Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 283
Filtrar
1.
Int Immunopharmacol ; 133: 112073, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38636372

RESUMEN

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.


Asunto(s)
Ivermectina , Ratones Endogámicos BALB C , Miocarditis , Factor de Transcripción ReIA , Animales , Miocarditis/tratamiento farmacológico , Miocarditis/virología , Ratones , Ivermectina/uso terapéutico , Ivermectina/farmacología , Células RAW 264.7 , Masculino , Factor de Transcripción ReIA/metabolismo , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Citocinas/metabolismo , beta Carioferinas/metabolismo , Modelos Animales de Enfermedad , Enfermedades Autoinmunes/tratamiento farmacológico , Humanos , Miocardio/patología , Miocardio/metabolismo
2.
Antiviral Res ; 215: 105621, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37156267

RESUMEN

Group B Coxsackieviruses (CVB) are non-enveloped small RNA viruses in the genus Enterovirus, family Picornaviridae. CVB infection causes diverse conditions from common cold to myocarditis, encephalitis, and pancreatitis. No specific antiviral is available for the treatment of CVB infection. Anisomycin, a pyrrolidine-containing antibiotic and translation inhibitor, was reported to inhibit the replication of some picornaviruses. However, it is unknown if anisomycin can act as an antiviral against CVB infection. Here we observed that anisomycin showed potent inhibition on CVB type 3 (CVB3) infection with negligible cytotoxicity when applied at the early stage of virus infection. Mice infected with CVB3 showed markedly alleviated myocarditis with reduced viral replication. We found that CVB3 infection significantly increased the transcription of eukaryotic translation elongation factor 1 alpha 1 (eEF1A1). CVB3 replication was suppressed by EEF1A1 knockdown, while elevated by EEF1A1 overexpression. Similar to the effect of CVB3 infection, EEF1A1 transcription was increased in response to anisomycin treatment. However, eEF1A1 protein level was decreased with anisomycin treatment in a dose-dependent manner in CVB3-infected cells. Moreover, anisomycin promoted eEF1A1 degradation, which was inhibited by the treatment of chloroquine but not MG132. We demonstrated that eEF1A1 interacted with the heat shock cognate protein 70 (HSP70), and eEF1A1 degradation was inhibited by LAMP2A knockdown, implicating that eEF1A1 is degraded through chaperone-mediated autophagy. Taken together, we demonstrated that anisomycin, which inhibits CVB replication through promoting the lysosomal degradation of eEF1A1, could be a potential antiviral candidate for the treatment of CVB infection.


Asunto(s)
Infecciones por Coxsackievirus , Miocarditis , Ratones , Animales , Humanos , Anisomicina/farmacología , Factor 1 de Elongación Peptídica/metabolismo , Factor 1 de Elongación Peptídica/farmacología , Antivirales/farmacología , Antivirales/metabolismo , Enterovirus Humano B , Lisosomas/metabolismo , Replicación Viral , Infecciones por Coxsackievirus/tratamiento farmacológico , Células HeLa
3.
J Appl Biomed ; 21(1): 15-22, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37016776

RESUMEN

Myocardial fibrosis is the most serious complication of viral myocarditis (VMC). This study aimed to investigate the therapeutic benefits and underlying mechanisms of lentivirus-mediated human tissue kallikrein gene transfer in myocardial fibrosis in VMC mice. We established VMC mouse model via intraperitoneal injection with Coxsackie B3 virus. The effect was then assessed after treatment with vehicle, the empty lentiviral vectors (EZ.null), and the vectors expressing hKLK1 (EZ.hKLK1) via tail vein injection for 30 days, respectively. The results showed that administering EZ.hKLK1 successfully induced hKLK1 overexpression in mouse heart. Compared with EZ.null treatment, EZ.hKLK1 administration significantly reduced the heart/weight ratio, improved cardiac function, and ameliorated myocardial inflammation in VMC mice, suggesting that hKLK1 overexpression alleviates VMC in mice. EZ.hKLK1 administration also significantly abrogated the increased myocardial collagen content, type I/III collagen ratio, TGF-ß1 mRNA and protein expression in VMC mice, suggesting that hKLK1 overexpression reduces collagen accumulation and blunts TGF-ß1 signaling in the hearts of VMC mice. In conclusion, our results suggest that hKLK1 alleviates myocardial fibrosis in VMC mice, possibly by downregulating TGF-ß1 expression.


Asunto(s)
Cardiomiopatías , Infecciones por Coxsackievirus , Miocarditis , Ratones , Humanos , Animales , Miocarditis/tratamiento farmacológico , Miocarditis/metabolismo , Factor de Crecimiento Transformador beta1/genética , Colágeno/metabolismo , Colágeno/uso terapéutico , Colágeno Tipo I/genética , Colágeno Tipo I/uso terapéutico , Infecciones por Coxsackievirus/terapia , Infecciones por Coxsackievirus/tratamiento farmacológico , Fibrosis , Colágeno Tipo III/uso terapéutico
4.
Food Funct ; 14(9): 4129-4142, 2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37042256

RESUMEN

Lactiplantibacillus plantarum is a lactic acid bacterium widely used in food production. Coxsackievirus B3 (CVB3) is an important human pathogen associated with acute pancreatitis development, and no antiviral therapeutics or vaccines are approved to treat or prevent its infection. However, whether L. plantarum could inhibit CVB3 infection remains unclear. Here, L. plantarum FLPL05 showed antiviral activity against CVB3 infection in vivo and in vitro. Pretreatment with L. plantarum FLPL05 reduced serum amylase levels, CVB3 viral load in the pancreas, serum pro-inflammatory cytokine levels, and macrophage infiltration in CVB3-infected mice. In mice, L. plantarum FLPL05 inhibited CVB3-induced pancreas apoptosis via the B cell leukemia/lymphoma 2 (BCL2)/BCL2-associated X protein (BAX)/caspase-3 (CASP3) signaling pathway. Furthermore, L. plantarum FLPL05 reduced CVB3 replication, protected cells from the cytopathic effect of CVB3 infection, and inhibited cell apoptosis. Moreover, L. plantarum FLPL05's exopolysaccharide (EPS) had activity against CVB3 in vitro, reducing the CVB3 titer and improving cell activity. Therefore, L. plantarum FLPL05 pretreatment improved CVB3-induced pancreatitis by partially reversing pancreatitis, which might be associated with EPS. Consequently, L. plantarum FLPL05 could be a potential probiotic with antiviral activity against CVB3.


Asunto(s)
Infecciones por Coxsackievirus , Pancreatitis , Humanos , Ratones , Animales , Caspasa 3/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Enterovirus Humano B/metabolismo , Enfermedad Aguda , Pancreatitis/tratamiento farmacológico , Transducción de Señal , Infecciones por Coxsackievirus/tratamiento farmacológico , Antivirales/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
5.
J Virol ; 97(3): e0143322, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36916989

RESUMEN

Cathelicidin antimicrobial peptides (mouse, CRAMP; human, LL-37) have broad-spectrum antiviral activities against enveloped viruses, but their mechanisms of action against nonenveloped viruses remain to be elucidated. Coxsackievirus B3 (CVB3), a member of nonenveloped virus belonging to the Enterovirus genus of Picornaviridae, is an important pathogen of viral myocarditis and dilated cardiomyopathy. Here, we observed that cardiac CRAMP expression was significantly upregulated in mice after CVB3 infection. The administration of CRAMP or LL-37 markedly suppressed CVB3 infection in mice, and CRAMP deficiency increased the susceptibility of mice to CVB3. CRAMP and LL-37 inhibited CVB3 replication in primary cardiomyocytes. However, they did not inactivate CVB3 particles and did not regulate the response of cardiomyocytes against CVB3 infection. Intriguingly, they inhibited CVB3 transmission through the exosome, but not virus receptor. In detail, CRAMP and LL-37 directly induced the lysis of exosomes by interfering with exosomal heat shock protein 60 (HSP60) and then blocked the diffusion of exosomes to recipient cells and inhibited the establishment of productive infection by exosomes. In addition, the interaction of CRAMP and LL-37 with HSP60 simultaneously inhibited HSP60-induced apoptosis in cardiomyocytes and reduced HSP60-enhanced CVB3 replication. Our findings reveal a novel mechanism of cathelicidins against viral infection and provide a new therapeutic strategy for CVB3-induced viral myocarditis. IMPORTANCE The relative mechanisms that cathelicidin antimicrobial peptides use to influence nonenveloped virus infection are unclear. We show here that cathelicidin antimicrobial peptides (CRAMP and LL-37) directly target exosomal HSP60 to destroy exosomes, which in turn block the diffusion of exosomes to recipient cardiomyocytes and reduced HSP60-induced apoptosis, thus restricting coxsackievirus B3 infection. Our results provide new insights into the mechanisms cathelicidin antimicrobial peptides use against viral infection.


Asunto(s)
Catelicidinas , Infecciones por Coxsackievirus , Exosomas , Miocitos Cardíacos , Animales , Humanos , Ratones , Apoptosis/efectos de los fármacos , Catelicidinas/administración & dosificación , Chaperonina 60/antagonistas & inhibidores , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B/fisiología , Exosomas/efectos de los fármacos , Miocarditis , Miocitos Cardíacos/efectos de los fármacos , Replicación Viral
6.
Microbiol Spectr ; 11(1): e0424822, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36541788

RESUMEN

Umifenovir, a broad-spectrum nonnucleoside antiviral drug, has a promising efficacy against coxsackievirus B4 (CVB4) infection, but its mechanism remains unclear. CVB4 is a common human single-stranded RNA virus that belongs to the Picornaviridae family and the Enterovirus genus. Enterovirus can cause severe diseases, such as meningitis, myocarditis, pancreatitis, insulin-dependent diabetes, and several other diseases, in both adults and children. We have previously demonstrated the critical role of interleukin 10 (IL-10) in promoting CVB4 infection and the downregulation of IL-10 by umifenovir. To further explore the underlying mechanisms of umifenovir, we characterized the epigenetic regulation of IL-10 in IL-10 knockout RAW264.7 cells and a BALB/c mouse splenocyte model. Mechanistically, we found that umifenovir inhibited CVB4-activated IL-10 by enhancing the methylation level of the repressive histones H3K9me3 and H3K27me3 while reducing the acetylation level of the activating histone H3K9ac in the promoter region of the IL-10 gene. Furthermore, using a chromosome conformation capture approach, we discovered that CVB4 infection activated the IL-10 gene by forming an intrachromosomal interaction between the IL-10 gene promoter and an intronic enhancer of the downstream MK2 (mitogen-activated protein kinase [MAPK]-activated protein kinase 2 [MAPKAPK2]) gene, a critical component of the p38-MAPK signaling pathway, which is required for IL-10 gene expression. However, umifenovir treatment abolished this spatial conformation and chromatin interaction, thus reducing the continuous expression of IL-10 and subsequent CVB4 replication. In conclusion, this study reveals a novel epigenetic mechanism by which umifenovir controls CVB4 infections, thus laying a theoretical foundation for therapeutic use of umifenovir. IMPORTANCE Viral infections are major threats to human health because of their strong association with a variety of inflammation-related diseases, especially cancer. Many antiviral drugs are performing poorly in treating viral infections. This is probably due to the immunosuppressive effect of highly expressed IL-10, which is caused by viral infection. Umifenovir is a broad-spectrum antiviral drug. Our recent studies showed that umifenovir has a significant inhibitory effect on CVB4 infection and can reduce IL-10 expression caused by CVB4. However, another antiviral drug, rupintrivir, showed good antiviral activity but had no effect on the expression of IL-10. This suggests that the regulation of IL-10 expression is a key part of the antiviral mechanism of umifenovir. Therefore, due to the dual function of the inhibition of CVB4 replication and the regulation of immune antiviral pathway, the mechanism of umifenovir is of great value to study.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus Humano B , Interleucina-10 , Animales , Niño , Humanos , Ratones , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Coxsackievirus/tratamiento farmacológico , Infecciones por Coxsackievirus/metabolismo , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/metabolismo , Epigénesis Genética , Interleucina-10/metabolismo , Interleucina-10/farmacología , Enterovirus Humano B/efectos de los fármacos
7.
Rev Med Virol ; 33(1): e2406, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36371612

RESUMEN

Coxsackieviruses B (CVB) are small, non-enveloped, single-stranded RNA viruses belonging to the Enterovirus genus of the Picornaviridae family. They are common worldwide and cause a wide variety of human diseases ranging from those having relatively mild symptoms to severe acute and chronic pathologies such as cardiomyopathy and type 1 diabetes. The development of safe and effective strategies to combat these viruses remains a challenge. The present review outlines current approaches to control CVB infections and associated diseases. Various drugs targeting viral or host proteins involved in viral replication as well as vaccines have been developed and shown potential to prevent or combat CVB infections in vitro and in vivo in animal models. Repurposed drugs and alternative strategies targeting miRNAs or based on plant extracts and probiotics and their derivatives have also shown antiviral effects against CVB. In addition, clinical trials with vaccines and drugs are underway and offer hope for the prevention or treatment of CVB-induced diseases.


Asunto(s)
Infecciones por Coxsackievirus , Diabetes Mellitus Tipo 1 , Infecciones por Enterovirus , Enterovirus , Animales , Humanos , Infecciones por Coxsackievirus/tratamiento farmacológico , Infecciones por Coxsackievirus/prevención & control , Infecciones por Enterovirus/complicaciones , Enterovirus Humano B , Diabetes Mellitus Tipo 1/complicaciones
8.
Int Immunopharmacol ; 113(Pt A): 109343, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36308891

RESUMEN

OBJECTIVE: To investigate whether interleukin-37 (IL-37) could directly inhibit the formation of neutrophil extracellular traps (NETs) in the early stage of acute viral myocarditis (VMC) and its potential mechanisms of action. METHODS: Acute VMC was induced by intraperitoneally injecting coxsackievirus B3 (CVB3)(103 TCID50) in mice on day 0. Mice were injected with AAV9-IL-37 or AAV9-NC through the caudal vein 1 week before intraperitoneal administration of CVB3. DNASE1 (50ug per mouse) was administered on days 0 to 7 to investigate the role of NETs formation during acute VMC. The severity of myocardial inflammation was evaluated by observing the general condition of mice and detecting cardiac histopathology. Moreover, neutrophils isolated from healthy human peripheral blood were stimulated by phorbol myristate acetate (PMA 100 nM) and treated with IL-37 (0.1 ng/ml) or BAY11-7082(2.5uM) in vitro. The production of NETs was detected by immunofluorescence labeled MPO and DNA. The expression of related proteins (IκBα, P-IκBα, NFκb, P-NFκb) was detected by Western blot. RESULTS: The results showed that, like DNASE1, IL-37 alleviates the symptoms in acute VMC induced by CVB3, reduces inflammatory cell infiltration, improves cardiac function, and inhibits the formation of NETs in the myocardium. Besides, both IL-37 and DNASE1 could effectively inhibit the activation of NFκb /IκBα. In the isolated peripheral blood neutrophils, the inhibitory effect of IL-37 on the formation of NETs and the activation of NFκb /IκBα was further confirmed. CONCLUSION: IL-37 has a protective effect on VMC by reducing the infiltration of inflammatory cells and inhibiting the formation of NETs at an early phase.


Asunto(s)
Infecciones por Coxsackievirus , Trampas Extracelulares , Lesiones Cardíacas , Miocarditis , Animales , Humanos , Ratones , Infecciones por Coxsackievirus/tratamiento farmacológico , Inhibidor NF-kappaB alfa , Trampas Extracelulares/metabolismo , Ratones Endogámicos BALB C , Miocarditis/metabolismo , Interleucinas , Enterovirus Humano B
10.
Virus Res ; 317: 198816, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35598772

RESUMEN

Group B coxsackievirus is an enterovirus that can cause a variety of diseases, including myocarditis, aseptic meningitis, and hand, foot, and mouth disease. Currently, there is no effective antiviral drug against this virus. In this study, we used a cytopathic effect-based viral inhibition assay to screen an FDA-approved drug library and found that doxepin hydrochloride had potential antiviral activity. Doxepin hydrochloride exhibited strong antiviral activity against coxsackievirus B types 1-3 with a 50% inhibitory concentration of 10.12 ± 0.85 µM. Moreover, doxepin hydrochloride did not exert antiviral activity against other enteroviruses, including enterovirus A71 (subtypes BrCr/C4) and coxsackievirus A (subtypes 6/10/16). Furthermore, doxepin hydrochloride inhibited virus replication in the early stage of the infection cycle rather than affecting the entry or assembly process. In addition, a few mechanism-related pharmacophores were discovered through gene association network analysis. These findings identify a possible lead compound for treating coxsackievirus B infection and simultaneously offer valuable clues for drug repositioning.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Coxsackievirus/tratamiento farmacológico , Doxepina/farmacología , Doxepina/uso terapéutico , Enterovirus Humano A/fisiología , Enterovirus Humano B , Humanos , Replicación Viral
11.
Int J Biochem Cell Biol ; 146: 106208, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35381374

RESUMEN

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.


Asunto(s)
Infecciones por Coxsackievirus , Miocarditis , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia , Homólogo de la Proteína 1 Relacionada con la Autofagia , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/tratamiento farmacológico , Infecciones por Coxsackievirus/metabolismo , Ratones , Ratones Endogámicos BALB C , Miocarditis/tratamiento farmacológico , Trehalosa/farmacología , Trehalosa/uso terapéutico
12.
Pediatr. aten. prim ; 24(93)ene. - mar. 2022. ilus
Artículo en Español | IBECS | ID: ibc-210333

RESUMEN

El eccema coxsackium es una dermatosis infecciosa caracterizada por lesiones papulovesiculosas, eccematosas e incluso costrosas de predominio en extremidades, nalgas y región perioral. Suele aparecer en pacientes con afectación cutánea previa, como es el caso de la dermatitis atópica de los niños. El germen causante más frecuentemente aislado es el Coxsackie A6. Está considerado como una forma atípica de la enfermedad mano-pie-boca y es importante un correcto diagnóstico diferencial para evitar tratamientos innecesarios (AU)


Eczema coxsackium is an infectious dermatosis characterized by papulovesicular, ezzematous and even crusty lesions predominantly on the extremities, buttocks and perioral region. It usually appears in patients with previous skin involvement, as in the case of atopic dermatitis in children. The most frequently isolated causative germ is Coxsackie A6. It is considered an atypical form of Hand, Foot and Mouth Disease and a correct differential diagnosis is important to avoid unnecessary treatments. (AU)


Asunto(s)
Humanos , Masculino , Lactante , Preescolar , Eccema/virología , Dermatitis Atópica/virología , Infecciones por Coxsackievirus/diagnóstico , Infecciones por Coxsackievirus/tratamiento farmacológico
13.
Virol Sin ; 36(6): 1575-1584, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34581960

RESUMEN

Coxsackievirus B1 (CVB1) is a leading causative agent of severe infectious diseases in humans and has been reported to be associated with outbreaks of aseptic meningitis, myocarditis, and the development of chronic diseases such as type 1 diabetes mellitus (T1DM). There is no approved vaccine or effective antiviral therapy to treat CBV1 infection. And animal models to assess the effects of antiviral agents and vaccine remain limited. In this study, we established a neonatal mouse model of CVB1 using a clinically isolated strain to characterize the pathological manifestations of virus infection and to promote the development of vaccines and antiviral drugs against CVB1. One-day-old BALB/c mice were susceptible to CVB1 infection by intraperitoneal injection. Mice challenged with CVB1 at a low dose [10 median tissue culture infective dose (TCID50)] exhibited a series of clinical symptoms, such as inactivity, emaciation, limb weakness, hair thinning, hunching and even death. Pathological examination and tissue viral load analysis showed that positive signals of CVB1 were detected in the heart, spinal cord, limb muscle and kidney without pathological damage. Particularly, CVB1 had a strong tropism towards the pancreas, causing severe cellular necrosis with inflammatory infiltration, and was spread by viraemia. Notably, the monoclonal antibody (mAb) 6H5 and antisera elicited from CVB1-vaccinated mice effectively protected the mice from CVB1 infection in the mouse model. In summary, the established neonatal mouse model is an effective tool for evaluating the efficacy of CVB1 antiviral reagents and vaccines.


Asunto(s)
Infecciones por Coxsackievirus , Vacunas Virales , Animales , Animales Recién Nacidos , Antivirales/farmacología , Infecciones por Coxsackievirus/tratamiento farmacológico , Modelos Animales de Enfermedad , Ratones
14.
Int Heart J ; 62(4): 900-909, 2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34234076

RESUMEN

Virus myocarditis (VMC) is a common cardiovascular disease and a major cause of sudden death in young adults. However, there is still a lack of effective treatments. Our previous studies found that calpain activation was involved in VMC pathogenesis. This study aims to explore the underlying mechanisms further. Neonatal rat cardiomyocytes (NRCMs) and transgenic mice overexpressing calpastatin (Tg-CAST), the endogenous calpain inhibitor, were used to establish VMC model. Hematoxylin and eosin and Masson staining revealed inflammatory cell infiltration and fibrosis. An ELISA array detected myocardial injury. Cardiac function was measured using echocardiography. CVB3 replication was assessed by capsid protein VP1. Apoptosis was measured by TUNEL staining, flow cytometry, and western blot. The endoplasmic reticulum (ER) stress-related proteins were detected by western blot. Our data showed that CVB3 infection resulted in cardiac injury, as evidenced by increased inflammatory responses and fibrosis, which induced myocardial apoptosis. Inhibiting calpain, both by PD150606 and calpastatin overexpression, could attenuate these effects. Furthermore, ER stress was activated during CVB3 infection. However, calpain inhibition could downregulate some ER stress-associated protein levels such as GRP78, pancreatic ER kinase-like ER kinase (PERK), and inositol-requiring enzyme-1α (IRE-1α), and ER stress-related apoptotic factors, during CVB3 infection. In conclusion, calpain inhibition attenuated CVB3-induced myocarditis by suppressing ER stress, thereby inhibiting cardiomyocyte apoptosis.


Asunto(s)
Acrilatos/uso terapéutico , Calpaína/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Miocarditis/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Acrilatos/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Calpaína/antagonistas & inhibidores , Infecciones por Coxsackievirus/tratamiento farmacológico , Infecciones por Coxsackievirus/metabolismo , Evaluación Preclínica de Medicamentos , Chaperón BiP del Retículo Endoplásmico , Enterovirus Humano B , Ratones Transgénicos , Miocarditis/tratamiento farmacológico , Miocarditis/virología , Ratas Sprague-Dawley
15.
Mediators Inflamm ; 2021: 5551578, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093086

RESUMEN

BACKGROUND: The functional characteristics of NLRP3 in the pathogenesis of coxsackievirus B3- (CVB3-) induced viral myocarditis (VMC) have not been fully elucidated, and the targeted therapeutic effect of NLRP3 or its related pathway in VMC has not been reported. METHOD: In this work, the change patterns of NLRP3- and Th17-related factors were detected during the pathological process of CVB3-induced VMC in Balb/c mice. The correlation between NLRP3 and Th17 cells during the VMC process was analyzed by Spearman test. The coculture system of spleen CD4+ T and bone marrow CD11c+ DC cells was set to explore the orchestration of NLRP3 and Th17 in the pathological development of VMC in vitro. Anti-IL-1ß antibody or NLRP3-/- Balb/c were used to block the NLRP3 pathway indirectly and directly to analyze the NLRP3-targeting therapeutic value. RESULTS: The change patterns of NLRP3- and Th17-related molecules in the whole pathological process of mouse CVB3-induced VMC were described. Through Spearman correlation analysis, it was confirmed that there was a close correlation between NLRP3 and Th17 cells in the whole pathological process of VMC. And the interaction mode between NLRP3 and Th17 was preliminarily explored in the cell experiment in vitro. Under the intervention of an anti-IL-1ß antibody or NLRP3 knockout, the survival rate of the intervention group was significantly improved, the degree of myocardial inflammation and fibrosis was significantly alleviated, and the content of myocardial IL-17 and spleen Th17 was also significantly decreased. CONCLUSION: Our findings demonstrated a key role of the NLRP3 inflammasome and its close relationship with Th17 in the pathological progression of CVB3-induced VMC and suggested a possible positive feedback-like mutual regulation mechanism between the NLRP3 inflammasome and Th17 in vitro and in the early stage of CVB3 infection. Taking NLRP3 as a new starting point, it provides a new target and idea for the prevention and treatment of CVB3-induced VMC.


Asunto(s)
Infecciones por Coxsackievirus/tratamiento farmacológico , Infecciones por Coxsackievirus/virología , Enterovirus Humano B , Miocarditis/tratamiento farmacológico , Miocarditis/terapia , Miocarditis/virología , Proteína con Dominio Pirina 3 de la Familia NLR/biosíntesis , Células Th17/citología , Animales , Antígeno CD11c/biosíntesis , Linfocitos T CD4-Positivos/citología , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Genotipo , Humanos , Inmunohistoquímica , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Neoplasias Laríngeas/metabolismo , Ratones , Ratones Endogámicos BALB C , Miocarditis/inmunología , Bazo/metabolismo
16.
Acta Virol ; 65(1): 58-67, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33827222

RESUMEN

Coxsackie virus B3 (CVB3) is believed to be a major cause of viral myocarditis, with virus-induced apoptosis playing an important role in pathogenesis. The purpose of this study was to characterize the antiviral activity of a novel fluoronucleoside analogue, N-cyclopropyl-4'-azido-2'-deoxy-2'-fluoro-ß-D-cytidine (NCC), against CVB3 in vitro and in vivo, and to establish whether NCC inhibits apoptosis in infected cells. In this study, HeLa cells infected with CVB3 were treated with NCC. Cell viability and apoptosis were examined. Caspase-3 and Bcl-2 levels were monitored by real-time RT PCR and Western blot analysis. For in vivo studies, BALB/c mice infected with CVB3 were treated with NCC daily. Serum markers of myocardial injury and histological studies were measured to examine myocardial injury on day 8 post-infection. To measure apoptosis, levels of Bcl-2 and caspase-3 were examined by immunohistochemistry and real-time RT-PCR. We found that NCC inhibited virus-mediated cytopathic effects in HeLa cells with an EC50 of 116.60 ± 0.32 µM. In infected mice, administration of NCC (2 mg/kg) decreased the activities of serum creatine kinase and lactic dehydrogenase, inhibited the replication of CVB3 and alleviated damage to the heart. Importantly, NCC suppressed CVB3-induced apoptosis in HeLa cells and affected the expression of apoptosis-related factors in infected mice. Together, our results demonstrate that NCC exerts significant antiviral activities against CVB3. We conclude that NCC is a potential therapeutic agent for the treatment of viral myocarditis. Keywords: coxsackie virus B3; viral myocarditis; N-cyclopropyl-4'-azido-2'-deoxy-2'-fluoro-ß-D-cytidine; apoptosis.


Asunto(s)
Infecciones por Coxsackievirus , Miocarditis , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Miocarditis/tratamiento farmacológico , Replicación Viral
17.
Arch Virol ; 166(7): 1869-1875, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33877422

RESUMEN

Although chemotherapy is generally indicated for treatment of enterovirus infections, antivirals are currently not used in clinical practice. The use of monotherapy is the main reason for this unfavourable state. This is related to the fact that enterovirus progeny consist of innumerable quasispecies, allowing the virus to develop drug resistance quickly. Here, we present a consecutive alternating administration (CAA) treatment scheme for combining enterovirus inhibitors. Applying the CAA approach with a combination of pleconaril (capsid binder), guanidine HCl (viral 2C inhibitor), and oxoglaucine (PI4KB inhibitor) (PGO) was found to be effective in the treatment of newborn mice infected with a massive inoculum (20 MLD50) of the coxsackievirus B3 cardiotropic Woodruff or neurotropic Nancy strain. In addition to preventing drug resistance, the CAA approach resulted in the parallel development of increased susceptibility to the compounds in the PGO combination. These observations demonstrate the therapeutic potential of the CAA approach for treatment of enterovirus infections.


Asunto(s)
Antivirales/administración & dosificación , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B/efectos de los fármacos , Animales , Farmacorresistencia Viral/efectos de los fármacos , Quimioterapia Combinada/métodos , Humanos , Ratones , Ratones Endogámicos ICR , Resultado del Tratamiento
18.
Basic Res Cardiol ; 116(1): 7, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523326

RESUMEN

A preclinical model of troponin I-induced myocarditis (AM) revealed a prominent role of the immunoproteasome (ip), the main immune cell-resident proteasome isoform, in heart-directed autoimmunity. Viral infection of the heart is a known trigger of cardiac autoimmunity, with the ip enhancing systemic inflammatory responses after infection with a cardiotropic coxsackievirusB3 (CV). Here, we used ip-deficient A/J-LMP7-/- mice to investigate the role of ip-mediated effects on adaptive immunity in CV-triggered myocarditis and found no alteration of the inflammatory heart tissue damage or cardiac function in comparison to wild-type controls. Aiming to define the impact of the systemic inflammatory storm under the control of ip proteolysis during CV infection, we targeted the ip in A/J mice with the inhibitor ONX 0914 after the first cycle of infection, when systemic inflammation has set in, well before cardiac inflammation. During established acute myocarditis, the ONX 0914 treatment group had the same reduction in cardiac output as the controls, with inflammatory responses in heart tissue being unaffected by the compound. Based on these findings and with regard to the known anti-inflammatory role of ONX 0914 in CV infection, we conclude that the efficacy of ip inhibitors for CV-triggered myocarditis in A/J mice relies on their immunomodulatory effects on the systemic inflammatory reaction.


Asunto(s)
Antiinflamatorios/farmacología , Infecciones por Coxsackievirus/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Células Mieloides/efectos de los fármacos , Miocarditis/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Animales , Células Cultivadas , Infecciones por Coxsackievirus/enzimología , Infecciones por Coxsackievirus/inmunología , Modelos Animales de Enfermedad , Enterovirus Humano B/inmunología , Enterovirus Humano B/patogenicidad , Interacciones Huésped-Patógeno , Inflamación/enzimología , Inflamación/inmunología , Inflamación/virología , Masculino , Ratones Noqueados , Células Mieloides/enzimología , Células Mieloides/inmunología , Células Mieloides/virología , Miocarditis/enzimología , Miocarditis/inmunología , Miocarditis/virología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/virología , Complejo de la Endopetidasa Proteasomal/genética , Proteolisis
19.
Acta Virol ; 65(4): 411-419, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34978843

RESUMEN

The effects of double combinations of enterovirus (EV) replication inhibitors against Coxsackieviruses B1 (neurotropic Connecticut-5 strain) and B3 (cardiotropic Woodruff and neurotropic Nancy strains) were tested in cell culture experiments. Compounds with different mechanisms of action were studied: pleconaril, guanidine.HCl, MDL-860 and oxoglaucine. A three-dimensional method was applied for determining the character of the combined effect. The study determined several synergistic double combinations: guanidine.HCL + pleconaril or MDL-860 against Coxsackievirus B1; MDL-860 + each of the other EV replication inhibitors and guanidine.HCl + pleconaril against the cardiotropic Woodruff strain of Coxsackievirus B3; MDL-860 + oxoglaucine against the neurotropic Nancy strain of Coxsackievirus B3. No increased cytotoxicity was manifested in any of the tested double combinations. Keywords: antivirals; combination activity; Coxsackieviruses.


Asunto(s)
Infecciones por Coxsackievirus , Infecciones por Enterovirus , Enterovirus , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B , Infecciones por Enterovirus/tratamiento farmacológico , Humanos
20.
J Med Virol ; 93(6): 3581-3589, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33336842

RESUMEN

Berberine (BBR), an isoquinoline alkaloid isolated from Rhizoma coptidis, is reported to possess antiviral activity. Our previous study has shown that BBR alleviates coxsackievirus B3 (CVB3) replication in HeLa cells. However, the anti-CVB3 activity of BBR is still unclear in vivo. In this study, we explored the effect of BBR on CVB3-induced viral myocarditis in mice. These results demonstrated the beneficial effect of BBR on alleviating CVB3-induced myocarditis in vivo, which sheds new light on the utility of BBR as a therapeutic strategy against CVB3-induced viral myocarditis.


Asunto(s)
Antivirales/uso terapéutico , Berberina/uso terapéutico , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B/efectos de los fármacos , Inflamación/tratamiento farmacológico , Miocarditis/tratamiento farmacológico , Replicación Viral/efectos de los fármacos , Animales , Infecciones por Coxsackievirus/complicaciones , Modelos Animales de Enfermedad , Enterovirus Humano B/fisiología , Células HeLa , Corazón/efectos de los fármacos , Corazón/virología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Extractos Vegetales/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...