Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.095
Filter
1.
PeerJ ; 12: e17809, 2024.
Article in English | MEDLINE | ID: mdl-39329140

ABSTRACT

Background: The growing concern of antibiotic-resistant microbial strains worldwide has prompted the need for alternative methods to combat microbial resistance. Biofilm formation poses a significant challenge to antibiotic efficiency due to the difficulty of penetrating antibiotics through the sticky microbial aggregates. Drug repurposing is an innovative technique that aims to expand the use of non-antibiotic medications to address this issue. The primary objective of this study was to evaluate the antimicrobial properties of Diltiazem HCl, a 1,5-benzothiazepine Ca2 + channel blocker commonly used as an antihypertensive agent, against four pathogenic bacteria and three pathogenic yeasts, as well as its antiviral activity against the Coxsackie B4 virus (CoxB4). Methods: To assess the antifungal and antibacterial activities of Diltiazem HCl, the well diffusion method was employed, while crystal violet staining was used to determine the anti-biofilm activity. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) colorimetric assay was utilized to evaluate the antiviral activity of Diltiazem HCl against the CoxB4 virus. Results: This study revealed that Diltiazem HCl exhibited noticeable antimicrobial properties against Gram-positive bacteria, demonstrating the highest inhibition of Staphylococcus epidermidis, followed by Staphylococcus aureus. It effectively reduced the formation of biofilms by 95.1% and 90.7% for S. epidermidis, and S. aureus, respectively. Additionally, the antiviral activity of Diltiazem HCl was found to be potent against the CoxB4 virus, with an IC50 of 35.8 ± 0.54 µg mL-1 compared to the reference antiviral Acyclovir (IC50 42.71 ± 0.43 µg mL-1). Conclusion: This study suggests that Diltiazem HCl, in addition to its antihypertensive effect, may also be a potential treatment option for infections caused by Gram-positive bacteria and the CoxB4 viruses, providing an additional off-target effect for Diltiazem HCl.


Subject(s)
Antiviral Agents , Biofilms , Diltiazem , Drug Repositioning , Microbial Sensitivity Tests , Diltiazem/pharmacology , Biofilms/drug effects , Antiviral Agents/pharmacology , Humans , Anti-Bacterial Agents/pharmacology , Enterovirus B, Human/drug effects , Staphylococcus aureus/drug effects , Calcium Channel Blockers/pharmacology , Staphylococcus epidermidis/drug effects , Anti-Infective Agents/pharmacology , Antifungal Agents/pharmacology , Gram-Positive Bacteria/drug effects
2.
Theranostics ; 14(13): 5303-5315, 2024.
Article in English | MEDLINE | ID: mdl-39267792

ABSTRACT

Rationale: Parkin (an E3 ubiquitin protein ligase) is an important regulator of mitophagy. However, the role of Parkin in viral myocarditis (VMC) remains unclear. Methods: Coxsackievirus B3 (CVB3) infection was induced in mice to create VMC. Cardiac function and inflammatory response were evaluated by echocardiography, histological assessment, and molecular analyses. AAV9 (adeno-associated virus 9), transmission electron microscopy (TEM) and western blotting were used to investigate the mechanisms by which Parkin regulates mitophagy and cardiac inflammation. Results: Our data indicated that Parkin- and BNIP3 (BCL2 interacting protein 3 like)-mediated mitophagy was activated in VMC mice and neonatal rat cardiac myocytes (NRCMs) infected with CVB3, which blocked autophagic flux by inhibiting autophagosome-lysosome fusion. Parkin silencing aggravated mortality and accelerated the development of cardiac dysfunction in CVB3-treated mice. While silencing of Parkin did not significantly increase inflammatory response through activating NF-κB pathway and production of inflammatory cytokines post-VMC, the mitophagy activity were reduced, which stimulated the accumulation of damaged mitochondria. Moreover, Parkin silencing exacerbated VMC-induced apoptosis. We consistently found that Parkin knockdown disrupted mitophagy activity and inflammatory response in NRCMs. Conclusion: This study elucidated the important role of Parkin in maintaining cardiac function and inflammatory response by regulating mitophagy activity and the NF-κB pathway during acute VMC. Although the functional impact of mitophagy remains unclear, our findings suggest that Parkin silencing may accelerate VMC development.


Subject(s)
Coxsackievirus Infections , Mitophagy , Myocarditis , Myocytes, Cardiac , Ubiquitin-Protein Ligases , Animals , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Myocarditis/virology , Myocarditis/metabolism , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/virology , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Male , Rats , Enterovirus B, Human/physiology , Apoptosis , Disease Models, Animal , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , NF-kappa B/metabolism , Mice, Inbred BALB C
3.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273444

ABSTRACT

The ongoing COVID-19 pandemic, caused by SARS-CoV-2, continues to pose significant global health challenges. The results demonstrated that GB-2 at 200 µg/mL effectively increased the population of 293T-ACE2 cells with low RBD binding for both SARS-CoV-2 Omicron EG.5.1 and HV.1 variants by dual-color flow cytometry, indicating its ability to inhibit virus attachment. Further investigation revealed that (+)-catechin at 25 and 50 µg/mL did not significantly alter the ACE2-RBD interaction for the EG.5.1 variant. In contrast, theaflavin showed inhibitory effects at both 25 and 50 µg/mL for EG.5.1, while only the higher concentration was effective for HV.1. Notably, theaflavin 3-gallate exhibited a potent inhibition of ACE2-RBD binding for both variants at both concentrations tested. Molecular docking studies provided insight into the binding mechanisms of theaflavin and theaflavin 3-gallate with the RBD of EG.5.1 and HV.1 variants. Both compounds showed favorable docking scores, with theaflavin 3-gallate demonstrating slightly lower scores (-8 kcal/mol) compared to theaflavin (-7 kcal/mol) for both variants. These results suggest stable interactions between the compounds and key residues in the RBD, potentially explaining their inhibitory effects on virus attachment. In conclusion, GB-2, theaflavin, and theaflavin 3-gallate demonstrate significant potential as inhibitors of the ACE2-RBD interaction in Omicron variants, highlighting their therapeutic promise against COVID-19. However, these findings are primarily based on computational and in vitro studies, necessitating further in vivo research and clinical trials to confirm their efficacy and safety in humans.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antiviral Agents , Biflavonoids , Catechin , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Humans , Biflavonoids/pharmacology , Biflavonoids/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Molecular Docking Simulation , HEK293 Cells , COVID-19/virology , COVID-19 Drug Treatment , Virus Attachment/drug effects , Enterovirus B, Human/drug effects , Gallic Acid/analogs & derivatives
4.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39273613

ABSTRACT

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.


Subject(s)
Coxsackievirus Infections , Disease Models, Animal , MicroRNAs , Myocarditis , Animals , Myocarditis/metabolism , Myocarditis/virology , Myocarditis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Male , Mice , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/genetics , Coxsackievirus Infections/virology , Enterovirus B, Human , Biomarkers/metabolism , Sex Characteristics , Cytokines/metabolism , Cytokines/genetics , Myocardium/metabolism , Myocardium/pathology , Inflammation/genetics , Inflammation/metabolism , Sex Factors , Gene Expression Regulation
5.
J Gen Virol ; 105(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39311842

ABSTRACT

Non-polio enteroviruses (NPEV) cause significant disease worldwide. Population-based sero-surveillance, by measuring antibodies against specific NPEV types, provides additional information on past circulation and the prediction for future upsurges. Virus neutralisation assays (VNA), the current method of choice for measuring NPEV type specific antibodies, are not entirely standardised. Via the European Non-Polio Enterovirus Network, we organised a VNA quality assessment in which twelve laboratories participated. We provided five echovirus (E) types (E1, E18, E30 G2, E30 G6 and E6) and intravenous immunoglobulins (IVIG) as a sample for the NPEV VNA quality assessment. Differences in VNA protocols and neutralising Ab (nAb) titres were found between the participating laboratories with geometric coefficients of variation ranging from 10.3-62.9 %. Mixed-effects regression analysis indicated a small but significant effect of type of cell line used. Harmonisation of cell line passage number, however, did not improve variation between laboratories. Calibration by making use of a reference sample, reduced variation between laboratories but differences in nAb titres remained higher than two log2 dilution steps. In conclusion, sero-surveillance data from different laboratories should be compared with caution and standardised protocols are needed.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Enterovirus B, Human , Neutralization Tests , Europe , Humans , Antibodies, Viral/blood , Antibodies, Viral/immunology , Neutralization Tests/methods , Neutralization Tests/standards , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Enterovirus B, Human/immunology , Echovirus Infections/virology , Echovirus Infections/epidemiology , Echovirus Infections/immunology , Seroepidemiologic Studies , Enterovirus Infections/virology , Enterovirus Infections/immunology
6.
Viruses ; 16(9)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39339923

ABSTRACT

Enteroviruses such as coxsackievirus B3 are identified as a common cause of viral myocarditis, but the potential mechanism of its replication and pathogenesis are largely unknown. The genomes of a variety of viruses contain N6-methyladenosine (m6A), which plays important roles in virus replication. Here, by using the online bioinformatics tools SRAMP and indirect immunofluorescence assay (IFA), we predict that the CVB3 genome contains m6A sites and found that CVB3 infection could alter the expression and cellular localization of m6A-related proteins. Moreover, we found that 3-deazaadenosine (3-DAA), an m6A modification inhibitor, significantly decreased CVB3 replication. We also observed that the m6A methyltransferases methyltransferase-like protein 3 (METTL3) and METTL14 play positive roles in CVB3 replication, whereas m6A demethylases fat mass and obesity-associated protein (FTO) or AlkB homolog 5 (ALKBH5) have opposite effects. Knockdown of the m6A binding proteins YTH domain family protein 1 (YTHDF1), YTHDF2 and YTHDF3 strikingly decreased CVB3 replication. Finally, the m6A site mutation in the CVB3 genome decreased the replication of CVB3 compared with that in the CVB3 wild-type (WT) strain. Taken together, our results demonstrated that CVB3 could exploit m6A modification to promote viral replication, which provides new insights into the mechanism of the interaction between CVB3 and the host.


Subject(s)
Adenosine , Enterovirus B, Human , Methyltransferases , RNA-Binding Proteins , Virus Replication , Adenosine/analogs & derivatives , Adenosine/metabolism , Virus Replication/drug effects , Enterovirus B, Human/physiology , Enterovirus B, Human/genetics , Enterovirus B, Human/drug effects , Humans , Methyltransferases/metabolism , Methyltransferases/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Coxsackievirus Infections/virology , HeLa Cells , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , Host-Pathogen Interactions , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Genome, Viral
7.
Viruses ; 16(9)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39339932

ABSTRACT

Enteroviruses are a genus of small RNA viruses that are responsible for approximately one billion global infections annually. These infections range in severity from the common cold and flu-like symptoms to more severe diseases, such as viral myocarditis, pancreatitis, and neurological disorders, that continue to pose a global health challenge with limited therapeutic strategies currently available. In the current study, we sought to understand the interaction between coxsackievirus B3 (CVB3), which is a model enterovirus, and macrophage cells, as there is limited understanding of how this virus interacts with macrophage innate immune cells. Our study demonstrated that CVB3 can robustly activate macrophages without apparent viral replication in these cells. We also showed that myeloid cells lacked the viral entry receptor coxsackievirus and adenovirus receptor (CAR). However, the expression of exogenous CAR in RAW264.7 macrophages was unable to overcome the viral replication deficit. Interestingly, the CAR expression was associated with altered inflammatory responses during prolonged infection. Additionally, we identified the autophagy protein LC3 as a novel stimulus for macrophage activation. These findings provide new insights into the mechanisms of CVB3-induced macrophage activation and its implications for viral pathogenesis.


Subject(s)
Coxsackie and Adenovirus Receptor-Like Membrane Protein , Enterovirus B, Human , Macrophage Activation , Macrophages , Virus Internalization , Virus Replication , Animals , Enterovirus B, Human/physiology , Macrophages/virology , Macrophages/immunology , Mice , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , RAW 264.7 Cells , Coxsackievirus Infections/virology , Coxsackievirus Infections/immunology , Humans , Autophagy , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Mice, Inbred C57BL
8.
Viruses ; 16(9)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39339978

ABSTRACT

Group B Coxsackieviruses (CVB) are one of the causative pathogens of myocarditis, which may progress to cardiomyopathy. The pathogenesis of CVB is not fully understood, and effective antiviral therapy is not available. N-acetylcysteine (NAC), the classic antioxidant, has been used in clinical practice for several decades to treat various medical conditions. In this study, the anti-CVB effect of NAC was investigated. We show that NAC dramatically suppressed viral replication and alleviated cardiac injury induced by CVB3. To further study the antiviral mechanism of NAC, RNA-sequencing was performed for CVB3-infected cells with NAC treatment. We found that eukaryotic elongation factor 1 alpha 1 (EEF1A1) is one of the most upregulated genes in CVB3-infected cells. However, EEF1A2, the highly homologous isoform of EEF1A1, remains unchanged. EEF1A1 expression was significantly suppressed by NAC treatment in CVB3-infected cells, while EEF1A2 was not affected. eEF1A1 knockdown significantly inhibited CVB3 replication, implicating that eEF1A1 facilitates viral replication. Importantly, we show that eEF1A1, which was not expressed in the myocardia of newborn mice, was significantly upregulated by CVB3 infection. NAC markedly downregulated the expression of eEF1A1 but not eEF1A2 in the myocardia of CVB3-infected mice. Furthermore, NAC accelerated eEF1A1 degradation by promoting autophagy in CVB3-infected cells. We show that p62, one of the critical adaptors of autophagic targets, interacts with eEF1A1 and was downregulated in CVB3-infected cells upon NAC treatment. Taken together, this study demonstrated that NAC shows a potent anti-CVB effect through the downregulation of eEF1A1.


Subject(s)
Acetylcysteine , Coxsackievirus Infections , Down-Regulation , Enterovirus B, Human , Peptide Elongation Factor 1 , Virus Replication , Peptide Elongation Factor 1/metabolism , Peptide Elongation Factor 1/genetics , Virus Replication/drug effects , Enterovirus B, Human/drug effects , Enterovirus B, Human/physiology , Animals , Acetylcysteine/pharmacology , Humans , Mice , Coxsackievirus Infections/drug therapy , Coxsackievirus Infections/virology , Down-Regulation/drug effects , Antiviral Agents/pharmacology , Cell Line , Myocarditis/virology , Myocarditis/drug therapy , Male
9.
J Virol ; 98(9): e0080524, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194244

ABSTRACT

Coxsackievirus group B3 (CVB3) belongs to the genus Enteroviruses of the family Picornaviridae and is the main pathogen underlying viral myocarditis (VMC). No specific therapeutic is available for this condition. Argininosuccinate synthase 1 (ASS1) is a key enzyme in the urea cycle that converts citrulline and aspartic acid to argininosuccinate. Here, we found that CVB3 and its capsid protein VP2 inhibit the autophagic degradation of ASS1 and that CVB3 consumes citrulline to upregulate ASS1, triggers urea cycle metabolic reprogramming, and then activates macrophages to develop pro-inflammatory polarization, thereby promoting the occurrence and development of VMC. Conversely, citrulline supplementation to prevent depletion can downregulate ASS1, rescue macrophage polarization, and alleviate the pathogenicity of VMC. These findings provide a new perspective on the occurrence and development of VMC, revealing ASS1 as a potential new target for treating this disease. IMPORTANCE: Viral myocarditis (VMC) is a common and potentially life-threatening myocardial inflammatory disease, most commonly caused by CVB3 infection. So far, the pathogenesis of VMC caused by CVB3 is mainly focused on two aspects: one is the direct myocardial injury caused by a large number of viral replication in the early stage of infection, and the other is the local immune cell infiltration and inflammatory damage of the myocardium in the adaptive immune response stage. There are few studies on the early innate immunity of CVB3 infection in myocardial tissue, but the appearance of macrophages in the early stage of CVB3 infection suggests that they can play a regulatory role as early innate immune response cells in myocardial tissue. Here, we discovered a possible new mechanism of VMC caused by CVB3, revealed new drug targets for anti-CVB3, and discovered the therapeutic potential of citrulline for VMC.


Subject(s)
Argininosuccinate Synthase , Coxsackievirus Infections , Enterovirus B, Human , Macrophages , Myocarditis , Myocarditis/virology , Myocarditis/metabolism , Myocarditis/immunology , Myocarditis/pathology , Enterovirus B, Human/physiology , Animals , Macrophages/virology , Macrophages/metabolism , Macrophages/immunology , Mice , Coxsackievirus Infections/virology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/metabolism , Argininosuccinate Synthase/metabolism , Humans , Male , Inflammation/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocardium/immunology , Capsid Proteins/metabolism , Capsid Proteins/immunology , Metabolic Reprogramming
10.
Commun Biol ; 7(1): 969, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122806

ABSTRACT

Serine proteases are important environmental contributors of enterovirus biocontrol. However, the structural features of molecular interaction accounting for the susceptibility of enteroviruses to proteases remains unexplained. Here, we describe the molecular mechanisms involved in the recruitment of serine proteases to viral capsids. Among the virus types used, coxsackievirus A9 (CVA9), but not CVB5 and echovirus 11 (E11), was inactivated by Subtilisin A in a host-independent manner, while Bovine Pancreatic Trypsin (BPT) only reduced CVA9 infectivity in a host-dependent manner. Predictive interaction models of each protease with capsid protomers indicate the main targets as internal disordered protein (IDP) segments exposed either on the 5-fold vertex (DE loop VP1) or at the 5/2-fold intersection (C-terminal end VP1) of viral capsids. We further show that a functional binding protease/capsid depends on both the strength and the evolution over time of protease-VP1 complexes, and lastly on the local adaptation of proteases on surrounding viral regions. Finally, we predicted three residues on CVA9 capsid that trigger cleavage by Subtilisin A, one of which may act as a sensor residue contributing to enzyme recognition on the DE loop. Overall, this study describes an important biological mechanism involved in enteroviruses biocontrol.


Subject(s)
Capsid Proteins , Capsid , Serine Proteases , Capsid/metabolism , Serine Proteases/metabolism , Serine Proteases/chemistry , Serine Proteases/genetics , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Humans , Enterovirus/enzymology , Enterovirus/physiology , Animals , Enterovirus B, Human/physiology , Enterovirus B, Human/enzymology
11.
BMJ Case Rep ; 17(8)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39216881

ABSTRACT

Acute viral myocarditis and hyperthyroidism can present with acute coronary syndrome. However, the link between hyperthyroidism and myocarditis has hardly been explored apart from a small collection of published case reports. We report a case where a patient presents with severe chest pain and was found to have concomitant severe coronary vasospasm and acute myocarditis and was diagnosed with Graves' disease.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , Graves Disease , Myocarditis , Humans , Myocarditis/virology , Myocarditis/diagnosis , Coxsackievirus Infections/complications , Coxsackievirus Infections/diagnosis , Graves Disease/complications , Male , Chest Pain/etiology , Adult , Electrocardiography , Antithyroid Agents/therapeutic use , Acute Disease , Coronary Vasospasm/complications , Coronary Vasospasm/diagnosis , Middle Aged
12.
Int J Mol Sci ; 25(16)2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39201692

ABSTRACT

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.


Subject(s)
Enterovirus B, Human , Myocarditis , Pancreatitis , STAT1 Transcription Factor , STAT3 Transcription Factor , Virus Replication , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Myocarditis/virology , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Animals , Pancreatitis/metabolism , Pancreatitis/virology , Pancreatitis/pathology , Pancreatitis/genetics , Enterovirus B, Human/physiology , Mice , Humans , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/genetics , Cell Nucleus/metabolism , Male , Active Transport, Cell Nucleus , Gene Expression Regulation , Acute Disease , Cell Line , Signal Transduction
13.
Nat Cardiovasc Res ; 3(1): 76-93, 2024 01.
Article in English | MEDLINE | ID: mdl-39195892

ABSTRACT

Viral myocarditis is characterized by infiltration of mononuclear cells essential for virus elimination. GPR15 has been identified as a homing receptor for regulatory T cells in inflammatory intestine diseases, but its role in inflammatory heart diseases is still elusive. Here we show that GPR15 deficiency impairs coxsackievirus B3 elimination, leading to adverse cardiac remodeling and dysfunction. Delayed recruitment of regulatory T cells in GPR15-deficient mice was accompanied by prolonged persistence of cytotoxic and regulatory T cells. In addition, RNA sequencing revealed prolonged inflammatory response and altered chemotaxis in knockout mice. In line, we identified GPR15 and its ligand GPR15L as an important chemokine receptor-ligand pair for the recruitment of regulatory and cytotoxic T cells. In summary, the insufficient virus elimination might be caused by a delayed recruitment of T cells as well as delayed interferon-γ expression, resulting in a prolonged inflammatory response and an adverse outcome in GPR15-deficient mice.


Subject(s)
Coxsackievirus Infections , Disease Models, Animal , Enterovirus B, Human , Mice, Knockout , Myocarditis , Receptors, G-Protein-Coupled , Animals , Myocarditis/immunology , Myocarditis/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/immunology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/genetics , Enterovirus B, Human/immunology , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Acute Disease , Interferon-gamma/metabolism , Mice , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Male , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Myocardium/metabolism , Myocardium/immunology , Myocardium/pathology , Signal Transduction
14.
Acta Crystallogr F Struct Biol Commun ; 80(Pt 8): 183-190, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39052022

ABSTRACT

Enteroviruses cause a wide range of disorders with varying presentations and severities, and some enteroviruses have emerged as serious public health concerns. These include Coxsackievirus B3 (CVB3), an active causative agent of viral myocarditis, and Coxsackievirus B4 (CVB4), which may accelerate the progression of type 1 diabetes. The 3C proteases from CVB3 and CVB4 play important roles in the propagation of these viruses. In this study, the 3C proteases from CVB3 and CVB4 were expressed in Escherichia coli and purified by affinity chromatography and gel-filtration chromatography. The crystals of the CVB3 and CVB4 3C proteases diffracted to 2.10 and 2.01 Šresolution, respectively. The crystal structures were solved by the molecular-replacement method and contained a typical chymotrypsin-like fold and a conserved His40-Glu71-Cys147 catalytic triad. Comparison with the structures of 3C proteases from other enteroviruses revealed high similarity with minor differences, which will guide the design of 3C-targeting inhibitors with broad-spectrum properties.


Subject(s)
3C Viral Proteases , Amino Acid Sequence , Cysteine Endopeptidases , Enterovirus B, Human , Models, Molecular , Viral Proteins , 3C Viral Proteases/chemistry , Crystallography, X-Ray , Enterovirus B, Human/enzymology , Enterovirus B, Human/chemistry , Enterovirus B, Human/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Catalytic Domain , Humans , Protein Conformation , Cloning, Molecular
15.
J Med Virol ; 96(8): e29831, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39072815

ABSTRACT

Coxsackievirus B3 (CVB3) triggers viral myocarditis, with no effective vaccine yet. This fecal-oral transmitted pathogen has prompted interest in mucosal immunization strategies to impede CVB3 spread. We developed a new attenuated vaccine strain, named CVB3(mu). The potential of CVB3(mu) to stimulate mucosal immune protection remains to be elucidated. This study evaluates the attenuation characteristics of CVB3(mu) via a rapid evolution cellular model and RNA sequencing. Its temperature sensitivity and safety were evaluated through in vitro and in vivo experiments. The mucosal immunity protection of CVB3(mu) was assessed via intranasal immunization in Balb/c mice. The results indicate that CVB3(mu) exhibits temperature sensitivity and forms smaller plaques. It sustains fewer genetic mutations and still possesses certain attenuated traits up to the 25th passage, in comparison to CVB3(WT). Intranasal immunization elicited a significant serum neutralizing antibodies, and a substantial sIgA response in nasal washes. In vivo trials revealed CVB3(mu) protection in adult mice and passive protection in suckling mice against lethal CVB3(WT) challenges. In conclusion, CVB3(mu), a live attenuated intranasal vaccine, provides protection involving humoral and mucosal immunity, making it a promising candidate to control CVB3 spread and infection.


Subject(s)
Administration, Intranasal , Antibodies, Neutralizing , Antibodies, Viral , Coxsackievirus Infections , Enterovirus B, Human , Immunity, Mucosal , Mice, Inbred BALB C , Vaccines, Attenuated , Viral Vaccines , Animals , Enterovirus B, Human/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/prevention & control , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Mice , Immunoglobulin A, Secretory/immunology , Humans , Female , Disease Models, Animal
16.
J Nat Prod ; 87(7): 1735-1745, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38976838

ABSTRACT

Heterologous expression of an atr terpenoid gene cluster derived from Streptomyces atratus Gö66 in S. albus J1074 led to the discovery of three novel labdane diterpenoids featuring an unprecedented 6/6/5-fused tricyclic skeleton, designated as atralabdans A-C (1-3), along with a known compound, labdanmycin A. Compounds 1-3 were identified through extensive spectroscopic analysis, including NMR calculations with DP4+ probability analysis, and a comparative assessment of experimental and theoretical electronic circular dichroism (ECD) spectra. A plausible biosynthetic pathway for these compounds was proposed. Compounds 1-3 exhibited inhibitory activity against the human neurotropic coxsackievirus B3 (CVB3); 1 was the most potent, surpassing the positive control ribavirin with a higher therapeutic index.


Subject(s)
Antiviral Agents , Soil Microbiology , Streptomyces , Streptomyces/chemistry , Streptomyces/genetics , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Molecular Structure , Diterpenes/pharmacology , Diterpenes/chemistry , Humans , Enterovirus B, Human/drug effects , Multigene Family
17.
Sci Rep ; 14(1): 17634, 2024 07 31.
Article in English | MEDLINE | ID: mdl-39085360

ABSTRACT

Self-amplifying RNAs (saRNAs) are versatile vaccine platforms that take advantage of a viral RNA-dependent RNA polymerase (RdRp) to amplify the messenger RNA (mRNA) of an antigen of interest encoded within the backbone of the viral genome once inside the target cell. In recent years, more saRNA vaccines have been clinically tested with the hope of reducing the vaccination dose compared to the conventional mRNA approach. The use of N1-methyl-pseudouridine (1mΨ), which enhances RNA stability and reduces the innate immune response triggered by RNAs, is among the improvements included in the current mRNA vaccines. In the present study, we evaluated the effects of this modified nucleoside on various saRNA platforms based on different viruses. The results showed that different stages of the replication process were affected depending on the backbone virus. For TNCL, an insect virus of the Alphanodavirus genus, replication was impaired by poor recognition of viral RNA by RdRp. In contrast, the translation step was severely abrogated in coxsackievirus B3 (CVB3), a member of the Picornaviridae family. Finally, the effects of 1mΨ on Semliki forest virus (SFV), were not detrimental in in vitro studies, but no advantages were observed when immunogenicity was tested in vivo.


Subject(s)
RNA, Viral , Virus Replication , RNA, Viral/genetics , Animals , Replicon/genetics , Pseudouridine/metabolism , Positive-Strand RNA Viruses/genetics , Humans , Semliki forest virus/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , RNA Stability , Enterovirus B, Human/genetics , Enterovirus B, Human/physiology
18.
Virology ; 597: 110156, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38981316

ABSTRACT

This study aims to elucidate the role of TIP30 (30 KDa HIV-1 TAT-Interacting Protein) in the progression of coxsackievirus B3 (CVB3)-induced viral myocarditis. TIP30 knockout and wildtype mice were intraperitoneally infected with CVB3 and evaluated at day 7 post-infection. HeLa cells were transfected with TIP30 lentiviral particles and subsequently infected with CVB3 to evaluate viral replication, cellular pathogenesis, and mechanistic target of rapamycin complex 1 (mTORC1) signaling. Deletion of the TIP30 gene heightened heart virus titers and mortality rates in mice with CVB3-induced myocarditis, exacerbating cardiac damage and fibrosis, and elevating pro-inflammatory factors level. In vitro experiments demonstrated the modulation of mTORC1 signaling by TIP30 during CVB3 infection in HeLa cells. TIP30 overexpression mitigated CVB3-induced cellular pathogenesis and VP1 expression, with rapamycin, an mTOR1 inhibitor, reversing these effects. These findings suggest TIP30 plays a critical protective role against CVB3-induced myocarditis by regulating mTORC1 signaling.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , Mechanistic Target of Rapamycin Complex 1 , Mice, Knockout , Myocarditis , Signal Transduction , Animals , Humans , Male , Mice , Coxsackievirus Infections/virology , Coxsackievirus Infections/metabolism , Disease Models, Animal , Enterovirus B, Human/physiology , HeLa Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocarditis/virology , Myocarditis/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Virus Replication
19.
PLoS Biol ; 22(7): e3002709, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39012844

ABSTRACT

RNA viruses have notoriously high mutation rates due to error-prone replication by their RNA polymerase. However, natural selection concentrates variability in a few key viral proteins. To test whether this stems from different mutation tolerance profiles among viral proteins, we measured the effect of >40,000 non-synonymous mutations across the full proteome of coxsackievirus B3 as well as >97% of all possible codon deletions in the nonstructural proteins. We find significant variation in mutational tolerance within and between individual viral proteins, which correlated with both general and protein-specific structural and functional attributes. Furthermore, mutational fitness effects remained stable across cell lines, suggesting selection pressures are mostly conserved across environments. In addition to providing a rich dataset for understanding virus biology and evolution, our results illustrate that incorporation of mutational tolerance data into druggable pocket discovery can aid in selecting targets with high barriers to drug resistance.


Subject(s)
Enterovirus B, Human , Mutation , Proteome , Enterovirus B, Human/genetics , Proteome/metabolism , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Genetic Fitness , Virus Replication/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
20.
Viruses ; 16(7)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39066174

ABSTRACT

After the first phase of the COVID-19 pandemic in Europe, a new highly pathogenic variant of echovirus 11 (E11) was detected. The aim of this study was to analyze the genetic diversity of Polish E11 environmental and clinical strains circulating between 2017 and 2023 as well as compare them with E11 strains isolated from severe neonatal sepsis cases reported in Europe between 2022 and 2023. Additionally, the study explores the effectiveness of environmental monitoring in tracking the spread of new variants. For this purpose, the complete sequences of the VP1 capsid protein gene were determined for 266 E11 strains isolated in Poland from 2017 to 2023, and phylogenetic analysis was performed. In the years 2017-2023, a significant increase in the detection of E11 strains was observed in both environmental and clinical samples in Poland. The Polish E11 strains represented three different genotypes, C3, D5 and E, and were characterized by a high diversity. In Poland, the intensive circulation of the new variant E11, responsible for severe neonatal infections with a high mortality in Europe, was detected in the years 2022-2023. This investigation demonstrates the important role of environmental surveillance in the tracking of enteroviruses circulation, especially in settings with limited clinical surveillance.


Subject(s)
COVID-19 , Enterovirus B, Human , Phylogeny , SARS-CoV-2 , Poland/epidemiology , Humans , Enterovirus B, Human/genetics , Enterovirus B, Human/classification , Enterovirus B, Human/isolation & purification , COVID-19/epidemiology , COVID-19/virology , SARS-CoV-2/genetics , SARS-CoV-2/classification , SARS-CoV-2/isolation & purification , Genotype , Genetic Variation , Capsid Proteins/genetics , Infant, Newborn , Echovirus Infections/epidemiology , Echovirus Infections/virology , Pandemics
SELECTION OF CITATIONS
SEARCH DETAIL