , Myocarditis/virology , Mice , Coxsackievirus Infections/drug therapy , Coxsackievirus Infections/virology , Humans , Enterovirus B, Human/drug effects , HeLa Cells , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Male , Mice, Inbred BALB C , Inflammation/drug therapy , Inflammation/virology , Virus Replication/drug effects
12.
Int Immunopharmacol ; 133: 112073, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38636372

ABSTRACT

BACKGROUND: Myocarditis is an important clinical issue which lacks specific treatment by now. Ivermectin (IVM) is an inhibitor of importin α/ß-mediated nuclear translocation. This study aimed to explore the therapeutic effects of IVM on acute myocarditis. METHODS: Mouse models of coxsackie B3 virus (CVB3) infection-induced myocarditis and experimental autoimmune myocarditis (EAM) were established to evaluate the effects of IVM. Cardiac functions were evaluated by echocardiography and Millar catheter. Cardiac inflammatory infiltration was assessed by histological staining. Cytometric bead array and quantitative real-time PCR were used to detect the levels of pro-inflammatory cytokines. The macrophages and their M1/M2 polarization were analyzed via flow cytometry. Protein expression and binding were detected by co-immunoprecipitation, Western blotting and histological staining. The underlying mechanism was verified in vitro using CVB3-infected RAW264.7 macrophages. Cyclic polypeptide (cTN50) was synthesized to selectively inhibit the nuclear translocation of NF-κB/p65, and CVB3-infected RAW264.7 cells were treated with cTN50. RESULTS: Increased expression of importin ß was observed in both models. IVM treatment improved cardiac functions and reduced the cardiac inflammation associated with CVB3-myocarditis and EAM. Furthermore, the pro-inflammatory cytokine (IL-1ß/IL-6/TNF-α) levels were downregulated via the inhibition of the nuclear translocation of NF-κB/p65 in macrophages. IVM and cTN50 treatment also inhibited the nuclear translocation of NF-κB/p65 and downregulated the expression of pro-inflammatory cytokines in RAW264.7 macrophages. CONCLUSIONS: Ivermectin inhibits the nuclear translocation of NF-κB/p65 and the expression of major pro-inflammatory cytokines in myocarditis. The therapeutic effects of IVM on viral and non-viral myocarditis models suggest its potential application in the treatment of acute myocarditis.


Subject(s)
Ivermectin , Mice, Inbred BALB C , Myocarditis , Transcription Factor RelA , Animals , Myocarditis/drug therapy , Myocarditis/virology , Mice , Ivermectin/therapeutic use , Ivermectin/pharmacology , RAW 264.7 Cells , Male , Transcription Factor RelA/metabolism , Coxsackievirus Infections/drug therapy , Enterovirus B, Human/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Cytokines/metabolism , beta Karyopherins/metabolism , Disease Models, Animal , Autoimmune Diseases/drug therapy , Humans , Myocardium/pathology , Myocardium/metabolism
13.
Nat Commun ; 15(1): 3481, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664417

ABSTRACT

Viral myocarditis, an inflammatory disease of the myocardium, is a significant cause of sudden death in children and young adults. The current coronavirus disease 19 pandemic emphasizes the need to understand the pathogenesis mechanisms and potential treatment strategies for viral myocarditis. Here, we found that TRIM29 was highly induced by cardiotropic viruses and promoted protein kinase RNA-like endoplasmic reticulum kinase (PERK)-mediated endoplasmic reticulum (ER) stress, apoptosis, and reactive oxygen species (ROS) responses that promote viral replication in cardiomyocytes in vitro. TRIM29 deficiency protected mice from viral myocarditis by promoting cardiac antiviral functions and reducing PERK-mediated inflammation and immunosuppressive monocytic myeloid-derived suppressor cells (mMDSC) in vivo. Mechanistically, TRIM29 interacted with PERK to promote SUMOylation of PERK to maintain its stability, thereby promoting PERK-mediated signaling pathways. Finally, we demonstrated that the PERK inhibitor GSK2656157 mitigated viral myocarditis by disrupting the TRIM29-PERK connection, thereby bolstering cardiac function, enhancing cardiac antiviral responses, and curbing inflammation and immunosuppressive mMDSC in vivo. Our findings offer insight into how cardiotropic viruses exploit TRIM29-regulated PERK signaling pathways to instigate viral myocarditis, suggesting that targeting the TRIM29-PERK axis could mitigate disease severity.


Subject(s)
Adenine , Endoplasmic Reticulum Stress , Indoles , Myocarditis , Myocytes, Cardiac , eIF-2 Kinase , Animals , Humans , Male , Mice , Adenine/analogs & derivatives , Apoptosis , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/virology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/pathology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/virology , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Virus Replication
14.
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
15.
Antiviral Res ; 217: 105702, 2023 09.
Article in English | MEDLINE | ID: mdl-37604350

ABSTRACT

Coxsackievirus B3 (CVB3) is one of the major pathogens of viral myocarditis, lacking specific anti-virus therapeutic options. Increasing evidence has shown an important involvement of the miR-17-92 cluster both in virus infection and cardiovascular development and diseases, while its role in CVB3-induced viral myocarditis remains unclear. In this study, we found that miR-19a and miR-19b were significantly up-regulated in heart tissues of CVB3-infected mice and exerted a significant facilitatory impact on CVB3 biosynthesis and replication, with a more pronounced effect observed in miR-19b, by targeting the encoding region of viral RNA-dependent RNA polymerase 3D (RdRp, 3Dpol) to increase viral genomic RNA stability. The virus-promoting effects were nullified by the synonymous mutations in the viral 3Dpol-encoding region, which corresponded to the seed sequence shared by miR-19a and miR-19b. In parallel, treatment with miR-19b antagomir not only resulted in a noteworthy suppression of CVB3 replication and infection in infected cells, but also demonstrated a significant reduction in the cardiac viral load of CVB3-infected mice, resulting in a considerable alleviation of myocarditis. Collectively, our study showed that CVB3-induced cardiac miR-19a/19b contributed to viral myocarditis via facilitating virus biosynthesis and replication, and targeting miR-19a/19b might represent a novel therapeutic target for CVB3-induced viral myocarditis.


Subject(s)
Enterovirus B, Human , MicroRNAs , Myocarditis , Myocardium , Virus Replication , Enterovirus B, Human/genetics , Enterovirus B, Human/physiology , Myocarditis/metabolism , Myocarditis/virology , Myocardium/metabolism , Myocardium/pathology , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Humans , Virus Replication/drug effects , Virus Replication/genetics , Genome, Viral , RNA-Dependent RNA Polymerase/genetics , Antagomirs/pharmacology , Mice, Inbred BALB C , HEK293 Cells , HeLa Cells , Mice , Animals
16.
Immunol Cell Biol ; 101(8): 735-745, 2023 09.
Article in English | MEDLINE | ID: mdl-37253434

ABSTRACT

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.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , HMGB1 Protein , Myocarditis , Animals , Mice , Apoptosis/genetics , HMGB1 Protein/metabolism , Mice, Inbred BALB C , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/virology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Tumor Suppressor Protein p53/metabolism , Coxsackievirus Infections/immunology
17.
Artif Organs ; 47(2): 396-407, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36269688

ABSTRACT

BACKGROUND: The impact of etiologies of acute fulminant myocarditis (AFM), which requires extracorporeal membrane oxygenation (ECMO), on clinical outcomes remains unknown. This study aimed to investigate the risk factors for ECMO weaning and mortality among patients with AFM due to viral etiologies in a tertiary referral medical center. METHODS: We included 33 adults with AFM who received ECMO and were admitted between January 2002 and January 2021. General demographics, laboratory data, echocardiography findings, and long-term outcomes were analyzed for confirmed viral etiology and unconfirmed etiology groups. RESULTS: The overall hospital survival rate was 54.5%. The age, sex, severity of the hemodynamic condition, and cardiac rhythm were similar between the two groups. Multivariate Cox regression analysis revealed that a confirmed viral etiology (HR 4.201, 95% CI 1.061-16.666), peri-ECMO renal replacement therapy (RRT) (HR 9.804, 1.140-83.333) and a high positive end-expiratory pressure (PEEP) in the ventilator settings at 24 h after ECMO (HR 1.479, 1.020-2.143) were significant prognostic factors for in-hospital mortality. Peri-ECMO RRT was also a significant negative prognostic factor for successful ECMO weaning (OR 0.061, 0.006-0.600) in the multivariate logistic model. CONCLUSIONS: Among AFM patients receiving ECMO support, RRT use was associated with a decreased chance of survival to ECMO weaning. Multiple organ dysfunction and a high PEEP were also predictive of a lower chance of hospital survival. Those with a confirmed diagnosis of viral myocarditis may require more medical attention due to the higher risk of hospital mortality than those without a definite diagnosis.


Subject(s)
Extracorporeal Membrane Oxygenation , Myocarditis , Adult , Humans , Myocarditis/diagnosis , Myocarditis/therapy , Myocarditis/virology , Retrospective Studies , Treatment Outcome , Virus Diseases
18.
Rev. esp. cardiol. (Ed. impr.) ; 75(9): 735-747, sept. 2022. ilus, tab, graf
Article in Spanish | IBECS | ID: ibc-208226

ABSTRACT

Introducción y objetivos Las secuelas cardiacas tras la infección por SARS-CoV-2 todavía están poco documentadas. Se realizó un estudio transversal en trabajadores sanitarios para estudiar la prevalencia de afección pericárdica y miocárdica tras la infección por SARS-CoV-2. Métodos Se estudió a 139 trabajadores sanitarios con infección previa confirmada por SARS-CoV-2. Los participantes se sometieron a evaluación clínica, electrocardiograma, laboratorio, incluido el perfil de células inmunitarias, y resonancia magnética cardiaca (RMC). El diagnóstico clínico de pericarditis se realizó ante la presencia de los criterios clásicos y el diagnóstico clínico de miocarditis ante la presencia de al menos 2 criterios de RMC. Resultados La mediana de edad fue de 52 (41–57) años, el 71,9% eran mujeres, y el 16,5% había sido hospitalizado previamente por neumonía por COVID-19. En la evaluación (10,4 [9,3–11,0] semanas después de los síntomas de infección), todos los participantes presentaban estabilidad hemodinámica. El 41,7% presentaba dolor torácico, disnea o palpitaciones; el 49,6%, alteraciones electrocardiográficas; el 7,9%, elevación de NT-proBNP; el 0,7%, elevación de troponina; y el 60,4%, alteraciones en la RMC (AU)


Introduction and objectives The cardiac sequelae of SARS-CoV-2 infection are still poorly documented. We conducted a cross-sectional study in healthcare workers to report evidence of pericardial and myocardial involvement after SARS-CoV-2 infection. Methods We studied 139 healthcare workers with confirmed past SARS-CoV-2 infection. Participants underwent clinical assessment, electrocardiography, and laboratory tests, including immune cell profiling and cardiac magnetic resonance (CMR). Clinically suspected pericarditis was diagnosed when classic criteria were present and clinically suspected myocarditis was based on the combination of at least 2 CMR criteria. Results Median age was 52 (41-57) years, 71.9% were women, and 16.5% were previously hospitalized for COVID-19 pneumonia. On examination (10.4 [9.3-11.0] weeks after infection-like symptoms), participants showed hemodynamic stability. Chest pain, dyspnea or palpitations were present in 41.7% participants, electrocardiographic abnormalities in 49.6%, NT-proBNP elevation in 7.9%, troponin in 0.7%, and CMR abnormalities in 60.4%. A total of 30.9% participants met criteria for either pericarditis and/or myocarditis: isolated pericarditis was diagnosed in 5.8%, myopericarditis in 7.9%, and isolated myocarditis in 17.3%. Most participants (73.2%) showed altered immune cell counts in blood, particularly decreased eosinophil (27.3%; P<.001) and increased cytotoxic T cell numbers (17.3%; P <.001). Clinically suspected pericarditis was associated (P <.005) with particularly elevated cytotoxic T cells and decreased eosinophil counts, while participants diagnosed with clinically suspected myopericarditis or myocarditis had lower (P <.05) neutrophil counts, natural killer-cells, and plasma cells. Conclusions Pericardial and myocardial involvement with clinical stability are frequent after SARS-CoV-2 infection and are associated with specific immune cell profiles (AU)


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Arrhythmias, Cardiac/virology , Coronavirus Infections/complications , Myocarditis/virology , Health Personnel , Pericarditis/virology , Coronavirus Infections/epidemiology , Myocarditis/epidemiology , Pericarditis/epidemiology , Cross-Sectional Studies , Spain/epidemiology
19.
J Biomed Sci ; 29(1): 55, 2022 Jul 31.
Article in English | MEDLINE | ID: mdl-35909127

ABSTRACT

BACKGROUND: Infections by viruses including severe acute respiratory syndrome coronavirus 2 could cause organ inflammations such as myocarditis, pneumonia and encephalitis. Innate immunity to viral nucleic acids mediates antiviral immunity as well as inflammatory organ injury. However, the innate immune mechanisms that control viral induced organ inflammations are unclear. METHODS: To understand the role of the E3 ligase TRIM18 in controlling viral myocarditis and organ inflammation, wild-type and Trim18 knockout mice were infected with coxsackievirus B3 for inducing viral myocarditis, influenza A virus PR8 strain and human adenovirus for inducing viral pneumonia, and herpes simplex virus type I for inducing herpes simplex encephalitis. Mice survivals were monitored, and heart, lung and brain were harvested for histology and immunohistochemistry analysis. Real-time PCR, co-immunoprecipitation, immunoblot, enzyme-linked immunosorbent assay, luciferase assay, flow cytometry, over-expression and knockdown techniques were used to understand the molecular mechanisms of TRIM18 in regulating type I interferon (IFN) production after virus infection in this study. RESULTS: We find that knockdown or deletion of TRIM18 in human or mouse macrophages enhances production of type I IFN in response to double strand (ds) RNA and dsDNA or RNA and DNA virus infection. Importantly, deletion of TRIM18 protects mice from viral myocarditis, viral pneumonia, and herpes simplex encephalitis due to enhanced type I IFN production in vivo. Mechanistically, we show that TRIM18 recruits protein phosphatase 1A (PPM1A) to dephosphorylate TANK binding kinase 1 (TBK1), which inactivates TBK1 to block TBK1 from interacting with its upstream adaptors, mitochondrial antiviral signaling (MAVS) and stimulator of interferon genes (STING), thereby dampening antiviral signaling during viral infections. Moreover, TRIM18 stabilizes PPM1A by inducing K63-linked ubiquitination of PPM1A. CONCLUSIONS: Our results indicate that TRIM18 serves as a negative regulator of viral myocarditis, lung inflammation and brain damage by downregulating innate immune activation induced by both RNA and DNA viruses. Our data reveal that TRIM18 is a critical regulator of innate immunity in viral induced diseases, thereby identifying a potential therapeutic target for treatment.


Subject(s)
Encephalitis, Herpes Simplex , Myocarditis , Ubiquitin-Protein Ligases , Virus Diseases , Animals , Antiviral Agents , Humans , Immunity, Innate , Inflammation/genetics , Mice , Myocarditis/genetics , Myocarditis/virology , Protein Phosphatase 2C , RNA , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...