Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Circ Res ; 130(1): 48-66, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34763521

ABSTRACT

BACKGROUND: Patients with acute myocardial infarction suffer systemic metabolic dysfunction via incompletely understood mechanisms. Adipocytes play critical role in metabolic homeostasis. The impact of acute myocardial infarction upon adipocyte function is unclear. Small extracellular vesicles (sEVs) critically contribute to organ-organ communication. Whether and how small extracellular vesicle mediate post-MI cardiomyocyte/adipocyte communication remain unknown. METHODS: Plasma sEVs were isolated from sham control (Pla-sEVSham) or 3 hours after myocardial ischemia/reperfusion (Pla-sEVMI/R) and incubated with adipocytes for 24 hours. Compared with Pla-sEVSham, Pla-sEVMI/R significantly altered expression of genes known to be important in adipocyte function, including a well-known metabolic regulatory/cardioprotective adipokine, APN (adiponectin). Pla-sEVMI/R activated 2 (PERK-CHOP and ATF6 [transcription factor 6]-EDEM [ER degradation enhancing alpha-mannosidase like protein 1] pathways) of the 3 endoplasmic reticulum (ER) stress pathways in adipocytes. These pathological alterations were also observed in adipocytes treated with sEVs isolated from adult cardiomyocytes subjected to in vivo myocardial ischemia/reperfusion (MI/R) (Myo-sEVMI/R). Bioinformatic/RT-qPCR analysis demonstrates that the members of miR-23-27-24 cluster are significantly increased in Pla-sEVMI/R, Myo-sEVMI/R, and adipose tissue of MI/R animals. Administration of cardiomyocyte-specific miR-23-27-24 sponges abolished adipocyte miR-23-27-24 elevation in MI/R animals, supporting the cardiomyocyte origin of adipocyte miR-23-27-24 cluster. In similar fashion to Myo-sEVMI/R, a miR-27a mimic activated PERK-CHOP and ATF6-EDEM-mediated ER stress. Conversely, a miR-27a inhibitor significantly attenuated Myo-sEVMI/R-induced ER stress and restored APN production. RESULTS: An unbiased approach identified EDEM3 (ER degradation enhancing alpha-mannosidase like protein 3) as a novel downstream target of miR-27a. Adipocyte EDEM3 deficiency phenocopied multiple pathological alterations caused by Myo-sEVMI/R, whereas EDEM3 overexpression attenuated Myo-sEVMI/R-resulted ER stress. Finally, administration of GW4869 or cardiomyocyte-specific miR-23-27-24 cluster sponges attenuated adipocyte ER stress, improved adipocyte endocrine function, and restored plasma APN levels in MI/R animals. CONCLUSIONS: We demonstrate for the first time that MI/R causes significant adipocyte ER stress and endocrine dysfunction by releasing miR-23-27-24 cluster-enriched small extracellular vesicle. Targeting small extracellular vesicle-mediated cardiomyocyte-adipocyte pathological communication may be of therapeutic potential to prevent metabolic dysfunction after MI/R.


Subject(s)
Adipocytes/metabolism , Cell Communication , Endoplasmic Reticulum Stress , Extracellular Vesicles/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Activating Transcription Factor 6/metabolism , Adiponectin/metabolism , Animals , Male , Membrane Proteins/metabolism , Mice , MicroRNAs/metabolism , Transcription Factor CHOP/metabolism , eIF-2 Kinase/metabolism
2.
Circulation ; 141(12): 968-983, 2020 03 24.
Article in English | MEDLINE | ID: mdl-31918577

ABSTRACT

BACKGROUND: Diabetes mellitus exacerbates myocardial ischemia/reperfusion (MI/R) injury by incompletely understood mechanisms. Adipocyte dysfunction contributes to remote organ injury. However, the molecular mechanisms linking dysfunctional adipocytes to increased MI/R injury remain unidentified. The current study attempted to clarify whether and how small extracellular vesicles (sEV) may mediate pathological communication between diabetic adipocytes and cardiomyocytes, exacerbating MI/R injury. METHODS: Adult male mice were fed a normal or a high-fat diet for 12 weeks. sEV (from diabetic serum, diabetic adipocytes, or high glucose/high lipid-challenged nondiabetic adipocytes) were injected intramyocardially distal of coronary ligation. Animals were subjected to MI/R 48 hours after injection. RESULTS: Intramyocardial injection of diabetic serum sEV in the nondiabetic heart significantly exacerbated MI/R injury, as evidenced by poorer cardiac function recovery, larger infarct size, and greater cardiomyocyte apoptosis. Similarly, intramyocardial or systemic administration of diabetic adipocyte sEV or high glucose/high lipid-challenged nondiabetic adipocyte sEV significantly exacerbated MI/R injury. Diabetic epididymal fat transplantation significantly increased MI/R injury in nondiabetic mice, whereas administration of a sEV biogenesis inhibitor significantly mitigated MI/R injury in diabetic mice. A mechanistic investigation identified that miR-130b-3p is a common molecule significantly increased in diabetic serum sEV, diabetic adipocyte sEV, and high glucose/high lipid-challenged nondiabetic adipocyte sEV. Mature (but not primary) miR-130b-3p was significantly increased in the diabetic and nondiabetic heart subjected to diabetic sEV injection. Whereas intramyocardial injection of a miR-130b-3p mimic significantly exacerbated MI/R injury in nondiabetic mice, miR-130b-3p inhibitors significantly attenuated MI/R injury in diabetic mice. Molecular studies identified AMPKα1/α2, Birc6, and Ucp3 as direct downstream targets of miR-130b-3p. Overexpression of these molecules (particularly AMPKα2) reversed miR-130b-3p induced proapoptotic/cardiac harmful effect. Finally, miR-130b-3p levels were significantly increased in plasma sEV from patients with type 2 diabetes mellitus. Incubation of cardiomyocytes with diabetic patient sEV significantly exacerbated ischemic injury, an effect blocked by miR-130b-3p inhibitor. CONCLUSIONS: We demonstrate for the first time that miR-130b-3p enrichment in dysfunctional adipocyte-derived sEV, and its suppression of multiple antiapoptotic/cardioprotective molecules in cardiomyocytes, is a novel mechanism exacerbating MI/R injury in the diabetic heart. Targeting miR-130b-3p mediated pathological communication between dysfunctional adipocytes and cardiomyocytes may be a novel strategy attenuating diabetic exacerbation of MI/R injury.


Subject(s)
Adipocytes/metabolism , Diabetes Mellitus, Experimental/physiopathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Animals , Humans , Male , Mice
3.
Circulation ; 140(9): 751-764, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31220942

ABSTRACT

BACKGROUND: Myocardial ischemia-reperfusion (MI/R) injury is a significant clinical problem without effective therapy. Unbiased omics approaches may reveal key MI/R mediators to initiate MI/R injury. METHODS: We used a dynamic transcriptome analysis of mouse heart exposed to various MI/R periods to identify S100a8/a9 as an early mediator. Using loss/gain-of-function approaches to understand the role of S100a8/a9 in MI/R injury, we explored the mechanisms through transcriptome and functional experiment. Dynamic serum S100a8/a9 levels were measured in patients with acute myocardial infarction before and after percutaneous coronary intervention. Patients were prospectively followed for the occurrence of major adverse cardiovascular events. RESULTS: S100a8/a9 was identified as the most significantly upregulated gene during the early reperfusion stage. Knockout of S100a9 markedly decreased cardiomyocyte death and improved heart function, whereas hematopoietic overexpression of S100a9 exacerbated MI/R injury. Transcriptome/functional studies revealed that S100a8/a9 caused mitochondrial respiratory dysfunction in cardiomyocytes. Mechanistically, S100a8/a9 downregulated NDUF gene expression with subsequent mitochondrial complex I inhibition via Toll-like receptor 4/Erk-mediated Pparg coactivator 1 alpha/nuclear respiratory factor 1 signaling suppression. Administration of S100a9 neutralizing antibody significantly reduced MI/R injury and improved cardiac function. Finally, we demonstrated that serum S100a8/a9 levels were significantly increased 1 day after percutaneous coronary intervention in patients with acute myocardial infarction, and elevated S100a8/a9 levels were associated with the incidence of major adverse cardiovascular events. CONCLUSIONS: Our study identified S100a8/a9 as a master regulator causing cardiomyocyte death in the early stage of MI/R injury via the suppression of mitochondrial function. Targeting S100a8/a9-intiated signaling may represent a novel therapeutic intervention against MI/R injury. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov. Unique identifier: NCT03752515.


Subject(s)
Apoptosis , Calgranulin B/metabolism , Mitochondria/metabolism , Myocardial Reperfusion Injury/pathology , Animals , Antibodies, Neutralizing/administration & dosage , Calgranulin A/blood , Calgranulin B/genetics , Calgranulin B/immunology , Disease Models, Animal , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Heart Failure/etiology , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/diagnosis , Myocardial Infarction/surgery , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Percutaneous Coronary Intervention , Signal Transduction
4.
Circulation ; 135(21): 2041-2057, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28249877

ABSTRACT

BACKGROUND: Hypertensive ventricular remodeling is a common cause of heart failure. However, the molecular mechanisms regulating ventricular remodeling remain poorly understood. METHODS: We used a discovery-driven/nonbiased approach to identify increased activating transcription factor 3 (ATF3) expression in hypertensive heart. We used loss/gain of function approaches to understand the role of ATF3 in heart failure. We also examined the mechanisms through transcriptome, chromatin immunoprecipitation sequencing analysis, and in vivo and in vitro experiments. RESULTS: ATF3 expression increased in murine hypertensive heart and human hypertrophic heart. Cardiac fibroblast cells are the primary cell type expressing high ATF3 levels in response to hypertensive stimuli. ATF3 knockout (ATF3KO) markedly exaggerated hypertensive ventricular remodeling, a state rescued by lentivirus-mediated/miRNA-aided cardiac fibroblast-selective ATF3 overexpression. Conversely, conditional cardiac fibroblast cell-specific ATF3 transgenic overexpression significantly ameliorated ventricular remodeling and heart failure. We identified Map2K3 as a novel ATF3 target. ATF3 binds with the Map2K3 promoter, recruiting HDAC1, resulting in Map2K3 gene-associated histone deacetylation, thereby inhibiting Map2K3 expression. Genetic Map2K3 knockdown rescued the profibrotic/hypertrophic phenotype in ATF3KO cells. Last, we demonstrated that p38 is the downstream molecule of Map2K3 mediating the profibrotic/hypertrophic effects in ATF3KO animals. Inhibition of p38 signaling reduced transforming growth factor-ß signaling-related profibrotic and hypertrophic gene expression, and blocked exaggerated cardiac remodeling in ATF3KO cells. CONCLUSIONS: Our study provides the first evidence that ATF3 upregulation in cardiac fibroblasts in response to hypertensive stimuli protects the heart by suppressing Map2K3 expression and subsequent p38-transforming growth factor-ß signaling. These results suggest that positive modulation of cardiac fibroblast ATF3 may represent a novel therapeutic approach against hypertensive cardiac remodeling.


Subject(s)
Activating Transcription Factor 3/metabolism , Fibroblasts/enzymology , Heart Failure/prevention & control , Hypertrophy, Left Ventricular/prevention & control , MAP Kinase Kinase 3/metabolism , Myocardium/enzymology , Ventricular Function, Left , Ventricular Remodeling , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylation , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Angiotensin II , Animals , Binding Sites , Cells, Cultured , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Genetic Predisposition to Disease , Heart Failure/enzymology , Heart Failure/etiology , Heart Failure/physiopathology , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , Hypertension/chemically induced , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/physiopathology , MAP Kinase Kinase 3/genetics , Male , Mice, Knockout , Myocardium/pathology , Phenotype , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
Cells ; 11(5)2022 02 23.
Article in English | MEDLINE | ID: mdl-35269401

ABSTRACT

Background The impairment of the inner blood-retinal barrier (iBRB) increases the pathological development of diabetic retinopathy (DR), a severe complication in diabetic patients. Identifying approaches to preserving iBRB integrity and function is a significant challenge in DR. C1q/tumor necrosis factor-related protein-3 (CTRP3) is a newly discovered adipokine and a vital biomarker, predicting DR severity. We sought to determine whether and how CTRP3 affects the pathological development of non-proliferative diabetic retinopathy (NPDR). Methods To clarify the pathophysiologic progress of the blood-retinal barrier in NPDR and explore its potential mechanism, a mouse Type 2 diabetic model of diabetic retinopathy was used. The capillary leakage was assessed by confocal microscope with fluorescent-labeled protein in vivo. Furthermore, the effect of CTRP3 on the inner blood-retinal barrier (iBRB) and its molecular mechanism was clarified. Results The results demonstrated that CTRP3 protects iBRB integrity and resists the vascular permeability induced by DR. Mechanistically, the administration of CTRP3 activates the AMPK signaling pathway and enhances the expression of Occludin and Claudin-5 (tight junction protein) in vivo and in vitro. Meanwhile, CTRP3 improves the injury of human retinal endothelial cells (HRMECs) induced by high glucose/high lipids (HG/HL), and its protective effects are AMPK-dependent. Conclusions In summary, we report, for the first time, that CTRP3 prevents diabetes-induced retinal vascular permeability via stabilizing the tight junctions of the iBRB and through the AMPK-dependent Occludin/Claudin-5 signaling pathway, thus critically affecting the development of NPDR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , AMP-Activated Protein Kinases/metabolism , Animals , Blood-Retinal Barrier , Claudin-5 , Complement C1q/metabolism , Diabetes Mellitus/metabolism , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Humans , Mice , Occludin , Tight Junctions/metabolism
7.
J Gerontol A Biol Sci Med Sci ; 73(8): 1010-1017, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29415197

ABSTRACT

Previous studies have demonstrated that aging promotes myocardial apoptosis after ischemia/reperfusion, via unknown specific mechanisms. The present study investigates the potential relationship between lncRNAs and aging-related apoptosis by lncRNA/mRNA microarray technology. The results indicate aging increased myocardial lncRNA ENSMUST00000134285 and mMAPK11, confirmed by both bioinformatics analysis and polymerase chain reaction (PCR). Mouse cardiomyocytes were subjected to gene manipulation (ENSMUST00000134285 knockdown and overexpression). Knockdown of ENSMUST00000134285 inhibited MAPK11 activity and increased the myocardial apoptotic ratio (determined by TUNEL staining and caspase activity assays) after hypoxia/reoxygenation. Conversely, overexpression of ENSMUST00000134285 increased MAPK11 activity and decreased the myocardial apoptotic ratio. Furthermore, luciferase reporter assay revealed that miR760 may be a mediator between ENSMUST00000134285 and mMAPK11. We have provided evidence that lncRNAs are the important regulatory molecules in aging-mediated effects upon apoptosis. The apoptosis regulatory effects of aging are complex. Except apoptosis-promoting effects, aging could also inhibit myocardial apoptosis after hypoxia or ischemia. Further studies investigating the mechanisms that aging inhibit myocardial apoptosis after hypoxia/ischemia.


Subject(s)
Aging/metabolism , Mitogen-Activated Protein Kinase 11/metabolism , Myocardium/metabolism , RNA, Long Noncoding/physiology , Animals , Apoptosis , Blotting, Western , Mice , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
8.
Eur J Emerg Med ; 21(5): 364-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24368406

ABSTRACT

OBJECTIVES: Previous studies have reported that civilian transport is a mortality risk factor in low-resource communities. Few studies have analyzed the chief complaints associated with mortality involving civilian transport after an earthquake.Therefore, the present study was conducted to determine whether mortality resulting from medical professional transport differs from that involving civilian transport, and if so, the chief complaints associated with mortality involving civilian transport after the Wen-chuan earthquake. METHODS: A hospital-based case-control study was conducted. Cases included all victims transported by civilians to West China Hospital from the disaster area (n=473). Controls included all victims transported by medical professionals to West China Hospital (n=1452). We further analyze six potential chief complaints of death to clarify the specific contributing chief complaints associated with mortality involving civilian transport. RESULTS: Civilian transport is associated with significantly greater mortality compared with medical professional transport (Pearson's χ-test: P<0.05). Patients with altered mental status had the greatest risk of death [odds ratio (OR)=4.552, 95% confidence interval (CI)=2.165-9.572], followed by patients with trunk injury (OR=2.517, 95% CI=1.251-5.066), and finally patients with shortness of breath (OR=2.345, 95% CI=1.040-5.288). CONCLUSION: Altered mental status, trunk injury, and shortness of breath were the significant chief complaints associated with mortality involving civilian transport to the hospital after the Wen-chuan earthquake. Our data suggest that patients with any of these complaints should be transported by medical professionals, not civilians, to the nearest hospital for treatment.


Subject(s)
Disasters/statistics & numerical data , Earthquakes/mortality , Transportation of Patients/statistics & numerical data , Adult , Case-Control Studies , China/epidemiology , Emergency Medical Services/statistics & numerical data , Female , Humans , Male , Middle Aged , Transportation of Patients/methods , Transportation of Patients/standards , Wounds and Injuries/mortality
9.
Nat Commun ; 5: 3303, 2014.
Article in English | MEDLINE | ID: mdl-24526256

ABSTRACT

Interferon regulatory factor 8 (IRF8) is known to affect the innate immune response, for example, by regulating the differentiation and function of immune cells. However, whether IRF8 can influence cardiac hypertrophy is unknown. Here we show that IRF8 levels are decreased in human dilated/hypertrophic cardiomyopathic hearts and in murine hypertrophic hearts. Mice overexpressing Irf8 specifically in the heart are resistant to aortic banding (AB)-induced cardiac hypertrophy, whereas mice lacking IRF8 either globally or specifically in cardiomyocytes develop an aggravated phenotype induced by pressure overload. Mechanistically, we show that IRF8 directly interacts with NFATc1 to prevent NFATc1 translocation and thus inhibits the hypertrophic response. Inhibition of NFATc1 ameliorates the cardiac abnormalities in IRF8(-/-) mice after AB. In contrast, constitutive activation of NFATc1 nullifies the protective effects of IRF8 on cardiac hypertrophy in IRF8-overexpressing mice. Our results indicate that IRF8 is a potential therapeutic target in pathological cardiac hypertrophy.


Subject(s)
Calcineurin/metabolism , Interferon Regulatory Factors/metabolism , Animals , Calcineurin/genetics , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Female , Humans , Interferon Regulatory Factors/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Oligopeptides/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL