Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.714
Filtrar
1.
J Zhejiang Univ Sci B ; 25(5): 422-437, 2024 May 15.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38725341

RESUMEN

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


Asunto(s)
Enterovirus Humano B , Exosomas , Ferroptosis , Células Madre Mesenquimatosas , MicroARNs , Miocitos Cardíacos , Transducción de Señal , Proteína Smad2 , Cordón Umbilical , Células Madre Mesenquimatosas/metabolismo , Exosomas/metabolismo , Animales , Humanos , Ratones , Proteína Smad2/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Enterovirus Humano B/fisiología , Miocitos Cardíacos/metabolismo , Cordón Umbilical/citología , Infecciones por Coxsackievirus/metabolismo , Masculino , Miocarditis/metabolismo , Miocarditis/virología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo
2.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38696536

RESUMEN

Major 5'-terminally deleted (5'TD) RNA forms of group-B coxsackievirus (CVB-5'TD) has been associated with myocarditis in both mice and humans. Although it is known that interferon-ß (IFN-ß) signaling is critical for an efficient innate immune response against CVB-induced myocarditis, the link between CVB-5'TD RNA forms and type I IFN signaling in cardiomyocytes remains to be explored. In a mouse model of CVB3/28-induced myocarditis, major early-emerging forms of CVB-5'TD RNA have been characterized as replicative viral populations that impair IFN-ß production in the heart. Synthetic CVB3/28 RNA forms mimicking each of these major 5'TD virus populations were transfected in mice and have been shown to modulate innate immune responses in the heart and to induce myocarditis in mice. Remarkably, transfection of synthetic viral RNA with deletions in the secondary structures of the 5'-terminal CVB3 RNA domain I, modifying stem-loops "b", "c" or "d", were found to impair IFN-ß production in human cardiomyocytes. In addition, the activation of innate immune response by Poly(I:C), was found to restore IFN-ß production and to reduce the burden of CVB-5'TD RNA-forms in cardiac tissues, thereby reducing the mortality rate of infected mice. Overall, our results indicate that major early-emerging CVB3 populations deleted in the domain I of genomic RNA, in the 5' noncoding region, modulate the activation of the type I IFN pathway in cardiomyocytes and induce myocarditis in mice. These findings shed new light on the role of replicative CVB-5'TD RNA forms as key pathophysiological factors in CVB-induced human myocarditis.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus Humano B , Interferón Tipo I , Miocarditis , Miocitos Cardíacos , ARN Viral , Miocarditis/virología , Miocarditis/inmunología , Miocarditis/genética , Animales , Miocitos Cardíacos/virología , Miocitos Cardíacos/metabolismo , Ratones , Enterovirus Humano B/inmunología , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/virología , Infecciones por Coxsackievirus/genética , Interferón Tipo I/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Humanos , Inmunidad Innata , Transducción de Señal , Interferón beta/metabolismo , Interferón beta/genética , Interferón beta/inmunología , Masculino , Regiones no Traducidas 5'
3.
Front Immunol ; 15: 1380697, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38715608

RESUMEN

The Corona Virus Disease (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), has quickly spread worldwide and resulted in significant morbidity and mortality. Although most infections are mild, some patients can also develop severe and fatal myocarditis. In eukaryotic RNAs, 5-methylcytosine (m5C) is a common kind of post-transcriptional modification, which is involved in regulating various biological processes (such as RNA export, translation, and stability maintenance). With the rapid development of m5C modification detection technology, studies related to viral m5C modification are ever-increasing. These studies have revealed that m5C modification plays an important role in various stages of viral replication, including transcription and translation. According to recent studies, m5C methylation modification can regulate SARS-CoV-2 infection by modulating innate immune signaling pathways. However, the specific role of m5C modification in SARS-CoV-2-induced myocarditis remains unclear. Therefore, this review aims to provide insights into the molecular mechanisms of m5C methylation in SARS-CoV-2 infection. Moreover, the regulatory role of NSUN2 in viral infection and host innate immune response was also highlighted. This review may provide new directions for developing therapeutic strategies for SARS-CoV-2-associated myocarditis.


Asunto(s)
COVID-19 , Miocarditis , SARS-CoV-2 , Miocarditis/virología , Miocarditis/inmunología , Miocarditis/terapia , Miocarditis/genética , Humanos , COVID-19/inmunología , COVID-19/genética , COVID-19/terapia , SARS-CoV-2/fisiología , Metilación , 5-Metilcitosina/metabolismo , Inmunidad Innata , Tratamiento Farmacológico de COVID-19 , Animales , ARN Viral/genética , ARN Viral/metabolismo , Procesamiento Postranscripcional del ARN
4.
Sci Rep ; 14(1): 10289, 2024 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704437

RESUMEN

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


Asunto(s)
Fiebre Aftosa , Miocarditis , Animales , Bovinos , Miocarditis/veterinaria , Miocarditis/virología , Miocarditis/patología , Fiebre Aftosa/virología , Fiebre Aftosa/patología , Enfermedades de los Bovinos/virología , Enfermedades de los Bovinos/sangre , Enfermedades de los Bovinos/patología , Virus de la Fiebre Aftosa/patogenicidad , Virus de la Fiebre Aftosa/aislamiento & purificación , Animales Lactantes , Factores de Edad , Aspartato Aminotransferasas/sangre , Masculino , L-Lactato Deshidrogenasa/sangre
5.
Int Immunopharmacol ; 133: 112073, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38636372

RESUMEN

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


Asunto(s)
Ivermectina , Ratones Endogámicos BALB C , Miocarditis , Factor de Transcripción ReIA , Animales , Miocarditis/tratamiento farmacológico , Miocarditis/virología , Ratones , Ivermectina/uso terapéutico , Ivermectina/farmacología , Células RAW 264.7 , Masculino , Factor de Transcripción ReIA/metabolismo , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Citocinas/metabolismo , beta Carioferinas/metabolismo , Modelos Animales de Enfermedad , Enfermedades Autoinmunes/tratamiento farmacológico , Humanos , Miocardio/patología , Miocardio/metabolismo
6.
Nat Commun ; 15(1): 3481, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664417

RESUMEN

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


Asunto(s)
Adenina , Estrés del Retículo Endoplásmico , Indoles , Miocarditis , Miocitos Cardíacos , eIF-2 Quinasa , Animales , Humanos , Masculino , Ratones , Adenina/análogos & derivados , Apoptosis , eIF-2 Quinasa/metabolismo , eIF-2 Quinasa/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/virología , Miocarditis/metabolismo , Miocarditis/patología , Miocardio/patología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/virología , Miocitos Cardíacos/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Replicación Viral
7.
Virus Res ; 339: 199250, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-37865350

RESUMEN

Evidence is emerging on the roles of long noncoding RNAs (lncRNAs) as regulatory factors in a variety of viral infection processes, but the mechanisms underlying their functions in coxsackievirus group B type3 (CVB3)-induced acute viral myocarditis have not been explicitly delineated. We previously demonstrated that CVB3 infection decreases miRNA-21 expression; however, lncRNAs that regulate the miRNA-21-dependent CVB3 disease process have yet to be identified. To evaluate lncRNAs upstream of miRNA-21, differentially expressed lncRNAs in CVB3-infected mouse hearts were identified by microarray analysis and lncRNA/miRNA-21 interactions were predicted bioinformatically. MEG3 was identified as a candidate miRNA-21-interacting lncRNA upregulated in CVB3-infected mouse hearts. MEG3 expression was verified to be upregulated in HeLa cells 48 h post CVB3 infection and to act as a competitive endogenous RNA of miRNA-21. MEG3 knockdown resulted in the upregulation of miRNA-21, which inhibited CVB3 replication by attenuating P38-MAPK signaling in vitro and in vivo. Knockdown of MEG3 expression before CVB3 infection inhibited viral replication in mouse hearts and alleviated cardiac injury, which improved survival. Furthermore, the knockdown of CREB5, which was predicted bioinformatically to function upstream of MEG3, was demonstrated to decrease MEG3 expression and CVB3 viral replication. This study identifies the function of the lncRNA MEG3/miRNA-21/P38 MAPK axis in the process of CVB3 replication, for which CREB5 could serve as an upstream modulator.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus , MicroARNs , Miocarditis , ARN Largo no Codificante , Virosis , Animales , Humanos , Ratones , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/genética , Enterovirus/genética , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Células HeLa/virología , MicroARNs/genética , MicroARNs/metabolismo , Miocarditis/genética , Miocarditis/metabolismo , Miocarditis/virología , ARN Largo no Codificante/genética , Replicación Viral
8.
Antiviral Res ; 217: 105702, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37604350

RESUMEN

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


Asunto(s)
Enterovirus Humano B , MicroARNs , Miocarditis , Miocardio , Replicación Viral , Enterovirus Humano B/genética , Enterovirus Humano B/fisiología , Miocarditis/metabolismo , Miocarditis/virología , Miocardio/metabolismo , Miocardio/patología , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Humanos , Replicación Viral/efectos de los fármacos , Replicación Viral/genética , Genoma Viral , ARN Polimerasa Dependiente del ARN/genética , Antagomirs/farmacología , Ratones Endogámicos BALB C , Células HEK293 , Células HeLa , Ratones , Animales
9.
Immunol Cell Biol ; 101(8): 735-745, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37253434

RESUMEN

Coxsackievirus B3 (CVB3)-induced viral myocarditis (VMC) is characterized by immune cell infiltration and myocardial damage. High mobility group box 1 (HMGB1) is a highly conserved nuclear DNA-binding protein that participates in DNA replication, transcriptional regulation, repair response and inflammatory response in different disease models. To investigate the exact function of HMGB1 in CVB3-induced VMC, we crossed Hmgb1-floxed (Hmgb1f/f ) mice with mice carrying a suitable Cre recombinase transgenic strain to achieve conditional inactivation of the Hmgb1 gene in a cardiomyocyte-specific manner and to establish myocarditis. In this study, we found that cardiomyocyte-specific Hmgb1-deficient (Hmgb1f/f TgCre/+ ) mice exhibited exacerbated myocardial injury. Hmgb1-deficient cardiomyocytes may promote early apoptosis via the p53-mediated Bax mitochondrial pathway, as evidenced by the higher localization of p53 protein in the cytosol of Hmgb1-deficient cardiomyocytes upon CVB3 infection. Moreover, cardiomyocyte Hmgb1-deficient mice are more susceptible to cardiac dysfunction after infection. This study provides new insights into HMGB1 in VMC pathogenesis and a strategy for appropriate blocking of HMGB1 in the clinical treatment of VMC.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus Humano B , Proteína HMGB1 , Miocarditis , Animales , Ratones , Apoptosis/genética , Proteína HMGB1/metabolismo , Ratones Endogámicos BALB C , Miocarditis/inmunología , Miocarditis/patología , Miocarditis/virología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteína p53 Supresora de Tumor/metabolismo , Infecciones por Coxsackievirus/inmunología
10.
Artif Organs ; 47(2): 396-407, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36269688

RESUMEN

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


Asunto(s)
Oxigenación por Membrana Extracorpórea , Miocarditis , Adulto , Humanos , Miocarditis/diagnóstico , Miocarditis/terapia , Miocarditis/virología , Estudios Retrospectivos , Resultado del Tratamiento , Virosis
11.
J Biomed Sci ; 29(1): 55, 2022 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-35909127

RESUMEN

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


Asunto(s)
Encefalitis por Herpes Simple , Miocarditis , Ubiquitina-Proteína Ligasas , Virosis , Animales , Antivirales , Humanos , Inmunidad Innata , Inflamación/genética , Ratones , Miocarditis/genética , Miocarditis/virología , Proteína Fosfatasa 2C , ARN , Ubiquitina-Proteína Ligasas/genética
12.
Signal Transduct Target Ther ; 7(1): 57, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35197452

RESUMEN

The coronavirus disease 2019 (COVID-19) is a highly transmissible disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that poses a major threat to global public health. Although COVID-19 primarily affects the respiratory system, causing severe pneumonia and acute respiratory distress syndrome in severe cases, it can also result in multiple extrapulmonary complications. The pathogenesis of extrapulmonary damage in patients with COVID-19 is probably multifactorial, involving both the direct effects of SARS-CoV-2 and the indirect mechanisms associated with the host inflammatory response. Recognition of features and pathogenesis of extrapulmonary complications has clinical implications for identifying disease progression and designing therapeutic strategies. This review provides an overview of the extrapulmonary complications of COVID-19 from immunological and pathophysiologic perspectives and focuses on the pathogenesis and potential therapeutic targets for the management of COVID-19.


Asunto(s)
Lesión Renal Aguda/complicaciones , COVID-19/complicaciones , Síndrome de Liberación de Citoquinas/complicaciones , Coagulación Intravascular Diseminada/complicaciones , Linfopenia/complicaciones , Miocarditis/complicaciones , Embolia Pulmonar/complicaciones , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/virología , Anticoagulantes/uso terapéutico , Antivirales/uso terapéutico , COVID-19/inmunología , COVID-19/virología , Ensayos Clínicos como Asunto , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Coagulación Intravascular Diseminada/tratamiento farmacológico , Coagulación Intravascular Diseminada/inmunología , Coagulación Intravascular Diseminada/virología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/virología , Humanos , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Linfopenia/tratamiento farmacológico , Linfopenia/inmunología , Linfopenia/virología , Miocarditis/tratamiento farmacológico , Miocarditis/inmunología , Miocarditis/virología , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/inmunología , Embolia Pulmonar/virología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/inmunología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/patogenicidad , Tratamiento Farmacológico de COVID-19
13.
Diagn Pathol ; 17(1): 31, 2022 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177093

RESUMEN

BACKGROUND: Despite a reported cardiac injury in patients with new coronavirus infection, the possibility and specifics of genuine viral myocarditis in COVID-19 remains not fully clear. PURPOSE: To study the presence of SARS-CoV-2 in the myocardium and the morphological properties of myocarditis in patients with severe coronavirus infection (COVID-19). METHODS: Autopsy data of eight elderly patients (75.6 ± 7.4 years), four male and four female, with severe new coronavirus infection were studied. The lifetime diagnosis of COVID-19 is based on a positive result of the PCR study. The inclusion criterion was the presence of morphological signs of myocarditis according to the Dallas criteria. A standard histological examination included staining by hematoxylin and eosin, toluidin blue and Van Gieson. An immunohistochemical study was performed using antibodies to CD3, CD 68, CD20, perforin, toll-like receptor (TLR) types 4 and 9. PCR in real-time was performed to determine the viral RNA in the myocardium. RESULTS: All patients had severe bilateral viral pneumonia. In all cases, myocarditis was not clinically diagnosed. Morphological examination of the heart found signs of active lymphocytic myocarditis. PCR identified the SARS-Cov2 RNA in all cases. There were also signs of destructive coronaritis in all cases, thrombovasculitis, lymphocytic pericarditis (in 3 cases) and endocarditis (in 2 cases). The absence of neutrophils confirms the aseptic nature of inflammation. An immunohistochemical study showed the CD3-positive T lymphocytes in the infiltrates. Increased expression of TLR type 4 and less 9 was also detected. CONCLUSION: Morphological and immunohistochemical evidence of myocarditis in COVID-19 was presented. Lymphocytic infiltrations and positive PCR confirm the viral nature of inflammation. Myocarditis in COVID-19 is also characterized by coronaritis with microvascular thrombosis and associated with lymphocytic endo- and pericarditis.


Asunto(s)
COVID-19/patología , Miocarditis/patología , Neumonía Viral/patología , SARS-CoV-2/aislamiento & purificación , Anciano , Anciano de 80 o más Años , Autopsia , COVID-19/complicaciones , COVID-19/diagnóstico , COVID-19/virología , Femenino , Corazón/virología , Humanos , Inmunohistoquímica , Inflamación , Linfocitos/patología , Masculino , Persona de Mediana Edad , Miocarditis/complicaciones , Miocarditis/diagnóstico , Miocarditis/virología , Miocardio/patología , Neumonía Viral/diagnóstico , Neumonía Viral/virología , SARS-CoV-2/genética
15.
Cells ; 11(4)2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35203260

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is an extremely contagious disease whereby the virus damages the host's respiratory tract via entering through the ACE2 receptor. Cardiovascular disorder is being recognized in the majority of COVID-19 patients; yet, the relationship between SARS-CoV-2 and heart failure has not been established. In the present study, SARS-CoV-2 infection was induced in the monkey model. Thereafter, heart tissue samples were collected, and pathological changes were analyzed in the left ventricular tissue by hematoxylin and eosin, trichrome, and immunohistochemical staining specific to T lymphocytes and macrophages. The findings revealed that SARS-CoV-2 infection induces several pathological changes in the heart, which cause cardiomyocyte disarray, mononuclear infiltrates of inflammatory cells, and hypertrophy. Furthermore, collagen-specific staining showed the development of cardiac fibrosis in the interstitial and perivascular regions in the hearts of infected primates. Moreover, the myocardial tissue samples displayed multiple foci of inflammatory cells positive for T lymphocytes and macrophages within the myocardium. These findings suggest the progression of the disease, which can lead to the development of severe complications, including heart failure. Additionally, SARS-CoV-2 antigen staining detected the presence of virus particles in the myocardium. Thus, we found that SARS-CoV-2 infection is characterized by an exaggerated inflammatory immune response in the heart, which possibly contributes to myocardial remodeling and subsequent fibrosis.


Asunto(s)
COVID-19/inmunología , Insuficiencia Cardíaca/fisiopatología , Corazón/fisiopatología , Animales , Chlorocebus aethiops , Corazón/virología , Insuficiencia Cardíaca/virología , Ventrículos Cardíacos/fisiopatología , Ventrículos Cardíacos/virología , Sistema Inmunológico/patología , Macaca mulatta , Miocarditis/virología , Miocardio/metabolismo , SARS-CoV-2/patogenicidad
16.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35163412

RESUMEN

Protein kinase B2 (AKT2) is involved in various cardiomyocyte signaling processes, including those important for survival and metabolism. Coxsackievirus B3 (CVB3) is one of the most common pathogens that cause myocarditis in humans. The role of AKT2 in CVB3 infection is not yet well understood. We used a cardiac-specific AKT2 knockout (KO) mouse to determine the role of AKT2 in CVB3-mediated myocarditis. CVB3 was injected intraperitoneally into wild-type (WT) and KO mice. The mice's survival rate was recorded: survival in KO mice was significantly decreased compared with WT mice (WT vs. KO: 73.3 vs. 27.1%). Myocardial damage and inflammation were significantly increased in the hearts of KO mice compared with those of WT mice. Moreover, from surface ECG, AKT2 KO mice showed a prolonged atria and ventricle conduction time (PR interval, WT vs. KO: 47.27 ± 1.17 vs. 64.79 ± 7.17 ms). AKT2 deletion induced severe myocarditis and cardiac dysfunction due to CVB3 infection. According to real-time PCR, the mRNA level of IL-1, IL-6, and TNF-α decreased significantly in KO mice compared with WT mice on Days 5 after infection. In addition, innate immune response antiviral effectors, Type I interferon (interferon-α and ß), and p62, were dramatically suppressed in the heart of KO mice. In particular, the adult cardiac myocytes isolated from the heart showed high induction of TLR4 protein in KO mice in comparison with WT. AKT2 deletion suppressed the activation of Type I interferon and p62 transcription in CVB3 infection. In cardiac myocytes, AKT2 is a key signaling molecule for the heart from damage through the activation of innate immunity during acute myocarditis.


Asunto(s)
Enterovirus Humano B/inmunología , Infecciones por Enterovirus/inmunología , Inmunidad Innata , Miocarditis/inmunología , Miocardio/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Enfermedad Aguda , Animales , Enterovirus Humano B/genética , Infecciones por Enterovirus/genética , Células HeLa , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/virología , Ratones , Ratones Noqueados , Miocarditis/genética , Miocarditis/virología , Proteínas Proto-Oncogénicas c-akt/genética
17.
Viruses ; 14(2)2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35215893

RESUMEN

INTRODUCTION: This study investigated the spontaneous clinical course of patients with endomyocardial biopsy (EMB)-proven lymphocytic myocarditis and cardiac human herpesvirus 6 (HHV6) DNA presence, and the effectiveness of steroid-based intervention in HHV6-positive patients. RESULTS: 756 heart failure (HF) patients underwent an EMB procedure to determine the underlying cause of unexplained HF. Low levels of HHV6 DNA, detectable by nested PCR only, were found in 10.4% of the cases (n = 79) of which 62% (n = 49) showed myocardial inflammation. The spontaneous course of patients with EMB-proven HHV6 DNA-associated lymphocytic myocarditis (n = 26) showed significant improvements in the left ventricular ejection fraction (LVEF) and clinical symptoms, respectively, in 15/26 (60%) patients, 3-12 months after disease onset. EMB mRNA expression of components of the NLRP3 inflammasome pathway and protein analysis of cardiac remodeling markers, analyzed by real-time PCR and MALDI mass spectrometry, respectively, did not differ between HHV6-positive and -negative patients. In another cohort of patients with ongoing symptoms related to lymphocytic myocarditis associated with cardiac levels of HHV6-DNA copy numbers <500 copies/µg cardiac DNA, quantified by real-time PCR, the efficacy and safety of steroid-based immunosuppression for six months was investigated. Steroid-based immunosuppression improved the LVEF (≥5%) in 8/10 patients and reduced cardiac inflammation in 7/10 patients, without an increase in cardiac HHV6 DNA levels in follow-up EMBs. CONCLUSION: Low HHV6 DNA levels are frequently detected in the myocardium, independent of inflammation. In patients with lymphocytic myocarditis with low levels of HHV6 DNA, the spontaneous clinical improvement is nearby 60%. In selected symptomatic patients with cardiac HHV6 DNA copy numbers less than 500 copies/µg cardiac DNA and without signs of an active systemic HHV6 infection, steroid-based therapy was found to be effective and safe. This finding needs to be further confirmed in large, randomized trials.


Asunto(s)
Herpesvirus Humano 6/fisiología , Inmunosupresores/administración & dosificación , Miocarditis/tratamiento farmacológico , Miocarditis/virología , Infecciones por Roseolovirus/tratamiento farmacológico , Infecciones por Roseolovirus/virología , Esteroides/administración & dosificación , Adulto , Anciano , Biopsia , Estudios de Cohortes , ADN Viral/genética , Femenino , Dosificación de Gen , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/aislamiento & purificación , Humanos , Masculino , Persona de Mediana Edad , Miocarditis/inmunología , Miocarditis/fisiopatología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Infecciones por Roseolovirus/inmunología , Infecciones por Roseolovirus/fisiopatología , Volumen Sistólico
18.
Open Heart ; 9(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35046125

RESUMEN

OBJECTIVES: To determine the prevalence and types of viral pathogens in the myocardium of patients presenting with clinically suspected myocarditis in South Africa. METHOD: This is a prospective cross-sectional study. Consecutive adults presenting to a single tertiary centre in South Africa between August 2017 and January 2021 who fulfilled the European Society of Cardiology's diagnostic criteria for clinically suspected myocarditis and who had undergone the appropriate investigations, including cardiac MRI (CMR) and endomyocardial biopsy (EMB), were included. RESULTS: One hundred and two patients with clinically suspected myocarditis were enrolled. Acute myocarditis (AM) was confirmed by CMR or EMB in 82 (80.39%) patients. Viral genomes were detected by PCR in EMB specimens of 50 patients with AM. Parvovirus B19 (PVB19) was the most frequently detected virus, in 37 as monoinfection and 4 as coinfection. This was followed by Epstein-Barr virus (n=6), human herpesvirus 6 (n=2) and human bocavirus (n=1). PVB19 was also detected in 9 patients with no evidence of AM on CMR or EMB. CONCLUSION: Viral myocarditis is the most common form of myocarditis in South Africa. Local viral prevalence appears to be similar those of the developed world. The clinical significance and pathogenic role of PVB19 remains questioned, and its local background prevalence will have to be further investigated.


Asunto(s)
Infecciones por Virus de Epstein-Barr/epidemiología , Herpesvirus Humano 4/genética , Imagen por Resonancia Cinemagnética/métodos , Miocarditis/epidemiología , Miocardio/patología , Adulto , Biopsia , Estudios Transversales , Infecciones por Virus de Epstein-Barr/diagnóstico , Infecciones por Virus de Epstein-Barr/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miocarditis/diagnóstico , Miocarditis/virología , Prevalencia , Estudios Prospectivos , Sudáfrica/epidemiología
19.
BMC Cardiovasc Disord ; 22(1): 10, 2022 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-35034631

RESUMEN

OBJECTIVE: To investigate the effect of A20 and how A20 is regulated in viral myocarditis (VMC). METHODS: BABL/C mice, primary neonatal rat cardiomyocytes and H9c2 cells were infected with Coxsackie virus B3 (CVB3) to establish animal and cellular models of VMC. H&E staining revealed the pathologic condition of myocardium. ELISA measured the serum levels of creatine kinase, creatine kinase isoenzyme and cardiac troponin I. The effects of A20, miR-1a-3p and ADAR1 were investigated using gain and loss of function approaches. ELISA measured the levels of IL-6, IL-18 and TNF-α in serum or cell culture supernatant. TUNEL staining and flow cytometry assessed the apoptosis of myocardium and cardiomyocytes, respectively. RNA-binding protein immunoprecipitation and dual-luciferase reporter assays verified the binding between A20 and miR-1a-3p. Co-immunoprecipitation assay verified the binding between ADAR1 and Dicer. RESULTS: A20 was underexpressed and miR-1a-3p was overexpressed in the myocardium of VMC mice as well as in CVB3-infected cardiomyocytes. Overexpression of A20 suppressed cardiomyocyte inflammation and apoptosis in vivo and in vitro. miR-1a-3p promoted CVB3-induced inflammation and apoptosis in cardiomyocytes by binding to A20. The expression of miR-1a-3p was regulated by ADAR1. ADAR1 promoted the slicing of miR-1a-3p precursor by binding to Dicer. CONCLUSION: A20, regulated by ADAR1/miR-1a-3p, suppresses inflammation and cardiomyocyte apoptosis in VMC.


Asunto(s)
Adenosina Desaminasa/genética , Regulación de la Expresión Génica , Miocarditis/genética , Miocitos Cardíacos/metabolismo , ARN/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Adenosina Desaminasa/biosíntesis , Animales , Animales Recién Nacidos , Apoptosis , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Miocarditis/patología , Miocarditis/virología , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología , Proteínas Nucleares , Ratas , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/biosíntesis
20.
Am J Emerg Med ; 51: 150-155, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34739868

RESUMEN

BACKGROUND: Most COVID-19 infections result in a viral syndrome characterized by fever, cough, shortness of breath, and myalgias. A small but significant proportion of patients develop severe COVID-19 resulting in respiratory failure. Many of these patients also develop multi-organ dysfunction as a byproduct of their critical illness. Although heart failure can be a part of this, there also appears to be a subset of patients who have primary cardiac collapse from COVID-19. OBJECTIVE: Conduct a systematic review of COVID-19-associated myocarditis, including clinical presentation, risk factors, and prognosis. DISCUSSION: Our review demonstrates two distinct etiologies of primary acute heart failure in surprisingly equal incidence in patients with COVID-19: viral myocarditis and Takotsubo cardiomyopathy. COVID myocarditis, Takotsubo cardiomyopathy, and severe COVID-19 can be clinically indistinguishable. All can present with dyspnea and evidence of cardiac injury, although in myocarditis and Takotsubo this is due to primary cardiac dysfunction as compared to respiratory failure in severe COVID-19. CONCLUSION: COVID-19-associated myocarditis differs from COVID-19 respiratory failure by an early shock state. However, not all heart failure from COVID-19 is from direct viral infection; some patient's develop takotsubo cardiomyopathy. Regardless of etiology, steroids may be a beneficial treatment, similar to other critically ill COVID-19 patients. Evidence of cardiac injury in the form of ECG changes or elevated troponin in patients with COVID-19 should prompt providers to consider concurrent myocarditis.


Asunto(s)
COVID-19/complicaciones , Miocarditis/virología , Disnea , Insuficiencia Cardíaca/virología , Humanos , Insuficiencia Respiratoria/virología , Factores de Riesgo , Cardiomiopatía de Takotsubo/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA