1
|
Obal D, Liu Y. Opioid Preconditioning in Heart Failure: New Frontier or Old Dog? Anesth Analg 2025:00000539-990000000-01152. [PMID: 39908198 DOI: 10.1213/ane.0000000000007388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Affiliation(s)
- Detlef Obal
- From the Department of Anesthesia, University of Iowa, Iowa City, Iowa
| | - Yu Liu
- Stanford University School of Medicine, Stanford, California
| |
Collapse
|
2
|
Guo C, Pan X, Dou M, Wu J, Chen X, Wang B, Zhu R, Xu S, Peng W, Wu C, He S, Zhang S, Zhang Y, Jin S. The activated caveolin-3/μ-opioid receptor complex drives morphine-induced rescue therapy in failing hearts. Br J Pharmacol 2025; 182:651-669. [PMID: 39427683 DOI: 10.1111/bph.17326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/19/2024] [Accepted: 07/13/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Opioid analgesics can alleviate ischaemia/reperfusion (I/R) injury in chronic heart failure. However, the underlying mechanisms and targets remain unknown. Here, we investigate if caveolin-3 (Cav3) interacts with μ opioid receptors and if Cav3-μ receptor interactions play a role in morphine-induced cardioprotection in failing hearts. EXPERIMENTAL APPROACH Cav3 and μ receptor proteins in human and rat heart tissue were determined by western blot, immunofluorescence and co-immunoprecipitation. Methyl-β-cyclodextrin (MβCD), a destroyer of caveolae, and AAV-Cav3 shRNA were used to reduce Cav3 expression in failing rat hearts. CTOP, a specific μ antagonist, was administrated before morphine preconditioning in perfused failing heart models of myocardial I/R injury. KEY RESULTS Levels of Cav3 and μ receptor proteins were significantly higher in human and rat myocardial tissues with heart failure than in control tissues. Cav3 and μ receptor expression levels were positively correlated with disease severity. The signal of the cardiac Cav3 protein was colocalized with μ receptor in both the human and rat heart sections. Disruption of caveolae in the failing heart by either MβCD or AAV-Cav3 shRNA significantly inhibits morphine-induced phosphorylation of ERK1/2 and cardioprotection. Administration of CTOP substantially reduced Cav3 expression and morphine-induced cardioprotective effect in heart failure. CONCLUSION AND IMPLICATIONS Our data suggest that up-regulation of the Cav3/μ receptor complex is critical for morphine protection of the failing heart against I/R injury by regulating the ERK1/2 pathway. The activated Cav3/μ receptor complex is an understudied therapeutic target for opioid treatment of heart failure and ischaemic insult.
Collapse
Affiliation(s)
- Chengxiao Guo
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Xinxin Pan
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Mengyun Dou
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Juan Wu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinyu Chen
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Baoli Wang
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Rui Zhu
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Shijin Xu
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Wenyi Peng
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Chao Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Shufang He
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Ye Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Shiyun Jin
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Biswal N, Harish R, Roshan M, Samudrala S, Jiao X, Pestell RG, Ashton AW. Role of GPCR Signaling in Anthracycline-Induced Cardiotoxicity. Cells 2025; 14:169. [PMID: 39936961 PMCID: PMC11817789 DOI: 10.3390/cells14030169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 02/13/2025] Open
Abstract
Anthracyclines are a class of chemotherapeutics commonly used to treat a range of cancers. Despite success in improving cancer survival rates, anthracyclines have dose-limiting cardiotoxicity that prevents more widespread clinical utility. Currently, the therapeutic options for these patients are limited to the iron-chelating agent dexrazoxane, the only FDA-approved drug for anthracycline cardiotoxicity. However, the clinical use of dexrazoxane has failed to replicate expectations from preclinical studies. A limited list of GPCRs have been identified as pathogenic in anthracycline-induced cardiotoxicity, including receptors (frizzled, adrenoreceptors, angiotensin II receptors) previously implicated in cardiac remodeling in other pathologies. The RNA sequencing of iPSC-derived cardiac myocytes from patients has increased our understanding of the pathogenic mechanisms driving cardiotoxicity. These data identified changes in the expression of novel GPCRs, heterotrimeric G proteins, and the regulatory pathways that govern downstream signaling. This review will capitalize on insights from these experiments to explain aspects of disease pathogenesis and cardiac remodeling. These data provide a cornucopia of possible unexplored potential pathways by which we can reduce the cardiotoxic side effects, without compromising the anti-cancer effects, of doxorubicin and provide new therapeutic options to improve the recovery and quality of life for patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Nimish Biswal
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Ritika Harish
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
| | - Minahil Roshan
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Sathvik Samudrala
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
| | - Xuanmao Jiao
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
| | - Richard G. Pestell
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Anthony W. Ashton
- School of Medicine, Xavier University at Aruba, Oranjestad, Aruba (X.J.); (R.G.P.)
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Wynnewood, PA 19096, USA;
- Division of Perinatal Research, Kolling Institute of Medical Research, University of Sydney, St Leonards, NSW 2065, Australia
- Division of Cardiovascular Medicine, Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| |
Collapse
|
4
|
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
|
5
|
Peng L, Li S, Cai H, Chen X, Tang Y. Ginsenoside Rg1 treats chronic heart failure by downregulating ERK1/2 protein phosphorylation. In Vitro Cell Dev Biol Anim 2024; 60:1085-1098. [PMID: 39251466 DOI: 10.1007/s11626-024-00960-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
In this study, we investigated the potential therapeutic mechanism of ginsenoside Rg1 (GRg1) in chronic heart failure (CHF), focusing on its regulation of ERK1/2 protein phosphorylation. H9c2 cardiomyocytes and SD rats were divided into the control group, CHF (ADR) group, and CHF+ginsenoside Rg1 group using an isolated cardiomyocyte model and an in vivo CHF rat model induced by adriamycin (ADR). Cell viability, proliferation, apoptosis, and the expression of relevant proteins were measured to assess the effects of GRg1. The results showed that treatment with GRg1 increased cell activity and proliferation, while significantly reducing levels of inflammatory and apoptotic factors compared to the CHF (ADR) group. Moreover, the CHF+ginsenoside Rg1 group exhibited higher levels of Bcl-2 mRNA and protein expression, as well as lower levels of Caspase3 and Bax mRNA and protein expression, compared to the CHF (ADR) group. Notably, the CHF+ginsenoside Rg1 group displayed decreased serum NT-proBNP levels and heart weight/body weight (HW/BW) index. Furthermore, the electrocardiogram of rats in the CHF+ginsenoside Rg1 group resembled that of rats in the control group. Overall, our findings suggested that GRg1 alleviated CHF by inhibiting ERK1/2 protein phosphorylation, thereby inhibiting apoptosis, enhancing cell activity and proliferation, and reducing cardiac inflammatory responses.
Collapse
Affiliation(s)
- Liqi Peng
- First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Shaodong Li
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Huzhi Cai
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Xueliang Chen
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yanping Tang
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
6
|
Tan Y, Li J, Nie Y, Zheng Z. Novel Approach for Cardioprotection: In Situ Targeting of Metformin via Conductive Hydrogel System. Polymers (Basel) 2024; 16:2226. [PMID: 39125251 PMCID: PMC11314979 DOI: 10.3390/polym16152226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/18/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Ischemia/reperfusion (I/R) injury following myocardial infarction is a major cause of cardiomyocyte death and impaired cardiac function. Although clinical data show that metformin is effective in repairing cardiac I/R injury, its efficacy is hindered by non-specific targeting during administration, a short half-life, frequent dosing, and potential adverse effects on the liver and kidneys. In recent years, injectable hydrogels have shown substantial potential in overcoming drug delivery challenges and treating myocardial infarction. To this end, we developed a natural polymer hydrogel system comprising methacryloylated chitosan and methacryloylated gelatin modified with polyaniline conductive derivatives. In vitro studies demonstrated that the optimized hydrogel exhibited excellent injectability, biocompatibility, biodegradability, suitable mechanical properties, and electrical conductivity. Incorporating metformin into this hydrogel significantly extended the administration cycle, mitigated mitochondrial damage, decreased abnormal ROS production, and enhanced cardiomyocyte function. Animal experiments indicated that the metformin/hydrogel system reduced arrhythmia incidence, infarct size, and improved cardiac mitochondrial and overall cardiac function, promoting myocardial repair in I/R injury. Overall, the metformin-loaded conductive hydrogel system effectively mitigates mitochondrial oxidative damage and improves cardiomyocyte function, thereby offering a theoretical foundation for the potential application of metformin in cardioprotection.
Collapse
Affiliation(s)
| | | | - Yali Nie
- Hunan Provincial Key Laboratory of Multi-Omics and Artificial Intelligence of Cardiovascular Diseases & Institute of Cardiovascular Disease & Department of Cardiology, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Zhi Zheng
- Hunan Provincial Key Laboratory of Multi-Omics and Artificial Intelligence of Cardiovascular Diseases & Institute of Cardiovascular Disease & Department of Cardiology, Hengyang Medical School, University of South China, Hengyang 421001, China
| |
Collapse
|
7
|
Sun M, Mao S, Wu C, Zhao X, Guo C, Hu J, Xu S, Zheng F, Zhu G, Tao H, He S, Hu J, Zhang Y. Piezo1-Mediated Neurogenic Inflammatory Cascade Exacerbates Ventricular Remodeling After Myocardial Infarction. Circulation 2024; 149:1516-1533. [PMID: 38235590 DOI: 10.1161/circulationaha.123.065390] [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: 08/01/2023] [Accepted: 12/19/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Heart failure is associated with a high rate of mortality and morbidity, and ventricular remodeling invariably precedes heart failure. Ventricular remodeling is fundamentally driven by mechanotransduction that is regulated by both the nervous system and the immune system. However, it remains unknown which key molecular factors govern the neuro/immune/cardio axis that underlies mechanotransduction during ventricular remodeling. Here, we investigated whether the mechanosensitive Piezo cation channel-mediated neurogenic inflammatory cascade underlies ventricular remodeling-related mechanotransduction. METHODS By ligating the left coronary artery of rats to establish an in vivo model of chronic myocardial infarction (MI), lentivirus-mediated thoracic dorsal root ganglion (TDRG)-specific Piezo1 knockdown rats and adeno-associated virus-PHP.S-mediated TDRG neuron-specific Piezo1 knockout mice were used to investigate whether Piezo1 in the TDRG plays a functional role during ventricular remodeling. Subsequently, neutralizing antibody-mediated TDRG IL-6 (interleukin-6) inhibition rats and adeno-associated virus-PHP.S-mediated TDRG neuron-specific IL-6 knockdown mice were used to determine the mechanism underlying neurogenic inflammation. Primary TDRG neurons were used to evaluate Piezo1 function in vitro. RESULTS Expression of Piezo1 and IL-6 was increased, and these factors were functionally activated in TDRG neurons at 4 weeks after MI. Both knockdown of TDRG-specific Piezo1 and deletion of TDRG neuron-specific Piezo1 lessened the severity of ventricular remodeling at 4 weeks after MI and decreased the level of IL-6 in the TDRG or heart. Furthermore, inhibition of TDRG IL-6 or knockdown of TDRG neuron-specific IL-6 also ameliorated ventricular remodeling and suppressed the IL-6 cascade in the heart, whereas the Piezo1 level in the TDRG was not affected. In addition, enhanced Piezo1 function, as reflected by abundant calcium influx induced by Yoda1 (a selective agonist of Piezo1), led to increased release of IL-6 from TDRG neurons in mice 4 weeks after MI. CONCLUSIONS Our findings point to a critical role for Piezo1 in ventricular remodeling at 4 weeks after MI and reveal a neurogenic inflammatory cascade as a previously unknown facet of the neuronal immune signaling axis underlying mechanotransduction.
Collapse
Affiliation(s)
- Meiyan Sun
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Weifang Medical University, China (M.S.)
| | - Sui Mao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Chao Wu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Xiaoyong Zhao
- Department of Anesthesiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China (X.Z.)
| | - Chengxiao Guo
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Jun Hu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Shijin Xu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, China (F.Z., G.Z.)
| | - Guoqing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, China (F.Z., G.Z.)
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Shufang He
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| | - Ji Hu
- Laboratory of Stress Neurobiology, School of Life Science and Technology, ShanghaiTech University, China (J.H.)
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
- Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China (M.S., S.M., C.W., C.G., J.H., S.X., H.T., S.H., Y.Z.)
| |
Collapse
|
8
|
Hu J, Song Y, Huang X, Li C, Jin X, Cen L, Zhang C, Ding B, Lian J. Opioids-Induced Long QT Syndrome: A Challenge to Cardiac Health. Cardiovasc Toxicol 2024; 24:472-480. [PMID: 38630336 PMCID: PMC11076354 DOI: 10.1007/s12012-024-09853-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/28/2024] [Indexed: 05/08/2024]
Abstract
The challenge posed by opioid overdose has become a significant concern for health systems due to the complexities associated with drug prohibition, widespread clinical use, and potential abuse. In response, healthcare professionals have primarily concentrated on mitigating the hallucinogenic and respiratory depressant consequences of opioid overdose to minimize associated risks. However, it is crucial to acknowledge that most opioids possess the capacity to prolong the QT interval, particularly in cases of overdose, thereby potentially resulting in severe ventricular arrhythmias and even sudden death if timely intervention is not implemented. Consequently, alongside addressing the typical adverse effects of opioids, it is imperative to consider their cardiotoxicity. To enhance comprehension of the correlation between opioids and arrhythmias, identify potential targets for prompt intervention, and mitigate the hazards associated with clinical utilization, an exploration of the interaction between drugs and ion channels, as well as their underlying mechanisms, becomes indispensable. This review primarily concentrates on elucidating the impact of opioid drugs on diverse ion channels, investigating recent advancements in this domain, and attaining a deeper understanding of the mechanisms underlying the prolongation of the QT interval by opioid drugs, along with potential interventions.
Collapse
Affiliation(s)
- Jiale Hu
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
| | - Yongfei Song
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, No. 378 Dongqing Road, Yinzhou District, Ningbo, 315000, Zhejiang, China
| | - Xiaoyan Huang
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, No. 378 Dongqing Road, Yinzhou District, Ningbo, 315000, Zhejiang, China
| | - Chongrong Li
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
| | - Xiaojun Jin
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
| | - Lichao Cen
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
| | - Chuanjin Zhang
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
| | - Beilei Ding
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China
| | - Jiangfang Lian
- Department of Cardiology, Ningbo University Health Science Center Affiliated Lihuili Hospital, Ningbo University, Zhejiang, China.
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, No. 378 Dongqing Road, Yinzhou District, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
9
|
Liu L, Sun Y, Wang Y, Xin J, Chen W. [D-Ala2, D-Leu5]-enkephalin (DADLE) provides protection against myocardial ischemia reperfusion injury by inhibiting Wnt/β-Catenin pathway. BMC Cardiovasc Disord 2024; 24:115. [PMID: 38373914 PMCID: PMC10877899 DOI: 10.1186/s12872-024-03790-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Acute myocardial infarction is one of the leading causes of death worldwide. Myocardial ischemia reperfusion (MI/R) injury occurs immediately after the coronary reperfusion and aggravates myocardial ischemia. Whether the Wnt/β-Catenin pathway is involved in the protection against MI/R injury by DADLE has not been evaluated. Therefore, the present study aimed to investigate the protective effect of DADLE against MI/R injury in a mouse model and to further explore the association between DADLE and the Wnt/β-Catenin pathway. METHODS Forty-four mice were randomly allocated to four groups: Group Control (PBS Control), Group D 0.25 (DADLE 0.25 mg/kg), Group D 0.5 (DADLE 0.5 mg/kg), and Group Sham. In the control and DADLE groups, myocardial ischemia injury was induced by occluding the left anterior descending coronary artery (LAD) for 45 min. PBS and DADLE were administrated, respectively, 5 min before reperfusion. The sham group did not go through LAD occlusion. 24 h after reperfusion, functions of the left ventricle were assessed through echocardiography. Myocardial injury was evaluated using TTC double-staining and HE staining. Levels of myocardial enzymes, including CK-MB and LDH, in the serum were determined using ELISA kits. Expression of caspase-3, TCF4, Wnt3a, and β-Catenin was evaluated using the Western blot assay. RESULTS The infarct area was significantly smaller in the DADLE groups than in the control group (P < 0.01). The histopathology score and serum levels of myocardial enzymes were significantly lower in the DADLE groups than in the control group (P < 0.01). DADLE significantly improved functions of the left ventricle (P < 0.01), decreased expression of caspase-3 (P < 0.01), TCF4 (P < 0.01), Wnt3a (P < 0.05), and β-Catenin (P < 0.01) compared with PBS. CONCLUSIONS The present study showed that DADLE protected the myocardium from MI/R through suppressing the expression of caspase-3, TCF4, Wnt3a, and β-Catenin and consequently improving functions of the left ventricle in I/R model mice. The TCF4/Wnt/β-Catenin signaling pathway might become a therapeutic target for MI/R treatment.
Collapse
Affiliation(s)
- Linwen Liu
- Department of Cardiology, Shanghai Fourth People's Hospital Affiliated to Tongji University, 1279 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Yawu Sun
- Department of Cardiology, Shanghai Fourth People's Hospital Affiliated to Tongji University, 1279 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Yang Wang
- Department of Pathology, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China
| | - Jun Xin
- Department of Ultrasonics, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, 200434, China
| | - Wei Chen
- Department of Cardiology, Shanghai Fourth People's Hospital Affiliated to Tongji University, 1279 Sanmen Road, Hongkou District, Shanghai, 200434, China.
| |
Collapse
|
10
|
Gumede NAC, Khathi A. The Role of Pro-Opiomelanocortin Derivatives in the Development of Type 2 Diabetes-Associated Myocardial Infarction: Possible Links with Prediabetes. Biomedicines 2024; 12:314. [PMID: 38397916 PMCID: PMC10887103 DOI: 10.3390/biomedicines12020314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Myocardial infarction is a major contributor to CVD-related mortality. T2DM is a risk factor for MI. Stress activates the HPA axis, SNS, and endogenous OPS. These POMC derivatives increase the blood glucose and cardiovascular response by inhibiting the PI3K/AkT insulin signaling pathway and increasing cardiac contraction. Opioids regulate the effect of the HPA axis and SNS and they are cardioprotective. The chronic activation of the stress response may lead to insulin resistance, cardiac dysfunction, and MI. Stress and T2DM, therefore, increase the risk of MI. T2DM is preceded by prediabetes. Studies have shown that prediabetes is associated with an increased risk of MI because of inflammation, hyperlipidemia, endothelial dysfunction, and hypertension. The HPA axis is reported to be dysregulated in prediabetes. However, the SNS and the OPS have not been explored during prediabetes. The effect of prediabetes on POMC derivatives has yet to be fully explored and understood. The impact of stress and prediabetes on the cardiovascular response needs to be investigated. This study sought to review the potential impact of prediabetes on the POMC derivatives and pathways that could lead to MI.
Collapse
Affiliation(s)
- Nompumelelo Anna-Cletta Gumede
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban X54001, South Africa;
| | | |
Collapse
|
11
|
Yi S, Cao H, Zheng W, Wang Y, Li P, Wang S, Zhou Z. Targeting the opioid remifentanil: Protective effects and molecular mechanisms against organ ischemia-reperfusion injury. Biomed Pharmacother 2023; 167:115472. [PMID: 37716122 DOI: 10.1016/j.biopha.2023.115472] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/18/2023] Open
Abstract
Opioids are widely used in clinical practice by activating opioid receptors (OPRs), but their clinical application is limited by a series of side effects. Researchers have been making tremendous efforts to promote the development and application of opioids. Fortunately, recent studies have identified the additional effects of opioids in addition to anesthesia and analgesia, particularly in terms of organ protection against ischemia-reperfusion (I/R) injury, with unique advantages. I/R injury in vital organs not only leads to cell dysfunction and structural damage but also induces acute and chronic organ failure, even death. Early prevention and appropriate therapeutic targets for I/R injury are crucial for organ protection. Opioids have shown cardioprotective effects for over 20 years, especially remifentanil, a derivative of fentanyl, which is a new ultra-short-acting opioid analgesic widely used in clinical anesthesia induction and maintenance. In this review, we provide current knowledge about the physiological effects related to OPR-mediated organ protection, focusing on the protective effect and mechanism of remifentanil on I/R injury in the heart and other vital organs. Herein, we also explored the potential application of remifentanil in clinical I/R injury. These findings provide a theoretical basis for the use of remifentanil to inhibit or alleviate organ I/R injury during the perioperative period and provide insights for opioid-induced human organ protection and drug development.
Collapse
Affiliation(s)
- Shuyuan Yi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China; Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China; School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Hong Cao
- Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China
| | - Weilei Zheng
- Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Shoushi Wang
- Department of Anaesthesiology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao 266042, China.
| | - Zhixia Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
12
|
Degro CE, Jiménez-Vargas NN, Tsang Q, Yu Y, Guzman-Rodriguez M, Alizadeh E, Hurlbut D, Reed DE, Lomax AE, Stein C, Bunnett NW, Vanner SJ. Evolving acidic microenvironments during colitis provide selective analgesic targets for a pH-sensitive opioid. Pain 2023; 164:2501-2515. [PMID: 37326658 PMCID: PMC10731875 DOI: 10.1097/j.pain.0000000000002956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 04/25/2023] [Indexed: 06/17/2023]
Abstract
ABSTRACT Targeting the acidified inflammatory microenvironment with pH-sensitive opioids is a novel approach for managing visceral pain while mitigating side effects. The analgesic efficacy of pH-dependent opioids has not been studied during the evolution of inflammation, where fluctuating tissue pH and repeated therapeutic dosing could influence analgesia and side effects. Whether pH-dependent opioids can inhibit human nociceptors during extracellular acidification is unexplored. We studied the analgesic efficacy and side-effect profile of a pH-sensitive fentanyl analog, (±)- N -(3-fluoro-1-phenethylpiperidine-4-yl)- N -phenyl propionamide (NFEPP), during the evolution of colitis induced in mice with dextran sulphate sodium. Colitis was characterized by granulocyte infiltration, histological damage, and acidification of the mucosa and submucosa at sites of immune cell infiltration. Changes in nociception were determined by measuring visceromotor responses to noxious colorectal distension in conscious mice. Repeated doses of NFEPP inhibited nociception throughout the course of disease, with maximal efficacy at the peak of inflammation. Fentanyl was antinociceptive regardless of the stage of inflammation. Fentanyl inhibited gastrointestinal transit, blocked defaecation, and induced hypoxemia, whereas NFEPP had no such side effects. In proof-of-principle experiments, NFEPP inhibited mechanically provoked activation of human colonic nociceptors under acidic conditions mimicking the inflamed state. Thus, NFEPP provides analgesia throughout the evolution of colitis with maximal activity at peak inflammation. The actions of NFEPP are restricted to acidified layers of the colon, without common side effects in normal tissues. N -(3-fluoro-1-phenethylpiperidine-4-yl)- N -phenyl propionamide could provide safe and effective analgesia during acute colitis, such as flares of ulcerative colitis.
Collapse
Affiliation(s)
- Claudius E. Degro
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
- Department of General and Visceral Surgery, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | | | - Quentin Tsang
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
| | - Yang Yu
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
| | - Mabel Guzman-Rodriguez
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
| | - Elahe Alizadeh
- Queen’s Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, Ontario, Canada
| | - David Hurlbut
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
- Department of Pathology and Molecular Medicine, Queeńs University, Kingston, Ontario, Canada
| | - David E. Reed
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
| | - Alan E. Lomax
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
| | - Christoph Stein
- Department of Experimental Anaesthesiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Nigel W. Bunnett
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, USA
- Department of Neuroscience and Physiology, Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York, USA
| | - Stephen J. Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queeńs University, Kingston, Ontario, Canada
| |
Collapse
|
13
|
de Sousa JT, Dihl RR, Menezes Boaretto FB, Garcia ALH, Grivicich I, da Silva J, Picada JN. Morphine decreases cytotoxicity and mutagenicity of doxorubicin in vitro: Implications for cancer chemotherapy. Chem Biol Interact 2023; 382:110652. [PMID: 37524295 DOI: 10.1016/j.cbi.2023.110652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/21/2023] [Accepted: 07/29/2023] [Indexed: 08/02/2023]
Abstract
Morphine is the most common opioid analgesic administered to treat pain in patients undergoing cancer chemotherapy. This study aimed to evaluate the cytotoxic and mutagenic effects of morphine alone and in combination with doxorubicin (Dox), an antineoplastic agent largely used in patients with solid cancers. Cytotoxicity was evaluated in neuroblastoma (SH-SY5Y) and fibroblast (V79) cells using 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) colorimetric assay while mutagenicity was assessed using the Salmonella/microsome assay in the absence and in the presence of S9 mix. Morphine showed a cytotoxic effect mainly on SH-SY5Y cells and reduced the cytotoxic effects of Dox when evaluated in a co-treatment procedure. In the Salmonella/microsome assay, it was observed that morphine did not induce mutations and, in fact, decreased the mutagenic effects induced by Dox in TA98 and TA102 strains in the absence of metabolic activation. Furthermore, in the presence of metabolic activation, no induction of mutations was observed with morphine. In conclusion, morphine decreased Dox cytotoxicity in both neuronal and non-neuronal cells and showed antimutagenic effects in the TA102 strain which detects mutagens inducing DNA oxidative damages. However, morphine decreased frameshift mutations induced by Dox in non-cytotoxic concentrations, an effect suggesting interference of Dox intercalation activity that could decrease its chemotherapeutic efficacy. These compelling findings highlight the importance of conducting further studies to explore the potential implications of co-administering morphine and Dox during cancer chemotherapy.
Collapse
Affiliation(s)
- Jayne Torres de Sousa
- Laboratory of Genetic Toxicology, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, RS, Brazil; Laboratory of Genetic Toxicity and Cellular Toxic-Genetic Analysis, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, Brazil
| | - Rafael Rodrigues Dihl
- Laboratory of Genetic Toxicity and Cellular Toxic-Genetic Analysis, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, Brazil
| | - Fernanda Brião Menezes Boaretto
- Laboratory of Genetic Toxicology, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, RS, Brazil
| | - Ana Leticia Hilário Garcia
- Laboratory of Genetic Toxicology, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, RS, Brazil; Laboratory of Genetics Toxicology, La Salle University, Av. Victor Barreto, 2288, 92010-000, Canoas, RS, Brazil
| | - Ivana Grivicich
- Laboratory of Cancer Biology, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, RS, Brazil
| | - Juliana da Silva
- Laboratory of Genetic Toxicology, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, RS, Brazil; Laboratory of Genetics Toxicology, La Salle University, Av. Victor Barreto, 2288, 92010-000, Canoas, RS, Brazil
| | - Jaqueline Nascimento Picada
- Laboratory of Genetic Toxicology, Graduate Program in Molecular and Cellular Biology Applied to Health, Lutheran University of Brazil, Av. Farroupilha 8001, 92425-900, Canoas, RS, Brazil.
| |
Collapse
|
14
|
Coluzzi F, LeQuang JAK, Sciacchitano S, Scerpa MS, Rocco M, Pergolizzi J. A Closer Look at Opioid-Induced Adrenal Insufficiency: A Narrative Review. Int J Mol Sci 2023; 24:ijms24054575. [PMID: 36902007 PMCID: PMC10003084 DOI: 10.3390/ijms24054575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Among several opioid-associated endocrinopathies, opioid-associated adrenal insufficiency (OIAI) is both common and not well understood by most clinicians, particularly those outside of endocrine specialization. OIAI is secondary to long-term opioid use and differs from primary adrenal insufficiency. Beyond chronic opioid use, risk factors for OIAI are not well known. OIAI can be diagnosed by a variety of tests, such as the morning cortisol test, but cutoff values are not well established and it is estimated that only about 10% of patients with OIAI will ever be properly diagnosed. This may be dangerous, as OIAI can lead to a potentially life-threatening adrenal crisis. OIAI can be treated and for patients who must continue opioid therapy, it can be clinically managed. OIAI resolves with opioid cessation. Better guidance for diagnosis and treatment is urgently needed, particularly in light of the fact that 5% of the United States population has a prescription for chronic opioid therapy.
Collapse
Affiliation(s)
- Flaminia Coluzzi
- Department Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy
- Unit of Anaesthesia, Intensive Care, and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy
- Correspondence:
| | | | - Salvatore Sciacchitano
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, 00166 Rome, Italy
| | - Maria Sole Scerpa
- Unit of Anaesthesia, Intensive Care, and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Monica Rocco
- Unit of Anaesthesia, Intensive Care, and Pain Medicine, Sant’Andrea University Hospital, 00189 Rome, Italy
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | | |
Collapse
|
15
|
Dziemitko S, Harasim-Symbor E, Chabowski A. How do phytocannabinoids affect cardiovascular health? An update on the most common cardiovascular diseases. Ther Adv Chronic Dis 2023; 14:20406223221143239. [PMID: 36636553 PMCID: PMC9830002 DOI: 10.1177/20406223221143239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 11/17/2022] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular disease (CVD) causes millions of deaths worldwide each year. Despite the great progress in therapies available for patients with CVD, some limitations, including drug complications, still exist. Hence, the endocannabinoid system (ECS) was proposed as a new avenue for CVDs treatment. The ECS components are widely distributed through the body, including the heart and blood vessels, thus the action of its endogenous and exogenous ligands, in particular, phytocannabinoids play a key role in various pathological states. The cardiovascular action of cannabinoids is complex as they affect vasculature and myocardium directly via specific receptors and exert indirect effects through the central and peripheral nervous system. The growing interest in phytocannabinoid studies, however, has extended the knowledge about their molecular targets as well as therapeutical properties; nonetheless, some areas of their actions are not yet fully recognized. Researchers have reported various cannabinoids, especially cannabidiol, as a promising approach to CVDs; hence, the purpose of this review is to summarize and update the cardiovascular actions of the most potent phytocannabinoids and the potential therapeutic role of ECS in CVDs, including ischemic reperfusion injury, arrhythmia, heart failure as well as hypertension.
Collapse
Affiliation(s)
- Sylwia Dziemitko
- Department of Physiology, Medical University of
Bialystok, Bialystok 15-222, Poland
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of
Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of
Bialystok, Bialystok, Poland
| |
Collapse
|
16
|
Luo Z, Hu X, Wu C, Chan J, Liu Z, Guo C, Zhu R, Zhang L, Zhang Y, Jin S, He S. Plasma exosomes generated by ischaemic preconditioning are cardioprotective in a rat heart failure model. Br J Anaesth 2023; 130:29-38. [PMID: 36347723 PMCID: PMC9875906 DOI: 10.1016/j.bja.2022.08.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Exosomes released into the plasma after brief cardiac ischaemia mediate subsequent cardioprotection. Whether donor exosomes can provide cardioprotection to recipients with chronic heart failure, which confers the highest perioperative risk, is unknown. We examined whether ischaemic preconditioning (IPC)-induced plasma exosomes exerted cardioprotection after their transfer from normal donors to post-infarcted failing hearts. METHODS Plasma exosomes were obtained from adult rats after IPC or sham. An exosome inhibitor GW4869 was administrated before IPC in an in vivo model of ischaemia/reperfusion (I/R) injury in normal rats. The IPC exosomes or control exosomes from normal donor rats were perfused to the normal or post-infarcted failing rat hearts before ischaemia in Langendorff perfusion experiments. Infarct size, cardiac enzymes, cardiac function, and pro-survival kinases were quantified. RESULTS The IPC stimulus increased the release of exosomes, whereas GW4869 inhibited the rise of plasma exosomes. Pre-treatment with GW4869 reversed IPC-mediated cardioprotection against in vivo I/R injury. In the Langendorff perfusion experiments, IPC exosomes from normal donor rats reduced mean infarct size from 41.05 (1.87)% to 31.43 (1.81)% and decreased lactate dehydrogenase activity in the post-infarcted failing rat hearts. IPC exosomes but not control exosomes activated pro-survival kinases in the heart tissues. CONCLUSIONS Ischaemic preconditioning-induced exosomes from normal rats can restore cardioprotection in heart failure after myocardial infarction, which is associated with activation of pro-survival protein kinases. These results suggest a potential perioperative therapeutic role for ischaemic preconditioning-induced exosomes.
Collapse
Affiliation(s)
- Zhaofei Luo
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Xudong Hu
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Chao Wu
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jinzhong Chan
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Zhong Liu
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Chengxiao Guo
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Rui Zhu
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Li Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Ye Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Shiyun Jin
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.
| | - Shufang He
- Department of Anesthesiology, Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.
| |
Collapse
|
17
|
Rousseau B, Murugan S, Palagani A, Sarkar DK. Beta 2 adrenergic receptor and mu opioid receptor interact to potentiate the aggressiveness of human breast cancer cell by activating the glycogen synthase kinase 3 signaling. Breast Cancer Res 2022; 24:33. [PMID: 35568869 PMCID: PMC9107672 DOI: 10.1186/s13058-022-01526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Opioid and beta-adrenergic receptors are recently shown to cross talk via formation of receptor heterodimers to control the growth and proliferation of breast cancer cells. However, the underlying cell signaling mechanism remained unclear. METHODS To determine the effect of the interaction of the two systems in breast cancer, we employed triple-negative breast cancer cell lines MDA-MB-231 and MDA-MB-468, CRISPR or chemical inhibition or activation of beta-adrenergic receptors (B2AR) and mu-opioid receptors (MOR) gene, and PCR array technology and studied aggressive tumor phenotype and signaling cascades. RESULTS We show here that in triple-negative breast cancer cells, the reduction in expression B2AR and MOR by genetic and pharmacological tools leads to a less aggressive phenotype of triple-negative breast cancer cells in vitro and in animal xenografts. Genomic analysis indicates the glycogen synthase kinase 3 (GSK3) pathway as a possible candidate messenger system involved in B2AR and MOR cross talk. GSK3 inactivation in MDA-MB-231 and MDA-MB-468 cells induced similar phenotypic changes as the inhibition of B2AR and/or MOR, while a GSK3 activation by wortmannin reversed the effects of B2AR and/or MOR knockdown on these cells. GSK3 inactivation also prevents B2AR agonist norepinephrine or MOR agonist DAMGO from affecting MDA-MB-231 and MDA-MB-468 cell proliferation. CONCLUSIONS These data confirm a role of B2AR and MOR interaction in the control of breast cancer cell growth and identify a possible role of the GSK3 signaling system in mediation of these two receptors' cross talk. Screening for ligands targeting B2AR and MOR interaction and/or the GSK3 system may help to identify novel drugs for the prevention of triple-negative breast cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Bénédicte Rousseau
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Sengottuvelan Murugan
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Ajay Palagani
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA
| | - Dipak K Sarkar
- Endocrine Research Program, Department of Animal Sciences, Rutgers, The State University of New Jersey, 67 Poultry Farm Lane, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
18
|
Wang C, Sun S, Jiao J, Yu X, Huang S. Effects of nalbuphine on the cardiotoxicity of ropivacaine in rats. Fundam Clin Pharmacol 2022; 36:811-817. [PMID: 35373856 DOI: 10.1111/fcp.12778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/15/2022] [Accepted: 04/01/2022] [Indexed: 11/28/2022]
Abstract
When combined with nalbuphine, local anesthetics show a longer duration of nerve block without increasing complications. However, no evidence is available concerning the effect of nalbuphine on the cardiotoxicity of local anesthetics. The objective of this work is to investigate whether nalbuphine pretreatment can increase the lethal dose threshold of ropivacaine in rats. Anesthetized Sprague Dawley rats were pretreated with different doses of nalbuphine (0.4, 0.8, 1.5, 3.0, 5.0 mg/kg) or NS (normal saline, negative control) or 30% LE (lipid emulsion, positive control) 2 ml/kg/min for 5 min (n = 6). Then 0.5% ropivacaine was infused at a rate of 2.5 mg/kg/min until asystole occurs. Time of arrhythmia, 50% mean arterial pressure- and 50% heart rate-reduction, and asystole were recorded, and ropivacaine doses were calculated. Nalbuphine (0.4-5.0 mg/kg) did not affect ropivacaine-induced arrhythmia, 50% mean arterial pressure-reduction and 50% heart rate-reduction, and asystole in rats compared with NS pre-treatment. The asystole dose threshold (in milligrams per kilogram) of group LE was higher than that of group NS (NS 28.25(6.32) vs. LE, 41.58(10.65); P = 0.04; 95% confidence interval 0.23 to 26.45), while thresholds of arrhythmia, 50% mean arterial pressure-reduction, and 50% heart rate-reduction were not affected by LE. Nalbuphine doses of 0.4-5.0 mg/kg pretreatment did not increase the threshold of ropivacaine cardiotoxicity compared with NS control; 30% LE increases the lethal dose threshold of ropivacaine in rats.
Collapse
Affiliation(s)
- Chenran Wang
- Department of Anesthesia, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Shen Sun
- Department of Anesthesia, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Jing Jiao
- Department of Anesthesia, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Xinhua Yu
- Division of Epidemiology, Biostatistics and Environmental Health, Scholl of Public Health, University of Memphis, Memphis, Tennessee, USA
| | - Shaoqiang Huang
- Department of Anesthesia, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
19
|
Kolpakov AR, Knyazev RA. Endogenous Cardiotonics: Search And Problems. Cardiovasc Hematol Disord Drug Targets 2021; 21:95-103. [PMID: 33874876 DOI: 10.2174/1871529x21666210419121807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/04/2021] [Accepted: 02/15/2021] [Indexed: 11/22/2022]
Abstract
Medicinal preparations currently used for the treatment of patients with chronic cardiac failure involve those that reduce the heart load (vasodilators, diuretics, beta-blockers, and angiotensin-converting enzyme (ACE) inhibitors). Cardiotonic drugs with the cAMP-dependent mechanism are unsuitable for long-term administration due to the intensification of metabolic processes and an increase in the oxygen demand of the myocardium and all tissues of the body. For many years, digoxin has remained the only preparation enhancing the efficiency of myocardial performance. The detection of digoxin and ouabain in intact animals has initiated a search for other compounds with cardiotonic activity. The review summarizes current data on the effect exerted on the heart performance by endogenous compounds, from simple, such as NO and CO, to steroids, fatty acids, polypeptides, and proteins. Controversial questions and problems with the introduction of scientific achievements into clinical practice are discussed. The results obtained by the authors and their colleagues after many years of studies on the cardiotropic properties of serum lipoproteins are also reported. The experimentally established cardiotonic activity of apoprotein A-1, which is accompanied by a decrease in the relative consumption of oxygen, maybe of great interest.
Collapse
Affiliation(s)
- Arkady R Kolpakov
- Institute of Biochemistry of Federal Research Center for Fundamental and Translational Medicine, Novosibirsk. Russian Federation
| | - Roman A Knyazev
- Institute of Biochemistry of Federal Research Center for Fundamental and Translational Medicine, Novosibirsk. Russian Federation
| |
Collapse
|
20
|
Qazmooz HA, Smesam HN, Mousa RF, Al-Hakeim HK, Maes M. Trace element, immune and opioid biomarkers of unstable angina, increased atherogenicity and insulin resistance: Results of machine learning. J Trace Elem Med Biol 2021; 64:126703. [PMID: 33338984 DOI: 10.1016/j.jtemb.2020.126703] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/21/2020] [Accepted: 12/01/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Aberrations in endothelial cells, immune and oxidative pathways are associated with atherosclerosis (ATS) and unstable angina (UA). The role of trace elements, minerals, and the endogenous opioid system (EOS) in UA are less well established. METHODS We measured lipid, insulin resistance (IR), and immune, trace element (copper and zinc), mineral (magnesium, calcium), EOS (β-endorphin and mu-opioid receptor (MOR)) and antioxidant (vitamin D3) biomarkers in patients with ATS (n = 60) and UA (n = 60) and healthy controls (n = 58). RESULTS ATS patients showed increased atherogenic and IR indices, IL-6, IL-10, β-endorphin, copper and magnesium, and lower zinc than healthy controls. Logistic regression showed that UA was significantly discriminated from ATS without UA with an accuracy of 85.5 % using calcium, IL-10, β-endorphin, MOR, triglycerides, IR (all positively), and copper and vitamin D3 (inversely). Neural networks showed that UA was discriminated from ATS without UA with an area under the ROC curve of 0.942 using MOR, β-endorphin, calcium, insulin resistance, vitamin D3 and copper as input variables. We found that 50.0 % of the variance in IR was explained by the regression on copper, IL-10, IL-6 (all positively), and zinc (inversely), while 32.9 % of the variance in the atherogenic index of plasma was explained by copper, IL-10 (both positively), and magnesium (inversely). CONCLUSION UA is not only mediated by insulin resistance, atherogenicity, and immune disorders, but also by aberrations in the endogenous opioid system and trace elements as well as lowered antioxidant levels. Copper appears to play a key role in IR and atherogenicity.
Collapse
Affiliation(s)
| | | | - Rana Fadhil Mousa
- A biochemist at the Faculty of Veterinary Medicine, University of Kerbala, Iraq.
| | | | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria; School of Medicine, IMPACT Strategic Research Centre, Deakin University, PO Box 281, Geelong, VIC, 3220, Australia.
| |
Collapse
|
21
|
Effects of morphine preconditioning on TRPV1 sensitization and ERK1/2 phosphorylation induced by TGFβ1 in neurocytes. Neuroreport 2021; 32:339-344. [PMID: 33661801 DOI: 10.1097/wnr.0000000000001603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Myocardium ischemia-reperfusion injury (IRI) is the major cause of cardiac dysfunction. While intrathecal morphine preconditioning (MPC) can alleviate IRI in animal model, the molecular processes underlying IRI and MPC remain elusive. This study aims to test whether pretreatment with morphine can ameliorate the increased activity of transient receptor potential vanilloid 1 (TRPV1) induced by transforming growth beta1 (TGFβ1) in cultured dorsal root ganglion neurons as a model of the effects of cardiac ischemia on nociceptive primary afferent neurons. METHODS To simulate the effect of MPC on dorsal root ganglia (DRG) neurons during myocardial IRI in vivo, the cells were pretreated with morphine for 10 min, followed by wash-out for 30 min before TGFβ1 was added. Afterwards, DRG neurons and N2a cells in all groups were stimulated by capsaicin, and the inward current induced by capsaicin were detected by whole-cell recording on DRG neurons; the expression of TRPV1, phosphorylated (p) TRPV1, ERK1/2, and pERK1/2 were detected by western blot in N2a cells. RESULTS In comparison with cells with normal culture, the inward current was enhanced of cells incubated with TGFβ1 (P < 0.05), and the relative expression of TRPV1, pTRPV1, and pERK1/2 was upregulated as well (P < 0.05); In comparison with cells incubated with TGFβ1, the inward current induced by capsaicin were decreased by pretreatment with morphine (P < 0.05), Moreover, the relative expression of TRPV1, pTRPV1, and pERK1/2 were also reduced by MPC (P < 0.05). CONCLUSION MPC inhibits TRPV1 sensitized by TGFβ1 in DRG cells, and the mechanism might be associated with the downregulation of pERK1/2 expression.
Collapse
|
22
|
Wu LN, Hu R, Yu JM. Morphine and myocardial ischaemia-reperfusion. Eur J Pharmacol 2020; 891:173683. [PMID: 33121952 DOI: 10.1016/j.ejphar.2020.173683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
Coronary heart disease (CHD) is a cardiovascular disease with high mortality and disability worldwide. The main pathological manifestation of CHD is myocardial injury due to ischaemia-reperfusion, resulting in the death of cardiomyocytes (apoptosis and necrosis) and the occurrence of cardiac failure. Morphine is a nonselective opioid receptor agonist that has been commonly used for analgesia and to treat ischaemic heart disease. The present review focused on morphine-induced protection in an animal model of myocardial ischaemia-reperfusion and chronic heart failure and the effects of morphine on ST segment elevation myocardial infarction (STEMI) patients who underwent pre-primary percutaneous coronary intervention (pre-PPCI) or PPCI. The signalling pathways involved are also briefly described.
Collapse
Affiliation(s)
- Li-Ning Wu
- Institutions: Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
| | - Rui Hu
- Institutions: Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
| | - Jun-Ma Yu
- Institutions: Department of Anesthesiology, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China.
| |
Collapse
|
23
|
Pappritz K, Van Linthout S. Opioid-Induced Immunomodulation: Consequences for the Experimental Coxsackievirus B3-Induced Myocarditis Model. BIOLOGY 2020; 9:biology9100335. [PMID: 33066118 PMCID: PMC7650777 DOI: 10.3390/biology9100335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 01/18/2023]
Abstract
Simple Summary Myocarditis is an inflammatory disorder of the heart mainly caused by viruses. To investigate viral myocarditis, the Coxsackievirus B3 (CVB3)-induced myocarditis model is the experimental model used since more than sixty years. In the pathogeneses of viral myocarditis, the subtle balance between pro-and anti-inflammatory immune responses is of great importance for disease manifestation. Parallel to the infection of the heart, experimental CVB3-induced myocarditis results in an infection of the pancreas, causing a severe burden for the challenged animals. In frame of animal welfare, application of analgesics is mandatory. So far, positive as well as negative effects of opioids on the immune system have been described. However, the impact of opioid application on the pathogenesis of experimental CVB3-induced myocarditis has not been investigated yet. Since examinations on disease pathways and new treatment options rely on established models to generate reproducible data, applicability of opioids in experimental CVB3-induced myocarditis needs to be carefully evaluated. For this purpose, we summarized published studies for 13 different opioids and discussed their potential impact on the CVB3-induced myocarditis model. Abstract Myocarditis is an inflammatory disorder of the heart predominantly caused by infectious agents. Since more than sixty years, the Coxsackievirus B3 (CVB3)-induced myocarditis mouse model is the experimental model used to investigate viral myocarditis. The pathogenesis of viral myocarditis is conceptually a multiphase process, initiated by the infection of cardiomyocytes, followed by activation of the immune system, and resulting in myocardial fibrosis and left ventricular dysfunction. In parallel to the direct infection of the heart, CVB3 replicates in lymphatic organs such as the pancreas. Due to infection of the pancreas, the model of experimental CVB3-induced myocarditis is estimated as a severe burden for the challenged animals. Application of analgesics in frame of the animal welfare act (European directive 2010/63/EU) is more and more becoming a matter of debate. For this purpose, we summarized published studies for 13 different opioids and discussed their potential impact on CVB3-induced myocarditis. In addition, with this summary we also want to provide guidance for researchers beyond the myocarditis field to estimate the impact of opioids on the immune system for their specific model. In the literature, both immunosuppressive as well as immune-activating effects of opioids have been described, but examinations in experimental CVB3-induced myocarditis have still not been reported so far. Based on the existing publications, administration of opioids in experimental CVB3-induced myocarditis might result in more severe disease progression, including higher mortality, or a less pronounced myocarditis model, failing to be used for the establishment of new treatment options. Taken together, the applicability of opioids in experimental CVB3-induced myocarditis and in inflammatory models in general needs to be carefully evaluated and further investigated.
Collapse
Affiliation(s)
- Kathleen Pappritz
- Campus Virchow Klinikum (CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10115 Berlin, Germany
- Correspondence: ; Tel.: +49-(0)30-450539509
| | - Sophie Van Linthout
- Campus Virchow Klinikum (CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10115 Berlin, Germany
| |
Collapse
|
24
|
Abstract
This paper is the forty-first consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2018 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (2), the roles of these opioid peptides and receptors in pain and analgesia in animals (3) and humans (4), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (5), opioid peptide and receptor involvement in tolerance and dependence (6), stress and social status (7), learning and memory (8), eating and drinking (9), drug abuse and alcohol (10), sexual activity and hormones, pregnancy, development and endocrinology (11), mental illness and mood (12), seizures and neurologic disorders (13), electrical-related activity and neurophysiology (14), general activity and locomotion (15), gastrointestinal, renal and hepatic functions (16), cardiovascular responses (17), respiration and thermoregulation (18), and immunological responses (19).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY, 11367, United States.
| |
Collapse
|
25
|
Rajani SF, Faghihi M, Imani A. Post-infarct morphine treatment reduces apoptosis and myofibroblast density in a rat model of cardiac ischemia-reperfusion. Eur J Pharmacol 2020; 887:173590. [PMID: 32976827 DOI: 10.1016/j.ejphar.2020.173590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/05/2020] [Accepted: 09/18/2020] [Indexed: 11/30/2022]
Abstract
Following myocardial ischemia, the cardiac tissue undergoes both, physiological and pathological changes to compensate the initial loss of function. Long-term continuous adjustments often take a drastic picture indicated by deteriorated ventricular function. Morphine is commonly used for rescuing patients suffering a heart attack. Recent results from our laboratory showed the anti-remodeling potential of morphine. Here, we explored the effect of morphine treatment on gelatinolytic activity, apoptosis and myofibroblast density. The male Sprague - Dawley rats underwent ischemia via ligation of left anterior descending coronary artery and received morphine (3 mg/kg; i.p.) for five consecutive days. Seven days post-MI, morphine led to significant reduction in MMP - 2 activity, apoptotic cell death and fibroblast density. Morphine also reduced MI-induced rise in serum pro-oxidant antioxidant balance and nitrite levels on day 28th following the surgery. These results provide mechanistic insight for morphine - induced anti-remodeling effects.
Collapse
Affiliation(s)
- Sulail Fatima Rajani
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology, Jinnah Medical & Dental College, Sohail University, Karachi, Pakistan.
| | - Mahdieh Faghihi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Imani
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Cataldi M, Cignarelli A, Giallauria F, Muscogiuri G, Barrea L, Savastano S, Colao A. Cardiovascular effects of antiobesity drugs: are the new medicines all the same? INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2020; 10:14-26. [PMID: 32714509 DOI: 10.1038/s41367-020-0015-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Waiting for a definite answer from well-designed randomized prospective clinical trials, the impact of the new antiobesity drugs -liraglutide, bupropion/naltrexone, phentermine/topiramate and lorcaserin- on cardiovascular outcomes remains uncertain. What has been learned from previous experience with older medicines is that antiobesity drugs may influence cardiovascular health not only causing weight reduction but also through direct actions on the cardiovascular system. Therefore, in the present review, we examine what is known, mainly from preclinical investigations, about the cardiovascular pharmacology of the new antiobesity medicines with the aim of highlighting potential mechanistic differences. We will show that the two active substances of the bupropion/naltrexone combination both exert beneficial and unwanted cardiovascular effects. Indeed, bupropion exerts anti-inflammatory effects but at the same time it does increase heart rate and blood pressure by potentiating catecholaminergic neurotransmission, whereas naltrexone reduces TLR4-dependent inflammation and has potential protective effects in stroke but also impairs cardiac adaption to ischemia and the beneficial opioid protective effects mediated in the endothelium. On the contrary, with the only exception of a small increase in heat rate, liraglutide only exerts favorable cardiovascular effects by protecting myocardium and brain from ischemic damage, improving heart contractility, lowering blood pressure and reducing atherogenesis. As far as the phentermine/topiramate combination is concerned, no direct cardiovascular beneficial effect is expected for phentermine (as this drug is an amphetamine derivative), whereas topiramate may exert cardioprotective and neuroprotective effects in ischemia and anti-inflammatory and antiatherogenic actions. Finally, lorcaserin, a selective 5HT2C receptor agonist, does not seem to exert significant direct effects on the cardiovascular system though at very high concentrations this drug may also interact with other serotonin receptor subtypes and exert unwanted cardiovascular effects. In conclusion, the final effect of the new antiobesity drugs on cardiovascular outcomes will be a balance between possible (but still unproved) beneficial effects of weight loss and "mixed" weight-independent drug-specific effects. Therefore comparative studies will be required to establish which one of the new medicines is more appropriate in patients with specific cardiovascular diseases.
Collapse
Affiliation(s)
- Mauro Cataldi
- Department of Neuroscience, Reproductive Sciences and Dentistry, Division of Pharmacology, Federico II University of Naples, Naples, Italy
| | - Angelo Cignarelli
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, Internal Medicine (Metabolic and Cardiac Rehabilitation Unit), Federico II University of Naples, Naples, Italy
| | - Giovanna Muscogiuri
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Luigi Barrea
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Silvia Savastano
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Unit of Endocrinology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | | |
Collapse
|
27
|
Fu LW, Tjen-A-Looi SC, Barvarz S, Guo ZL, Malik S. Role of opioid receptors in modulation of P2X receptor-mediated cardiac sympathoexcitatory reflex response. Sci Rep 2019; 9:17224. [PMID: 31748569 PMCID: PMC6868205 DOI: 10.1038/s41598-019-53754-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/31/2019] [Indexed: 01/15/2023] Open
Abstract
Myocardial ischemia evokes powerful reflex responses through activation of vagal and sympathetic afferents in the heart through the release of ischemic metabolites. We have demonstrated that extracellular ATP stimulates cardiac sympathetic afferents through P2 receptor-mediated mechanism, and that opioid peptides suppress these afferents' activity. However, the roles of both P2 receptor and endogenous opioids in cardiac sympathoexcitatory reflex (CSR) responses remain unclear. We therefore hypothesized that activation of cardiac P2 receptor evokes CSR responses by stimulating cardiac sympathetic afferents and these CSR responses are modulated by endogenous opioids. We observed that intrapericardial injection of α,β-methylene ATP (α,β-meATP, P2X receptor agonist), but not ADP (P2Y receptor agonist), caused a graded increase in mean arterial pressure in rats with sinoaortic denervation and vagotomy. This effect of α,β-meATP was abolished by blockade of cardiac neural transmission with intrapericardial procaine treatment and eliminated by intrapericardial A-317491, a selective P2X2/3 and P2X3 receptor antagonist. Intrapericardial α,β-meATP also evoked CSR response in vagus-intact rats. Furthermore, the P2X receptor-mediated CSR responses were enhanced by intrapericardial naloxone, a specific opioid receptor antagonist. These data suggest that stimulation of cardiac P2X2/3 and P2X3, but not P2Y receptors, powerfully evokes CSR responses through activation of cardiac spinal afferents, and that endogenous opioids suppress the P2X receptor-mediated CSR responses.
Collapse
Affiliation(s)
- Liang-Wu Fu
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA.
| | - Stephanie C Tjen-A-Looi
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| | - Sherwin Barvarz
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| | - Zhi-Ling Guo
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| | - Shaista Malik
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| |
Collapse
|
28
|
Palomino J, Echavarria R, Franco-Acevedo A, Moreno-Carranza B, Melo Z. Opioids Preconditioning Upon Renal Function and Ischemia-Reperfusion Injury: A Narrative Review. ACTA ACUST UNITED AC 2019; 55:medicina55090522. [PMID: 31443610 PMCID: PMC6780949 DOI: 10.3390/medicina55090522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023]
Abstract
Kidneys have an important role in regulating water volume, blood pressure, secretion of hormones and acid-base and electrolyte balance. Kidney dysfunction derived from acute injury can, under certain conditions, progress to chronic kidney disease. In the late stages of kidney disease, treatment is limited to replacement therapy: Dialysis and transplantation. After renal transplant, grafts suffer from activation of immune cells and generation of oxidant molecules. Anesthetic preconditioning has emerged as a promising strategy to ameliorate ischemia reperfusion injury. This review compiles some significant aspects of renal physiology and discusses current understanding of the effects of anesthetic preconditioning upon renal function and ischemia reperfusion injury, focusing on opioids and its properties ameliorating renal injury. According to the available evidence, opioid preconditioning appears to reduce inflammation and reactive oxygen species generation after ischemia reperfusion. Therefore, opioid preconditioning represents a promising strategy to reduce renal ischemia reperfusion injury and, its application on current clinical practice could be beneficial in events such as acute renal injury and kidney transplantation.
Collapse
Affiliation(s)
- Julio Palomino
- School of Medicine, Universidad Durango-Santander, Hermosillo 83165, Mexico
| | - Raquel Echavarria
- CONACyT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800 Col. Independencia, Guadalajara 44340, Jalisco, Mexico
| | | | | | - Zesergio Melo
- CONACyT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800 Col. Independencia, Guadalajara 44340, Jalisco, Mexico.
| |
Collapse
|
29
|
Exercise-induced cardiac opioid system activation attenuates apoptosis pathway in obese rats. Life Sci 2019; 231:116542. [PMID: 31176781 DOI: 10.1016/j.lfs.2019.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
AIM To compare the effect of 150 min vs. 300 min of weekly moderate intensity exercise training on the activation of the opioid system and apoptosis in the hearts of a diet-induced obesity model. METHODS Male Wistar rats were fed with either control (CON) or high fat (HF) diet for 32 weeks. At the 20th week, HF group was subdivided into sedentary, low (LEV, 150 min·week-1) or high (HEV, 300 min·week-1) exercise volume. After 12 weeks of exercise, body mass gain, adiposity index, systolic blood pressure, cardiac morphometry, apoptosis biomarkers and opioid system expression were evaluated. RESULTS Sedentary animals fed with HF presented pathological cardiac hypertrophy and higher body mass gain, systolic blood pressure and adiposity index than control group. Both exercise volumes induced physiological cardiac hypertrophy, restored systolic blood pressure and improved adiposity index, but only 300 min·week-1 reduced body mass gain. HF group exhibited lower proenkephalin, PI3K, ERK and GSK-3β expression, and greater activated caspase-3 expression than control group. Compared to HF, no changes in the cardiac opioid system were observed in the 150 min·week-1 of exercise training, while 300 min·week-1 showed greater proenkephalin, DOR, KOR, MOR, Akt, ERK and GSK-3β expression, and lower activated caspase-3 expression. CONCLUSION 300 min·week-1 of exercise training triggered opioid system activation and provided greater cardioprotection against obesity than 150 min·week-1. Our findings provide translational aspect with clinical relevance about the critical dose of exercise training necessary to reduce cardiovascular risk factors caused by obesity.
Collapse
|
30
|
Rajani SF, Imani A, Faghihi M, Izad M, Kardar GA, Salehi Z. Post-infarct morphine treatment mitigates left ventricular remodeling and dysfunction in a rat model of ischemia-reperfusion. Eur J Pharmacol 2019; 847:61-71. [PMID: 30684466 DOI: 10.1016/j.ejphar.2019.01.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 02/07/2023]
Abstract
Following myocardial infarction, the heart undergoes a series of dramatic compensations which may later form a maladaptive picture characterized by ventricular dilation and pump failure. Among several opioid agents, morphine has been shown to confer protection against reperfusion injury and infarct size. Here, we sought to study the cardioprotective effect of post-infarct morphine treatment against left ventricular adverse remodeling. We induced myocardial infarction in male Sprague - Dawley rats by ligating left anterior descending artery and then, treated these animals with three different doses of morphine -0.3, 3 and 10 mg/kg (i.p.). The echocardiographic evaluation depicted improved cardiac performance and lesser chamber dilation in the animals that had received 3 mg/kg of morphine. Next, we studied the effect of 3 mg/kg morphine administration on left ventricular hemodynamics, infarct size, tissue architecture, changes in lung and heart weight, circulating TNF-α level and post-MI mRNA expression of collagen-1, collagen-3, TGF-β, TNF-α, MMP-2 and MMP-9. Five-day morphine administration markedly improved LV function, and also reduced infarct size, myocyte hypertrophy, fibrosis, index of infarct expansion, heart weight and serum TNF-α level. Moreover, morphine alleviated MI-induced increase in wet and dry lung weight. Morphine also altered the mRNA expression of fibrosis-related genes, TNF-α, MMP-2 and MMP-9. In conclusion, post-infarct morphine treatment can mitigate adverse remodeling and cardiac dysfunction after MI. Beside analgesic effect, we may be able to harvest benefits from the antifibrotic and anti-remodeling action of morphine in patients with the acute coronary syndrome.
Collapse
Affiliation(s)
- Sulail Fatima Rajani
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Alireza Imani
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdieh Faghihi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maryam Izad
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Gholam Ali Kardar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Immunology, Asthma & Allergy Research Institute (IAARI), Tehran, Iran.
| | - Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Cheng XY, Chen C, He SF, Huang CX, Zhang L, Chen ZW, Zhang Y. Spinal NGF induces anti-intrathecal opioid-initiated cardioprotective effect via regulation of TRPV1 expression. Eur J Pharmacol 2018; 844:145-155. [PMID: 30529472 DOI: 10.1016/j.ejphar.2018.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 11/26/2022]
Abstract
Evidences from previous studies confirmed that intrathecal morphine preconditioning (ITMP) reduces the cardiac injury of ischemia-reperfusion (IR) via the central nervous system. However, the molecular mechanism is not fully understood. The breath of central nerve growth factor (NGF) during nociceptive transmission has been well documented, and little is known about the significance of NGF in myocardial injury of IR and intrathecal morphine-induced cardioprotection. To address these questions, we over-expressed or silenced NGF in the spinal cord by using intrathecal injection of lentivirus-NGF or shRNA respectively, accompanied by ITMP in the IR rat model. The levels of NGF and tropomyosin receptor kinase A (Trka) as well as transient receptor potential vanilloid 1 (TRPV1) in the T2-6 spinal cord were evaluated. The results showed that cardiac damage indicators induced by IR, including the increased infarct size, arrhythmia score and serum troponin levels were attenuated after ITMP. However, overexpression of spinal NGF significantly reversed these decreases, as well as reduced the expression and phosphorylation of TRPV1 that was elicited by ITMP. Conversely, silencing of spinal NGF enhanced ITMP-induced cardioprotective effects. Phosphorylation and expression of TRPV1 in the spinal cord were significantly decreased after regional NGF silencing. These findings suggested that the cardioprotective effects of ITMP may implement by mediating through spinal NGF expression, wherein it involves the nociceptor TRPV1. NGF may act as a potential therapeutic target in the development of new agents for the treatment of cardiac injury induced by IR.
Collapse
Affiliation(s)
- Xue-Ying Cheng
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Chen Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Shu-Fang He
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Chun-Xia Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Li Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhi-Wu Chen
- Department of Pharmacology, Anhui Medical University, Hefei, China.
| | - Ye Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
32
|
Melo Z, Ishida C, Goldaraz MDLP, Rojo R, Echavarria R. Novel Roles of Non-Coding RNAs in Opioid Signaling and Cardioprotection. Noncoding RNA 2018; 4:ncrna4030022. [PMID: 30227648 PMCID: PMC6162605 DOI: 10.3390/ncrna4030022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease (CVD) is a significant cause of morbidity and mortality across the world. A large proportion of CVD deaths are secondary to coronary artery disease (CAD) and myocardial infarction (MI). Even though prevention is the best strategy to reduce risk factors associated with MI, the use of cardioprotective interventions aimed at improving patient outcomes is of great interest. Opioid conditioning has been shown to be effective in reducing myocardial ischemia-reperfusion injury (IRI) and cardiomyocyte death. However, the molecular mechanisms behind these effects are under investigation and could provide the basis for the development of novel therapeutic approaches in the treatment of CVD. Non-coding RNAs (ncRNAs), which are functional RNA molecules that do not translate into proteins, are critical modulators of cardiac gene expression during heart development and disease. Moreover, ncRNAs such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are known to be induced by opioid receptor activation and regulate opioid signaling pathways. Recent advances in experimental and computational tools have accelerated the discovery and functional characterization of ncRNAs. In this study, we review the current understanding of the role of ncRNAs in opioid signaling and opioid-induced cardioprotection.
Collapse
Affiliation(s)
- Zesergio Melo
- CONACyT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800 Col. Independencia, Guadalajara 44340, Jalisco, Mexico.
| | - Cecilia Ishida
- Programa de Genomica Computacional, Centro de Ciencias Genomicas, Universidad Nacional Autonoma de Mexico, Cuernavaca 62210, Morelos, Mexico.
| | - Maria de la Paz Goldaraz
- Departamento de Anestesiologia, Hospital de Especialidades UMAE CMNO, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico.
| | - Rocio Rojo
- Departamento de Anestesiologia, Hospital de Especialidades UMAE CMNO, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico.
| | - Raquel Echavarria
- CONACyT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800 Col. Independencia, Guadalajara 44340, Jalisco, Mexico.
| |
Collapse
|