Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Biomed Pharmacother ; 143: 112121, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34474346

ABSTRACT

Obesity has been recognized as a major risk factor for the development of chronic cardiomyopathy, which is associated with increased cardiac inflammation, fibrosis, and apoptosis. We previously developed an anti-inflammatory compound C66, which prevented inflammatory diabetic complications via targeting JNK. In the present study, we have tested the hypothesis that C66 could prevent obesity-induced cardiomyopathy by suppressing JNK-mediated inflammation. High-fat diet (HFD)-induced obesity mouse model and palmitic acid (PA)-challenged H9c2 cells were used to develop inflammatory cardiomyopathy and evaluate the protective effects of C66. Our data demonstrate a protective effect of C66 against obesity-induced cardiac inflammation, cardiac hypertrophy, fibrosis, and dysfunction, overall providing cardio-protection. C66 administration attenuates HFD-induced myocardial inflammation by inhibiting NF-κB and JNK activation in mouse hearts. In vitro, C66 prevents PA-induced myocardial injury and apoptosis in H9c2 cells, accompanied with inhibition against PA-induced JNK/NF-κB activation and inflammation. The protective effect of C66 is attributed to its potential to inhibit JNK activation, which led to reduced pro-inflammatory cytokine production and reduced apoptosis in cardiomyocytes both in vitro and in vivo. In summary, C66 provides significant protection against obesity-induced cardiac dysfunction, mainly by inhibiting JNK activation and JNK-mediated inflammation. Our data indicate that inhibition of JNK is able to provide significant protection against obesity-induced cardiac dysfunction.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Benzylidene Compounds/pharmacology , Cardiomyopathies/prevention & control , Cyclohexanones/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Myocarditis/prevention & control , Myocytes, Cardiac/drug effects , Obesity/complications , Animals , Cardiomyopathies/enzymology , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cell Line , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Male , Mice, Inbred C57BL , Myocarditis/enzymology , Myocarditis/etiology , Myocarditis/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Palmitic Acid/toxicity , Rats , Signal Transduction
2.
J Cardiovasc Pharmacol ; 77(5): 570-577, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33657050

ABSTRACT

ABSTRACT: This study aimed to evaluate the protective effect of quercetin and its in-depth mechanism in TNF-α-stimulated cardiomyocytes. The differential expression of TNF-alpha (TNF-α) and signal transducer and activator of transcription 1 (STAT1) was analyzed based on the GEO database. H9c2 cells were stimulated with TNF-α to simulate myocarditis. Cell counting kit-8 assay and flow cytometry assay were performed to detect the cell viability and apoptosis. ELISA was used to measure the levels of proinflammatory cytokines (IL-6 and IL-17A) and anti-inflammatory cytokine (IL-10). STAT1 expression was downregulated by transfection with si-STAT1, and its expression was detected using quantitative real-time polymerase chain reaction and Western blot. Western blot was also performed to assess the expression of the mitogen-activated protein kinase (MAPK) pathway-related factors. In this article, TNF-α was highly expressed in patients with myocarditis, and TNF-α (20 µg/mL) declined the viability of H9c2 cells. Quercetin pretreatment partially alleviated the decrease of cell viability, the increase of apoptosis, and the release of inflammatory cytokines (IL-10, IL-6, and IL-17A) induced by TNF-α. In addition, TNF-α increased STAT1 expression, but quercetin prevented the TNF-α-increased STAT1 level. Remarkably, knockdown of STAT1 enhanced the protective effect of quercetin on TNF-α-injured H9c2 cells. Moreover, quercetin restrained the TNF-α-induced activation of the MAPK pathway. Also, the inhibitory effect of quercetin on the pathway was aggravated by STAT1 lacking. In summing, quercetin plays a protective role in TNF-α-stimulated H9c2 cell injury, which may be related to the regulation of STAT1 and MAPK pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Myocarditis/drug therapy , Myocytes, Cardiac/drug effects , Quercetin/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Case-Control Studies , Cell Line , Humans , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Myocarditis/enzymology , Myocarditis/genetics , Myocarditis/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Rats , STAT1 Transcription Factor/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/toxicity
3.
Basic Res Cardiol ; 116(1): 7, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33523326

ABSTRACT

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


Subject(s)
Anti-Inflammatory Agents/pharmacology , Coxsackievirus Infections/drug therapy , Inflammation/drug therapy , Myeloid Cells/drug effects , Myocarditis/drug therapy , Myocytes, Cardiac/drug effects , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Cells, Cultured , Coxsackievirus Infections/enzymology , Coxsackievirus Infections/immunology , Disease Models, Animal , Enterovirus B, Human/immunology , Enterovirus B, Human/pathogenicity , Host-Pathogen Interactions , Inflammation/enzymology , Inflammation/immunology , Inflammation/virology , Male , Mice, Knockout , Myeloid Cells/enzymology , Myeloid Cells/immunology , Myeloid Cells/virology , Myocarditis/enzymology , Myocarditis/immunology , Myocarditis/virology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/virology , Proteasome Endopeptidase Complex/genetics , Proteolysis
4.
Commun Biol ; 3(1): 562, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037313

ABSTRACT

Myocardial inflammation has recently been recognized as a distinct feature of cardiac hypertrophy and heart failure. HectD3, a HECT domain containing E3 ubiquitin ligase has previously been investigated in the host defense against infections as well as neuroinflammation; its cardiac function however is still unknown. Here we show that HectD3 simultaneously attenuates Calcineurin-NFAT driven cardiomyocyte hypertrophy and the pro-inflammatory actions of LPS/interferon-γ via its cardiac substrates SUMO2 and Stat1, respectively. AAV9-mediated overexpression of HectD3 in mice in vivo not only reduced cardiac SUMO2/Stat1 levels and pathological hypertrophy but also largely abolished macrophage infiltration and fibrosis induced by pressure overload. Taken together, we describe a novel cardioprotective mechanism involving the ubiquitin ligase HectD3, which links anti-hypertrophic and anti-inflammatory effects via dual regulation of SUMO2 and Stat1. In a broader perspective, these findings support the notion that cardiomyocyte growth and inflammation are more intertwined than previously anticipated.


Subject(s)
Cardiomegaly/metabolism , Myocarditis/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Calcineurin/metabolism , Cardiomegaly/enzymology , Cardiomegaly/prevention & control , Humans , Immunoprecipitation , Mice , Microscopy, Fluorescence , Myocarditis/enzymology , Myocarditis/prevention & control , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , RAW 264.7 Cells , Rats , Rats, Wistar , STAT1 Transcription Factor/metabolism , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitin-Protein Ligases/physiology
5.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32515469

ABSTRACT

BACKGROUND: Genistein is widely used as a pharmacological compound as well as a food additive. However, the pharmaceutical effects of Genistein on myocarditis and its potential mechanisms have not been studied in detail. METHODS: H9c2 cells were continuously stimulated by lipopolysaccharide (LPS) for 12 h to simulate the in vitro model of myocarditis injury. DrugBank, String, and GEO dataset were used to investigate specific genes that interacting with Genistein. KEGG and GO enrichment analysis were employed to explore Myc-related signaling pathways. Biological behaviors of H9c2 cells were observed with the support of cell counting kit-8, MTT and flow cytometry. Expression levels of cytokines including TNF-α and ILs were evaluated by enzyme-linked immunosorbent assay. Western blot was applied to detect the expression of Myc and MAPK pathway related proteins. RESULTS: Genistein alleviated the damage of H9c2 cells subjected to LPS from the perspective of elevating cells growth ability, and inhibiting cells apoptosis and inflammatory response. Through bioinformatics analysis, we identified Myc as the potential target of Genistein in myocarditis, and MAPK as the signaling pathway. Significantly, Myc was highly up-regulated in myocarditis samples. More importantly, by performing biological experiments, we discovered that Genistein relieved H9c2 cells apoptosis and inflammatory reaction which caused by LPS stimulation through inhibiting Myc expression. Additionally, the marked augmentation of p-P38 MAPK and p-JNK expression in LPS-induced cardiomyocyte model were blocked by Genistein and si-Myc. CONCLUSIONS: Our research revealed that Myc mediated the protective effects of Genistein on H9c2 cells damage caused by LPS partly through modulation of MAPK/JNK signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Genistein/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/toxicity , Myocarditis/prevention & control , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-myc/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Cytokines/metabolism , Inflammation Mediators/metabolism , Myocarditis/enzymology , Myocarditis/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Phosphorylation , Rats , Signal Transduction
6.
Circ Heart Fail ; 13(6): e006573, 2020 06.
Article in English | MEDLINE | ID: mdl-32493060

ABSTRACT

BACKGROUND: Previous studies have shown beneficial effects of acute infusion of the primary ketone body, ß-hydroxybutyrate, in heart failure (HF). However, whether chronic elevations in circulating ketones are beneficial remains unknown. METHODS: To chronically elevate circulating ketones in mice, we deleted the expression of the ketolytic, rate-limiting-enzyme, SCOT (succinyl-CoA:3-ketoacid-CoA transferase 1; encoded by Oxct1), in skeletal muscle. Tamoxifen-inducible skeletal muscle-specific Oxct1Muscle-/- knockout (n=32) mice and littermate controls (wild type; WT; n=35) were subjected to transverse aortic constriction (TAC) surgery to induce HF. RESULTS: Deletion of SCOT in skeletal, but not cardiac muscle resulted in elevated concentrations of fasted circulating ß-hydroxybutyrate in knockout mice compared with WT mice (P=0.030). Five weeks following TAC, WT mice progressed to HF, whereas knockout mice with elevated fasting circulating ketones were largely protected from the TAC-induced effects observed in WT mice (ejection fraction, P=0.011; mitral E/A, P=0.012). Furthermore, knockout mice with TAC had attenuated expression of markers of sterile inflammation and macrophage infiltration, which were otherwise elevated in WT mice subjected to TAC. Lastly, addition of ß-hydroxybutyrate to isolated hearts was associated with reduced NLRP3 (nucleotide-binding domain-like receptor protein 3)-inflammasome activation, which has been previously shown to play a role in contributing to HF-induced cardiac inflammation. CONCLUSIONS: These data show that chronic elevation of circulating ketones protects against the development of HF that is associated with the ability of ß-hydroxybutyrate to directly reduce inflammation. These beneficial effects of ketones were associated with reduced cardiac NLRP3 inflammasome activation, suggesting that ketones may modulate cardiac inflammation via this mechanism.


Subject(s)
3-Hydroxybutyric Acid/blood , Coenzyme A-Transferases/deficiency , Heart Failure/prevention & control , Myocarditis/prevention & control , Myocardium/enzymology , Animals , Coenzyme A-Transferases/genetics , Disease Models, Animal , Fibrosis , Heart Failure/blood , Heart Failure/enzymology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/blood , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Inflammasomes/metabolism , Isolated Heart Preparation , Male , Mice, Knockout , Myocarditis/blood , Myocarditis/enzymology , Myocarditis/physiopathology , Myocardium/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Up-Regulation , Ventricular Dysfunction, Left/blood , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left , Ventricular Remodeling
7.
Cell Mol Biol Lett ; 25: 6, 2020.
Article in English | MEDLINE | ID: mdl-32099552

ABSTRACT

BACKGROUND: Viral myocarditis (VMC) is a type of cardiac inflammation that is generally caused by coxsackievirus B3 (CVB3) infection. Several MicroRNAs (miRNAs) are known to play crucial roles in VMC pathogenesis. MiR-15 is reportedly associated with myocardial injury, inflammatory responses and viral infection. Whether miR-15 affects the occurrence and development of VMC remains largely unknown. The roles of miR-15 and their underlying mechanisms in CVB3-stimulated H9c2 cells were assessed in this study. METHODS: We infected H9c2 cells with CVB3 to establish a VMC cellular model. We then determined the effects of miR-15 inhibition on three cardiomyocyte injury markers: lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB) and cardiac troponin-I (cTn-I). The impact on CVB3-induced cell apoptosis and pro-inflammatory cytokines was also investigated. The effects of miR-15 inhibition on NLRP3 inflammasome activation were also assessed. The target relationship between miR-15 and NOD-like receptor X1 (NLRX1) was determined using a luciferase reporter assay. RESULTS: MiR-15 expression was significantly upregulated in H9c2 cells after CVB3 infection. Inhibition of miR-15 significantly decreased the CVB3-induced levels of LDH, CK-MB and cTn-I. It also elevated cell viability, reduced CVB3-induced cell apoptosis and decreased the generation of the interleukins IL-1ß, IL-6 and IL-18. Furthermore, we determined that miR-15 inhibition suppressed NLRP3 inflammasome activation by downregulating NLRP3 and caspase-1 p20 expression. We found a direct target relationship between miR-15 and NLRX1. Additionally, inhibition of NLRX1 reversed the protective effects of miR-15 inhibition against CVB3-induced myocardial cell injury by regulating the NLRP3 inflammasome. CONCLUSION: Our results indicate that miR-15 inhibition alleviates CVB3-induced myocardial inflammation and cell injury. This may be partially due to NLRX1-mediated NLRP3 inflammasome inactivation.


Subject(s)
Apoptosis/genetics , Coxsackievirus Infections/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Myocarditis/metabolism , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cell Line , Cell Survival/genetics , Coxsackievirus Infections/genetics , Creatine Kinase, MB Form/metabolism , Cytokines/metabolism , Inflammation/genetics , L-Lactate Dehydrogenase/metabolism , Myocarditis/enzymology , Myocarditis/genetics , Myocarditis/virology , Myocytes, Cardiac/enzymology , Rats , Up-Regulation
9.
Front Immunol ; 11: 603437, 2020.
Article in English | MEDLINE | ID: mdl-33519812

ABSTRACT

Viral myocarditis is caused by a viral infection and characterized by the inflammation of the myocardium. Coxsackievirus B3 (CVB3) infection is one of the most common among the infections caused by this virus. The host's early innate immune response to CVB3 infection particularly depends on the functions of type I interferons (IFNs). In this study, we report that a host microRNA, miR-30a, was upregulated by CVB3 to facilitate its replication. We demonstrated that miR-30a was a potent negative regulator of IFN-I signaling by targeting tripartite motif protein 25 (TRIM25). In addition, we found that TRIM25 overexpression significantly suppressed CVB3 replication, whereas TRIM25 knockdown increased viral titer and VP1 protein expression. MiR-30a inhibits the expression of TRIM25 and TRIM25-mediated retinoic acid-inducible gene (RIG)-I ubiquitination to suppress IFN-ß activation and production, thereby resulting in the enhancement of CVB3 replication. These results indicate the proviral role of miR-30a in modulating CVB3 infection for the first time. This not only provides a new strategy followed by CVB3 in order to modulate IFN-I-mediated antiviral immune responses by engaging host miR-30a but also improves our understanding of its pathogenesis.


Subject(s)
Coxsackievirus Infections/virology , Enterovirus B, Human/growth & development , Interferon-beta/metabolism , MicroRNAs/metabolism , Myocarditis/virology , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Virus Replication , Animals , Capsid Proteins/metabolism , Coxsackievirus Infections/enzymology , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , DEAD Box Protein 58/metabolism , Enterovirus B, Human/immunology , Enterovirus B, Human/metabolism , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Interferon-beta/genetics , Mice, Inbred BALB C , MicroRNAs/genetics , Myocarditis/enzymology , Myocarditis/genetics , Myocarditis/immunology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/virology , Receptors, Immunologic/metabolism , Signal Transduction , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Up-Regulation
10.
J Cardiovasc Pharmacol ; 73(3): 186-194, 2019 03.
Article in English | MEDLINE | ID: mdl-30839512

ABSTRACT

OBJECTIVE: To investigate whether phenylephrine (PE) inhibits sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury through the PI3K/Akt signaling pathway. METHODS: A rat model of sepsis was established by cecal ligation and puncture. PE and/or wortmannin (a PI3K inhibitor) were administered to investigate the role of PI3K/Akt signaling in mediating the effects of PE on inhibiting sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury. Hematoxylin-eosin staining, echocardiography, and Langendorff system were used to examine the myocardial injury and function. The concentrations of TNF-α and IL-6 were analyzed by enzyme-linked immunosorbent assay. Intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), myeloperoxidase, mitochondria-related fusion/fission proteins, and PI3K/Akt signaling pathway-associated proteins were analyzed by Western blotting. RESULTS: PE improved the cardiac function and survival in septic rats. PE decreased TNF-α, IL-6, ICAM-1, VCAM-1, and myeloperoxidase contents in the myocardium of septic rats. Meanwhile, PE increased the fusion-related proteins and decreased the fission-related proteins in the myocardial mitochondria of septic rats. On the other hand, PE activated the PI3K/Akt signaling pathway in the cecal ligation and puncture-treated rats, and all the protective effects of PE were abolished by wortmannin. CONCLUSIONS: PE attenuated sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury through the PI3K/Akt signaling pathway.


Subject(s)
Mitochondria, Heart/drug effects , Mitochondrial Dynamics/drug effects , Myocarditis/prevention & control , Myocytes, Cardiac/drug effects , Phenylephrine/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/drug therapy , Animals , Disease Models, Animal , Inflammation Mediators/metabolism , Isolated Heart Preparation , Male , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Mitochondrial Proteins/metabolism , Myocarditis/enzymology , Myocarditis/etiology , Myocarditis/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Peroxidase/metabolism , Rats, Sprague-Dawley , Sepsis/complications , Signal Transduction , Stroke Volume/drug effects , Ventricular Function, Left
11.
Cardiovasc Ther ; 36(6): e12477, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30380183

ABSTRACT

AIMS: To investigate whether there exists a cardio-protective effect of Fasudil, a selective Rho kinase (ROCK) inhibitor, in an experimental murine model of acute viral myocarditis. METHODS: Male BALB/c mice were randomly assigned to three groups: control, myocarditis treated with placebo and myocarditis treated with Fasudil (n = 40 animals per group). Myocarditis was established by intraperitoneal injection with coxsackievirus B3 (CVB3). Twenty-four hours after infection, Fasudil was intraperitoneally administered for 14 consecutive days. Twenty mice were randomly selected from each group to monitor a 14-day survival rate. On day 7 and day 14, eight surviving mice from each group were sacrificed and their hearts and blood were obtained to perform serological and histological examinations. Expression of ROCKs, IL-17, IL-1b, TNFα, RORgt, and Foxp3 were quantified with RT-PCR. Plasma levels of TNF alpha, IL-1 beta, and IL-17 were measured by ELISA. In addition, protein levels of IL-17 and ROCK2 in cardiac tissues were analyzed with Western blot. RESULTS: Fasudil treatment significantly increased survival, attenuated myocardial necrotic lesions, reduced CVB3 replication and expression of ROCK2 and IL-17 in the infected hearts. This treatment also imposed a T-cell subpopulation shift, from Th17 to Treg, in cardiac tissues. CONCLUSIONS: ROCK pathway inhibition was cardio-protective in viral myocarditis with increased survival, decreased viral replication, and inflammatory response. These findings suggest that Fasudil might be a potential therapeutic agent for patients with viral myocarditis.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , Coxsackievirus Infections/prevention & control , Enterovirus B, Human/drug effects , Myocarditis/prevention & control , Myocardium/enzymology , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Coxsackievirus Infections/enzymology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Cytokines/blood , Disease Models, Animal , Enterovirus B, Human/growth & development , Inflammation Mediators/blood , Male , Mice, Inbred BALB C , Myocarditis/enzymology , Myocarditis/pathology , Myocarditis/virology , Myocardium/pathology , Necrosis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Th1 Cells/drug effects , Th1 Cells/metabolism , Th1 Cells/virology , Viral Load , Virus Replication/drug effects , rho-Associated Kinases/metabolism
12.
Basic Res Cardiol ; 113(4): 29, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29905892

ABSTRACT

In the cardiomyocyte, CaMKII has been identified as a nodal influencer of excitation-contraction and also excitation-transcription coupling. Its activity can be regulated in response to changes in intracellular calcium content as well as after several post-translational modifications. Some of the effects mediated by CaMKII may be considered adaptive, while effects of sustained CaMKII activity may turn into the opposite and are detrimental to cardiac integrity and function. As such, CaMKII has long been noted as a promising target for pharmacological inhibition, but the ubiquitous nature of CaMKII has made it difficult to target CaMKII specifically where it is detrimental. In this review, we provide a brief overview of the physiological and pathophysiological properties of CaMKII signaling, but we focus on the physiological and adaptive functions of CaMKII. Furthermore, special consideration is given to the emerging role of CaMKII as a mediator of inflammatory processes in the heart.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Chagas Cardiomyopathy/enzymology , Inflammation Mediators/metabolism , Myocarditis/enzymology , Myocardium/enzymology , Animals , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Chagas Cardiomyopathy/genetics , Chagas Cardiomyopathy/pathology , Chagas Cardiomyopathy/physiopathology , Enzyme Activation , Gene Expression Regulation, Enzymologic , Humans , Myocarditis/genetics , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/pathology , Necrosis , Protein Conformation , Signal Transduction , Structure-Activity Relationship
13.
Cardiovasc Diabetol ; 16(1): 61, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28476142

ABSTRACT

BACKGROUND: Diastolic dysfunction (DD), a hallmark of obesity and primary defect in heart failure with preserved ejection fraction, is a predictor of future cardiovascular events. We previously reported that linagliptin, a dipeptidyl peptidase-4 inhibitor, improved DD in Zucker Obese rats, a genetic model of obesity and hypertension. Here we investigated the cardioprotective effects of linagliptin on development of DD in western diet (WD)-fed mice, a clinically relevant model of overnutrition and activation of the renin-angiotensin-aldosterone system. METHODS: Female C56Bl/6 J mice were fed an obesogenic WD high in fat and simple sugars, and supplemented or not with linagliptin for 16 weeks. RESULTS: WD induced oxidative stress, inflammation, upregulation of Angiotensin II type 1 receptor and mineralocorticoid receptor (MR) expression, interstitial fibrosis, ultrastructural abnormalities and DD. Linagliptin inhibited cardiac DPP-4 activity and prevented molecular impairments and associated functional and structural abnormalities. Further, WD upregulated the expression of TRAF3IP2, a cytoplasmic adapter molecule and a regulator of multiple inflammatory mediators. Linagliptin inhibited its expression, activation of its downstream signaling intermediates NF-κB, AP-1 and p38-MAPK, and induction of multiple inflammatory mediators and growth factors that are known to contribute to development and progression of hypertrophy, fibrosis and contractile dysfunction. Linagliptin also inhibited WD-induced collagens I and III expression. Supporting these in vivo observations, linagliptin inhibited aldosterone-mediated MR-dependent oxidative stress, upregulation of TRAF3IP2, proinflammatory cytokine, and growth factor expression, and collagen induction in cultured primary cardiac fibroblasts. More importantly, linagliptin inhibited aldosterone-induced fibroblast activation and migration. CONCLUSIONS: Together, these in vivo and in vitro results suggest that inhibition of DPP-4 activity by linagliptin reverses WD-induced DD, possibly by targeting TRAF3IP2 expression and its downstream inflammatory signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cardiomyopathies/prevention & control , Diet, Western/adverse effects , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Linagliptin/pharmacology , Myocarditis/prevention & control , Myocardium/enzymology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cardiomyopathies/enzymology , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Cells, Cultured , Diastole , Disease Models, Animal , Down-Regulation , Female , Fibrosis , Mice, Inbred C57BL , Myocarditis/enzymology , Myocarditis/etiology , Myocarditis/physiopathology , Myocardium/ultrastructure , NF-kappa B/metabolism , Nitrosative Stress/drug effects , Obesity/etiology , Oxidative Stress/drug effects , Recovery of Function , Signal Transduction/drug effects , Time Factors , Transcription Factor AP-1/metabolism , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Biomed Pharmacother ; 88: 894-901, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28178619

ABSTRACT

OBJECTIVE: To observe the effect of QiShenYiQi pill (QSYQ) on myocardial collagen metabolism in experimental autoimmune myocarditis rats, and to explore its mechanism of action. METHODS: Lewis rats underwent the injection of myocardial myosin mixed with freund's complete adjuvant were randomized into three groups: model, valsartan and QSYQ groups. And we treated rats which were injected phosphate buffered saline (PBS) mixed with freund's complete adjuvant as control group. Rats were intervened and euthanized at 4 and 8 weeks. We use alkaline hydrolysis to detect the content of myocardial hydroxyproline (HYP), and ELISA to detect the level of serum procollagen type I carboxyterminal peptide (PICP), procollagen type III amino-terminal peptide (PIIINP), and collagen C telopeptide type I (CTX-I). Myocardial MMP-1 and TIMP-1 protein expression was detected by immunohistochemistry, and myocardial MMP-1 and TIMP-1 mRNA expression was detected by real-time qPCR. RESULTS: QSYQ reduced the content of myocardial HYP, and this reduction was greater over time. QSYQ also reduced the serum concentration of PICP, PIIINP, CTX-I and the PICP/PIIINP ratio, which further reduced over time, whereas its effect on lowering PICP was significantly greater than that of valsartan at 4 and 8 weeks, and lowering CTX-I was significantly greater than that of valsartan at 8 weeks. In addition, after 4 weeks, QSYQ enhanced the protein and mRNA expression of MMP-1 and TIMP-1, and its effect on highering TIMP-1 was significantly greater than that of valsartan, whereas there was no significant difference in the expression of myocardial MMP-1 or TIMP-1 at 8 weeks. QSYQ reduced the ratio of MMP-1/TIMP-1, which further reduced over time, and the effect of QYSQ was significantly greater than that of valsartan after 4 weeks. CONCLUSION: This study provides evidence that QSYQ can reduce the rate of myocardial collagen synthesis and degradation. It also effectively improved the degree of myocardial fibrosis in experimental autoimmune myocarditis rats and it had a tendency to have a greater effect with longer treatment duration, which is related to the mechanism of regulation of MMP-1 and TIMP-1 expression in the myocardial rat.


Subject(s)
Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Collagen/metabolism , Drugs, Chinese Herbal/therapeutic use , Myocarditis/drug therapy , Myocarditis/metabolism , Animals , Autoimmune Diseases/enzymology , Drugs, Chinese Herbal/pharmacology , Hydroxyproline/metabolism , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Myocarditis/enzymology , Myocardium/enzymology , Myocardium/metabolism , Myocardium/pathology , Proteolysis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred Lew , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
15.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(9): 984-91, 2016 Sep 28.
Article in Chinese | MEDLINE | ID: mdl-27640799

ABSTRACT

OBJECTIVE: To compare the changes in muscle enzyme between children with myocarditis and Duchene/Becker muscular dystrophy (DMD/BMD), and to seek the explanations for variation.
 METHODS: The retrospective analysis for 83 myocarditis children (myocarditis group) and 69 DMD/BMD children (DMD/BMD group), who were collected from Department of Pediatric of Shengjing Hospital affiliated to China Medical University since January 2008 to May 2015, was carried out. At the same time, 24 healthy children from the Department of Pediatric Development served as a control group. The examination indexes included creatine kinase (CK), creatine kinase-isoenzyme MB (CK-MB), creatine kinase isoenzyme MB mass (CK-MB mass), cardiac troponin I (cTnI) and high-sensitive-cTnT (hs-cTnT).
 RESULTS: 1) In the myocarditis group, the CK increased from 100 to 1 000 U/L, reached a peak after 5 days, which lasted for a week and then dropped to the normal; the CK-MB reached a peak after 5 to 7 days and dropped to the normal a month later; the CK-MB mass reached a peak on the first day and dropped to the normal after 3 weeks; the cTn reached to a peak after 5 days and dropped to the normal after about 17 days; hs-cTnT reached to a peak on the first day and dropped to the normal after about 19 days. 2) In the DMD/BMD group, the CK increased significantly and 27 cases had a CK value of more than 10 000 U/L. After the treatment for 1 to 2 weeks, their enzyme rose again after a slight drop. In terms of cTnI, 6 cases showed a moderate increase, 5 of them couldn't drop to the normal level until more than 3 weeks later; the hs-cTnT increased in the 45 cases, which lasted for more than 3 weeks in the 31 cases of them and showed a tendency of persisting increase.
 CONCLUSION: The cTnI and hs-cTnT rise significantly and possess wider observation window than CK and CK-MB mass in myocarditis children, with more sensitive and specific changes. The myocardial damage can occur before myasthenia and keep this trend for a long time in the DMD/BMD children. The trend of cTnI change in myocarditis children is similar to hs-cTnT, while hs-cTnT in DMD/BMD children is more sensitive than cTnI.


Subject(s)
Creatine Kinase, MB Form/metabolism , Creatine Kinase/metabolism , Muscular Dystrophy, Duchenne/enzymology , Myocarditis/enzymology , Troponin I/metabolism , Troponin T/metabolism , Biomarkers , Child , China , Creatine Kinase/blood , Creatine Kinase, MB Form/blood , Female , Humans , Male , Muscle Weakness/enzymology , Muscular Dystrophy, Duchenne/therapy , Myocarditis/therapy , Retrospective Studies , Time Factors , Troponin I/blood , Troponin T/blood
16.
Curr Res Transl Med ; 64(1): 21-7, 2016.
Article in English | MEDLINE | ID: mdl-27140596

ABSTRACT

BACKGROUND: Myocarditis, characterized by myocyte necrosis, fibrosis, and degeneration with mononuclear cell infiltration, always causes heart failure in patients. Phosphoinositide 3-kinase (PI3K) is a pivotal kinase known to regulate inflammatory responses in cardiac diseases. Although previous research has suggested that PI3K was involved in cardiac diseases such as myocardial infarction, it is still unclear whether the inhibition of PI3K is essential for the treatment of myosin-induced myocarditis. The aim of this study was to explore whether pharmacological blockade of PI3K is able to protect mice against experimental autoimmune myocarditis (EAM). MATERIALS AND METHODS: We used the cardiac myosin-induced murine EAM model to investigate the therapeutic effect of PI3K inhibitor LY294002 on autoimmune myocarditis in mice. RESULTS: LY294002 significantly alleviated EAM injury in mice, as indicated by the reduction of cardiac necrosis, inflammatory infiltrates, and CD3(+) T cells. LY294002 also decreased the expression of p-Akt upon cardiac myosin treatment in the cardiac tissue of the mice. In the present study, LY294002 resulted in a moderate reduction in absolute CD4(+) cell numbers and a significant decrease in the absolute numbers of CD8(+) cells. Consequently, LY294002 increased the CD4(+)/CD8(+) ratio compared with peptide treatment alone. CONCLUSION: This report provides evidence that PI3K inhibitor LY294002 has potent effects against cardiac injury caused by EAM, suggesting that it has therapeutic value for the treatment of myocarditis.


Subject(s)
Autoimmune Diseases/drug therapy , Chromones/therapeutic use , Morpholines/therapeutic use , Myocarditis/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/etiology , Autoimmune Diseases/pathology , Chromones/pharmacology , Cytokines/blood , Drug Evaluation, Preclinical , Female , Immunization , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Myocarditis/enzymology , Myocarditis/etiology , Myocarditis/pathology , Myocardium/immunology , Myocardium/pathology , Myosin Heavy Chains/immunology , Myosin Heavy Chains/toxicity , Peptide Fragments/immunology , Peptide Fragments/toxicity , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Ventricular Myosins/immunology , Ventricular Myosins/toxicity
17.
Clin Sci (Lond) ; 130(8): 625-41, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26795437

ABSTRACT

Fenofibrate (FF), as a peroxisome-proliferator-activated receptor α (PPARα) agonist, has been used clinically for decades to lower lipid levels. In the present study, we examined whether FF can be repurposed to prevent the pathogenesi of the heart in Type 1 diabetes and to describe the underlying mechanism of its action. Streptozotocin (STZ)-induced diabetic mice and their age-matched control mice were treated with vehicle or FF by gavage every other day for 3 or 6 months. FF prevented diabetes-induced cardiac dysfunction (e.g. decreased ejection fraction and hypertrophy), inflammation and remodelling. FF also increased cardiac expression of fibroblast growth factor 21 (FGF21) and sirtuin 1 (Sirt1) in non-diabetic and diabetic conditions. Deletion of FGF21 gene (FGF21-KO) worsened diabetes-induced pathogenic effects in the heart. FF treatment prevented heart deterioration in the wild-type diabetic mice, but could not do so in the FGF21-KO diabetic mice although the systemic lipid profile was lowered in both wild-type and FGF21-KO diabetic mice. Mechanistically, FF treatment prevented diabetes-impaired autophagy, reflected by increased microtubule-associated protein 1A/1B-light chain 3, in the wild-type diabetic mice but not in the FGF21-KO diabetic mice. Studies with H9C2 cells in vitro demonstrated that exposure to high glucose (HG) significantly increased inflammatory response, oxidative stress and pro-fibrotic response and also significantly inhibited autophagy. These effects of HG were prevented by FF treatment. Inhibition of either autophagy by 3-methyladenine (3MA) or Sirt1 by sirtinol (SI) abolished FF's prevention of HG-induced effects. These results suggested that FF could prevent Type 1 diabetes-induced pathological and functional abnormalities of the heart by increasing FGF21 that may up-regulate Sirt1-mediated autophagy.


Subject(s)
Autophagy/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetic Cardiomyopathies/prevention & control , Fenofibrate/pharmacology , Fibroblast Growth Factors/metabolism , Myocarditis/prevention & control , Myocardium/enzymology , Sirtuin 1/metabolism , Ventricular Remodeling/drug effects , Animals , Blood Glucose/metabolism , Cell Line , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/pathology , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Fibrosis , Histone Deacetylase Inhibitors/pharmacology , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/enzymology , Myocarditis/etiology , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/pathology , Oxidative Stress/drug effects , Rats , Signal Transduction/drug effects , Time Factors , Ventricular Function, Left/drug effects
18.
Biochim Biophys Acta ; 1863(7 Pt B): 1894-903, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26775585

ABSTRACT

Mammalian target of rapamycin (mTOR) is an evolutionary conserved kinase that senses the nutrient and energy status of cells, the availability of growth factors, stress stimuli and other cellular and environmental cues. It responds by regulating a range of cellular processes related to metabolism and growth in accordance with the available resources and intracellular needs. mTOR has distinct functions depending on its assembly in the structurally distinct multiprotein complexes mTORC1 or mTORC2. Active mTORC1 enhances processes including glycolysis, protein, lipid and nucleotide biosynthesis, and it inhibits autophagy. Reported functions for mTORC2 after growth factor stimulation are very diverse, are tissue and cell-type specific, and include insulin-stimulated glucose transport and enhanced glycogen synthesis. In accordance with its cellular functions, mTOR has been demonstrated to regulate cardiac growth in response to pressure overload and is also known to regulate cells of the immune system. The present manuscript presents recently obtained insights into mechanisms whereby mTOR may change anabolic, catabolic and stress response pathways in cardiomocytes and discusses how mTOR may affect inflammatory cells in the heart during hemodynamic stress. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Cardiomegaly/enzymology , Inflammation Mediators/metabolism , Myocarditis/enzymology , Myocytes, Cardiac/enzymology , TOR Serine-Threonine Kinases/metabolism , Animals , Cardiomegaly/drug therapy , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiovascular Agents/pharmacology , Humans , Myocarditis/drug therapy , Myocarditis/genetics , Myocarditis/pathology , Myocarditis/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Protein Biosynthesis , Protein Kinase Inhibitors/pharmacology , Proteolysis , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Ventricular Remodeling
19.
J Virol ; 89(20): 10512-23, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269170

ABSTRACT

UNLABELLED: Viral myocarditis, which is most prevalently caused by coxsackievirus B3 (CVB3), is a serious clinical condition characterized by excessive myocardial inflammation. Recent studies suggest that regulation of protein acetylation levels by inhibiting histone deacetylase (HDAC) activity modulates inflammatory response and shows promise as a therapy for several inflammatory diseases. However, the role of HDAC activity in viral myocarditis is still not fully understood. Here, we aim to investigate the role of HDAC activity in viral myocarditis and its underlying mechanism. CVB3-infected BALB/c mice were treated with the HDAC inhibitor (HDACI) suberoylanilide hydroxamic acid (SAHA) or trichostatin A (TSA). We found inhibition of HDAC activity aggravated rather than ameliorated the severity of CVB3-induced myocarditis, which was contrary to our expectations. The aggravated myocarditis by HDACI treatment seemed not to be caused by an elevated inflammatory response but by the increased CVB3 replication. Further, it was revealed that the increased CVB3 replication was closely associated with the HDACI-enhanced autophagosome formation. Inhibition of autophagosome formation by wortmannin or ATG5 short hairpin RNA dramatically suppressed the HDACI-increased CVB3 replication. The increased viral replication subsequently elevated CVB3-induced myocardial apoptosis. Conversely, inhibition of CVB3 replication and ensuing myocardial apoptosis by the antiviral drug ribavirin significantly reversed the HDACI-aggravated viral myocarditis. In conclusion, we elucidate that the inhibition of HDAC activity increases CVB3 replication and ensuing myocardial apoptosis, resulting in aggravated viral myocarditis. Possible adverse consequences of administering HDACI should be considered in patients infected (or coinfected) with CVB3. IMPORTANCE: Viral myocarditis, which is most prevalently caused by CVB3, is characterized by excessive myocardial inflammation. Inhibition of HDAC activity was originally identified as a powerful anti-cancer therapeutic strategy and was recently found to be implicated in the regulation of inflammatory response. HDACI has been demonstrated to be efficacious in animal models of several inflammatory diseases. Thus, we hypothesize that inhibition of HDAC activity also protects against CVB3-induced viral myocarditis. Surprisingly, we found inhibition of HDAC activity enhanced myocardial autophagosome formation, which led to the elevated CVB3 viral replication and ensuing increased myocardial apoptosis. Viral myocarditis was eventually aggravated rather than ameliorated by HDAC inhibition. In conclusion, we elucidate the role of HDAC activity in viral myocarditis. Moreover, given the importance of HDACI in preclinical and clinical treatments, the possible unfavorable effect of HDACI should be carefully evaluated in patients infected with viruses, including CVB3.


Subject(s)
Coxsackievirus Infections/virology , Enterovirus B, Human/pathogenicity , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylases/genetics , Myocarditis/virology , Virus Replication/drug effects , Androstadienes/pharmacology , Animals , Antiviral Agents/pharmacology , Apoptosis/drug effects , Autophagy-Related Protein 5 , Coxsackievirus Infections/enzymology , Coxsackievirus Infections/genetics , Coxsackievirus Infections/pathology , Disease Models, Animal , Enterovirus B, Human/drug effects , Enterovirus B, Human/physiology , Gene Expression Regulation , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Myocarditis/enzymology , Myocarditis/genetics , Myocarditis/pathology , Myocardium/enzymology , Myocardium/pathology , Phagosomes/chemistry , Phagosomes/drug effects , Phagosomes/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribavirin/pharmacology , Virus Replication/genetics , Vorinostat , Wortmannin
20.
Circ Res ; 117(6): 502-512, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26199323

ABSTRACT

RATIONALE: The development of molecular imaging approaches that assess specific immunopathologic mechanisms can advance the study of myocarditis. OBJECTIVE: This study validates a novel molecular imaging tool that enables the in vivo visualization of granzyme B activity, a major effector of cytotoxic CD8+ T lymphocytes. METHODS AND RESULTS: We synthesized and optimized a fluorogenic substrate capable of reporting on granzyme B activity and examined its specificity ex vivo in mice hearts with experimental cytotoxic CD8+ T lymphocyte-mediated myocarditis using fluorescence reflectance imaging, validated by histological examination. In vivo experiments localized granzyme B activity in hearts with acute myocarditis monitored by fluorescent molecular tomography in conjunction with coregistered computed tomography imaging. A model anti-inflammatory intervention (dexamethasone administration) in vivo reduced granzyme B activity (vehicle versus dexamethasone: 504±263 versus 194±77 fluorescence intensities in hearts; P=0.002). CONCLUSIONS: Molecular imaging of granzyme B activity can visualize T cell-mediated myocardial injury and monitor the response to an anti-inflammatory intervention.


Subject(s)
Granzymes/metabolism , Myocarditis/enzymology , Myocarditis/immunology , Animals , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , Enzyme Activation/physiology , Fluorescent Dyes/analysis , Granzymes/analysis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...