1
|
Hao G, Chen X, Fang Z, He Y, Liu M, Arora V, Dua A, Sun Z, Zhou B, Zheng G, Zuo L, Chen H, Zhu H, Dong Y. Association between prescription opioid use and heart failure: Cohort studies and Mendelian randomization analysis. Int J Cardiol 2024; 413:132404. [PMID: 39074619 DOI: 10.1016/j.ijcard.2024.132404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Prescription opioid use (POU) has been shown to lead to cardiovascular disease (CVD), but its association with heart failure has not been well studied. We investigated the potential causal association between POU and HF using cohort studies and Mendelian Randomization (MR) analysis. METHODS Initially, we examined the longitudinal association between POU and HF using the data from the Health and Retirement Study (HRS) and the UK biobank. Next, we employed a two-sample MR analysis using summary statistics from genome-wide association studies (GWAS) to assess the potential causal associations between POU and HF. RESULTS During a median of 3.8 and 13.8 years of follow-up, there were 441(8.04 per 1000 person-year) and 16,170 (3.96 per 1000 person-year) HF cases in the HRS and the UK biobank, respectively. After adjusting for covariates, participants who used prescription opioids had a 32% increased risk of developing HF, compared with non-users (HR = 1.32, 95%CI: 1.26-1.38, P < 0.001). In the MR analysis, summary statistics for POU were obtained from 78,808 UK Biobank study participants, and summary data for HF were obtained from 218,792 participants of a European population. A causal effect of genetic liability for POU on an increased risk of HF (OR = 1.16, 95% CI = 1.06, 1.27, P = 0.001) was suggested. The results were generally consistent in the sensitivity analysis, and no pleiotropy or heterogeneity were observed. CONCLUSIONS POU is associated with a high risk of HF. Our findings provide new insight into prescription opioid use among populations at risk of heart failure. More studies are needed to validate our results and further investigate the underlying mechanisms.
Collapse
Affiliation(s)
- Guang Hao
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Xia Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Zhenger Fang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Yunbiao He
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Mingliang Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Vishal Arora
- Department of Medicine: Cardiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Anterpreet Dua
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Zhuo Sun
- Department of Anesthesiology and Perioperative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Biying Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Guangjun Zheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Lei Zuo
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Haiyan Chen
- Control and Prevention, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Haidong Zhu
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yanbin Dong
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
2
|
Ganekal P, Vastrad B, Vastrad C, Kotrashetti S. Identification of biomarkers, pathways, and potential therapeutic targets for heart failure using next-generation sequencing data and bioinformatics analysis. Ther Adv Cardiovasc Dis 2023; 17:17539447231168471. [PMID: 37092838 PMCID: PMC10134165 DOI: 10.1177/17539447231168471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Heart failure (HF) is the most common cardiovascular diseases and the leading cause of cardiovascular diseases related deaths. Increasing molecular targets have been discovered for HF prognosis and therapy. However, there is still an urgent need to identify novel biomarkers. Therefore, we evaluated biomarkers that might aid the diagnosis and treatment of HF. METHODS We searched next-generation sequencing (NGS) dataset (GSE161472) and identified differentially expressed genes (DEGs) by comparing 47 HF samples and 37 normal control samples using limma in R package. Gene ontology (GO) and pathway enrichment analyses of the DEGs were performed using the g: Profiler database. The protein-protein interaction (PPI) network was plotted with Human Integrated Protein-Protein Interaction rEference (HiPPIE) and visualized using Cytoscape. Module analysis of the PPI network was done using PEWCC1. Then, miRNA-hub gene regulatory network and TF-hub gene regulatory network were constructed by Cytoscape software. Finally, we performed receiver operating characteristic (ROC) curve analysis to predict the diagnostic effectiveness of the hub genes. RESULTS A total of 930 DEGs, 464 upregulated genes and 466 downregulated genes, were identified in HF. GO and REACTOME pathway enrichment results showed that DEGs mainly enriched in localization, small molecule metabolic process, SARS-CoV infections, and the citric acid tricarboxylic acid (TCA) cycle and respiratory electron transport. After combining the results of the PPI network miRNA-hub gene regulatory network and TF-hub gene regulatory network, 10 hub genes were selected, including heat shock protein 90 alpha family class A member 1 (HSP90AA1), arrestin beta 2 (ARRB2), myosin heavy chain 9 (MYH9), heat shock protein 90 alpha family class B member 1 (HSP90AB1), filamin A (FLNA), epidermal growth factor receptor (EGFR), phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), cullin 4A (CUL4A), YEATS domain containing 4 (YEATS4), and lysine acetyltransferase 2B (KAT2B). CONCLUSIONS This discovery-driven study might be useful to provide a novel insight into the diagnosis and treatment of HF. However, more experiments are needed in the future to investigate the functional roles of these genes in HF.
Collapse
Affiliation(s)
- Prashanth Ganekal
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. College of Pharmacy, Gadag, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, #253, Bharthinagar, Dharwad 580001, India
| | | |
Collapse
|
3
|
Wang C, Sun S, Jiao J, Yu X, Huang S. Effects of delta-opioid receptor agonist pretreatment on the cardiotoxicity of bupivacaine in rats. BMC Anesthesiol 2022; 22:19. [PMID: 35021986 PMCID: PMC8753886 DOI: 10.1186/s12871-022-01568-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 01/04/2022] [Indexed: 11/27/2022] Open
Abstract
Background Delta-opioid receptor is widely expressed in human and rodent hearts, and has been proved to protect cardiomyocytes against ischemia/reperfusion and heart failure. The antagonist of delta-opioid receptor could block the rescue effect of lipid emulsion against local anesthetic cardiotoxicity. However, no evidence is available for the direct effect of delta-opioid-receptor agonists on the cardiotoxicity of local anesthetics. Methods Anesthetized Sprague Dawley rats were divided into five groups. Group NS received 2 ml·kg−1·min−1 normal saline, group LE received 2 ml·kg−1·min−1 30% lipid emulsion and group BW received 0.1, 1.0, or 5.0 mg/kg BW373U86, a delta-opioid-receptor agonist, for 5 min. Then 0.5% bupivacaine was infused intravenously at a rate of 3.0 mg·kg−1·min−1 until asystole. The time of arrhythmia, 50% mean arterial pressure-, 50% heart rate-reduction and asystole were recorded, and the dose of bupivacaine at each time point was calculated. Results All three different doses of BW373U86 did not affect the arrhythmia, 50% mean arterial pressure-reduction, 50% heart rate-reduction and asystole dose of bupivacaine compared with group NS. 30% LE significantly increased the bupivacaine threshold of 50% mean arterial pressure-reduction (17.9 [15.4–20.7] versus 7.2 [5.9–8.7], p = 0.018), 50% heart rate-reduction (18.7 ± 4.2 versus 8.8 ± 1.7, p < 0.001) and asystole (26.5 [21.0–29.1] versus 11.3 [10.7–13.4], p = 0.008) compared with group NS. There was no difference between group LE and group NS in the arrhythmia dose of bupivacaine (9.9 [8.9–11.7] versus 5.6 [4.5–7.0], p = 0.060). Conclusions Our data show that BW373U86 does not affect the cardiotoxicity of bupivacaine compared with NS control in rats. 30% LE pretreatment protects the myocardium against bupivacaine-induced cardiotoxicity.
Collapse
|
4
|
Dehe L, Shaqura M, Nordine M, Habazettl H, von Kwiatkowski P, Schluchter H, Shakibaei M, Mousa SA, Schäfer M, Treskatsch S. Chronic Naltrexone Therapy Is Associated with Improved Cardiac Function in Volume Overloaded Rats. Cardiovasc Drugs Ther 2021; 35:733-743. [PMID: 33484395 PMCID: PMC8266787 DOI: 10.1007/s10557-020-07132-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Myocardial opioid receptors were demonstrated in animals and humans and seem to colocalize with membranous and sarcolemmal calcium channels of the excitation-contraction coupling in the left ventricle (LV). Therefore, this study investigated whether blockade of the cardiac opioid system by naltrexone would affect cardiac function and neurohumoral parameters in Wistar rats with volume overload-induced heart failure. METHODS Volume overload in Wistar rats was induced by an aortocaval fistula (ACF). Left ventricular cardiac opioid receptors were identified by immunohistochemistry and their messenger ribonucleic acid (mRNA) as well as their endogenous ligand mRNA quantified by real-time polymerase chain reaction (RT-PCR). Following continuous delivery of either the opioid receptor antagonist naltrexone or vehicle via minipumps (n = 5 rats each), hemodynamic and humoral parameters were assessed 28 days after ACF induction. Sham-operated animals served as controls. RESULTS In ACF rats mu-, delta-, and kappa-opioid receptors colocalized with voltage-gated L-type Ca2+ channels in left ventricular cardiomyocytes. Chronic naltrexone treatment of ACF rats reduced central venous pressure (CVP) and left ventricular end-diastolic pressure (LVEDP), and improved systolic and diastolic left ventricular functions. Concomitantly, rat brain natriuretic peptide (rBNP-45) and angiotensin-2 plasma concentrations which were elevated during ACF were significantly diminished following naltrexone treatment. In parallel, chronic naltrexone significantly reduced mu-, delta-, and kappa-opioid receptor mRNA, while it increased the endogenous opioid peptide mRNA compared to controls. CONCLUSION Opioid receptor blockade by naltrexone leads to improved LV function and decreases in rBNP-45 and angiotensin-2 plasma levels. In parallel, naltrexone resulted in opioid receptor mRNA downregulation and an elevated intrinsic tone of endogenous opioid peptides possibly reflecting a potentially cardiodepressant effect of the cardiac opioid system during volume overload.
Collapse
Affiliation(s)
- Lukas Dehe
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Mohammed Shaqura
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Michael Nordine
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Helmut Habazettl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology Campus Charité Mitte, Chariteplatz 1, 10117, Berlin, Germany
| | - Petra von Kwiatkowski
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Helena Schluchter
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilians-Universität München, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Shaaban A Mousa
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Michael Schäfer
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Sascha Treskatsch
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
5
|
Su JY, Zhang RF, Dong YX, Yang MH, Yin XM, Gao LJ, Li HH, Xia YL, Yang YZ. Preprodynorphin gene mutation causes progressive cardiac conduction disease: A whole-exome analysis of a pedigree. Life Sci 2019; 219:74-81. [PMID: 30611784 DOI: 10.1016/j.lfs.2018.12.062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/18/2018] [Accepted: 12/31/2018] [Indexed: 10/27/2022]
Abstract
AIMS Progressive cardiac conduction disease (PCCD) is a rare heart disease that usually shows familial inheritance. Potential genetic risk factors for PCCD have been mostly limited to genes that encode ion channels, cardiac transcription factors, T-box transcription factors, gap junction proteins, energy metabolism regulators and structural proteins. MAIN METHODS Subjects in the present study came from a family who exhibited the autosomal dominant inheritance of PCCD. The primary proband had syncope and an electrocardiogram typical for PCCD, which started in the left bundle branch block, and passed to the atrioventricular block. The patient received a permanent pacemaker in 2013. Pathogenic mutations in the proband's family were identified using whole-exome sequencing and Sanger sequencing. KEY FINDINGS The results for the family members were verified using Sanger sequencing, while the results for healthy unrelated individuals were verified using SNaPShot. All patients in the family shared two adjacent missense mutations in the preprodynorphin (PDYN) gene (c.581A > T, c.580G > C; p.D194L). SIGNIFICANCE The PDYN double mutation c.581A > T and c.580G > C (p.D194L) may be linked to the onset of familial PCCD. The effects of these mutations on electrophysiology require further investigation.
Collapse
Affiliation(s)
- Jian-Yao Su
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Rong-Feng Zhang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Ying-Xue Dong
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Ming-Hui Yang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiao-Meng Yin
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lian-Jun Gao
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Disease, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yun-Long Xia
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Yan-Zong Yang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
6
|
Chen LT, Jiang CY. Bioinformatics analysis of sex differences in arrhythmogenic right ventricular cardiomyopathy. Mol Med Rep 2019; 19:2238-2244. [PMID: 30664203 PMCID: PMC6390068 DOI: 10.3892/mmr.2019.9873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited disease that exhibits sex differences on clinical presentation. The present study aimed to investigate the sex differences associated with ARVC by conducting an integrated bioinformatics analysis. The GSE29819 gene expression dataset was downloaded from the Gene Expression Omnibus database. The online analytical tool GEO2R was then used to screen for differentially expressed genes (DEGs), which were subsequently processed using enrichment analysis and protein‑protein interaction (PPI) network construction. Functional annotation of the DEGs was determined using ClueGO. The PPI network was constructed with Search Tool for the Retrieval of Interacting Genes, and was visualized with Cytoscape to identify the modules and hub genes. Compared with the female group, a total of 1,188 DEGs, of which 915 were upregulated and 273 were downregulated, were identified in the male group. The enrichment analysis revealed that in KEGG pathways, the upregulated DEGs were substantially enriched in the 'nicotine addiction' pathways, whereas the downregulated DEGS were mainly enriched in the 'ECM‑receptor interaction' and 'protein digestion and absorption' pathways. The PPI network contained 899 nodes and 1,627 edges, among which four significant modules were identified. In addition, kininogen 1, lysophosphatidic acid receptor 5, formyl peptide receptor (FPR) 2, adenylate cyclase 2, γ‑aminobutyric acid type B receptor subunit 2, FPR1, hydroxycarboxylic acid receptor 1, prostaglandin E receptor 3, cannabinoid receptor 1 and proenkephalin were identified as the top 10 hub genes. The key genes and related pathways identified in this study provide genetic insight into the diversity in phenotypes between female and male patients with ARVC, and may facilitate therapeutic individualization.
Collapse
Affiliation(s)
- Lai-Te Chen
- Department of Cardiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Chen-Yang Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
7
|
Sheng M, Zhang G, Wang J, Yang Q, Zhao H, Cheng X, Xu Z. Remifentanil Induces Cardio Protection Against Ischemia/Reperfusion Injury by Inhibiting Endoplasmic Reticulum Stress Through the Maintenance of Zinc Homeostasis. Anesth Analg 2018; 127:267-276. [PMID: 29771714 DOI: 10.1213/ane.0000000000003414] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although it is well known that remifentanil (Rem) elicits cardiac protection against ischemia/reperfusion (I/R) injury, the underlying mechanism remains unclear. This study tested if Rem can protect the heart from I/R injury by inhibiting endoplasmic reticulum (ER) stress through the maintenance of zinc (Zn) homeostasis. METHODS Isolated rat hearts were subjected to 30 minutes of regional ischemia followed by 2 hours of reperfusion. Rem was given by 3 consecutive 5-minute infusions, and each infusion was followed by a 5-minute drug-free perfusion before ischemia. Total Zn concentrations in cardiac tissue, cardiac function, infarct size, and apoptosis were assessed. H9c2 cells were subjected to 6 hours of hypoxia and 2 hours of reoxygenation (hypoxia/reoxygenation [H/R]), and Rem was given for 30 minutes before hypoxia. Metal-responsive transcription factor 1 (MTF1) overexpression plasmids were transfected into H9c2 cells 48 hours before hypoxia. Intracellular Zn level, cell viability, and mitochondrial injury parameters were evaluated. A Zn chelator N,N,N',N'-tetrakis-(2-pyridylmethyl) ethylenediamine (TPEN) or an ER stress activator thapsigargin was administrated during in vitro and ex vivo studies. The regulatory molecules related to Zn homeostasis and ER stress in cardiac tissue, and cardiomyocytes were analyzed by Western blotting. RESULTS Rem caused significant reversion of Zn loss from the heart (Rem + I/R versus I/R, 9.43 ± 0.55 vs 7.53 ± 1.18; P < .05) by suppressing the expression of MTF1 and Zn transporter 1 (ZnT1). The inhibited expression of ER stress markers after Rem preconditioning was abolished by TPEN. Rem preconditioning improved the cardiac function accompanied by the reduction of infarct size (Rem + I/R versus I/R, 21% ± 4% vs 40% ± 6%; P < .05). The protective effects of Rem could be reserved by TPEN and thapsigargin. Similar effects were observed in H9c2 cells exposed to H/R. In addition, MTF1 overexpression blocked the inhibitory effects of Rem on ZnT1 expression and ER stress at reoxygenation. Rem attenuated the collapse of mitochondrial membrane potential (ΔΨm) and the generation of mitochondrial reactive oxygen species by inhibiting ER stress via cardiac Zn restoration (Rem + H/R versus H/R, 79.57% ± 10.62% vs 58.27% ± 4.32%; P < .05). CONCLUSIONS Rem maintains Zn homeostasis at reperfusion by inhibiting MTF1 and ZnT1 expression, leading to the attenuation of ER stress and cardiac injury. Our findings provide a promising therapeutic approach for managing acute myocardial I/R injury.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cation Transport Proteins/genetics
- Cation Transport Proteins/metabolism
- Cell Line
- Cytoprotection
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Disease Models, Animal
- Endoplasmic Reticulum Stress/drug effects
- Homeostasis
- Isolated Heart Preparation
- Male
- Membrane Potential, Mitochondrial/drug effects
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/physiopathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Rats, Wistar
- Reactive Oxygen Species/metabolism
- Remifentanil/pharmacology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Ventricular Function, Left/drug effects
- Zinc/metabolism
- Transcription Factor MTF-1
Collapse
Affiliation(s)
- Mingwei Sheng
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
- Department of Anesthesiology, Tianjin First Center Hospital, Tianjin, China
| | - Ge Zhang
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jiannan Wang
- Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Qing Yang
- Department of Cardiology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Huanhuan Zhao
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xinxin Cheng
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Zhelong Xu
- From the Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
8
|
Abstract
The World Health Organization suggests that the cardiovascular diseases (CVDs) are the major cause of mortality and account for two-thirds of the deaths all over the world. These diseases kill about 17 million people every year and 3 in every 10 deaths are due to these diseases. The past decade has seen considerable improvements in diagnosis as well as treatment of various heart diseases. Various new therapeutic targets are being identified through in-depth knowledge of the disease mechanisms which has favored the testing of new strategies leading to newer treatment options. Opioid peptides and G-protein-coupled opioid receptors (ORs) have been previously studied widely in terms of central nervous system actions in mitigating the pain and drug abuse. The OR agonism or antagonism induces cytoprotective states in the myocardium, rendering these receptors as an attractive target for protection of heart from the fatal heart diseases. The opioids can provide an extended window of protection of the heart from various diseases. Although the mechanisms may not be fully understood, they seem to play a crucial role in various CVDs such as hypertension, hyperlipidemia, ischemic heart disease myocardial ischemia, and congestive heart failure. Since these compounds are already being used in acute and chronic pain, soon these compounds might be approved for use as cardioprotective agents. The following review focuses on the new information acquired on the role of the ORs in various CVDs.
Collapse
Affiliation(s)
- Hemangi Rawal
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | | |
Collapse
|
9
|
Abstract
This paper is the thirty-ninth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2016 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and CUNY Neuroscience Collaborative, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
10
|
He SF, Jin SY, Yang W, Pan YL, Huang J, Zhang SJ, Zhang L, Zhang Y. Cardiac μ-opioid receptor contributes to opioid-induced cardioprotection in chronic heart failure. Br J Anaesth 2018; 121:26-37. [PMID: 29935580 DOI: 10.1016/j.bja.2017.11.110] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/10/2017] [Accepted: 12/23/2017] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND The therapeutic potential of cardiac μ-opioid receptors in ischaemia-reperfusion (I/R) injury during opioid-modulating diseases, such as heart failure, is unknown. We aimed to explore the changes of cardiac μ-opioid receptor expression during heart failure, and its role in opioid-induced cardioprotection. METHODS Rats received doxorubicin (DOX) or were subjected to coronary artery ligation to induce heart failure, or received normal saline (NS) as control. Hearts from NS or DOX rats were isolated and subjected to myocardial ischaemia and reperfusion in an in vitro perfusion system. The opioid [D-Ala,2N-MePhe,4 Gly-ol]-enkephalin (DAMGO), with a high μ-opioid receptor specificity, morphine, and remifentanil were administrated before I/R with or without opioid receptor antagonists, or an extracellular signal-regulated kinase (ERK) inhibitor. RESULTS Cardiac μ-opioid receptor mRNA concentrations were 3.2 times elevated in DOX-treated rats compared with NS rats, while cardiac μ-opioid receptor protein concentrations showed 6.1- and 3.5-fold increases in DOX-treated and post-infarcted rats, respectively. DAMGO reduced I/R-caused infarct size, expressed as the ratio of area at risk, from 0.50 (0.04) to 0.25 (0.03) in failing rat hearts, but had no effect on infarct size in control hearts. DAMGO promoted phosphorylation of ERK and glycogen synthase kinase (GSK)-3β only in failing hearts. DAMGO-mediated cardioprotection was blocked by an ERK inhibitor. The μ-opioid receptor antagonist D-Pen-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) prevented morphine- and remifentanil-induced cardioprotection and phosphorylation of ERK and GSK-3β in failing hearts. In contrast, δ- and κ-opioid receptor selective antagonists were less potent than CTOP in the failing hearts. CONCLUSIONS Cardiac μ-opioid receptors were substantially up-regulated during heart failure, which increased DAMGO-induced cardioprotection against I/R injury.
Collapse
Affiliation(s)
- S F He
- Department of Anaesthesiology, The Second Hospital of Anhui Medical University, Hefei, China
| | - S Y Jin
- Department of Anaesthesiology, The Second Hospital of Anhui Medical University, Hefei, China
| | - W Yang
- Department of Anaesthesiology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Y L Pan
- Department of Anaesthesiology, The Second Hospital of Anhui Medical University, Hefei, China
| | - J Huang
- Department of Anaesthesiology, The Second Hospital of Anhui Medical University, Hefei, China
| | - S J Zhang
- Department of Ultrasound, The Second Hospital of Anhui Medical University, Hefei, China
| | - L Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Y Zhang
- Department of Anaesthesiology, The Second Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
11
|
Abstract
The opioid system is activated in heart failure, which may be cardioprotective but may also be counter-regulatory. Recently, systemic proenkephalin activation has been investigated in various conditions predicting mortality and kidney injury. In acute heart failure, proenkephalin independently predicts mortality and heart failure rehospitalization in addition to traditional risk markers. It also predicts worsening renal function, increasingly recognized as an important risk predictor for poor outcome in heart failure. This article explores the role of enkephalins and delta-opioid receptors in the heart, then reviews studies measuring proenkephalin levels in the circulation and their associations with prognosis.
Collapse
Affiliation(s)
- Daniel Chu Siong Chan
- Department of Cardiovascular Sciences, NIHR Biomedical Research Centre, University of Leicester, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK
| | - Thong Huy Cao
- Department of Cardiovascular Sciences, NIHR Biomedical Research Centre, University of Leicester, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK; Department of General Internal Medicine, University of Medicine and Pharmacy, Hong Bang Street, Ward 11, District 5, Ho Chi Minh City, Vietnam
| | - Leong Loke Ng
- Department of Cardiovascular Sciences, NIHR Biomedical Research Centre, University of Leicester, Glenfield Hospital, Groby Road, Leicester LE3 9QP, UK.
| |
Collapse
|
12
|
Treskatsch S, Shaqura M, Dehe L, Roepke TK, Shakibaei M, Schäfer M, Mousa SA. Evidence for MOR on cell membrane, sarcoplasmatic reticulum and mitochondria in left ventricular myocardium in rats. Heart Vessels 2015; 31:1380-8. [PMID: 26686371 DOI: 10.1007/s00380-015-0784-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/02/2015] [Indexed: 12/20/2022]
Abstract
Cardiac function is one important determinant to maintain tissue oxygenation and is thus highly regulated. In this context, it is interesting that centrally mediated opioidergic influence on cardiac function has long been known. Only recently, KOR and DOR have been found to be expressed in healthy left ventricular myocardium in rats and colocalized with parts of the excitation-contraction-coupling system. However, several comments in literature exist doubting the existence of MOR in cardiac tissue. We, therefore, aimed to detect MOR in rat left ventricular cardiomyocytes, and to evaluate whether MOR and POMC are regulated during heart failure. After IRB approval, heart failure was induced using a modified infrarenal aortocaval fistula (ACF) in male Wistar rats. All rats of the control and ACF group were characterized by their morphometrics and hemodynamics and the existence of MOR and POMC was investigated by means of radioligand binding, double immunofluorescence confocal analysis, RT-PCR and Western blot. Membrane MOR selective binding sites were detected in the left ventricular myocardium, however, they were lower in abundance than KOR- and DOR-specific binding sites and B max of MOR could not be determined. In left ventricular cardiomyocytes, MOR colocalized with parts of the excitation-coupling mechanism, e.g., Cav1.2 of the cell membrane and invaginated T-tubules as well as the ryanodine receptor of the sarcoplasmatic reticulum. More importantly, MOR strongly colocalized with mitochondria of left ventricular cardiomyocytes. Volume overload was not associated with an altered expression of MOR and POMC on both mRNA and protein level. These findings provide evidence for the existence of MOR on the cell membrane, sarcoplasmatic reticulum and mitochondria in left ventricular cardiomyocytes in rats. However, heart failure does not result in an altered expression of the cardiac MOR-opioid system. Thus, MOR agonist treatment-commonly used in the clinical setting-might directly affect cardiac function, which needs to be evaluated in greater detail in the near future.
Collapse
Affiliation(s)
- Sascha Treskatsch
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Mohammed Shaqura
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Lukas Dehe
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Torsten K Roepke
- Department of Cardiology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilians-Universität München, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Michael Schäfer
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Shaaban A Mousa
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|