Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 695
Filtrar
1.
Int J Mol Sci ; 24(6)2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36982385

RESUMEN

The coxsackievirus and adenovirus receptor (CAR) is very well known as an epithelial tight junction and cardiac intercalated disc protein; it mediates attachment and infection via the coxsackievirus B3 (CVB3) and type 5 adenovirus. Macrophages play important roles in early immunity during viral infections. However, the role of CAR in macrophages is not well studied in relation to CVB3 infection. In this study, the function of CAR was observed in the Raw264.7 mouse macrophage cell line. CAR expression was stimulated by treatment with lipopolysaccharide (LPS) and tumor necrosis factor-α (TNF-α). In thioglycollate-induced peritonitis, the peritoneal macrophage was activated and CAR expression was increased. The macrophage-specific CAR conditional knockout mice (KO) were generated from lysozyme Cre mice. The expression of inflammatory cytokine (IL-1ß and TNF-α) was attenuated in the KO mice's peritoneal macrophage after LPS treatment. In addition, the virus was not replicated in CAR-deleted macrophages. The organ virus replication was not significantly different in both wild-type (WT) and KO mice at days three and seven post-infection (p.i). However, the inflammatory M1 polarity genes (IL-1ß, IL-6, TNF-α and MCP-1) were significantly increased, with increased rates of myocarditis in the heart of KO mice compared to those of WT mice. In contrast, type1 interferon (IFN-α and ß) was significantly decreased in the heart of KO mice. Serum chemokine CXCL-11 was increased in the KO mice at day three p.i. compared to the WT mice. The attenuation of IFN-α and ß in macrophage CAR deletion induced higher levels of CXCL-11 and more increased CD4 and CD8 T cells in KO mice hearts compared to those of WT mice at day seven p.i. These results demonstrate that macrophage-specific CAR deletion increased the macrophage M1 polarity and myocarditis in CVB3 infection. In addition, chemokine CXCL-11 expression was increased, and stimulated CD4 and CD8 T cell activity. Macrophage CAR may be important for the regulation of innate-immunity-induced local inflammation in CVB3 infection.


Asunto(s)
Infecciones por Coxsackievirus , Miocarditis , Ratones , Animales , Miocarditis/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Lipopolisacáridos/metabolismo , Enterovirus Humano B/metabolismo , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/patología , Macrófagos/metabolismo , Ratones Noqueados
2.
Int J Exp Pathol ; 103(4): 149-155, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35363404

RESUMEN

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.


Asunto(s)
Infecciones por Coxsackievirus , Miocarditis , Animales , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/patología , Modelos Animales de Enfermedad , Enterovirus Humano B/genética , Fibrosis , Inflamación/patología , Ratones , Ratones Endogámicos C3H , Miocarditis/patología , Miocardio/patología
3.
Inflamm Res ; 71(12): 1559-1576, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36301340

RESUMEN

BACKGROUND: Myocardial inflammation and apoptosis are key processes in coxsackievirus B3 (CVB3)-induced acute viral myocarditis (AVMC). Accumulating evidence reveals the essential roles of long noncoding RNAs (lncRNAs) in the pathogenesis of AVMC. Here, we aimed to evaluate the biological functions of a novel lncRNA guanylate-binding protein 9 (lncGBP9) in AVMC progression and further explore its underlying mechanisms. METHODS: Initially, mouse models of AVMC were constructed by CVB3 infection. The expression and localization of lncGBP9 in heart tissues were analyzed using RT-qPCR and FISH. Adeno-associated virus serotype 9 (AAV9)-mediated lncGBP9 knockdown was then employed to clarify its roles in survival, cardiac function, and myocardial inflammation and apoptosis. Moreover, the mRNA and protein levels of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1ß) were detected by RT-qPCR and ELISA, and the regulation of lncGBP9 knockdown on the NF-κB signaling pathway was investigated by Western blotting. Using an in vitro model of HL-1 cardiomyocytes exposed to CVB3 infection, the effects of lncGBP9 knockdown on cell viability, inflammation, and apoptosis were further evaluated in vitro. RESULTS: Increased lncGBP9 expression was detected in the heart tissues of AVMC mice and CVB3-infected HL-1 cells, and was mainly located in the cytoplasm. Knockdown of lncGBP9 remarkably alleviated the severity of AVMC in CVB3-infected mice, as verified by improved cardiac function, and reduced myocardial inflammation and apoptosis. Additionally, lncGBP9 knockdown suppressed the NF-κB signaling pathway and consequently reduced productions of pro-inflammatory cytokines in vivo. In vitro functional assays further confirmed that lncGBP9 knockdown promoted cell viability, inhibited cell apoptosis, and reduced pro-inflammatory cytokines release in CVB3-infected HL-1 cells through suppressing NF-κB activation. CONCLUSIONS: Collectively, lncGBP9 knockdown exerts anti-inflammatory and anti-apoptotic effects in CVB3-induced AVMC, which may be mediated in part via NF-κB signaling pathway. These findings highlight lncGBP9 as an attractive target for therapeutic interventions.


Asunto(s)
Infecciones por Coxsackievirus , Miocarditis , Ratones , Animales , Miocarditis/genética , FN-kappa B/metabolismo , Enterovirus Humano B/metabolismo , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Transducción de Señal , Inflamación/metabolismo , Apoptosis , Citocinas/metabolismo , Ratones Endogámicos BALB C
4.
Cell Biol Toxicol ; 38(3): 487-504, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34365571

RESUMEN

Cardiomyocyte apoptosis is critical for the development of viral myocarditis (VMC), which is one of the leading causes of cardiac sudden death in young adults. Our previous studies have demonstrated that elevated calpain activity is involved in the pathogenesis of VMC. This study aimed to further explore the underlying mechanisms. Neonatal rat cardiomyocytes (NRCMs) and transgenic mice overexpressing calpastatin were infected with coxsackievirus B3 (CVB3) to establish a VMC model. Apoptosis was detected with flow cytometry, TUNEL staining, and western blotting. Cardiac function was measured using echocardiography. Mitochondrial function was measured using ATP assays, JC-1, and MitoSOX. Mitochondrial morphology was observed using MitoTracker staining and transmission electron microscopy. Colocalization of dynamin-related protein 1 (Drp-1) in mitochondria was examined using immunofluorescence. Phosphorylation levels of Drp-1 at Ser637 site were determined using western blotting analysis. We found that CVB3 infection impaired mitochondrial function as evidenced by increased mitochondrial ROS production, decreased ATP production and mitochondrial membrane potential, induced myocardial apoptosis and damage, and decreased myocardial function. These effects of CVB3 infection were attenuated by inhibition of calpain both by PD150606 treatment and calpastatin overexpression. Furthermore, CVB3-induced mitochondrial dysfunction was associated with the accumulation of Drp-1 in the outer membrane of mitochondria and subsequent increase in mitochondrial fission. Mechanistically, calpain cleaved and activated calcineurin A, which dephosphorylated Drp-1 at Ser637 site and promoted its accumulation in the mitochondria, leading to mitochondrial fission and dysfunction. In summary, calpain inhibition attenuated CVB3-induced myocarditis by reducing mitochondrial fission, thereby inhibiting cardiomyocyte apoptosis. Calpain is activated by CVB3 infection. Activated calpain cleaves calcineurin A and converts it to active form which could dephosphorylate Drp-1 at Ser637 site. Then, the active Drp-1 translocates from the cytoplasm to mitochondria and triggers excessive mitochondrial fission. Eventually, the balance of mitochondrial dynamics is broken, and apoptosis occurs.


Asunto(s)
Infecciones por Coxsackievirus , Miocarditis , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Calcineurina/metabolismo , Calcineurina/farmacología , Calpaína/metabolismo , Calpaína/farmacología , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Ratones , Dinámicas Mitocondriales , Miocarditis/metabolismo , Miocarditis/patología , Miocitos Cardíacos , Ratas
5.
Rev Med Virol ; 31(4): e2191, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33159700

RESUMEN

Type B coxsackieviruses (CV-B) frequently infect the central nervous system (CNS) causing neurological diseases notably meningitis and encephalitis. These infections occur principally among newborns and children. Epidemiological studies of patients with nervous system disorders demonstrate the presence of infectious virus, its components, or anti-CV-B antibodies. Some experimental studies conducted in vitro and in vivo support the potential association between CV-B and idiopathic neurodegenerative diseases such as amyotrophic lateral sclerosis and psychiatric illness such as schizophrenia. However, mechanisms explaining how CV-B infections may contribute to the genesis of CNS disorders remain unclear. The proposed mechanisms focus on the immune response following the viral infection as a contributor to pathogenesis. This review describes these epidemiological and experimental studies, the modes of transmission of CV-B with an emphasis on congenital transmission, the routes used by CV-B to reach the brain parenchyma, and plausible mechanisms by which CV-B may induce CNS diseases, with a focus on potential immunopathogenesis.


Asunto(s)
Encéfalo/virología , Enfermedades del Sistema Nervioso Central/virología , Infecciones del Sistema Nervioso Central/virología , Infecciones por Coxsackievirus/diagnóstico , Enterovirus Humano B/aislamiento & purificación , Anticuerpos Antivirales , Enfermedades del Sistema Nervioso Central/epidemiología , Enfermedades del Sistema Nervioso Central/etiología , Niño , Infecciones por Coxsackievirus/patología , Enterovirus Humano B/inmunología , Humanos , Recién Nacido
6.
Pediatr Cardiol ; 43(5): 960-968, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35022808

RESUMEN

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.


Asunto(s)
Infecciones por Coxsackievirus , Cardiopatías Congénitas , Atresia Pulmonar , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/patología , Enterovirus Humano B/genética , Femenino , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/epidemiología , Humanos , Inmunoglobulina G , Inmunoglobulina M , Atresia Pulmonar/complicaciones
7.
Molecules ; 27(18)2022 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-36144851

RESUMEN

Viral myocarditis (VMC), which is defined as inflammation of the myocardium with consequent myocardial injury, may develop chronic disease eventually leading to dilated cardiomyopathy (DCM). Molecular mechanisms underlying the progression from acute VMC (aVMC), to chronic VMC (cVMC) and finally to DCM, are still unclear. Here, we established mouse models of VMC and DCM with Coxsackievirus B3 infection and conducted NMR-based metabolomic analysis of aqueous metabolites extracted from cardiac tissues of three histologically classified groups including aVMC, cVMC and DCM. We showed that these three pathological groups were metabolically distinct from their normal counterparts and identified three impaired metabolic pathways shared by these pathological groups relative to normal controls, including nicotinate and nicotinamide metabolism; alanine, aspartate and glutamate metabolism; and D-glutamine and D-glutamate metabolism. We also identified two extra impaired metabolic pathways in the aVMC group, including glycine, serine and threonine metabolism; and taurine and hypotaurine metabolism Furthermore, we identified potential cardiac biomarkers for metabolically distinguishing these three pathological stages from normal controls. Our results indicate that the metabolomic analysis of cardiac tissues can provide valuable insights into the molecular mechanisms underlying the progression from acute VMC to DCM.


Asunto(s)
Cardiomiopatía Dilatada , Infecciones por Coxsackievirus , Miocarditis , Niacina , Alanina , Animales , Ácido Aspártico , Biomarcadores , Cardiomiopatía Dilatada/metabolismo , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Enterovirus Humano B , Ácido Glutámico , Glutamina , Glicina , Ratones , Ratones Endogámicos BALB C , Miocarditis/metabolismo , Miocarditis/patología , Niacinamida , Serina , Taurina , Treonina
8.
Cell Physiol Biochem ; 55(6): 679-703, 2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34791861

RESUMEN

Viral diseases are a major threat to modern society and the global health system. It is therefore of utter relevance to understand the way viruses affect the host as a basis to find new treatment solutions. The understanding of viral myocarditis (VMC) is incomplete and effective treatment options are lacking. This review will discuss the mechanism, effects, and treatment options of the most frequent myocarditis-causing viruses namely enteroviruses such as Coxsackievirus B3 (CVB3) and Parvovirus B19 (PVB19) on the human heart. Thereby, we focus on: 1. Viral entry: CVB3 use Coxsackievirus-Adenovirus-Receptor (CAR) and Decay Accelerating Factor (DAF) to enter cardiac myocytes while PVB19 use the receptor globoside (Gb4) to enter cardiac endothelial cells. 2. Immune system responses: The innate immune system mediated by activated cardiac toll-like receptors (TLRs) worsen inflammation in CVB3-infected mouse hearts. Different types of cells of the adaptive immune system are recruited to the site of inflammation that have either protective or adverse effects during VMC. 3. Autophagy: CVB3 evades autophagosomal degradation and misuses the autophasomal pathway for viral replication and release. 4. Viral replication sites: CVB3 promotes the formation of double membrane vesicles (DMVs), which it uses as replication sites. PVB19 uses the host cell nucleus as the replication site and uses the host cell DNA replication system. 5. Cell cycle manipulation: CVB3 attenuates the cell cycle at the G1/S phase, which promotes viral transcription and replication. PVB19 exerts cell cycle arrest in the S phase using its viral endonuclease activity. 6. Regulation of apoptosis: Enteroviruses prevent apoptosis during early stages of infection and promote cell death during later stages by using the viral proteases 2A and 3C, and viroporin 2B. PVB19 promotes apoptosis using the non-structural proteins NS1 and the 11 kDa protein. 7. Energy metabolism: Dysregulation of respiratory chain complex expression, activity and ROS production may be altered in CVB3- and PVB19-mediated myocarditis. 8. Ion channel modulation: CVB3-expression was indicated to alter calcium and potassium currents in Xenopus laevis oocytes and rodent cardiomyocytes. The phospholipase 2-like activity of PVB19 may alter several calcium, potassium and sodium channels. By understanding the general pathophysiological mechanisms of well-studied myocarditis-linked viruses, we might be provided with a guideline to handle other less-studied human viruses.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Interacciones Huésped-Patógeno/inmunología , Miocarditis , Infecciones por Parvoviridae/inmunología , Parvovirus B19 Humano/fisiología , Replicación Viral , Infecciones por Coxsackievirus/patología , Humanos , Miocarditis/inmunología , Miocarditis/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología , Infecciones por Parvoviridae/patología , Receptores Virales/inmunología
9.
Int J Mol Sci ; 22(18)2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34576058

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda/genética , Infecciones por Coxsackievirus/genética , Enfermedad de Boca, Mano y Pie/genética , Edema Pulmonar/genética , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/virología , Animales , Apoptosis/genética , Claudina-5/genética , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , Citocinas/genética , Modelos Animales de Enfermedad , Células Endoteliales/patología , Células Endoteliales/virología , Enfermedad de Boca, Mano y Pie/complicaciones , Enfermedad de Boca, Mano y Pie/patología , Enfermedad de Boca, Mano y Pie/virología , Humanos , Ratones , Ocludina/genética , Edema Pulmonar/complicaciones , Edema Pulmonar/patología , Edema Pulmonar/virología , Uniones Estrechas/genética , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
10.
PLoS Pathog ; 14(1): e1006872, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29360865

RESUMEN

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.


Asunto(s)
Catepsina B/fisiología , Infecciones por Coxsackievirus/complicaciones , Enterovirus Humano B/fisiología , Inflamasomas/metabolismo , Miocarditis/virología , Piroptosis/fisiología , Animales , Caspasa 1/metabolismo , Catepsina B/genética , Infecciones por Coxsackievirus/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Activación Enzimática , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/inmunología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología
11.
Microb Pathog ; 140: 103965, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31904449

RESUMEN

Coxsackie B viruses (CV-B) are associated with several central nervous system (CNS) disorders. These viruses are predominantly transmitted by fecal-oral route but vertical transmission can also occur. This work attempted to study the immune response ensuing vertical transmission of CV-B to the brain, and its eventual implementation in the brain pathogenesis. To this end, pregnant Swiss albino mice were inoculated with CV-B4 E2 or with sterile medium for control animals. At different ages after birth, brains were collected and analyzed for virus infection, histopathological changes and immune response. Infectious particles were detected in offspring's brain which demonstrates vertical transmission of the virus. This infection is persistent since the long lasting detection of viral RNA in offspring's brain. Some pathological signs including meningitis, edema and accumulation of inflammatory cells within and surrounding the inflammatory areas were observed. Immunoflorescence staining unveiled the presence of T lymphocytes and microgliosis in the sites of lesion for a long period after birth. Multiplex cytokines measurement upon supernatants of in vitro mixed brain cells and extracted mononuclear cells from offspring's brain has demonstrated an elevated secretion of the pro-inflammatory cytokines TNFα, IL-6 and IFNα and the chemokines RANTES and MCP-1. Hence, vertical transmission of CV-B4 and its persistence within offspring's brain can lead to pathological features linked to increased and sustained immune response.


Asunto(s)
Encéfalo/patología , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/transmisión , Enterovirus Humano B/fisiología , Complicaciones Infecciosas del Embarazo/inmunología , Animales , Encéfalo/inmunología , Encéfalo/virología , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , Citocinas/genética , Citocinas/inmunología , Enterovirus Humano B/genética , Femenino , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Interferón-alfa/genética , Interferón-alfa/inmunología , Masculino , Ratones , Embarazo , Complicaciones Infecciosas del Embarazo/genética , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/virología , Linfocitos T/inmunología
12.
Heart Vessels ; 35(8): 1171-1179, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32328712

RESUMEN

Viral myocarditis (VMC) is a widely studied but poorly understood inflammatory cardiomyopathy which mainly affects children and young adults and results in adverse outcomes. Cardiomyocyte apoptosis was reported important in the progress of coxsackievirus B3 (CVB3)-induced VMC and the blocking of this process may contribute to the therapeutic effect towards VMC. Therefore, this study was designed to explore whether survivin, one of the strongest antiapoptotic proteins, can protect cardiomyocytes from apoptosis in VMC and to discover its related mechanisms. Here, the cultured neonatal mouse cardiomyocytes (NMCs) were exposed to CVB3 to establish the cell model of VMC and the results of Western Blot showed that the protein expression of survivin in CVB3-infected NMCs varied at different post-infection time. Lentivirus was next used to examine the function of survivin in CVB3-infected NMCs. TUNEL assay demonstrated that the overexpression of survivin interrupted CVB3-induced apoptosis. It was next examined whether caspase-3 and -9 were involved in the antiapoptotic pathway initiated by survivin via Western Blot. The results showed a reverse relationship between the protein expression of survivin and that of cleaved caspase-3 and cleaved caspase-9, suggesting that survivin may attenuate apoptosis through restraining the activity of caspase-3 and -9. Moreover, the supernatant fluid of cultured NMCs was extracted to detect the quantitation of released lactate dehydrogenase (LDH) and a sharp decrease was discovered in the survivin-overexpressed group compared to the CVB3-infected group, indicating a protective role of survivin in the cell model of CVB3-induced myocarditis. This study demonstrated that survivin was triggered by CVB3 infection in NMCs and survivin executed its antiapoptotic effects via caspase-3- and caspase-9-dependent signaling pathway.


Asunto(s)
Apoptosis , Infecciones por Coxsackievirus/metabolismo , Enterovirus Humano B/patogenicidad , Miocarditis/metabolismo , Miocitos Cardíacos/metabolismo , Survivin/metabolismo , Animales , Animales Recién Nacidos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Infecciones por Coxsackievirus/genética , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , Ratones Endogámicos BALB C , Miocarditis/genética , Miocarditis/patología , Miocarditis/virología , Miocitos Cardíacos/patología , Transducción de Señal , Survivin/genética , Factores de Tiempo
13.
J Cell Physiol ; 234(10): 18029-18040, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30843214

RESUMEN

Fingolimod (FTY720) after phosphorylation, as the ligand of sphingosine 1-phosphate receptors (S1PRs), plays an important role in cell proliferation and differentiation. In this article, FTY720 in the treatment of coxsackievirus B3 (CVB3)-induced viral myocarditis was closely related to apoptosis and AKT/caspase-3 apoptotic pathways. We found that CVB3 inhibited myocardial apoptosis at the early stage with upregulating p-AKT level and downregulating activated caspase-3 level for replication of virus progeny, whereas it promoted apoptosis at a late stage with downregulating p-AKT and upregulating activated caspase-3 for releasing the newly synthesized virus to spread. Interestingly, FTY720 could reverse this trend; it promoted apoptosis at an early stage and inhibited apoptosis at the late stage in vivo and vitro, which proved the antiviral effect. We also found that S1PR1, S1PR4, and S1PR5, rather than S1PR2 and S1PR3, were regulated by FTY720 in this process. The results confirmed that FTY720 alleviates CVB3-induced myocarditis and inhibits viral replication through regulating S1PRs and AKT/caspase-3 pathways with a bidirectional regulation of apoptosis.


Asunto(s)
Antivirales/farmacología , Caspasa 3/metabolismo , Infecciones por Coxsackievirus/prevención & control , Enterovirus Humano B/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Miocarditis/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología , Receptores de Esfingosina-1-Fosfato/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , Modelos Animales de Enfermedad , Enterovirus Humano B/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Masculino , Ratones Endogámicos BALB C , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/virología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología , Transducción de Señal , Receptores de Esfingosina-1-Fosfato/metabolismo , Factores de Tiempo
14.
Cell Physiol Biochem ; 53(1): 121-140, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31230428

RESUMEN

Infections with Coxsackievirus B3 and other members of the enterovirus genus are a common reason for myocarditis and sudden cardiac death in modern society. Despite intensive scientific efforts to cure enterovirus infections, there is still no standardized treatment option. The complexity of Coxsackievirus B3´s effects on the host cell make well defined studies on this topic very challenging. However, recent publications report newly found effects of CVB3´s structural and non-structural proteins on infected cells. For the first time, the viral capsid protein VP1 was shown to have direct influence on the viral life-cycle. By shortening the G0 and the G2 phase and simultaneously prolonging the G1 and G1-S phase, the translation of viral proteins is enhanced and the production of viable CVB3 particles is promoted. Coxsackievirus B3´s viroporin, protein 2B, was recently studied in more detail as well. Structural and physiological analyses identified two hydrophilic α-helices in the structure of 2B, enabling it to insert into cellular membranes of host cells. As main target of 2B the endoplasmatic reticulum was identified. The insertion of 2B into the ER membranes leads to an uncontrolled calcium outflow into the cytoplasm. Additional insertion of 2B into the cell membrane leads to host cell destabilization and in the end to release of viral progeny. The importance of the Coxsackievirus B3´s proteases 2A and 3C in pathogenicity is observed since years. Recently, DAP5 and eIf4G were identified as new cleavage targets for protease 2A. Cleavage of DAP-5 into DAP5-N and DAP5-C changes the gene expression of the host cell and promotes cell death. Additionally, protease 3C targets and cleaves procaspase 8 promoting the mitochondrial apoptosis pathway and cell death. Recent studies identified significant effects of CVB3 on mitochondria of infected cells. Mouse cardiomyocytes showed decreased activities of respiratory chain complexes I-III and changed transcription of important subunits of the complexes I-IV. A disrupted energy metabolism may be one of the main causes of cardiac insufficiency and death in CVB3 infected patients. In addition to a modified energy metabolism, CVB3 affects cardiac ion channels, KCNQ1 in particular. SGK1, which is an important mediator in KCNQ1 membrane insertions, is highly upregulated during CVB3 infections. This results in an increased insertion of KCNQ1 into the cell membrane of cardiac cells. Under stress conditions, this KCNQ1 overshoot may lead to a disturbed cardiac action potential and therefore to sudden cardiac death, as it is often observed in CVB3 infected persons.


Asunto(s)
Infecciones por Coxsackievirus/patología , Enterovirus Humano B/fisiología , Animales , Proteínas de la Cápside/metabolismo , Infecciones por Coxsackievirus/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Enterovirus Humano B/patogenicidad , Humanos , Canal de Potasio KCNQ1/metabolismo , Mitocondrias/metabolismo , Mitocondrias/virología , Proteínas no Estructurales Virales/metabolismo
15.
Basic Res Cardiol ; 114(2): 11, 2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30673858

RESUMEN

Coxsackieviruses of group B (CVB) are well-known causes of acute and chronic myocarditis. Chronic myocarditis can evolve into dilated cardiomyopathy (DCM) characterized by fibrosis and cardiac remodeling. Interleukin-1ß (IL-1ß) plays a decisive role in the induction of the inflammatory response as a consequence of viral replication. In this study, we analyzed the effects of IL-1ß neutralization on the transition of acute to chronic myocarditis in a mouse model of CVB3 myocarditis. Mice were treated with an anti-murine IL-1ß antibody as a surrogate for Canakinumab at different time points post CVB3 infection. Treatment was performed in the early phase (day 1-14 pi, day 3-14 pi) or at a later stage of myocarditis (day 14-28 pi). Subsequently, the hearts were examined histologically, immunohistochemically and by molecular biology. A significant reduction of viral replication, cardiac damage and inflammation was found after administration of the antibody in the early phase and in the later phase of infection. Furthermore, less collagen I deposition and a considerable reduction of fibrosis were found in antibody-treated mice. Using microarray analysis, a significant upregulation of various extracellular matrix and fibrosis-associated molecules was found in CVB3-infected mice, including TGF-ß, TIMP-1 and MMP12, as well as diverse matricellular proteins, whereas, these molecules were significantly downregulated in all IL-1ß antibody-treated infected mice. Neutralization of IL-1ß at different stages of enteroviral infection prevents the development of chronic viral myocarditis by reducing inflammation, interstitial fibrosis and adverse cardiac remodeling. These findings are relevant for the treatment of patients with acute and chronic myocarditis.


Asunto(s)
Interleucina-1beta/antagonistas & inhibidores , Miocarditis/patología , Remodelación Ventricular/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Enfermedad Crónica , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Enterovirus Humano B , Ratones , Miocarditis/metabolismo , Miocarditis/virología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
PLoS Pathog ; 13(12): e1006744, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29220410

RESUMEN

Nuclear factor of activated T cells 5 (NFAT5)/Tonicity enhancer binding protein (TonEBP) is a transcription factor induced by hypertonic stress in the kidney. However, the function of NFAT5 in other organs has rarely been studied, even though it is ubiquitously expressed. Indeed, although NFAT5 was reported to be critical for heart development and function, its role in infectious heart diseases has remained obscure. In this study, we aimed to understand the mechanism by which NFAT5 interferes with infection of Coxsackievirus B3 (CVB3), a major cause of viral myocarditis. Our initial results demonstrated that although the mRNA level of NFAT5 remained constant during CVB3 infection, NFAT5 protein level decreased because the protein was cleaved. Bioinformatic prediction and verification of the predicted site by site-directed mutagenesis experiments determined that the NFAT5 protein was cleaved by CVB3 protease 2A at Glycine 503. Such cleavage led to the inactivation of NFAT5, and the 70-kDa N-terminal cleavage product (p70-NFAT5) exerted a dominant negative effect on the full-length NFAT5 protein. We further showed that elevated expression of NFAT5 to counteract viral protease cleavage, especially overexpression of a non-cleavable mutant of NFAT5, significantly inhibited CVB3 replication. Ectopic expression of NFAT5 resulted in elevated expression of inducible nitric oxide synthase (iNOS), a factor reported to inhibit CVB3 replication. The necessity of iNOS for the anti-CVB3 effect of NFAT5 was supported by the observation that inhibition of iNOS blocked the anti-CVB3 effect of NFAT5. In a murine model of viral myocarditis, we observed that treatment with hypertonic saline or mannitol solution upregulated NFAT5 and iNOS expression, inhibited CVB3 replication and reduced tissue damage in the heart. Taken together, our data demonstrate that the anti-CVB3 activity of NFAT5 is impaired during CVB3 infection due to 2A-mediated cleavage of NFAT5. Thus induction of NFAT5 by hypertonic agents may be a promising strategy for the development of anti-CVB3 therapeutics.


Asunto(s)
Infecciones por Coxsackievirus/virología , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano B/enzimología , Miocarditis/virología , Miocitos Cardíacos/virología , Factores de Transcripción/metabolismo , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Enterovirus Humano B/inmunología , Enterovirus Humano B/fisiología , Regulación de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos A , Mutación , Miocarditis/inmunología , Miocarditis/metabolismo , Miocarditis/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/química , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteolisis , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Factores de Transcripción/genética , Replicación Viral
17.
J Med Virol ; 91(5): 886-889, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30570750

RESUMEN

Latency associated peptide (LAP) is a protein expressed on the membrane of some regulatory T cells (Treg). LAP+ Treg have a greater immunomodulatory effect than that of their negative counterparts. In this study, we presented the data on the proportion of LAP+ Treg out of CD4+ cells in mice with viral myocarditis, which we believed was more sensitive and specific than that of the ratio of total Treg in CD4+ cells. Comparing with the previously recognized total Treg, LAP+ Treg was a better biomarker on myocardial inflammation.


Asunto(s)
Infecciones por Coxsackievirus/diagnóstico , Miocarditis/diagnóstico , Péptidos/análisis , Precursores de Proteínas/análisis , Subgrupos de Linfocitos T/química , Linfocitos T Reguladores/química , Factor de Crecimiento Transformador beta/análisis , Animales , Infecciones por Coxsackievirus/patología , Modelos Animales de Enfermedad , Recuento de Linfocitos , Masculino , Ratones Endogámicos BALB C , Miocarditis/patología
18.
Int J Mol Sci ; 20(2)2019 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-30669342

RESUMEN

Adenosine deaminases acting on RNA (ADAR) are enzymes that regulate RNA metabolism through post-transcriptional mechanisms. ADAR1 is involved in a variety of pathological conditions including inflammation, cancer, and the host defense against viral infections. However, the role of ADAR1p150 in vascular disease remains unclear. In this study, we examined the expression of ADAR1p150 and its role in viral myocarditis (VMC) in a mouse model. VMC mouse cardiomyocytes showed significantly higher expression of ADAR1p150 compared to the control samples. Coimmunoprecipitation verified that ADAR1p150 forms a complex with Dicer in VMC. miRNA-222, which is involved in many cardiac diseases, is highly expressed in cardiomyocytes in VMC. In addition, the expression of miRNA-222 was promoted by ADAR1p150/Dicer. Among the target genes of miRNA-222, the expression of phosphatase-and-tensin (PTEN) protein was significantly reduced in VMC. By using a bioinformatics tool, we found a potential binding site of miRNA-222 on the PTEN gene's 3'-UTR, suggesting that miRNA-222 might play a regulatory role. In cultured cells, miR-222 suppressed PTEN expression. Our findings suggest that ADAR1p150 plays a key role in complexing with Dicer and promoting the expression of miRNA-222, the latter of which suppresses the expression of the target gene PTEN during VMC. Our work reveals a previously unknown role of ADAR1p150 in gene expression in VMC.


Asunto(s)
Adenosina Desaminasa/metabolismo , Infecciones por Coxsackievirus/complicaciones , Enterovirus Humano B , MicroARNs/genética , Miocarditis/etiología , Miocarditis/metabolismo , Fosfohidrolasa PTEN/genética , Ribonucleasa III/metabolismo , Animales , Supervivencia Celular/genética , Células Cultivadas , Infecciones por Coxsackievirus/patología , Infecciones por Coxsackievirus/virología , Modelos Animales de Enfermedad , Enterovirus Humano B/fisiología , Expresión Génica , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Inmunohistoquímica , Masculino , Ratones , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/virología , Unión Proteica , Interferencia de ARN
19.
Biochim Biophys Acta Mol Basis Dis ; 1864(8): 2579-2589, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29730342

RESUMEN

Acute viral myocarditis (VM), characterised by leukocyte infiltration and dysfunction of the heart, is an important cause of sudden cardiac death in young adults. Unfortunately, to date, the pathological mechanisms underlying cardiac failure in VM remain incompletely understood. In the current study, we investigated if acute VM leads to cardiac metabolic rewiring and if this process is driven by local inflammation. Transcriptomic analysis of cardiac biopsies from myocarditis patients and a mouse model of VM revealed prominent reductions in the expression of a multitude of genes involved in mitochondrial oxidative energy metabolism. In mice, this coincided with reductions in high-energy phosphate and NAD levels, as determined by Imaging Mass Spectrometry, as well as marked decreases in the activity, protein abundance and mRNA levels of various enzymes and key regulators of cardiac oxidative metabolism. Indicative of fulminant cardiac inflammation, NF-κB signalling and inflammatory cytokine expression were potently induced in the heart during human and mouse VM. In cultured cardiomyocytes, cytokine-mediated NF-κB activation impaired cardiomyocyte oxidative gene expression, likely by interfering with the PGC-1 (peroxisome proliferator-activated receptor (PPAR)-γ co-activator) signalling network, the key regulatory pathway controlling cardiomyocyte oxidative metabolism. In conclusion, we provide evidence that acute VM is associated with extensive cardiac metabolic remodelling and our data support a mechanism whereby cytokines secreted primarily from infiltrating leukocytes activate NF-κB signalling in cardiomyocytes thereby inhibiting the transcriptional activity of the PGC-1 network and consequently modulating myocardial energy metabolism.


Asunto(s)
Infecciones por Coxsackievirus/metabolismo , Enterovirus Humano B , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Musculares/metabolismo , Miocarditis/metabolismo , FN-kappa B/metabolismo , Enfermedad Aguda , Animales , Infecciones por Coxsackievirus/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Miocarditis/patología , Miocarditis/virología , PPAR gamma/metabolismo , Factores de Transcripción/metabolismo
20.
J Neurovirol ; 24(6): 780-785, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30291563

RESUMEN

Enteroviruses and Coxsackie viruses are common causes of aseptic meningitis and encephalitis in children. These infections usually have a benign, self-limited course. However, they can have a florid presentation in immunocompromised patients, such as neonates, patients exposed to immunosuppressive drugs, such as transplant recipients and patients with agammaglobulinemia. We present a rare case of rapidly progressive acute encephalopathy caused by Coxsackie meningoencephalitis and complicated by refractory status epilepticus in an immunocompetent adult male. Our case highlights the importance of having a broad differential in patients presenting with rapidly progressive acute encephalopathy. Although rare, an enterovirus infection caused by a Coxsackie virus subtype can have a severe presentation causing significant morbidity. This case, also underscores the importance of searching for underlying immunodeficiency in malignant presentations of common viral infections. Hence, rapidly progressive acute encephalopathy due to coxsackievirus can occur in immunocompetent individuals. Aggressive and systematic diagnostic and therapeutic approach in such severe cases can influence overall outcomes.


Asunto(s)
Infecciones por Coxsackievirus/patología , Meningoencefalitis/patología , Meningoencefalitis/virología , Progresión de la Enfermedad , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda