1
|
Trauzeddel RF, Rothe LM, Nordine M, Dehé L, Scholtz K, Spies C, Hadzidiakos D, Winterer G, Borchers F, Kruppa J, Treskatsch S. Influence of a chronic beta-blocker therapy on perioperative opioid consumption - a post hoc secondary analysis. BMC Anesthesiol 2024; 24:80. [PMID: 38413849 PMCID: PMC10898005 DOI: 10.1186/s12871-024-02456-2] [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: 10/29/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Beta-blocker (BB) therapy plays a central role in the treatment of cardiovascular diseases. An increasing number of patients with cardiovascular diseases undergoe noncardiac surgery, where opioids are an integral part of the anesthesiological management. There is evidence to suggest that short-term intravenous BB therapy may influence perioperative opioid requirements due to an assumed cross-talk between G-protein coupled beta-adrenergic and opioid receptors. Whether chronic BB therapy could also have an influence on perioperative opioid requirements is unclear. METHODS A post hoc analysis of prospectively collected data from a multicenter observational (BioCog) study was performed. Inclusion criteria consisted of elderly patients (≥ 65 years) undergoing elective noncardiac surgery as well as total intravenous general anesthesia without the use of regional anesthesia and duration of anesthesia ≥ 60 min. Two groups were defined: patients with and without BB in their regular preopreative medication. The administered opioids were converted to their respective morphine equivalent doses. Multiple regression analysis was performed using the morphine-index to identify independent predictors. RESULTS A total of 747 patients were included in the BioCog study in the study center Berlin. 106 patients fulfilled the inclusion criteria. Of these, 37 were on chronic BB. The latter were preoperatively significantly more likely to have arterial hypertension (94.6%), chronic renal failure (27%) and hyperlipoproteinemia (51.4%) compared to patients without BB. Both groups did not differ in terms of cumulative perioperative morphine equivalent dose (230.9 (BB group) vs. 214.8 mg (Non-BB group)). Predictive factors for increased morphine-index were older age, male sex, longer duration of anesthesia and surgery of the trunk. In a model with logarithmised morphine index, only gender (female) and duration of anesthesia remained predictive factors. CONCLUSIONS Chronic BB therapy was not associated with a reduced perioperative opioid consumption. TRIAL REGISTRATION This study was registered at ClinicalTrials.gov ( NCT02265263 ) on the 15.10.2014 with the principal investigator being Univ.-Prof. Dr. med. Claudia Spies.
Collapse
Affiliation(s)
- Ralf F Trauzeddel
- Department of Anesthesiology and Intensive Care Medicine, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Luisa M Rothe
- IS Global Campus Cliníc Rosselló, Barcelona Institute for Global Health, 132, 7è, Barcelona, 08036, Spain
| | - Michael Nordine
- Department of Anesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Lukas Dehé
- Department of Anesthesiology and Intensive Care Medicine, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Kathrin Scholtz
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Claudia Spies
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Daniel Hadzidiakos
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Georg Winterer
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Friedrich Borchers
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Jochen Kruppa
- Hochschule Osnabrück, University of Applied Sciences, Osnabrück, Germany
| | - Sascha Treskatsch
- Department of Anesthesiology and Intensive Care Medicine, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany.
| |
Collapse
|
2
|
The apelin/APJ signaling system and cytoprotection: Role of its cross-talk with kappa opioid receptor. Eur J Pharmacol 2022; 936:175353. [DOI: 10.1016/j.ejphar.2022.175353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
|
3
|
Lei Y, Li XX, Guo Z. Impact of timing of morphine treatment on infarct size in experimental animal model of acute myocardial ischemia and reperfusion. Eur J Pharmacol 2022; 928:175094. [PMID: 35714691 DOI: 10.1016/j.ejphar.2022.175094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
Morphine is generally used in clinical treatment for the patients who have not been effectively alleviated for chest pain after the treatment with nitrites or who contraindicate nitrite drugs. However, it was reported that the treatment with morphine in acute myocardial infarction or acute coronary artery syndromes induced increase in myocardial injury even increase of the mortality of the patients. After comparing the reported laboratory studies showing the cardioprotective effects and the clinical observations presenting the harmful consequences, we query whether the timing of the morphine treatment makes the difference in the prognosis of the ischemic/infarct myocardium. We found that intravenous injections of morphine (0.3 mg/kg) at 15 min before the acute myocardial ischemia, at 5 min and 20 min or 60 min after ligation of the coronary artery in separate groups of rats scheduled for acute myocardial ischemia, for 30 min or 90 min followed by reperfusion for 120 min, induced different results, reduction in the size of infarction, no effect and increases of the infarct sizes, respectively. The opioid μ- and kappa-receptors mediated the detrimental effect of morphine on the myocardial injury. The findings of this study suggest that administration of morphine may cause different consequences when used at different time in the pathology of acute myocardial ischemia and reperfusion. The underlying mechanisms in the pathology of acute myocardial ischemia warrant further study.
Collapse
Affiliation(s)
- Yi Lei
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Xiao-Xi Li
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anaesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Zheng Guo
- College of Anaesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anaesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), National Education Commission, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
4
|
Petrocelli G, Pampanella L, Abruzzo PM, Ventura C, Canaider S, Facchin F. Endogenous Opioids and Their Role in Stem Cell Biology and Tissue Rescue. Int J Mol Sci 2022; 23:3819. [PMID: 35409178 PMCID: PMC8998234 DOI: 10.3390/ijms23073819] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/25/2023] Open
Abstract
Opioids are considered the oldest drugs known by humans and have been used for sedation and pain relief for several centuries. Nowadays, endogenous opioid peptides are divided into four families: enkephalins, dynorphins, endorphins, and nociceptin/orphanin FQ. They exert their action through the opioid receptors (ORs), transmembrane proteins belonging to the super-family of G-protein-coupled receptors, and are expressed throughout the body; the receptors are the δ opioid receptor (DOR), μ opioid receptor (MOR), κ opioid receptor (KOR), and nociceptin/orphanin FQ receptor (NOP). Endogenous opioids are mainly studied in the central nervous system (CNS), but their role has been investigated in other organs, both in physiological and in pathological conditions. Here, we revise their role in stem cell (SC) biology, since these cells are a subject of great scientific interest due to their peculiar features and their involvement in cell-based therapies in regenerative medicine. In particular, we focus on endogenous opioids' ability to modulate SC proliferation, stress response (to oxidative stress, starvation, or damage following ischemia-reperfusion), and differentiation towards different lineages, such as neurogenesis, vasculogenesis, and cardiogenesis.
Collapse
Affiliation(s)
- Giovannamaria Petrocelli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (L.P.); (P.M.A.); (F.F.)
| | - Luca Pampanella
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (L.P.); (P.M.A.); (F.F.)
| | - Provvidenza M. Abruzzo
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (L.P.); (P.M.A.); (F.F.)
| | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (L.P.); (P.M.A.); (F.F.)
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)–Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (L.P.); (P.M.A.); (F.F.)
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)–Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (L.P.); (P.M.A.); (F.F.)
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)–Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| |
Collapse
|
5
|
Popov SV, Mukhomedzyanov AV, Tsibulnikov SY, Khaliuli I, Oeltgen PR, Prasad NR, Maslov LN. Activation of Peripheral Opioid Kappa1 Receptor Prevents Cardiac Reperfusion Injury. Physiol Res 2021; 70:523-531. [PMID: 34062075 PMCID: PMC8820547 DOI: 10.33549/physiolres.934646] [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: 01/19/2021] [Accepted: 04/13/2021] [Indexed: 11/25/2022] Open
Abstract
The role of opioid kappa1 and kappa2 receptors in reperfusion cardiac injury was studied. Male Wistar rats were subjected to a 45-min coronary artery occlusion followed by a 120-min reperfusion. Opioid kappa receptor agonists were administered intravenously 5 min before the onset of reperfusion, while opioid receptor antagonists were given 10 min before reperfusion. The average value of the infarct size/area at risk (IS/AAR) ratio was 43 - 48% in untreated rats. Administration of the opioid kappa1 receptor agonist (-)-U-50,488 (1 mg/kg) limited the IS/AAR ratio by 42%. Administration of the opioid kappa receptor agonist ICI 199,441 (0.1 mg/kg) limited the IS/AAR ratio by 41%. The non-selective opioid kappa receptor agonist (+)-U-50,488 (1 mg/kg) with low affinity for opioid kappa receptor, the peripherally acting opioid kappa2 receptor agonist ICI 204,448 (4 mg/kg) and the selective opioid ?2 receptor agonist GR89696 (0.1 mg/kg) had no effect on the IS/AAR ratio. Pretreatment with naltrexone, the peripherally acting opioid receptor antagonist naloxone methiodide, or the selective opioid kappa2 receptor antagonist nor-binaltorphimine completely abolished the infarct-reducing effect of (-)-U-50,488 and ICI 199,441. Pretreatment with the selective opioid ? receptor antagonist TIPP[psi] and the selective opioid µ receptor antagonist CTAP did not alter the infarct reducing effect of (-)-U-50,488 and ICI 199,441. Our study is the first to demonstrate the following: (a) the activation of opioid kappa2 receptor has no effect on cardiac tolerance to reperfusion; (b) peripheral opioid kappa1 receptor stimulation prevents reperfusion cardiac injury; (c) ICI 199,441 administration resulted in an infarct-reducing effect at reperfusion; (e) bradycardia induced by opioid kappa receptor antagonists is not dependent on the occupancy of opioid kappa receptor.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/toxicity
- Administration, Intravenous
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/toxicity
- Animals
- Arrhythmias, Cardiac/chemically induced
- Arrhythmias, Cardiac/physiopathology
- Disease Models, Animal
- Heart Rate/drug effects
- Male
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/prevention & control
- Myocardial Reperfusion Injury/metabolism
- Myocardial Reperfusion Injury/pathology
- Myocardial Reperfusion Injury/prevention & control
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Narcotic Antagonists/administration & dosage
- Piperazines/administration & dosage
- Pyrrolidines/administration & dosage
- Pyrrolidines/toxicity
- Rats, Wistar
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Signal Transduction
- Rats
Collapse
Affiliation(s)
- S V Popov
- Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, the Russian Academy of Sciences, Tomsk, Russia.
| | | | | | | | | | | | | |
Collapse
|
6
|
Liang Q, Huang X, Zeng C, Li D, Shi Y, Zhao G, Zhong M. BW373U86 upregulates autophagy by inhibiting the PI3K/Akt pathway and regulating the mTOR pathway to protect cardiomyocytes from hypoxia-reoxygenation injury. Can J Physiol Pharmacol 2020; 98:684-690. [PMID: 32955950 DOI: 10.1139/cjpp-2019-0684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to explore the protective effect of BW373U86 (a δ-opioid receptor (DOR) agonist) on ischemia-reperfusion (I/R) injury in rat cardiomyocytes and its underlying mechanism. Primary rat cardiomyocytes were cultured and pretreated with BW373U86 for intervention. The cardiomyocytes were cultured under the condition of 94% N2 and 5% CO2 for 24 h to perform hypoxia culture and conventionally cultured for 12 h to perform reoxygenation culture. The cell viability of cardiomyocytes was detected by an MTT assay (Sigma-Aldrich). The autophagy lysosome levels in cardiomyocytes were evaluated by acidic vesicular organelles with dansylcadaverine (MDC) staining (autophagy test kit, Kaiji Biology, kgatg001). The protein expression levels of LC3, p62, and factors in the PI3K/Akt/mTOR signaling pathway were detected by Western blot. Pretreatment with BW373U86 could improve the cell viability of cardiomyocytes with hypoxia-reoxygenation (H/R) injury (p < 0.05). Interestingly, after coculture of BW373U86 and PI3K inhibitor (3-methyladenine), the protein expression levels of p-Akt in cardiomyocytes were markedly increased in comparison with those in the BW373U86 group (p < 0.05). However, there were no significant differences in the protein expression levels of mTOR between the coculture group and the BW373U86 group (p > 0.05). BW373U86 upregulated autophagy to protect cardiomyocytes from H/R injury, which may be related to the PI3K/Akt/m TOR pathway.
Collapse
Affiliation(s)
- Qianyi Liang
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoling Huang
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Chaokun Zeng
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Dewei Li
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Yongyong Shi
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Gaofeng Zhao
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Min Zhong
- Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Anaesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
7
|
Canaider S, Facchin F, Tassinari R, Cavallini C, Olivi E, Taglioli V, Zannini C, Bianconi E, Maioli M, Ventura C. Intracrine Endorphinergic Systems in Modulation of Myocardial Differentiation. Int J Mol Sci 2019; 20:ijms20205175. [PMID: 31635381 PMCID: PMC6829321 DOI: 10.3390/ijms20205175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
A wide variety of peptides not only interact with the cell surface, but govern complex signaling from inside the cell. This has been referred to as an "intracrine" action, and the orchestrating molecules as "intracrines". Here, we review the intracrine action of dynorphin B, a bioactive end-product of the prodynorphin gene, on nuclear opioid receptors and nuclear protein kinase C signaling to stimulate the transcription of a gene program of cardiogenesis. The ability of intracrine dynorphin B to prime the transcription of its own coding gene in isolated nuclei is discussed as a feed-forward loop of gene expression amplification and synchronization. We describe the role of hyaluronan mixed esters of butyric and retinoic acids as synthetic intracrines, controlling prodynorphin gene expression, cardiogenesis, and cardiac repair. We also discuss the increase in prodynorphin gene transcription and intracellular dynorphin B afforded by electromagnetic fields in stem cells, as a mechanism of cardiogenic signaling and enhancement in the yield of stem cell-derived cardiomyocytes. We underline the possibility of using the diffusive features of physical energies to modulate intracrinergic systems without the needs of viral vector-mediated gene transfer technologies, and prompt the exploration of this hypothesis in the near future.
Collapse
Affiliation(s)
- Silvia Canaider
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Federica Facchin
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Riccardo Tassinari
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Claudia Cavallini
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Elena Olivi
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Valentina Taglioli
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Chiara Zannini
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Eva Bianconi
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| |
Collapse
|
8
|
Root-Bernstein R, Churchill B, Turke M, Subhramanyam UKT, Labahn J. Mutual Enhancement of Opioid and Adrenergic Receptors by Combinations of Opioids and Adrenergic Ligands Is Reflected in Molecular Complementarity of Ligands: Drug Development Possibilities. Int J Mol Sci 2019; 20:ijms20174137. [PMID: 31450631 PMCID: PMC6747318 DOI: 10.3390/ijms20174137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/07/2019] [Accepted: 08/22/2019] [Indexed: 11/16/2022] Open
Abstract
Crosstalk between opioid and adrenergic receptors is well characterized and due to interactions between second messenger systems, formation of receptor heterodimers, and extracellular allosteric binding regions. Both classes of receptors bind both sets of ligands. We propose here that receptor crosstalk may be mirrored in ligand complementarity. We demonstrate that opioids bind to adrenergic compounds with micromolar affinities. Additionally, adrenergic compounds bind with micromolar affinities to extracellular loops of opioid receptors while opioids bind to extracellular loops of adrenergic receptors. Thus, each compound type can bind to the complementary receptor, enhancing the activity of the other compound type through an allosteric mechanism. Screening for ligand complementarity may permit the identification of other mutually-enhancing sets of compounds as well as the design of novel combination drugs or tethered compounds with improved duration and specificity of action.
Collapse
Affiliation(s)
- Robert Root-Bernstein
- Department of Physiology, 567 Wilson Road, Room 2201 Biomedical and Physical Sciences Building, Michigan State University, East Lansing, MI 48824, USA.
| | - Beth Churchill
- Department of Physiology, 567 Wilson Road, Room 2201 Biomedical and Physical Sciences Building, Michigan State University, East Lansing, MI 48824, USA
| | - Miah Turke
- Department of Physiology, 567 Wilson Road, Room 2201 Biomedical and Physical Sciences Building, Michigan State University, East Lansing, MI 48824, USA
| | - Udaya K Tiruttani Subhramanyam
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607 Hamburg, Germany
- Forschungszentrum Juelich GmbH, ICS-6, 52425 Juelich, Germany
| | - Joerg Labahn
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607 Hamburg, Germany
- Forschungszentrum Juelich GmbH, ICS-6, 52425 Juelich, Germany
| |
Collapse
|
9
|
Reyes BAS, Zhang XY, Dufourt EC, Bhatnagar S, Valentino RJ, Van Bockstaele EJ. Neurochemically distinct circuitry regulates locus coeruleus activity during female social stress depending on coping style. Brain Struct Funct 2019; 224:1429-1446. [PMID: 30767070 DOI: 10.1007/s00429-019-01837-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/16/2019] [Indexed: 12/18/2022]
Abstract
Stress-related psychiatric diseases are nearly twice as prevalent in women compared to men. We recently showed in male rats that the resident-intruder model of social stress differentially engages stress-related circuitry that regulates norepinephrine-containing neurons of the locus coeruleus (LC) depending on coping strategy as determined by the latency to assume a defeat posture. Here, we determined whether this social stress had similar effects in female rats. LC afferents were retrogradely labeled with Fluorogold (FG) and rats had one or five daily exposures to an aggressive resident. Sections through the nucleus paragigantocellularis (PGi), a source of enkephalin (ENK) afferents to the LC, and central nucleus of the amygdala (CeA), a source of corticotropin-releasing factor (CRF) afferents to the LC, were processed for immunocytochemical detection of c-fos, a marker of neuronal activity, FG and ENK or CRF. Like male rats, female rats defeated with a relatively short latency (SL) in response to a single resident-intruder exposure and showed significant c-fos activation of LC neurons, PGi-ENK LC afferents, and CeA-CRF-LC afferents. With repeated exposure, some rats exhibited a long latency to defeat (LL). LC neurons and CeA-CRF-LC afferents were activated in SL rats compared to control and LL, whereas PGi-ENK LC afferents were not. Conversely, in LL rats, PGi-ENK LC and CeA-CRF-LC afferents were activated compared to controls but not LC neurons. CRF type 1 receptor (CRF1) and µ-opioid receptor (MOR) expression levels in LC were decreased in LL rats. Finally, electron microscopy showed a relative increase in MOR on the plasma membrane of LL rats and a relative increase in CRF1 on the plasma membrane of SL rats. Together, these results suggest that as is the case for males, social stress engages divergent circuitry to regulate the LC in female rats depending on coping strategy, with a bias towards CRF influence in more subordinate rats and opioid influence in less subordinate rats.
Collapse
Affiliation(s)
- Beverly A S Reyes
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA.
| | - Xiao-Yan Zhang
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Elsa C Dufourt
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Rita J Valentino
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Elisabeth J Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, 245 S. 15th Street, Philadelphia, PA, 19102, USA
| |
Collapse
|
10
|
Adrenergic Agonists Bind to Adrenergic-Receptor-Like Regions of the Mu Opioid Receptor, Enhancing Morphine and Methionine-Enkephalin Binding: A New Approach to "Biased Opioids"? Int J Mol Sci 2018; 19:ijms19010272. [PMID: 29342106 PMCID: PMC5796218 DOI: 10.3390/ijms19010272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/10/2018] [Accepted: 01/13/2018] [Indexed: 11/16/2022] Open
Abstract
Extensive evidence demonstrates functional interactions between the adrenergic and opioid systems in a diversity of tissues and organs. While some effects are due to receptor and second messenger cross-talk, recent research has revealed an extracellular, allosteric opioid binding site on adrenergic receptors that enhances adrenergic activity and its duration. The present research addresses whether opioid receptors may have an equivalent extracellular, allosteric adrenergic binding site that has similar enhancing effects on opioid binding. Comparison of adrenergic and opioid receptor sequences revealed that these receptors share very significant regions of similarity, particularly in some of the extracellular and transmembrane regions associated with adrenergic binding in the adrenergic receptors. Five of these shared regions from the mu opioid receptor (muOPR) were synthesized as peptides and tested for binding to adrenergic, opioid and control compounds using ultraviolet spectroscopy. Adrenergic compounds bound to several of these muOPR peptides with low micromolar affinity while acetylcholine, histamine and various adrenergic antagonists did not. Similar studies were then conducted with purified, intact muOPR with similar results. Combinations of epinephrine with methionine enkephalin or morphine increased the binding of both by about half a log unit. These results suggest that muOPR may be allosterically enhanced by adrenergic agonists.
Collapse
|
11
|
Abstract
The opioid receptor family, with associated endogenous ligands, has numerous roles throughout the body. Moreover, the delta opioid receptor (DORs) has various integrated roles within the physiological systems, including the cardiovascular system. While DORs are important modulators of cardiovascular autonomic balance, they are well-established contributors to cardioprotective mechanisms. Both endogenous and exogenous opioids acting upon DORs have roles in myocardial hibernation and protection against ischaemia-reperfusion (I-R) injury. Downstream signalling mechanisms governing protective responses alternate, depending on the timing and duration of DOR activation. The following review describes models and mechanisms of DOR-mediated cardioprotection, the impact of co-morbidities and challenges for clinical translation.
Collapse
Affiliation(s)
- Louise See Hoe
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, 4222, Australia
- Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, QLD, Australia
| | - Hemal H Patel
- VA San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, 4222, Australia.
| |
Collapse
|
12
|
Guajardo HM, Snyder K, Ho A, Valentino RJ. Sex Differences in μ-Opioid Receptor Regulation of the Rat Locus Coeruleus and Their Cognitive Consequences. Neuropsychopharmacology 2017; 42:1295-1304. [PMID: 27827371 PMCID: PMC5437881 DOI: 10.1038/npp.2016.252] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 12/04/2022]
Abstract
Stress-related neuropsychiatric pathologies are more prevalent in females compared with males. An important component of the stress response is activation of the locus coeruleus (LC)-norepinephrine system. Because LC activation is tempered by endogenous opioid release during stress, the magnitude of opioid regulation of the LC could determine stress vulnerability. Here we report convergent evidence for decreased μ-opioid receptor (MOR) function in the female rat LC. The selective MOR agonist, DAMGO (10 pg), completely inhibited LC discharge of male but not female rats and DAMGO (30 pg) produced no further inhibition of female LC neurons. Consistent with a decreased maximum DAMGO response, MOR protein and mRNA expression were decreased in female compared with male LC. These molecular and cellular sex differences were associated with sexually distinct effects of LC-MOR activation on cognitive processing in an operant strategy-shifting task. Although DAMGO (10 pg intra-LC) increased the number of trials to reach criterion for both sexes, it increased the duration to complete the task and the total number of errors selectively in males. Specifically, DAMGO increased premature responses, regressive errors, and random errors in males and perseverative errors in females. The sexually distinct cognitive consequences of activating LC-MOR may contribute to sex differences in opioid abuse patterns and may guide sex-specific therapies. Finally, given evidence that endogenous opioids restrain stress-induced LC activation and promote recovery of activity to pre-stress levels, decreased MOR function in the female LC could contribute to LC-NE overactivity that underlies the hyperarousal symptoms of stress-related psychiatric diseases.
Collapse
Affiliation(s)
- Herminio M Guajardo
- Neuroscience Graduate Group, The University of Pennsylvania, Philadelphia, PA, USA,The Children's Hospital of Philadelphia, Department of Anesthesiology and Critical Care Medicine, Philadelphia, PA, USA
| | - Kevin Snyder
- Neuroscience Graduate Group, The University of Pennsylvania, Philadelphia, PA, USA,The Children's Hospital of Philadelphia, Department of Anesthesiology and Critical Care Medicine, Philadelphia, PA, USA
| | - Andrew Ho
- The Children's Hospital of Philadelphia, Department of Anesthesiology and Critical Care Medicine, Philadelphia, PA, USA
| | - Rita J Valentino
- Neuroscience Graduate Group, The University of Pennsylvania, Philadelphia, PA, USA,The Children's Hospital of Philadelphia, Department of Anesthesiology and Critical Care Medicine, Philadelphia, PA, USA,The Children's Hospital of Philadelphia, 402D Abramson Pediatric Research Center, Philadelphia, PA 19104, USA, Tel: 215-590-0650; Fax: 215-590-3364, E-mail:
| |
Collapse
|
13
|
Maslov LN, Khaliulin I, Oeltgen PR, Naryzhnaya NV, Pei J, Brown SA, Lishmanov YB, Downey JM. Prospects for Creation of Cardioprotective and Antiarrhythmic Drugs Based on Opioid Receptor Agonists. Med Res Rev 2016; 36:871-923. [PMID: 27197922 PMCID: PMC5082499 DOI: 10.1002/med.21395] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/19/2022]
Abstract
It has now been demonstrated that the μ, δ1 , δ2 , and κ1 opioid receptor (OR) agonists represent the most promising group of opioids for the creation of drugs enhancing cardiac tolerance to the detrimental effects of ischemia/reperfusion (I/R). Opioids are able to prevent necrosis and apoptosis of cardiomyocytes during I/R and improve cardiac contractility in the reperfusion period. The OR agonists exert an infarct-reducing effect with prophylactic administration and prevent reperfusion-induced cardiomyocyte death when ischemic injury of heart has already occurred; that is, opioids can mimic preconditioning and postconditioning phenomena. Furthermore, opioids are also effective in preventing ischemia-induced arrhythmias.
Collapse
Affiliation(s)
| | - Igor Khaliulin
- School of Clinical SciencesUniversity of BristolBristolUK
| | | | | | - Jian‐Ming Pei
- Department of PhysiologyFourth Military Medical UniversityXi'anP. R. China
| | | | - Yury B. Lishmanov
- Research Institute for CardiologyTomskRussia
- National Research Tomsk Polytechnic University634050TomskRussia
| | | |
Collapse
|
14
|
Irwin MG, Wong GTC. Remifentanil and Opioid-Induced Cardioprotection. J Cardiothorac Vasc Anesth 2015; 29 Suppl 1:S23-6. [DOI: 10.1053/j.jvca.2015.01.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Indexed: 02/07/2023]
|
15
|
Fujita W, Gomes I, Devi LA. Revolution in GPCR signalling: opioid receptor heteromers as novel therapeutic targets: IUPHAR review 10. Br J Pharmacol 2015; 171:4155-76. [PMID: 24916280 DOI: 10.1111/bph.12798] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/27/2014] [Accepted: 04/16/2014] [Indexed: 12/24/2022] Open
Abstract
GPCRs can interact with each other to form homomers or heteromers. Homomers involve interactions with the same receptor type while heteromers involve interactions between two different GPCRs. These receptor-receptor interactions modulate not only the binding but also the signalling and trafficking properties of individual receptors. Opioid receptor heteromerization has been extensively investigated with the objective of identifying novel therapeutic targets that are as potent as morphine but without the side effects associated with chronic morphine use. In this context, studies have described heteromerization between the different types of opioid receptors and between opioid receptors and a wide range of GPCRs including adrenoceptors, cannabinoid, 5-HT, metabotropic glutamate and sensory neuron-specific receptors. Recent advances in the field involving the generation of heteromer-specific reagents (antibodies or ligands) or of membrane-permeable peptides that disrupt the heteromer interaction are helping to elucidate the physiological role of opioid receptor heteromers and the contribution of the partner receptor to the side effects associated with opioid use. For example, studies using membrane-permeable peptides targeting the heteromer interface have implicated μ and δ receptor heteromers in the development of tolerance to morphine, and heteromers of μ and gastrin-releasing peptide receptors in morphine-induced itch. In addition, a number of ligands that selectively target opioid receptor heteromers exhibit potent antinociception with a decrease in the side effects commonly associated with morphine use. In this review, we summarize the latest findings regarding the biological and functional characteristics of opioid receptor heteromers both in vitro and in vivo.
Collapse
Affiliation(s)
- Wakako Fujita
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | |
Collapse
|
16
|
Headrick JP, See Hoe LE, Du Toit EF, Peart JN. Opioid receptors and cardioprotection - 'opioidergic conditioning' of the heart. Br J Pharmacol 2015; 172:2026-50. [PMID: 25521834 PMCID: PMC4386979 DOI: 10.1111/bph.13042] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/18/2014] [Accepted: 12/09/2014] [Indexed: 12/21/2022] Open
Abstract
Ischaemic heart disease (IHD) remains a major cause of morbidity/mortality globally, firmly established in Westernized or 'developed' countries and rising in prevalence in developing nations. Thus, cardioprotective therapies to limit myocardial damage with associated ischaemia-reperfusion (I-R), during infarction or surgical ischaemia, is a very important, although still elusive, clinical goal. The opioid receptor system, encompassing the δ (vas deferens), κ (ketocyclazocine) and μ (morphine) opioid receptors and their endogenous opioid ligands (endorphins, dynorphins, enkephalins), appears as a logical candidate for such exploitation. This regulatory system may orchestrate organism and organ responses to stress, induces mammalian hibernation and associated metabolic protection, triggers powerful adaptive stress resistance in response to ischaemia/hypoxia (preconditioning), and mediates cardiac benefit stemming from physical activity. In addition to direct myocardial actions, central opioid receptor signalling may also enhance the ability of the heart to withstand I-R injury. The δ- and κ-opioid receptors are strongly implicated in cardioprotection across models and species (including anti-infarct and anti-arrhythmic actions), with mixed evidence for μ opioid receptor-dependent protection in animal and human tissues. A small number of clinical trials have provided evidence of cardiac benefit from morphine or remifentanil in cardiopulmonary bypass or coronary angioplasty patients, although further trials of subtype-specific opioid receptor agonists are needed. The precise roles and utility of this GPCR family in healthy and diseased human myocardium, and in mediating central and peripheral survival responses, warrant further investigation, as do the putative negative influences of ageing, IHD co-morbidities, and relevant drugs on opioid receptor signalling and protective responses.
Collapse
Affiliation(s)
- John P Headrick
- Heart Foundation Research Centre, Griffith Health Institute Griffith UniversitySouthport, Qld., Australia
| | - Louise E See Hoe
- Heart Foundation Research Centre, Griffith Health Institute Griffith UniversitySouthport, Qld., Australia
| | - Eugene F Du Toit
- Heart Foundation Research Centre, Griffith Health Institute Griffith UniversitySouthport, Qld., Australia
| | - Jason N Peart
- Heart Foundation Research Centre, Griffith Health Institute Griffith UniversitySouthport, Qld., Australia
| |
Collapse
|
17
|
Xu YC, Li RP, Xue FS, Cui XL, Wang SY, Liu GP, Yang GZ, Sun C, Liao X. κ-Opioid receptors are involved in enhanced cardioprotection by combined fentanyl and limb remote ischemic postconditioning. J Anesth 2015; 29:535-43. [DOI: 10.1007/s00540-015-1998-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/04/2015] [Indexed: 11/29/2022]
|
18
|
|
19
|
Gomes I, Fujita W, Chandrakala MV, Devi LA. Disease-specific heteromerization of G-protein-coupled receptors that target drugs of abuse. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 117:207-65. [PMID: 23663971 DOI: 10.1016/b978-0-12-386931-9.00009-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Drugs of abuse such as morphine or marijuana exert their effects through the activation of G-protein-coupled receptors (GPCRs), the opioid and cannabinoid receptors, respectively. Moreover, interactions between either of these receptors have been shown to be involved in the rewarding effects of drugs of abuse. Recent advances in the field, using a variety of approaches, have demonstrated that many GPCRs, including opioid, cannabinoid, and dopamine receptors, can form associations between different receptor subtypes or with other GPCRs to form heteromeric complexes. The formation of these complexes, in turn, leads to the modulation of the properties of individual protomers. The development of tools that can selectively disrupt GPCR heteromers as well as monoclonal antibodies that can selectively block signaling by specific heteromer pairs has indicated that heteromers involving opioid, cannabinoid, or dopamine receptors may play a role in various disease states. In this review, we describe evidence for opioid, cannabinoid, and dopamine receptor heteromerization and the potential role of GPCR heteromers in pathophysiological conditions.
Collapse
Affiliation(s)
- Ivone Gomes
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, USA
| | | | | | | |
Collapse
|
20
|
Maslov LN, Naryzhnaia NV, Tsibulnikov SY, Kolar F, Zhang Y, Wang H, Gusakova AM, Lishmanov YB. Role of endogenous opioid peptides in the infarct size-limiting effect of adaptation to chronic continuous hypoxia. Life Sci 2013; 93:373-9. [PMID: 23891777 DOI: 10.1016/j.lfs.2013.07.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/27/2013] [Accepted: 07/15/2013] [Indexed: 11/16/2022]
Abstract
AIMS The objective of this study was to examine the involvement of endogenous opioid peptides and opioid receptor (OR) subtypes in the cardioprotective effect of adaptation to chronic hypoxia in rats. MAIN METHODS Rats were exposed to continuous normobaric hypoxia (CNH; 12% oxygen) for 3 weeks. Myocardial ischemia was induced by 20-min coronary artery occlusion followed by 3-h reperfusion in anesthetized open-chest animals. Various OR antagonists were administered to rats prior to ischemia. The size of myocardial infarction and the incidence of ischemic ventricular arrhythmias were assessed. Myocardial and plasma concentrations of opioid peptides (met-enkephalin, β-endorphin, and endomorphins) were determined. KEY FINDINGS Adaptation to CNH significantly increased myocardial and plasma concentrations of opioids, potentiated their further elevation by ischemia/reperfusion, and reduced myocardial infarct size, but it did not affect the incidence of ischemic arrhythmias. The infarct size-limiting effect of CNH was abolished by OR antagonists naltrexone (non-selective), naloxone methiodide (non-selective peripherally acting), TIPP[ψ] (δ-OR), naltriben (δ2-OR), or CTAP (μ-OR), while BNTX (δ1-OR) and nor-binaltorphimine (κ-OR) had no effect. SIGNIFICANCE The results suggest that the infarct size-limiting effect afforded by adaptation to CNH is mediated by activation of peripheral δ2- and μ-ORs by elevated levels of endogenous opioid peptides.
Collapse
Affiliation(s)
- Leonid N Maslov
- Laboratory Experimental Cardiology, Research Institute for Cardiology, Siberian Branch of the Russian Academy of Medical Sciences, Tomsk 634012, Russia.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Pharmacological traits of delta opioid receptors: pitfalls or opportunities? Psychopharmacology (Berl) 2013; 228:1-18. [PMID: 23649885 PMCID: PMC3679311 DOI: 10.1007/s00213-013-3129-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/15/2013] [Indexed: 12/11/2022]
Abstract
RATIONALE Delta opioid receptors (DORs) have been considered as a potential target to relieve pain as well as treat depression and anxiety disorders and are known to modulate other physiological responses, including ethanol and food consumption. A small number of DOR-selective drugs are in clinical trials, but no DOR-selective drugs have been approved by the Federal Drug Administration and some candidates have failed in phase II clinical trials, highlighting current difficulties producing effective delta opioid-based therapies. Recent studies have provided new insights into the pharmacology of the DOR, which is often complex and at times paradoxical. OBJECTIVE This review will discuss the existing literature focusing on four aspects: (1) Two DOR subtypes have been postulated based on differences in pharmacological effects of existing DOR-selective ligands. (2) DORs are expressed ubiquitously throughout the body and central nervous system and are, thus, positioned to play a role in a multitude of diseases. (3) DOR expression is often dynamic, with many reports of increased expression during exposure to chronic stimuli, such as stress, inflammation, neuropathy, morphine, or changes in endogenous opioid tone. (4) A large structural variety in DOR ligands implies potential different mechanisms of activating the receptor. CONCLUSION The reviewed features of DOR pharmacology illustrate the potential benefit of designing tailored or biased DOR ligands.
Collapse
|
22
|
Karlsson LO, Bergh N, Li L, Bissessar E, Bobrova I, Gross GJ, Akyürek LM, Grip L. Dose-dependent cardioprotection of enkephalin analogue Eribis peptide 94 and cardiac expression of opioid receptors in a porcine model of ischaemia and reperfusion. Eur J Pharmacol 2011; 674:378-83. [PMID: 22119384 DOI: 10.1016/j.ejphar.2011.11.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 11/02/2011] [Accepted: 11/04/2011] [Indexed: 11/24/2022]
Abstract
Opioids confer cardioprotection after myocardial ischaemia and reperfusion. The primary aim of the present study was to evaluate the cardioprotective effect of different doses of enkephalin analogue Eribis peptide 94 (EP 94) in a porcine model of ischaemia and reperfusion. A secondary aim was to analyse the impact of ischaemia and reperfusion on the expression of opioid receptor subtypes in the porcine heart. Thirty-four anesthetised pigs underwent 40 min of balloon occlusion of the left anterior descending coronary artery followed by four hours of reperfusion. Pigs were given either vehicle (0.9% NaCl) or one of four doses of EP 94 (0.2, 1, 5 or 25 ug/kg at each administration, respectively), intravenously after 26, 33 and 40 min of ischaemia. Hearts were stained to quantify area at risk and infarct size. mRNA and protein expressions of the opioid receptor subtypes were detected with RT-PCR, immunoblotting and immunohistochemistry in the control and ischaemic/reperfused areas. There was a significant dose-response relationship between higher doses of EP 94 and reduced infarct size. Expression of κ- and δ-opioid receptors was detected at both mRNA and protein levels. In ischaemic/reperfused areas, an increased expression of mRNA for both receptors was observed, whereas only protein expression for the δ subtype was up-regulated. The μ-opioid receptor was not detected.
Collapse
Affiliation(s)
- Lars O Karlsson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Göteborg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Role of central and peripheral opioid receptors in the cardioprotection of intravenous morphine preconditioning. Ir J Med Sci 2011; 180:881-5. [DOI: 10.1007/s11845-011-0734-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 07/07/2011] [Indexed: 10/17/2022]
|
24
|
Kim JH, Jang YH, Chun KJ, Kim J, Park YH, Kim JS, Kim JM, Lee MY. Kappa-opioid receptor activation during reperfusion limits myocardial infarction via ERK1/2 activation in isolated rat hearts. Korean J Anesthesiol 2011; 60:351-6. [PMID: 21716908 PMCID: PMC3110294 DOI: 10.4097/kjae.2011.60.5.351] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 12/09/2010] [Accepted: 12/09/2010] [Indexed: 11/10/2022] Open
Abstract
Background We investigated whether p42/p44 extracellular signal-regulated kinases (ERK1/2) and/or phosphatidylinositol-3-OH kinase (PI3K)-Akt play a crucial role in cardioprotection by κ-opioid receptor (KOP) activation. Methods Langendorff perfused rat hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. Antagonists of ERK1/2 and PI3K were perfused in hearts treated with the KOP agonist U50488H (U50). Infarct size was measured after 2 h of reperfusion. The phosphorylation states of ERK1/2 and Akt by Western immunoblots were determined. Drugs were perfused for a period of 5 min before and 30 min after reperfusion. Results Inhibition of ERK1/2 (26.8 ± 2.9%, P < 0.05 vs. U50) but not PI3K (15.5 ± 1.1%, P > 0.05 vs. U50) completely abrogated the anti-infarct effect of U50488H. Western blot analysis revealed a significant increase in ERK1/2 but not Akt phsophorylation in U50488H-treated hearts as compared to control hearts when measured immediately after reperfusion. Conclusions KOP activation effectively reduces myocardial infarction. The anti-infarct effect of U50488H is mediated by the ERK1/2, but not the PI3K-Akt pathway.
Collapse
Affiliation(s)
- June Hong Kim
- Institute of Cardiovascular Research Center, Pusan National University Yangsan Hospital, Yangsan, Korea
| | | | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Jaiswal A, Kumar S, Seth S, Dinda AK, Maulik SK. Effect of U50,488H, a κ-opioid receptor agonist on myocardial α-and β-myosin heavy chain expression and oxidative stress associated with isoproterenol-induced cardiac hypertrophy in rat. Mol Cell Biochem 2010; 345:231-40. [DOI: 10.1007/s11010-010-0577-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 08/09/2010] [Indexed: 01/19/2023]
|
27
|
Wong GTC, Li R, Jiang LL, Irwin MG. Remifentanil post-conditioning attenuates cardiac ischemia-reperfusion injury via kappa or delta opioid receptor activation. Acta Anaesthesiol Scand 2010; 54:510-8. [PMID: 19878098 DOI: 10.1111/j.1399-6576.2009.02145.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Ischemic pre- or post-conditioning of the heart has been shown to involve opioid receptors. Remifentanil, an ultra-short-acting selective mu opioid receptor agonist in clinical use, pre-conditions the rat heart against ischemia-reperfusion injury. This study investigates whether remifentanil post-conditioning is also cardioprotective. METHODS Remifentanil post-conditioning (5-min infusion at 1, 5, 10 or 20 microg/kg/min) or ischemic post-conditioning (three cycles of a 10 s reperfusion interspersed with a 10 s ischemia) was induced in an open-chest rat heart model of ischemia and reperfusion injury, in the presence or absence of nor-binaltorphimine, naltrindole or CTOP, specific kappa, delta and mu opioid receptor antagonists, respectively. The same sequence of experiments was repeated in the isolated heart model using the maximal protective dose of remifentanil from the dose-response studies. RESULTS Both ischemic and remifentanil post-conditioning reduced the myocardial infarct size relative to the control group in both models. This cardioprotective effect for both post-conditioning regimes was prevented by the prior administration of nor-binaltorphimine and naltrindole but not CTOP. The sole administration of the antagonists had no effect on the size of myocardial infarction. CONCLUSIONS These results indicate that remifentanil post-conditioning protects the heart from ischemia-reperfusion injury to a similar extent as of ischemic post-conditioning. This protection involves kappa and delta but not mu opioid receptor activation. This drug has great potential as a clinical post-conditioning modality as it can be given in large doses without prolonged opioid-related side effects.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Blood Pressure/drug effects
- Blood Pressure/physiology
- Cardiotonic Agents
- Dose-Response Relationship, Drug
- Heart Rate/drug effects
- Heart Rate/physiology
- In Vitro Techniques
- Ischemic Preconditioning, Myocardial
- Male
- Myocardial Infarction/pathology
- Myocardial Reperfusion Injury/drug therapy
- Myocardial Reperfusion Injury/pathology
- Myocardium/pathology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Piperidines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, mu/antagonists & inhibitors
- Remifentanil
- Somatostatin/analogs & derivatives
- Somatostatin/pharmacology
Collapse
Affiliation(s)
- G T C Wong
- Department of Anaesthesiology, University of Hong Kong, Queen Mary Hospital, Hong Kong.
| | | | | | | |
Collapse
|
28
|
Gross ER, Hsu AK, Gross GJ. Acute methadone treatment reduces myocardial infarct size via the delta-opioid receptor in rats during reperfusion. Anesth Analg 2009; 109:1395-402. [PMID: 19843777 DOI: 10.1213/ane.0b013e3181b92201] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Methadone is an opioid agonist often given to manage acute and chronic pain. We sought to determine whether methadone compared with morphine dose dependently reduces myocardial infarct size (IS) and whether the mechanism is delta-opioid receptor mediated. Furthermore, we examined whether myocardial IS reduction varies with the timing of methadone administration or duration of induced ischemia. METHODS After surgical instrumentation, we divided male Sprague-Dawley rats into 3 sets. The first set was divided into groups, which received methadone (0.03-3 mg/kg), morphine (0.03-3 mg/kg), or water (placebo) 30 min before ischemia. Some animals of the first set also received the delta-opioid antagonist naltrindole (5 mg/kg) before methadone (0.3 mg/kg), morphine (0.3 mg/kg), or placebo administration. The second set of animals was divided into groups that received methadone (0.3 mg/kg) 5 min before reperfusion or 10 s after reperfusion. These 2 sets of animals were subjected to 30 min of myocardial ischemia by left anterior descending coronary artery occlusion and then 2 h of reperfusion. The third set of animals received placebo, methadone (0.3 mg/kg), or morphine (0.3 mg/kg) 5 min before reperfusion and were subjected to 45 min of ischemia by left anterior descending coronary artery occlusion with 2 h of reperfusion. Myocardial IS was assessed by staining myocardial tissue with triphenyltetrazolium chloride and expressed as a percentage of the area at risk (mean +/- sem). RESULTS Methadone or morphine administered before ischemia reduced myocardial IS. The greatest effect was achieved at a dose of 0.3 mg/kg (methadone, 46% +/- 1%, P < 0.001 and morphine, 47% +/- 1%, P < 0.001 versus placebo, 61% +/- 1%, respectively). Naltrindole (5 mg/kg) blocked methadone-induced (0.3 mg/kg) and morphine-induced (0.3 mg/kg) cardioprotection (naltrindole + methadone, 58% +/- 1%, P < 0.001 versus methadone; and naltrindole + morphine, 58 +/- 1%, P < 0.001 versus morphine). Methadone (0.3 mg/kg) reduced myocardial IS when given 5 min before reperfusion (46% +/- 1%, P < 0.001 versus placebo) but not 10 s after reperfusion (60% +/- 1%, P = 0.675 versus placebo). No significant myocardial IS differences were seen for placebo when comparing the 45-min ischemia group (64% +/- 1%) with the 30-min ischemia group (60% +/- 1%, P = 0.069). The longer ischemia time of 45 min abrogated methadone-induced IS reduction (64% +/- 2%, P = 0.867 versus 45-min ischemia placebo group) and morphine-induced IS reduction (65% +/- 1%, P = 0.836 versus 45-min ischemia placebo group). CONCLUSIONS These findings demonstrate that methadone and morphine produce similar myocardial IS-sparing effects that are delta-opioid receptor mediated and that are dependent on the duration of myocardial ischemia.
Collapse
Affiliation(s)
- Eric R Gross
- Department of Anesthesiology, Stanford University, Stanford, California, USA
| | | | | |
Collapse
|
29
|
Peng P, Huang LY, Li J, Fan R, Zhang SM, Wang YM, Hu YZ, Sun X, Kaye AD, Pei JM. Distribution of kappa-opioid receptor in the pulmonary artery and its changes during hypoxia. Anat Rec (Hoboken) 2009; 292:1062-7. [PMID: 19462402 DOI: 10.1002/ar.20911] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The present study evaluated the distribution of kappa-opioid receptors (kappa-ORs) in pulmonary arteries (PAs) in rats and investigated whether kappa-ORs are altered in PAs during hypoxia. An animal model of hypobaric/hypoxic pulmonary hypertension and a pulmonary artery smooth muscle cell (PASMC) model of hypoxia were utilized. Distribution of kappa-ORs was determined by fluorescence immunohistochemistry and changes in kappa-ORs expression in PAs and PASMCs were determined by fluorescence immunohistochemistry or Western blot techniques. The kappa-ORs were primarily distributed in the smooth muscle layer of the PAs and in the nucleus of PASMCs. The expression of the kappa-ORs were increased in PAs of rats subjected to hypoxia for 1-4 week (P < 0.01). Accordingly, the expression of kappa-ORs in PASMCs were also increased when subjected to hypoxia for 12-36 hr (P < 0.05). The present study has provided evidence for the first time of the precise location of kappa-ORs in PAs and PASMCs of rats and that hypoxia upregulates expression of kappa-ORs.
Collapse
Affiliation(s)
- Pai Peng
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Lishmanov YB, Maslov LN, Barzakh EI, Krylatov AV, Oltgen PR, Browne SA, Govindashvami M. Effects of activation of μ-, κ1-, δ1-, δ2-, and ORL1-receptors on heart resistance to the pathogenic action of delayed ischemia and reperfusion. BIOL BULL+ 2009. [DOI: 10.1134/s1062359009040062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Bolte C, Newman G, Schultz JEJ. Hypertensive state, independent of hypertrophy, exhibits an attenuated decrease in systolic function on cardiac kappa-opioid receptor stimulation. Am J Physiol Heart Circ Physiol 2009; 296:H967-75. [PMID: 19181965 DOI: 10.1152/ajpheart.00909.2008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Opioids/opiates are commonly administered to alleviate pain, unload the heart, or decrease breathlessness in patients with advanced heart failure. As such, it is important to evaluate whether the myocardial opioidergic system is altered in cardiac disease. A hamster model of spontaneous hypertension was investigated before the development of hypertension (1 mo of age) and in the hypertensive state (10 mo of age) to evaluate the effect of prolonged hypertension on myocardial opioidergic activity. Plasma beta-endorphin was decreased before the development of hypertension and in the hypertensive state (P < 0.05). There was no change in cardiac beta-endorphin content at either time point. No differences were detected in cardiac or plasma dynorphin A, Met-enkephalin, or Leu-enkephalin, or in cardiac peptide expression of kappa- or delta-opioid receptors. mu-Opioid receptor was not detected in either model. To determine how hypertension affects myocardial opioid signaling, the ex vivo work-performing heart was used to assess the cardiac response to opioid administration in healthy hearts and those subjected to chronic hypertension. Agonists selective for the kappa- and delta-opioid receptors, but not mu-opioid receptors, induced a concentration-dependent decrease in cardiac function. The decrease in left ventricular systolic pressure on administration of the kappa-opioid receptor-selective agonist, U50488H, was attenuated in hearts from hamsters subjected to chronic, untreated hypertension (P < 0.05) compared with control. These results show that peripheral and myocardial opioid expression and signaling are altered in hypertension.
Collapse
Affiliation(s)
- Craig Bolte
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | |
Collapse
|
32
|
van den Brink OWV, Delbridge LM, Rosenfeldt FL, Penny D, Esmore DS, Quick D, Kaye DM, Pepe S. Endogenous cardiac opioids: enkephalins in adaptation and protection of the heart. Heart Lung Circ 2008; 12:178-87. [PMID: 16352129 DOI: 10.1046/j.1444-2892.2003.00240.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opiates have been used for thousands of years in the form of opium for relief of pain or fever and to induce sleep. However, it was only in the 1970s that the endogenous ligands for the opiate receptors were identified and termed opioid peptides. Opioid peptides activate G protein-coupled receptors in the central and autonomic nervous system, with marked effects on the regulation of pain perception, body temperature, respiration, heart rate and blood pressure. Cardiovascular regulatory effects of endogenous opioids were initially considered to originate from neural centres in the central nervous system, facilitating a regulatory role in neuro-transmission, as demonstrated by the presynaptic co-release from sympathetic neurones of norepinephrine with enkephalin or acetylcholine with enkephalin. However, opioid peptides of myocardial origin have also recently been shown to play a key role in local regulation of the heart. This brief review highlights the key features of the enkephalin opioids in the heart and the current understanding of their role in development, ageing, cardioprotection, hypertension, hypertrophy, and heart failure.
Collapse
|
33
|
Morphine Postconditioning Protects Against Reperfusion Injury in the Isolated Rat Hearts. J Surg Res 2008; 145:287-94. [DOI: 10.1016/j.jss.2007.07.020] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Revised: 07/22/2007] [Accepted: 07/25/2007] [Indexed: 11/21/2022]
|
34
|
Lee YC, Jang YH, Kim JM, Kim AR, Kim CJ, Kim YN. Effect of A Kappa-opioid Receptor Agonist U50488H Given at Early Reperfusion Phase in Isolated Rat Hearts. Korean J Anesthesiol 2008. [DOI: 10.4097/kjae.2008.54.3.s29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Yong Cheol Lee
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Young Ho Jang
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Jin Mo Kim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Ae Ra Kim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Chan Jin Kim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Yoon Nyun Kim
- Department of Internal Medicine, School of Medicine, Keimyung University, Daegu, Korea
| |
Collapse
|
35
|
Hernández A, Sola MA, Domínguez B, Rochera MI, Bascuñana P, Gancedo V. [Is morphine still the analgesic of choice in acute myocardial infarction?]. REVISTA ESPANOLA DE ANESTESIOLOGIA Y REANIMACION 2008; 55:32-39. [PMID: 18333384 DOI: 10.1016/s0034-9356(08)70495-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Chest pain is the most common symptom of patients who present with ischemic heart disease. Morphine has traditionally been the drug of choice for managing chest pain in acute coronary syndrome (ACS) due to its high analgesic potency, though its physiological effects are poorly understood. Routinely used for managing chest pain, morphine is recommended in the 2002 guidelines of the American College of Cardiology/American Heart Association. This recommendation, however, is not based on a high level of scientific evidence but on expert opinion. Studies have found both for and against the use of morphine in ACS, suggesting that its benefits are perhaps not altogether clear. This review examines the pathophysiological effects of morphine and their cardiac implications, with special attention to a possible negative effect on ACS. We reviewed articles in the MEDLINE database from 1982 to 2006.
Collapse
Affiliation(s)
- A Hernández
- Servicio de Anestesiología y Reanimación, Hospital General Universitario Vall d'Hebron, Barcelona.
| | | | | | | | | | | |
Collapse
|
36
|
Lee YC, Jang YH, Kim JM, Kim AR, Lee SR, Kim YN, Hong JH. Ischemic Postconditioning Inhibits Mitochondrial Permeability Transition Pore via Opioid Receptor Activation in Intact Rat Heart. Korean J Anesthesiol 2008. [DOI: 10.4097/kjae.2008.54.3.320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Yong Cheol Lee
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Young Ho Jang
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Jin Mo Kim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Ae Ra Kim
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Seung Ryong Lee
- Department of Pharmacology, School of Medicine, Keimyung University, Daegu, Korea
| | - Yoon Nyun Kim
- Department of Internal Medicine, School of Medicine, Keimyung University, Daegu, Korea
| | - Ji Hee Hong
- Department of Anesthesiology and Pain Medicine, School of Medicine, Keimyung University, Daegu, Korea
| |
Collapse
|
37
|
Maslov LN, Lishmanov YB, Barzakh EI, Lasukova TV, Rice KK, Oeltgen PR. Negative inotropic and chronotropic effects of δ-opioid receptor antagonists are mediated via non-opioid receptors. Bull Exp Biol Med 2006; 141:420-3. [PMID: 17152360 DOI: 10.1007/s10517-006-0188-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Ten-minute perfusion of intact isolated rat heart with Krebs-Henseleit solution containing delta-opioid receptor agonists (DPDPE, (-)-TAN-67) or delta-opioid receptor antagonists (naltrindole, TIPP[psi], ICI 174,864) at a final concentration of 0.1 mg/liter decreased HR, blood pressure in the left ventricle, and the rates of myocardial contraction and relaxation. Intravenous injection of delta-agonists (DPDPE, (-)-TAN-67, deltorphin II) or delta-antagonists (naltrindole, TIPP[psi], ICI 174,864) decreased HR in narcotized rats. Naloxone and naltrexone produced no effect on contractility and HR both in vivo and in vitro. Preliminary injection of naloxone and naltrexone did not prevent the negative chronotropic effect of ICI 174,864 in vitro. The negative inotropic and chronotropic effects of delta-opioid receptor antagonists are mediated by unknown non-opioid receptors in the heart.
Collapse
Affiliation(s)
- L N Maslov
- State Research Institute of Cardiology, Tomsk Research Center, Siberian Division of the Russian Academy of Medical Sciences.
| | | | | | | | | | | |
Collapse
|
38
|
Xu W, Yoon SI, Huang P, Wang Y, Chen C, Chong PLG, Liu-Chen LY. Localization of the κ Opioid Receptor in Lipid Rafts. J Pharmacol Exp Ther 2006; 317:1295-306. [PMID: 16505160 DOI: 10.1124/jpet.105.099507] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Lipid rafts are microdomains of plasma membranes enriched in cholesterol and sphingolipids in the outer layer. We determined whether kappa opioid receptors (KOR) in human placenta and FLAG (DYKDDDDK)-tagged human KOR (FLAG-hKOR) expressed in Chinese hamster ovary (CHO) cells are localized in lipid rafts and whether changes in cholesterol contents affect hKOR properties and signaling. Lipid rafts were prepared from placenta membranes and CHO cells expressing FLAG-hKOR using the Na2CO3 method and fractionation through a sucrose density gradient. The majority of the KOR in the placenta and FLAG-hKOR in CHO cells, determined by [3H]diprenorphine binding and/or immunoblotting with an anti-FLAG antibody, was present in low-density fractions, coinciding with high levels of caveolin-1 and cholesterol, markers of lipid rafts, which indicated that the KOR is localized in lipid rafts. Pretreatment with 2% methyl beta-cyclodextrin (MCD) reduced cholesterol content by approximately 48% and changed the cells from spindle-shaped to spherical. MCD treatment disrupted lipid rafts, shifted caveolin-1 and FLAG-hKOR to higher density fractions, increased the affinity of (-)-(trans)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamide (U50,488H) for the hKOR, and greatly increased U50,488H-induced [35S]guanosine 5'-O-(3-thio)triphosphate binding and p42/44 mitogen-activated protein kinase phosphorylation. Cholesterol replenishment reversed all the MCD effects. Caveolin-1 immunoprecipitated with Galphai proteins and MCD treatment reduced caveolin-1 associated with Galphai proteins, which may contribute to the enhanced agonist-induced G protein activation. Caveolin-1 also immunoprecipitated with FLAG-hKOR, but MCD treatment had no effect on the association. Thus, the KOR is located in lipid rafts and its localization in the microdomains greatly affects coupling to G proteins.
Collapse
Affiliation(s)
- Wei Xu
- Department of Pharmacology, Center for Substance Abuse Research, Temple University School of Medicine, 3420 N. Broad St., Philadelphia, PA 19140, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Li Y, Saito Y, Suzuki M, Ueda H, Endo M, Maruyama K. Kyotorphin has a novel action on rat cardiac muscle. Biochem Biophys Res Commun 2006; 339:805-9. [PMID: 16325776 DOI: 10.1016/j.bbrc.2005.11.081] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Accepted: 11/14/2005] [Indexed: 10/25/2022]
Abstract
Several endogenous peptides for G-protein-coupled receptors have been found to play physiological roles in muscle contraction in addition to their well-demonstrated actions in other tissues. To further identify such peptides, we screened over 400 peptides using an isometric tension assay of rat papillary muscle. Here, we report that kyotorphin, which is known as an analgesic dipeptide, has a cardiac effect. Although kyotorphin had no effect on the twitch tension itself, it inhibited beta-adrenergic agonist isoprenaline-induced increases in twitch tension in a dose-dependent manner. Leu-Arg, a selective antagonist of kyotorphin, reversed this inhibitory effect. The inhibitory effect was also reversed by naloxone, an opioid receptor antagonist. These results suggest that kyotorphin may release opioid peptides from rat cardiac muscle and have an indirect regulatory role in beta-adrenergic action through cross-talk with opioid receptors.
Collapse
Affiliation(s)
- Yue Li
- Department of Pharmacology, Saitama Medical School, Moroyama 38, Iruma-gun, Saitama 350-0495, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Head BP, Patel HH, Roth DM, Lai NC, Niesman IR, Farquhar MG, Insel PA. G-protein-coupled Receptor Signaling Components Localize in Both Sarcolemmal and Intracellular Caveolin-3-associated Microdomains in Adult Cardiac Myocytes. J Biol Chem 2005; 280:31036-44. [PMID: 15961389 DOI: 10.1074/jbc.m502540200] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
This study tests the hypothesis that G-protein-coupled receptor (GPCR) signaling components involved in the regulation of adenylyl cyclase (AC) localize with caveolin (Cav), a protein marker for caveolae, in both cell-surface and intracellular membrane regions. Using sucrose density fractionation of adult cardiac myocytes, we detected Cav-3 in both buoyant membrane fractions (BF) and heavy/non-buoyant fractions (HF); beta2-adrenergic receptors (AR) in BF; and AC5/6, beta1-AR, M4-muscarinic acetylcholine receptors (mAChR), mu-opioid receptors, and Galpha(s) in both BF and HF. In contrast, M2-mAChR, Galpha(i3), and Galpha(i2) were found only in HF. Immunofluorescence microscopy showed co-localization of Cav-3 with AC5/6, Galpha(s), beta2-AR, and mu-opioid receptors in both sarcolemmal and intracellular membranes, whereas M2-mAChR were detected only intracellularly. Immunofluorescence of adult heart revealed a distribution of Cav-3 identical to that in isolated adult cardiac myocytes. Upon immunoelectron microscopy, Cav-3 co-localized with AC5/6 and Galpha(s) in sarcolemmal and intracellular vesicles, the latter closely allied with T-tubules. Cav-3 immunoprecipitates possessed components that were necessary and sufficient for GPCR agonist-promoted stimulation and inhibition of cAMP formation. The distribution of GPCR, G-proteins, and AC with Cav-3 in both sarcolemmal and intracellular T-tubule-associated regions indicates the existence of multiple Cav-3-localized cellular microdomains for signaling by hormones and drugs in the heart.
Collapse
MESH Headings
- Adenylyl Cyclases/metabolism
- Animals
- Biomarkers
- Caveolin 3
- Caveolins/metabolism
- Cell Fractionation
- Cells, Cultured
- Cyclic AMP/metabolism
- Fibroblasts/cytology
- Fibroblasts/metabolism
- GTP-Binding Protein alpha Subunits/metabolism
- Humans
- Intracellular Membranes/chemistry
- Intracellular Membranes/metabolism
- Intracellular Membranes/ultrastructure
- Isoenzymes/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Muscarinic M2/metabolism
- Receptors, Adrenergic, beta/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Muscarinic/metabolism
- Receptors, Opioid/metabolism
- Sarcolemma/chemistry
- Sarcolemma/metabolism
- Sarcolemma/ultrastructure
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Brian P Head
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093-0636, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Wu S, Wong MCY, Chen M, Cho CH, Wong TM. Role of opioid receptors in cardioprotection of cold-restraint stress and morphine. J Biomed Sci 2005; 11:726-31. [PMID: 15591768 DOI: 10.1007/bf02254356] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2004] [Accepted: 07/12/2004] [Indexed: 10/25/2022] Open
Abstract
Since cold exposure confers cardioprotection, the present study attempted to determine the role of opioid receptors (OR). Stress with cold exposure and restraint for 3 h, shown previously to induce peptic ulcer in a synergistic manner, attenuated infarct size induced by myocardial ischemia and reperfusion in the isolated perfused rat heart from 36.64 +/- 1.8 to 22.85 +/- 2.6%. This is similar to protecting the rat with morphine at 8 mg/kg, which also attenuated the infarct size from 36.26 +/- 1.6 to 20.30 +/- 2.1%. The effects of cold-restraint or morphine were abolished by naloxone, a non-selective OR antagonist; nor-binaltorphimine, a selective kappa-OR antagonist; naltrindole, a selective delta-OR antagonist, or CTOP, a selective mu-OR antagonist. The effects were also attenuated by blockade of protein kinase C or the mitochondrial K(ATP) channel. The finding is first evidence that all three OR subtypes mediate cardioprotection of cold-restraint stress in the rat.
Collapse
Affiliation(s)
- S Wu
- Department of Physiology, Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
42
|
Maslov LN, Lasukova TV, Bespalova ZD, Oldgen P, Rice KK, Nagase H. δ-opioid receptor antagonists exhibit properties of partial δ-receptor agonists in isolated perfused heart. Bull Exp Biol Med 2004; 138:376-9. [PMID: 15665949 DOI: 10.1007/s10517-005-0046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Perfusion of the isolated intact rat heart with Krebs-Henseleit solution containing agonists ((-)-TAN-67, DPDPE, and dalargin) or antagonists of delta-opioid receptors (naltrindole, TIPP[psi], and ICI 174,864) in a final concentration of 0.1 mg/liter was followed by a decrease in the heart rate, end-diastolic pressure, contraction rate, relaxation rate, and left ventricular developed pressure. Perfusion with a solution containing the delta-opioid receptor agonist DPDPE or delta-antagonists naltrindole, TIPP[psi], and ICI 174,864 before modeling of global ischemia increased the severity of reperfusion-induced contractile dysfunction in the myocardium. Our results suggest that delta-opioid receptor antagonists in vitro exhibit properties of partial delta-receptor agonists.
Collapse
MESH Headings
- Animals
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Enkephalin, Leucine/analogs & derivatives
- Enkephalin, Leucine/pharmacology
- Enkephalin, Leucine-2-Alanine/analogs & derivatives
- Enkephalin, Leucine-2-Alanine/pharmacology
- Heart/drug effects
- Heart/physiology
- In Vitro Techniques
- Male
- Myocardial Contraction/drug effects
- Myocardial Ischemia/physiopathology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Oligopeptides/pharmacology
- Perfusion
- Quinolines/pharmacology
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/physiology
Collapse
Affiliation(s)
- L N Maslov
- Institute of Cardiology, Tomsk Research Center, Siberian Division, Russian Academy of Medical Sciences.
| | | | | | | | | | | |
Collapse
|
43
|
?-opioid receptor antagonists exhibit properties of partial ?-receptor agonists in isolated perfused heart. Bull Exp Biol Med 2004. [DOI: 10.1007/s10517-004-0016-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Wang G, Wang H, Yang Y, Wong TM. κ-Opioid receptor stimulation inhibits growth of neonatal rat ventricular myocytes. Eur J Pharmacol 2004; 498:53-8. [PMID: 15363975 DOI: 10.1016/j.ejphar.2004.07.082] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2004] [Revised: 07/12/2004] [Accepted: 07/15/2004] [Indexed: 10/26/2022]
Abstract
The effects of trans-(+/-)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]-benzeneacetamide methanesulfonate salt (U50,488H), a selective kappa-opioid receptor agonist, on growth in neonatal ventricular myocytes were determined. In 15% serum culture medium, U50,488H at 0.1-1 microM significantly reduced the protein content, [3H]leucine uptake and cell size of the myocytes. The effect of U50,488H on protein content was abolished in the presence of 1 microM nor-binaltorphimine (nor-BNI), a selective kappa-opioid receptor antagonist. In a 0.4% serum medium, U50,488H at 0.1-1 microM had no effect on myocyte growth. Interestingly, 1 microM U50,488H abolished the stimulatory effects of 1 microM norepinephrine on protein content, [3H]leucine uptake and cell size of the myocytes in the low serum medium. The effect of U50,488H was abolished by 1 microM nor-BNI. With the exception of cell size, the effects of norepinephrine were completely abolished by blockade of both alpha- and beta-adrenoceptors, but only partially blocked by blockade of either adrenoceptors. These results provide first evidence that kappa-opioid receptor stimulation inhibits growth of the neonatal ventricular myocyte as a result of direct action as well as by inhibiting sympathetic stimulation of the heart. The stimulatory effects of sympathetic activity on growth occurs via both alpha- and beta-adrenoceptors.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- Adrenergic alpha-Antagonists/pharmacology
- Adrenergic beta-Antagonists/pharmacology
- Analgesics, Non-Narcotic/pharmacology
- Animals
- Animals, Newborn
- Cell Division/drug effects
- Cell Division/physiology
- Cell Size/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Heart Ventricles/cytology
- Heart Ventricles/drug effects
- Heart Ventricles/metabolism
- Leucine/pharmacokinetics
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Norepinephrine/pharmacology
- Prazosin/pharmacology
- Propranolol/pharmacology
- Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, kappa/metabolism
- Tritium
Collapse
Affiliation(s)
- Guijun Wang
- Department of Pharmacology, Jinzhou Medical College, Jinzhou, China
| | | | | | | |
Collapse
|
45
|
Ventura C, Zinellu E, Maninchedda E, Fadda M, Maioli M. Protein kinase C signaling transduces endorphin-primed cardiogenesis in GTR1 embryonic stem cells. Circ Res 2003; 92:617-22. [PMID: 12623877 DOI: 10.1161/01.res.0000065168.31147.5b] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The prodynorphin gene and its product, dynorphin B, have been found to promote cardiogenesis in embryonic cells by inducing the expression of GATA-4 and Nkx-2.5, two transcription factor-encoding genes essential for cardiogenesis. The molecular mechanism(s) underlying endorphin-induced cardiogenesis remain unknown. In the present study, we found that GTR1 embryonic stem (ES) cells expressed cell surface kappa opioid receptors, as well as protein kinase C (PKC)-alpha, -beta1, -beta2, -delta, -epsilon, and -zeta. Cardiac differentiation was associated with a marked increase in the Bmax value for a selective opioid receptor ligand and complex subcellular redistribution of selected PKC isozymes. PKC-alpha, -beta1, -beta2, -delta, and -epsilon all increased in the nucleus of ES-derived cardiac myocytes, compared with nuclei from undifferentiated cells. In both groups of cells, PKC-delta and -epsilon were mainly expressed at the nuclear level. The nuclear increase of PKC-alpha, -beta1, and -beta2 was due to a translocation from the cytosolic compartment. In contrast, the increase of both PKC-delta and PKC-epsilon in the nucleus of ES-derived cardiomyocytes occurred independently of enzyme translocation, suggesting changes in isozyme turnover and/or gene expression during cardiogenesis. No change in PKC-zeta expression was observed during cardiac differentiation. Opioid receptor antagonists prevented the nuclear increase of PKC-alpha, PKC-beta1, and PKC-beta2 and reduced cardiomyocyte yield but failed to affect the nuclear increase in PKC-delta and -epsilon. PKC inhibitors prevented the expression of cardiogenic genes and dynorphin B in ES cells and abolished their development into beating cardiomyocytes.
Collapse
Affiliation(s)
- Carlo Ventura
- Department of Biomedical Sciences, Center for Biotechnology Development and Biodiversity Research, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | | | | | | | | |
Collapse
|
46
|
Nakae Y, Fujita S, Namiki A. Modulation of myofilament Ca2+ densitivity by delta- and kappa-opioid agonists in intact guinea pig hearts. Anesth Analg 2003; 96:733-739. [PMID: 12598254 DOI: 10.1213/01.ane.0000050281.16556.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
UNLABELLED We investigated whether delta- and kappa-opioid agonists alter myocardial function, intracellular Ca(2+) concentration ([Ca(2+)](i)), and myofilament Ca(2+) sensitivity in intact guinea pig beating hearts and whether these effects are mediated by an opioid receptor. Intact guinea pig hearts were perfused with modified Krebs Ringer solution containing delta- (TAN-67) and kappa- (ICI-199441) opioid agonists in the absence and presence of delta- (BNTX) and kappa- (nor-BNI) opioid antagonists, respectively, while functional variables and [Ca(2+)](i) were recorded. TAN-67 (1 microM) and ICI-199441 (1 microM) decreased heart rate (P < 0.05). TAN-67 (1 microM) and ICI-199441 (1 micro M) decreased available [Ca(2+)](i) without changing developed left ventricular pressure (LVP) (P < 0.05). TAN-67 (1 microM) and ICI-199441 (1 microM) also caused a leftward shift in the curve of developed LVP as a function of available [Ca(2+)](i) (P < 0.05). ICI-199441 (1 microM) produced a steeper slope in the relation curve compared with baseline (P < 0.05). BNTX (1 microM) and nor-BNI (1 microM) blocked the effects of TAN-67 and ICI-199441, respectively. delta- and kappa-opioid agonists enhance myofilament Ca(2+) sensitivity despite decreasing available [Ca(2+)](i) in intact isolated guinea pig hearts, and these effects are mediated by delta- and kappa-opioid receptor stimulation. IMPLICATIONS Our results indicate that delta- and kappa-opioid agonists enhance myofilament Ca(2+) sensitivity despite decreasing available intracellular Ca(2+) concentrations in intact isolated guinea pig beating hearts, and these effects are mediated by delta- and kappa-opioid receptor stimulation.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Animals
- Benzylidene Compounds/pharmacology
- Blood Pressure/drug effects
- Calcium/pharmacology
- Coronary Circulation/drug effects
- Guinea Pigs
- Heart/drug effects
- Heart Rate/drug effects
- In Vitro Techniques
- Kinetics
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Pyrrolidines/pharmacology
- Quinolines/pharmacology
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Stimulation, Chemical
- Ventricular Function, Left/drug effects
Collapse
Affiliation(s)
- Yuri Nakae
- *Department of Anesthesiology, Sapporo Medical University School of Medicine; and †Departments of Anesthesiology and Critical Care Medicine, Asahikawa Medical College, Japan
| | | | | |
Collapse
|
47
|
Johnson-Davis KL, Hanson GR, Keefe KA. Lack of effect of kappa-opioid receptor agonism on long-term methamphetamine-induced neurotoxicity in rats. Neurotox Res 2003; 5:273-81. [PMID: 12835119 DOI: 10.1007/bf03033385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
High-dose methamphetamine treatment induces long-term deficits in central monoamine systems. However, the mechanisms underlying these effects are unknown. Previous work has shown that the Kappa-opioid receptor agonist U-69593 [(+)-(5alpha,7alpha,8b)-(+)-N-methyl-N[7-(1-pyrrolidinyl)-1-oxaspiro[4.5]dec-8-yl] benzeneacetamide] attenuates the neurotoxic effects of methamphetamine on extracellular dopamine levels in mice, suggesting that endogenous Kappa-opioid receptor ligands, such as dynorphin, may protect against methamphetamine-induced toxicity and play a role in mediating the long-term consequences of methamphetamine. To further examine the role that dynorphin systems play in methamphetamine-induced neurotoxicity, we administered to male rats a total of four injections of methamphetamine (7.5 mg/kg, s.c.), with a 2-h interval between each dose. Rats were pretreated with either the Kappa-agonist U-69593 (0.32 mg/kg, s.c.) or vehicle, 15 min prior to the first and third methamphetamine injection. Furthermore, cages containing the U-69593 + methamphetamine-treated rats were placed on heating pads for 30 min after the first U-69593 injection to prevent the drug from blocking methamphetamine-induced hyperthermia. Rats were sacrificed 7 days after treatment. Striatal dopamine and serotonin contents were decreased approximately 75% and 55%, respectively, in the methamphetamine-treated rats and approximately 88% and 65%, respectively, in rats receiving the U-69593 + methamphetamine combination. There was a approximately 20% mortality rate in the rats treated with methamphetamine compared to approximately 75% mortality rate in rats treated with both U-69593 and methamphetamine. A similar rate of mortality was observed when combining a different Kappa-agonist, U-50488 [trans-(-)-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl]benzeneacetamine], with methamphetamine. These data suggest that Kappa-agonists do not protect against methamphetamine-induced toxicity to monoamines in rats, and may potentiate mortality when co-administered with methamphetamine.
Collapse
Affiliation(s)
- Kamisha L Johnson-Davis
- Department of Pharmacology and Toxicology, University of Utah, 30 South 2000 East, Rm. 201, Salt Lake City, UT 84112, USA.
| | | | | |
Collapse
|
48
|
Wong TM, Wu S. Role of kappa opioid receptor in cardioprotection of preconditioning: implications in cardiac surgery. J Card Surg 2002; 17:462-4. [PMID: 12630549 DOI: 10.1111/j.1540-8191.2001.tb01178.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Kappa-opioid receptor mediates both immediate and delayed cardioprotection of ischemic/metabolic inhibition preconditioning. Both PKC and mitochondria ATP-sensitive K+ channel, mediate the protective action of kappa-opioid receptor stimulation. In delayed cardioprotection, PKC-epsilon may produce its effect via mitochondria ATP-sensitive K+ channel, and a stress inducible heat stress protein 70 may also be involved. Metabolic inhibition causes cell death which is accompanied by Ca2+ overload and mobilization of Ca2+ from sarcoplasmic reticulum to cytoplasm. The altered Ca2+ homeostasis is restored partially by kappa-opioid receptor or metabolic inhibition preconditioning. The finding indicates that opioids such as morphine may be used in the treatment of ischemic heart diseases and in open heart surgery.
Collapse
Affiliation(s)
- Tak Ming Wong
- Department of Physiology and Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | | |
Collapse
|
49
|
Wu D, Soong Y, Zhao GM, Szeto HH. A highly potent peptide analgesic that protects against ischemia-reperfusion-induced myocardial stunning. Am J Physiol Heart Circ Physiol 2002; 283:H783-91. [PMID: 12124228 DOI: 10.1152/ajpheart.00193.2002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We recently discovered an opioid peptide analgesic, 2',6'-dimethyltyrosine (Dmt)-D-Arg-Phe-Lys-NH(2) ([Dmt(1)]DALDA), that can protect against ischemia-induced myocardial stunning. In buffer-perfused hearts, 30-min global ischemia followed by reperfusion resulted in a significant increase in norepinephrine (NE) overflow immediately upon reperfusion and significant decline in contractile force (45%). Pretreatment with [Dmt(1)]DALDA before ischemia completely abolished myocardial stunning and significantly reduced NE overflow (68%). In contrast, pretreatment with morphine before ischemia only provided brief protection against myocardial stunning and no reduction in NE overflow. [Dmt(1)]DALDA inhibited [(3)H]NE uptake into cardiac synaptosomes in vitro (IC(50) = 3.9 microM), whereas morphine had no effect. Surprisingly, protection against myocardial stunning was apparent even when hearts were perfused with [Dmt(1)]DALDA only upon reperfusion, whereas reperfusion with morphine had no effect. Binding studies with [(3)H][Dmt(1)]DALDA revealed no high-affinity specific binding in cardiac membranes, suggesting that the cardioprotective actions of [Dmt(1)]DALDA are not mediated via opioid receptors. These findings suggest that [Dmt(1)]DALDA is a potent analgesic that may be useful for myocardial stunning resulting from cardiac interventions or myocardial ischemia.
Collapse
Affiliation(s)
- Dunli Wu
- Department of Pharmacology, Joan and Sanford I. Weill Medical College, Cornell University, 1300 York Avenue, New York, NY 10021, USA
| | | | | | | |
Collapse
|
50
|
Zhou JJ, Bian JS, Pei JM, Wu S, Li HY, Wong TM. Role of protein kinase C-epsilon in the development of kappa-opioid receptor tolerance to U50,488H in rat ventricular myocytes. Br J Pharmacol 2002; 135:1675-84. [PMID: 11934807 PMCID: PMC1573305 DOI: 10.1038/sj.bjp.0704640] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The role of protein kinase C-epsilon (PKC-epsilon) in the development of kappa-opioid receptor (kappa-OR) tolerance to the effects of trans-(+/-)-3,4-dichloro-N-methyl-N-(2-[1-pyrrolidinyl]cyclohexyl) (U50,488H), the selective agonist of kappa-OR, was determined in rat ventricular myocytes. Incubation of ventricular myocytes with 1 microM U50,488H for 24 h significantly attenuated the inhibitory effects of 30 microM U50,488H on the electrically-induced [Ca(2+)](i) transient and forskolin-stimulated cyclic AMP accumulation, indicating the development of tolerance to the kappa-OR agonist. Chronic treatment of ventricular myocytes with U50,488H also induced translocation of PKC-epsilon to the particulate fraction. On the other hand, administration of 30 microM U50,488H for 10 min induced translocation of PKC-alpha to the particulate fraction in naïve ventricular myocytes, but not in cells pretreated with 1 microM U50,488H for 24 h. In ventricular myocytes incubated for 24 h with 1 microM U50,488H together with 1 microM chelerythrine or 1 microM GF109203X, PKC inhibitors, or 0.1 microM epsilonV1-2 peptide, a selective inhibitor of PKC-epsilon, 30 microM U50,488H still produced the inhibitory effect on the electrically-induced [Ca(2+)](i) transient as it did in naïve ventricular myocytes. Chronic treatment of ventricular myocytes with U50,488H and chelerythrine also attenuated the development of tolerance to acute U50,488H on cyclic AMP accumulation. Cells exposed to chelerythrine, GF109203X, or epsilonV1-2 peptide alone did not show an altered [Ca(2+)](i) response to U50,488H. These results indicate that activation of PKC-epsilon is a critical step in the development of tolerance in the kappa-OR.
Collapse
Affiliation(s)
- Jing-Jun Zhou
- Department of Physiology and Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jin-Song Bian
- Department of Physiology and Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jian-Ming Pei
- Department of Physiology and Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Song Wu
- Department of Physiology and Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hong-Yu Li
- Department of Physiology and Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tak-Ming Wong
- Department of Physiology and Institute of Cardiovascular Science and Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Author for correspondence:
| |
Collapse
|