Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 945
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 97(1): e0142622, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36475766

RESUMO

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


Assuntos
Cardiomiopatia Dilatada , Infecções por Coxsackievirus , Enterovirus Humano B , Humanos , Antígenos Virais , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/virologia , Infecções por Coxsackievirus/complicações , Enterovirus Humano B/metabolismo , Genômica , Miócitos Cardíacos/virologia , Peptídeo Hidrolases , Infecção Persistente , RNA Viral/genética , Proteínas Virais/metabolismo , Replicação Viral
2.
Int J Mol Sci ; 25(3)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38339075

RESUMO

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


Assuntos
Doenças Autoimunes , Infecções por Coxsackievirus , Diabetes Mellitus Tipo 1 , Enterovirus , Miocardite , Infecções Estreptocócicas , Animais , Humanos , Doenças Autoimunes/complicações , Infecções por Coxsackievirus/complicações , Autoantígenos , Streptococcus , Infecções Estreptocócicas/complicações , Antígenos de Bactérias , Receptores de Antígenos de Linfócitos T
3.
PLoS Pathog ; 17(10): e1010018, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34695168

RESUMO

Parkinson's disease (PD) is one of the most common neurodegenerative diseases. PD is pathologically characterized by the death of midbrain dopaminergic neurons and the accumulation of intracellular protein inclusions called Lewy bodies or Lewy neurites. The major component of Lewy bodies is α-synuclein (α-syn). Prion-like propagation of α-syn has emerged as a novel mechanism in the progression of PD. This mechanism has been investigated to reveal factors that initiate Lewy pathology with the aim of preventing further progression of PD. Here, we demonstrate that coxsackievirus B3 (CVB3) infection can induce α-syn-associated inclusion body formation in neurons which might act as a trigger for PD. The inclusion bodies contained clustered organelles, including damaged mitochondria with α-syn fibrils. α-Syn overexpression accelerated inclusion body formation and induced more concentric inclusion bodies. In CVB3-infected mice brains, α-syn aggregates were observed in the cell body of midbrain neurons. Additionally, α-syn overexpression favored CVB3 replication and related cytotoxicity. α-Syn transgenic mice had a low survival rate, enhanced CVB3 replication, and exhibited neuronal cell death, including that of dopaminergic neurons in the substantia nigra. These results may be attributed to distinct autophagy-related pathways engaged by CVB3 and α-syn. This study elucidated the mechanism of Lewy body formation and the pathogenesis of PD associated with CVB3 infection.


Assuntos
Infecções por Coxsackievirus/complicações , Neurônios/patologia , Transtornos Parkinsonianos/patologia , Transtornos Parkinsonianos/virologia , alfa-Sinucleína/metabolismo , Animais , Encéfalo/patologia , Enterovirus Humano B , Humanos , Corpos de Lewy/patologia , Camundongos , Camundongos Transgênicos
4.
PLoS Pathog ; 17(2): e1008992, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33556114

RESUMO

Coxsackievirus B3 (CVB3) belongs to the genus Enterovirus of the family Picornaviridae and can cause acute acinar pancreatitis in adults. However, the molecular mechanisms of pathogenesis underlying CVB3-induced acute pancreatitis have remained unclear. In this study, we discovered that CVB3 capsid protein VP1 inhibited pancreatic cell proliferation and exerted strong cytopathic effects on HPAC cells. Through yeast two-hybrid, co-immunoprecipitation, and confocal microscopy, we show that Menage a trois 1 (MAT1), a subunit of the Cdk-Activating Kinase (CAK) complex involved in cell proliferation and transcription, is a novel interaction protein with CVB3 VP1. Moreover, CVB3 VP1 inhibited MAT1 accumulation and localization, thus interfering with its interaction with CDK7. Furthermore, CVB3 VP1 could suppress CAK complex enzymic phosphorylation activity towards RNA Pol II and CDK4/6, direct substrates of CAK. VP1 also suppresses phosphorylation of retinoblastoma protein (pRb), an indirect CAK substrate, especially at phospho-pRb Ser780 and phospho-pRb Ser807/811 residues, which are associated with cell proliferation. Finally, we present evidence using deletion mutants that the C-terminal domain (VP1-D8, 768-859aa) is the minimal VP1 region required for its interaction with MAT1, and furthermore, VP1-D8 alone was sufficient to arrest cells in G1/S phase as observed during CVB3 infection. Taken together, we demonstrate that CVB3 VP1 can inhibit CAK complex assembly and activity through direct interaction with MAT1, to block MAT1-mediated CAK-CDK4/6-Rb signaling, and ultimately suppress cell proliferation in pancreatic cells. These findings substantially extend our basic understanding of CVB3-mediated pancreatitis, providing strong candidates for strategic therapeutic targeting.


Assuntos
Proteínas do Capsídeo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Infecções por Coxsackievirus/complicações , Quinases Ciclina-Dependentes/metabolismo , Enterovirus Humano B/patogenicidade , Pancreatite/patologia , Fatores de Transcrição/metabolismo , Proteínas do Capsídeo/genética , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/genética , Diferenciação Celular , Infecções por Coxsackievirus/virologia , Quinases Ciclina-Dependentes/genética , Humanos , Pancreatite/metabolismo , Pancreatite/virologia , Fosforilação , Fatores de Transcrição/genética , Quinase Ativadora de Quinase Dependente de Ciclina
5.
Int J Exp Pathol ; 103(4): 149-155, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35363404

RESUMO

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


Assuntos
Infecções por Coxsackievirus , Miocardite , Animais , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/patologia , Modelos Animais de Doenças , Enterovirus Humano B/genética , Fibrose , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C3H , Miocardite/patologia , Miocárdio/patologia
6.
Pediatr Cardiol ; 43(5): 960-968, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35022808

RESUMO

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


Assuntos
Infecções por Coxsackievirus , Cardiopatias Congênitas , Atresia Pulmonar , Anticorpos Neutralizantes , Anticorpos Antivirais , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/patologia , Enterovirus Humano B/genética , Feminino , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/epidemiologia , Humanos , Imunoglobulina G , Imunoglobulina M , Atresia Pulmonar/complicações
7.
Virologie (Montrouge) ; 26(6): 415-430, 2022.
Artigo em Francês | MEDLINE | ID: mdl-36565260

RESUMO

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


Assuntos
Infecções por Coxsackievirus , Diabetes Mellitus Tipo 1 , Infecções por Enterovirus , Enterovirus , Humanos , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/patologia , Infecções por Coxsackievirus/complicações , Enterovirus Humano B/fisiologia , Infecções por Enterovirus/complicações , Infecções por Enterovirus/epidemiologia
8.
J Med Virol ; 93(6): 3581-3589, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33336842

RESUMO

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.


Assuntos
Antivirais/uso terapêutico , Berberina/uso terapêutico , Infecções por Coxsackievirus/tratamento farmacológico , Enterovirus Humano B/efeitos dos fármacos , Inflamação/tratamento farmacológico , Miocardite/tratamento farmacológico , Replicação Viral/efeitos dos fármacos , Animais , Infecções por Coxsackievirus/complicações , Modelos Animais de Doenças , Enterovirus Humano B/fisiologia , Células HeLa , Coração/efeitos dos fármacos , Coração/virologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Extratos Vegetais/uso terapêutico
9.
BMC Infect Dis ; 21(1): 1167, 2021 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-34794380

RESUMO

BACKGROUND: Certain types of enteroviruses, including coxsackieviruses, cause encephalitis, and other neurological complications. However, these pathogens rarely cause fatal infections, especially in immunocompetent infants. In this study, we present a rare case of acute encephalopathy caused by coxsackievirus A2 (CV-A2), which progressed rapidly in a previously healthy female child. CASE PRESENTATION: In June 2013, a 26-month-old female child from Kanagawa, Japan, was found unresponsive during sleep. She was healthy until that morning. Her temperature was 37 °C at 08:00. She was feeling fine and went to the nursery that same morning. However, her condition worsened around noon. Therefore, she went home and slept at around 13:00. Surprisingly, after 2 h, her parents checked on her and found that she was lying on her back and was not breathing. Hence, she was immediately taken to a hospital by ambulance, but she was declared dead on arrival at the hospital. Subsequently, pathological autopsy and pathogenetic analysis, including multiple pathogen detection real-time PCR, were conducted to investigate the cause of death. The examination results revealed that she had an infectious respiratory disease and acute encephalopathy due to a CV-A2 infection. CONCLUSIONS: Based on our findings, we concluded that a previously healthy girl who had no immediate history of underlying medical condition were susceptible to death by acute encephalopathy due to CV-A2 infections. We proposed this conclusion because the patient's condition progressed rapidly in less than 2 h and eventually led to her death. This is the first report on an acute encephalitis-dependent death in a child due to CV-A2 infection.


Assuntos
Encefalopatias , Infecções por Coxsackievirus , Encefalite , Autopsia , Criança , Pré-Escolar , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/diagnóstico , Família , Feminino , Humanos
10.
Cell Mol Life Sci ; 77(1): 179-194, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31172216

RESUMO

It has been suggested that the persistence of coxsackieviruses-B (CV-B) in pancreatic beta cells plays a role in the pathogenesis of type 1 diabetes (T1D). Yet, immunological effectors, especially natural killer (NK) cells, are supposed to clear virus-infected cells. Therefore, an evaluation of the response of NK cells to pancreatic beta cells persistently infected with CV-B4 was conducted. A persistent CV-B4 infection was established in 1.1B4 pancreatic beta cells. Infectious particles were found in supernatants throughout the culture period. The proportion of cells containing viral protein VP1 was low (< 5%), although a large proportion of cells harbored viral RNA (around 50%), whilst cell viability was preserved. HLA class I cell surface expression was downregulated in persistently infected cultures, but HLA class I mRNA levels were unchanged in comparison with mock-infected cells. The cytolytic activities of IL-2-activated non-adherent peripheral blood mononuclear cells (PBMCs) and of NK cells were higher towards persistently infected cells than towards mock-infected cells, as assessed by an LDH release assay. Impaired cytolytic activity of IL-2-activated non-adherent PBMCs from patients with T1D towards infected beta cells was observed. In conclusion, pancreatic beta cells persistently infected with CV-B4 can be lysed by NK cells, implying that impaired cytolytic activity of these effector cells may play a role in the persistence of CV-B in the host and thus in the viral pathogenesis of T1D.


Assuntos
Infecções por Coxsackievirus/complicações , Diabetes Mellitus Tipo 1/virologia , Enterovirus Humano B/imunologia , Células Secretoras de Insulina/virologia , Células Matadoras Naturais/imunologia , Adulto , Linhagem Celular , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/imunologia , Humanos , Imunidade Celular , Células Secretoras de Insulina/imunologia , Pessoa de Meia-Idade
11.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-34445539

RESUMO

BACKGROUND: Myocarditis is an inflammatory heart disease caused by viral infections that can lead to heart failure, and occurs more often in men than women. Since animal studies have shown that myocarditis is influenced by sex hormones, we hypothesized that endocrine disruptors, which interfere with natural hormones, may play a role in the progression of the disease. The human population is exposed to the endocrine disruptor bisphenol A (BPA) from plastics, such as water bottles and plastic food containers. METHODS: Male and female adult BALB/c mice were housed in plastic versus glass caging, or exposed to BPA in drinking water versus control water. Myocarditis was induced with coxsackievirus B3 on day 0, and the endpoints were assessed on day 10 post infection. RESULTS: We found that male BALB/c mice that were exposed to plastic caging had increased myocarditis due to complement activation and elevated numbers of macrophages and neutrophils, whereas females had elevated mast cell activation and fibrosis. CONCLUSIONS: These findings show that housing mice in traditional plastic caging increases viral myocarditis in males and females, but using sex-specific immune mechanisms.


Assuntos
Infecções por Coxsackievirus/complicações , Enterovirus Humano B/patogenicidade , Abrigo para Animais/estatística & dados numéricos , Miocardite/patologia , Plásticos/efeitos adversos , Animais , Infecções por Coxsackievirus/virologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/etiologia , Miocardite/virologia , Fatores Sexuais
12.
Int J Mol Sci ; 22(18)2021 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-34576058

RESUMO

Sporadic occurrences and outbreaks of hand, foot, and mouth disease (HFMD) caused by Coxsackievirus A2 (CVA2) have frequently reported worldwide recently, which pose a great challenge to public health. Epidemiological studies have suggested that the main cause of death in critical patients is pulmonary edema. However, the pathogenesis of this underlying comorbidity remains unclear. In this study, we utilized the 5-day-old BALB/c mouse model of lethal CVA2 infection to evaluate lung damage. We found that the permeability of lung microvascular was significantly increased after CVA2 infection. We also observed the direct infection and apoptosis of lung endothelial cells as well as the destruction of tight junctions between endothelial cells. CVA2 infection led to the degradation of tight junction proteins (e.g., ZO-1, claudin-5, and occludin). The gene transcription levels of von Willebrand factor (vWF), endothelin (ET), thrombomodulin (THBD), granular membrane protein 140 (GMP140), and intercellular cell adhesion molecule-1 (ICAM-1) related to endothelial dysfunction were all significantly increased. Additionally, CVA2 infection induced the increased expression of inflammatory cytokines (IL-6, IL-1ß, and MCP-1) and the activation of p38 mitogen-activated protein kinase (MAPK). In conclusion, the disruption of the endothelial barrier contributes to acute lung injury induced by CVA2 infection; targeting p38-MAPK signaling may provide a therapeutic approach for pulmonary edema in critical infections of HFMD.


Assuntos
Lesão Pulmonar Aguda/genética , Infecções por Coxsackievirus/genética , Doença de Mão, Pé e Boca/genética , Edema Pulmonar/genética , Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/virologia , Animais , Apoptose/genética , Claudina-5/genética , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/patologia , Infecções por Coxsackievirus/virologia , Citocinas/genética , Modelos Animais de Doenças , Células Endoteliais/patologia , Células Endoteliais/virologia , Doença de Mão, Pé e Boca/complicações , Doença de Mão, Pé e Boca/patologia , Doença de Mão, Pé e Boca/virologia , Humanos , Camundongos , Ocludina/genética , Edema Pulmonar/complicações , Edema Pulmonar/patologia , Edema Pulmonar/virologia , Junções Íntimas/genética , Junções Íntimas/patologia , Proteína da Zônula de Oclusão-1/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
13.
PLoS Pathog ; 14(1): e1006872, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29360865

RESUMO

Cathepsin B (CatB) is a cysteine proteolytic enzyme widely expressed in various cells and mainly located in the lysosomes. It contributes to the pathogenesis and development of many diseases. However, the role of CatB in viral myocarditis (VMC) has never been elucidated. Here we generated the VMC model by intraperitoneal injection of coxsackievirus B3 (CVB3) into mice. At day 7 and day 28, we found CatB was significantly activated in hearts from VMC mice. Compared with the wild-type mice receiving equal amount of CVB3, genetic ablation of CatB (Ctsb-/-) significantly improved survival, reduced inflammatory cell infiltration, decreased serum level of cardiac troponin I, and ameliorated cardiac dysfunction, without altering virus titers in hearts. Conversely, genetic deletion of cystatin C (Cstc-/-), which markedly enhanced CatB levels in hearts, distinctly increased the severity of VMC. Furthermore, compared with the control, we found the inflammasome was activated in the hearts of wild-type mice with VMC, which was attenuated in the hearts of Ctsb-/- mice but was further enhanced in Cstc-/- mice. Consistently, the inflammasome-initiated pyroptosis was reduced in Ctsb-/- mice hearts and further increased in Cstc-/- mice. These results suggest that CatB aggravates CVB3-induced VMC probably through activating the inflammasome and promoting pyroptosis. This finding might provide a novel strategy for VMC treatment.


Assuntos
Catepsina B/fisiologia , Infecções por Coxsackievirus/complicações , Enterovirus Humano B/fisiologia , Inflamassomos/metabolismo , Miocardite/virologia , Piroptose/fisiologia , Animais , Caspase 1/metabolismo , Catepsina B/genética , Infecções por Coxsackievirus/patologia , Modelos Animais de Doenças , Progressão da Doença , Ativação Enzimática , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/imunologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/virologia
14.
PLoS Pathog ; 14(8): e1007235, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30075026

RESUMO

During Coxsackievirus B3 (CVB3) infection hepatitis is a potentially life threatening complication, particularly in newborns. Studies with type I interferon (IFN-I) receptor (IFNAR)-deficient mice revealed a key role of the IFN-I axis in the protection against CVB3 infection, whereas the source of IFN-I and cell types that have to be IFNAR triggered in order to promote survival are still unknown. We found that CVB3 infected IFN-ß reporter mice showed effective reporter induction, especially in hepatocytes and only to a minor extent in liver-resident macrophages. Accordingly, upon in vitro CVB3 infection of primary hepatocytes from murine or human origin abundant IFN-ß responses were induced. To identify sites of IFNAR-triggering we performed experiments with Mx reporter mice, which upon CVB3 infection showed massive luciferase induction in the liver. Immunohistological studies revealed that during CVB3 infection MX1 expression of hepatocytes was induced primarily by IFNAR-, and not by IFN-III receptor (IFNLR)-triggering. CVB3 infection studies with primary human hepatocytes, in which either the IFN-I or the IFN-III axis was inhibited, also indicated that primarily IFNAR-, and to a lesser extent IFNLR-triggering was needed for ISG induction. Interestingly, CVB3 infected mice with a hepatocyte-specific IFNAR ablation showed severe liver cell necrosis and ubiquitous viral dissemination that resulted in lethal disease, as similarly detected in classical IFNAR-/- mice. In conclusion, we found that during CVB3 infection hepatocytes are major IFN-I producers and that the liver is also the organ that shows strong IFNAR-triggering. Importantly, hepatocytes need to be IFNAR-triggered in order to prevent virus dissemination and to assure survival. These data are compatible with the hypothesis that during CVB3 infection hepatocytes serve as important IFN-I producers and sensors not only in the murine, but also in the human system.


Assuntos
Infecções por Coxsackievirus , Enterovirus Humano B/imunologia , Hepatócitos/metabolismo , Interferon beta/genética , Fígado/patologia , Receptor de Interferon alfa e beta/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , Enterovirus Humano B/crescimento & desenvolvimento , Humanos , Interferon beta/metabolismo , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/virologia , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Vero , Carga Viral/genética , Carga Viral/imunologia
15.
Cell Biol Int ; 44(2): 488-498, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31631456

RESUMO

Cardiomyocyte apoptosis contributes to the development of coxsackievirus B3 (CVB3)-induced myocarditis, but the mechanism for the apoptosis by CVB3 infection remains unclear. Here, we showed that CVB3-induced endoplasmic reticulum (ER) stress response and apoptosis in cultured H9c2 cardiomyocytes. We found that Ca2+ -calmodulin-dependent kinase II (CaMKII) was activated by ER stress-dependent intracellular Ca2+ overload in the CVB3-infected H9c2 cardiomyocytes. Treatment with an inhibitor of ER stress, 4-phenylbutyric acid (4-PBA), attenuated intracellular Ca2+ accumulation indirectly and reduced CaMKII activity. Inhibition of CaMKII with pharmacological inhibitor (KN-93) or short hairpin RNA reduced CVB3-induced H9c2 apoptosis and repressed cytochrome c release from mitochondria to cytoplasm; whereas overexpression of the activated mutant of CaMKII (CaMKII-T287D) enhanced CVB3-induced H9c2 apoptosis and mitochondrial cytochrome c release, which could be alleviated by blocking of mitochondrial Ca2+ uniporter or mitochondrial permeability transition pore. Further in vivo investigation revealed that blocking of CaMKII with KN-93 prevented cardiomyocytes apoptosis and improved cardiac contractile function in CVB3-infected mouse heart. Collectively, these findings provide a novel evidence that CaMKII plays a vital role in the promotion of CVB3-induced cardiomyocyte apoptosis, which links ER stress and mitochondrial Ca2+ uptake.


Assuntos
Apoptose , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Infecções por Coxsackievirus/complicações , Estresse do Retículo Endoplasmático , Miócitos Cardíacos/patologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Infecções por Coxsackievirus/virologia , Enterovirus Humano B/isolamento & purificação , Ativação Enzimática , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/microbiologia , Transdução de Sinais
16.
J Immunol ; 200(1): 61-70, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158420

RESUMO

Coxsackievirus B infections are suspected environmental triggers of type 1 diabetes (T1D) and macrophage antiviral responses may provide a link to virus-induced T1D. We previously demonstrated an important role for NADPH oxidase (NOX)-derived superoxide production during T1D pathogenesis, as NOX-deficient NOD mice (NOD.Ncf1m1J ) were protected against T1D due, in part, to impaired proinflammatory TLR signaling in NOD.Ncf1m1J macrophages. Therefore, we hypothesized that loss of NOX-derived superoxide would dampen diabetogenic antiviral macrophage responses and protect from virus-induced diabetes. Upon infection with a suspected diabetogenic virus, Coxsackievirus B3 (CB3), NOD.Ncf1m1J mice remained resistant to virus-induced autoimmune diabetes. A concomitant decrease in circulating inflammatory chemokines, blunted antiviral gene signature within the pancreas, and reduced proinflammatory M1 macrophage responses were observed. Importantly, exogenous superoxide addition to CB3-infected NOD.Ncf1m1J bone marrow-derived macrophages rescued the inflammatory antiviral M1 macrophage response, revealing reduction-oxidation-dependent mechanisms of signal transducer and activator of transcription 1 signaling and dsRNA viral sensors in macrophages. We report that superoxide production following CB3 infection may exacerbate pancreatic ß cell destruction in T1D by influencing proinflammatory M1 macrophage responses, and mechanistically linking oxidative stress, inflammation, and diabetogenic virus infections.


Assuntos
Infecções por Coxsackievirus/imunologia , Diabetes Mellitus Tipo 1/imunologia , Enterovirus/imunologia , Células Secretoras de Insulina/imunologia , Macrófagos/imunologia , NADPH Oxidases/metabolismo , Superóxidos/metabolismo , Animais , Apoptose , Células Cultivadas , Quimiocinas/metabolismo , Infecções por Coxsackievirus/complicações , Diabetes Mellitus Tipo 1/etiologia , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos NOD , NADPH Oxidases/genética , Estresse Oxidativo , RNA Viral/imunologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
17.
J Infect Chemother ; 26(5): 516-519, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31983616

RESUMO

BACKGROUND: Fukuyama congenital muscular dystrophy (FCMD), which is characterized by generalized muscle weakness, hypotonia, and motor delay during early infancy, gradually progresses with advanced age. Although acute rhabdomyolysis following infection in patients with FCMD has occasionally been reported, no studies have investigated rhabdomyolysis following viral infection in FCMD patients during early infancy. CASE REPORT: We report the case of a 50-day-old girl with no apparent symptoms of muscular dystrophy who developed severe acute rhabdomyolysis caused by viral infection, resulting in quadriplegia and respiratory failure therefore requiring mechanical ventilation. Brain magnetic resonance imaging incidentally showed the typical characteristics of FCMD, and FCMD was confirmed by genetic analysis, which revealed a 3-kb retrotransposon insertion in one allele of the fukutin gene and a deep intronic splicing variant in intron 5 in another allele. The virus etiology was confirmed to be Coxsackie A4. CONCLUSION: We report a severe case of acute rhabdomyolysis with the earliest onset of symptoms due to the Coxsackie A4 virus in a patient with FCMD. The present findings indicate that physicians should consider FCMD with viral infection a differential diagnosis if the patient presents with acute rhabdomyolysis following a fever.


Assuntos
Infecções por Coxsackievirus/virologia , Enterovirus Humano A/patogenicidade , Rabdomiólise/virologia , Síndrome de Walker-Warburg/complicações , Doença Aguda , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/diagnóstico , Diagnóstico Diferencial , Enterovirus Humano A/genética , Enterovirus Humano A/isolamento & purificação , Feminino , Humanos , Lactente , Imageamento por Ressonância Magnética , Proteínas de Membrana/genética , Reação em Cadeia da Polimerase , Quadriplegia/etiologia , RNA Viral , Respiração Artificial , Insuficiência Respiratória/etiologia , Rabdomiólise/complicações , Rabdomiólise/diagnóstico , Índice de Gravidade de Doença , Síndrome de Walker-Warburg/diagnóstico , Síndrome de Walker-Warburg/virologia
18.
Int J Mol Sci ; 21(9)2020 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-32344926

RESUMO

AIMS: Coxsackievirus B3 (CVB3) is known to be an important cause of myocarditis and dilated cardiomyopathy. Enterovirus-2C (E2C) is a viral RNA helicase. It inhibits host protein synthesis. Based on these facts, we hypothesize that the inhibition of 2C may suppress virus replication and prevent enterovirus-mediated cardiomyopathy. METHODS AND RESULTS: We generated a chemically modified enterovirus-2C inhibitor (E2CI). From the in vitro assay, E2CI was showed strong antiviral effects. For in vivo testing, mice were treated with E2CI intraperitoneally injected daily for three consecutive days at a dose of 8mg/kg per day, after CVB3 post-infection (p.i) (CVB3 + E2CI, n = 33). For the infected controls (CVB3 only, n = 35), mice were injected with PBS (phosphate buffered saline) in a DBA/2 strain to establish chronic myocarditis. The four-week survival rate of E2CI-treated mice was significantly higher than that of controls (92% vs. 71%; p < 0.05). Virus titers and myocardial damage were significantly reduced in the E2CI treated group. In addition, echocardiography indicated that E2CI administration dramatically maintained mouse heart function compared to control at day 28 p.i chronic stage (LVIDD, 3.1 ± 0.08 vs. 3.9 ± 0.09, p < 0.01; LVDS, 2.0 ± 0.07 vs. 2.5 ± 0.07, p < 0.001; FS, 34.8 ± 1.6% vs. 28.5 ± 1.5%; EF, 67. 9 ± 2.9% vs. 54.7 ± 4.7%, p < 0.05; CVB3 + E2CI, n = 6 vs. CVB3, n = 4). Moreover, E2CI is effectively worked in human iPS (induced pluripotent stem cell) derived cardiomyocytes. CONCLUSION: Enterovirus-2C inhibitor (E2CI) was significantly reduced viral replication, chronic myocardium damage, and CVB3-induced mortality in DBA/2 mice. These results suggested that E2CI is a novel therapeutic agent for the treatment of enterovirus-mediated diseases.


Assuntos
Antivirais/farmacologia , Infecções por Coxsackievirus/tratamento farmacológico , Enterovirus Humano B/enzimologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miocardite/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , RNA Helicases/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Animais , Antivirais/síntese química , Antivirais/uso terapêutico , Cardiomiopatia Dilatada/etiologia , Cardiomiopatia Dilatada/prevenção & controle , Doença Crônica , Infecções por Coxsackievirus/complicações , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/fisiologia , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/virologia , Luciferases de Renilla/análise , Masculino , Camundongos , Camundongos Endogâmicos DBA , Miocardite/etiologia , Miocardite/virologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/virologia , Oxidiazóis/farmacologia , Oxidiazóis/uso terapêutico , Oxazóis/farmacologia , Oxazóis/uso terapêutico , Proteínas Recombinantes de Fusão/metabolismo , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/prevenção & controle , Replicação Viral/efeitos dos fármacos
19.
Am J Pathol ; 188(12): 2853-2862, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30273599

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that primarily affects motor neurons in the cerebral cortex, brainstem, and spinal cord, leading to progressive paralysis and eventual death. Approximately 95% of all ALS cases are sporadic without known causes. Enteroviruses have been suspected to play a role in ALS because of their ability to target motor neurons and to cause muscle weakness and paralysis. In vitro enteroviral infection results in cytoplasmic aggregation and cleavage of transactive response DNA binding protein-43, a pathologic hallmark of ALS. However, whether enteroviral infection can induce ALS-like pathologies in vivo remains to be characterized. In this study, neonatal BALB/C mice were intracranially inoculated with either a recombinant coxsackievirus B3 expressing enhanced green fluorescent protein or mock-infected for 2, 5, 10, 30, and 90 days. Histologic and immunohistochemical analysis of brain tissues demonstrated sustained inflammation (microglia and astrogliosis) and lesions in multiple regions of the brain (hippocampus, cerebral cortex, striatum, olfactory bulb, and putamen) in parallel with virus detection as early as 2 days for up to 90 days after infection. Most notably, ALS-like pathologies, including cytoplasmic mislocalization of transactive response DNA binding protein-43, p62-, and ubiquitin-positive inclusions, were observed in the areas of infection. These data provide the first pathologic evidence to support a possible link between enteroviral infection and ALS.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Encéfalo/imunologia , Infecções por Coxsackievirus/complicações , Citoplasma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Enterovirus Humano B/patogenicidade , Esclerose Lateral Amiotrófica/etiologia , Esclerose Lateral Amiotrófica/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/microbiologia , Células Cultivadas , Infecções por Coxsackievirus/virologia , Camundongos , Camundongos Endogâmicos BALB C , Transporte Proteico
20.
BMC Infect Dis ; 19(1): 285, 2019 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-30917800

RESUMO

BACKGROUND: Hand, foot and mouth disease (HFMD) is a transmissible infectious disease caused by human enteroviruses (EV). Here, we described features of children with severe HFMD caused by EV-A71 or coxsackievirus A16 (CV-A16) in Shanghai, China. METHODS: Severe EV-A71 or CV-A16 caused HFMD children admitted to the Xinhua Hospital from January 2014 and December 2016, were recruited retrospectively to the study. Symptoms and findings at the time of hospitalization, laboratory tests, treatments, length of stay and residual findings at discharge were systematically recorded and analyzed. RESULTS: Of 19,995 children visited clinic service with probable HFMD, 574 children (2.87%) were admitted, 234 children (40.76%) were confirmed with EV-A71 (90/574) or CV-A16 (144/574) disease. Most (91.02%) of the patients were under 5 years. Initial clinical symptoms of EV-A71 and CV-A16 cases were: fever > 39 °C in 81 (90%) and 119 (82.63%), vomiting in 31 (34.44%) and 28 (19.44%), myoclonic twitching in 19 (21.11%) and 11(7.64%), startle in 21 (23.33%) and 20 (13.69%), respectively. Serum levels of interleukin-1ß (IL-1ß), IL-2, IL-6, IL-8, interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α) were significantly upregulated in severe HFMD subjects. Forty-seven children (20.08%) treated with intravenous gamma globulin (IVIG) showed decreased duration of illness episodes. All children were discharged without complications. CONCLUSIONS: EV-A71 and CV-A16 accounted 40.76% of admitted HFMD during 2014 to 2016 in Xinhua Hospital. IVIG appeared to be beneficial in shortening the duration of illness episodes of severe HFMD.


Assuntos
Infecções por Coxsackievirus/epidemiologia , Infecções por Coxsackievirus/terapia , Infecções por Enterovirus/epidemiologia , Infecções por Enterovirus/terapia , Enterovirus , Doença de Mão, Pé e Boca/epidemiologia , Doença de Mão, Pé e Boca/terapia , Adolescente , Criança , Pré-Escolar , China/epidemiologia , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/diagnóstico , Enterovirus/fisiologia , Enterovirus Humano A/fisiologia , Infecções por Enterovirus/complicações , Infecções por Enterovirus/diagnóstico , Feminino , Doença de Mão, Pé e Boca/diagnóstico , Doença de Mão, Pé e Boca/microbiologia , Hospitalização/estatística & dados numéricos , Humanos , Lactente , Recém-Nascido , Masculino , Estudos Retrospectivos , Índice de Gravidade de Doença , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA