Skip to main content
Top
Published in: Cardiovascular Drugs and Therapy 4/2021

Open Access 01-08-2021 | Naltrexone | Routine Article

Chronic Naltrexone Therapy Is Associated with Improved Cardiac Function in Volume Overloaded Rats

Authors: Lukas Dehe, Mohammed Shaqura, Michael Nordine, Helmut Habazettl, Petra von Kwiatkowski, Helena Schluchter, Mehdi Shakibaei, Shaaban A. Mousa, Michael Schäfer, Sascha Treskatsch

Published in: Cardiovascular Drugs and Therapy | Issue 4/2021

Login to get access

Abstract

Purpose

Myocardial opioid receptors were demonstrated in animals and humans and seem to colocalize with membranous and sarcolemmal calcium channels of the excitation–contraction coupling in the left ventricle (LV). Therefore, this study investigated whether blockade of the cardiac opioid system by naltrexone would affect cardiac function and neurohumoral parameters in Wistar rats with volume overload-induced heart failure.

Methods

Volume overload in Wistar rats was induced by an aortocaval fistula (ACF). Left ventricular cardiac opioid receptors were identified by immunohistochemistry and their messenger ribonucleic acid (mRNA) as well as their endogenous ligand mRNA quantified by real-time polymerase chain reaction (RT-PCR). Following continuous delivery of either the opioid receptor antagonist naltrexone or vehicle via minipumps (n = 5 rats each), hemodynamic and humoral parameters were assessed 28 days after ACF induction. Sham-operated animals served as controls.

Results

In ACF rats mu-, delta-, and kappa-opioid receptors colocalized with voltage-gated L-type Ca2+ channels in left ventricular cardiomyocytes. Chronic naltrexone treatment of ACF rats reduced central venous pressure (CVP) and left ventricular end-diastolic pressure (LVEDP), and improved systolic and diastolic left ventricular functions. Concomitantly, rat brain natriuretic peptide (rBNP-45) and angiotensin-2 plasma concentrations which were elevated during ACF were significantly diminished following naltrexone treatment. In parallel, chronic naltrexone significantly reduced mu-, delta-, and kappa-opioid receptor mRNA, while it increased the endogenous opioid peptide mRNA compared to controls.

Conclusion

Opioid receptor blockade by naltrexone leads to improved LV function and decreases in rBNP-45 and angiotensin-2 plasma levels. In parallel, naltrexone resulted in opioid receptor mRNA downregulation and an elevated intrinsic tone of endogenous opioid peptides possibly reflecting a potentially cardiodepressant effect of the cardiac opioid system during volume overload.
Literature
1.
go back to reference Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016;37:2129–200.CrossRef Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016;37:2129–200.CrossRef
2.
go back to reference Pearse RM, Moreno RP, Bauer P, Pelosi P, Metnitz P, Spies C, et al. Mortality after surgery in Europe: a 7 day cohort study. Lancet. 2012;380:1059–65.PubMedPubMedCentral Pearse RM, Moreno RP, Bauer P, Pelosi P, Metnitz P, Spies C, et al. Mortality after surgery in Europe: a 7 day cohort study. Lancet. 2012;380:1059–65.PubMedPubMedCentral
3.
go back to reference Headrick JP, See Hoe LE, Toit Du EF, Peart JN. Opioid receptors and cardioprotection - “opioidergic conditioning” of the heart. Br J Pharmacol. 2015;172:2026–50.PubMedPubMedCentral Headrick JP, See Hoe LE, Toit Du EF, Peart JN. Opioid receptors and cardioprotection - “opioidergic conditioning” of the heart. Br J Pharmacol. 2015;172:2026–50.PubMedPubMedCentral
4.
go back to reference Goudra BG, Duggan M, Chidambaran V, et al. Anesthesiology. 1st ed: Springer; 2018. Goudra BG, Duggan M, Chidambaran V, et al. Anesthesiology. 1st ed: Springer; 2018.
5.
go back to reference Treskatsch S, Feldheiser A, Shaqura M, et al. Cellular localization and adaptive changes of the cardiac delta opioid receptor system in an experimental model of heart failure in rats. Heart Vessel. 2015;31:241–50. Treskatsch S, Feldheiser A, Shaqura M, et al. Cellular localization and adaptive changes of the cardiac delta opioid receptor system in an experimental model of heart failure in rats. Heart Vessel. 2015;31:241–50.
6.
go back to reference Treskatsch S, Shaqura M, Dehe L, et al. Evidence for MOR on cell membrane, sarcoplasmatic reticulum and mitochondria in left ventricular myocardium in rats. Heart Vessel. 2015;31:1380–8. Treskatsch S, Shaqura M, Dehe L, et al. Evidence for MOR on cell membrane, sarcoplasmatic reticulum and mitochondria in left ventricular myocardium in rats. Heart Vessel. 2015;31:1380–8.
7.
go back to reference Treskatsch S, Shaqura M, Dehe L, Feldheiser A, Roepke TK, Shakibaei M, et al. Upregulation of the kappa opioidergic system in left ventricular rat myocardium in response to volume overload: adaptive changes of the cardiac kappa opioid system in heart failure. Pharmacol Res. 2015;102:33–41.PubMed Treskatsch S, Shaqura M, Dehe L, Feldheiser A, Roepke TK, Shakibaei M, et al. Upregulation of the kappa opioidergic system in left ventricular rat myocardium in response to volume overload: adaptive changes of the cardiac kappa opioid system in heart failure. Pharmacol Res. 2015;102:33–41.PubMed
8.
go back to reference Sobanski P, Krajnik M, Shaqura M, Bloch-Boguslawska E, Schäfer M, Mousa SA. The presence of mu-, delta-, and kappa-opioid receptors in human heart tissue. Heart Vessel. 2014;29:855–63. Sobanski P, Krajnik M, Shaqura M, Bloch-Boguslawska E, Schäfer M, Mousa SA. The presence of mu-, delta-, and kappa-opioid receptors in human heart tissue. Heart Vessel. 2014;29:855–63.
9.
go back to reference Mousa SA, Shaqura M, Schäper J, Treskatsch S, Habazettl H, Schäfer M, et al. Developmental expression of δ-opioid receptors during maturation of the parasympathetic, sympathetic, and sensory innervations of the neonatal heart: early targets for opioid regulation of autonomic control. J Comp Neurol. 2011;519:957–71.PubMed Mousa SA, Shaqura M, Schäper J, Treskatsch S, Habazettl H, Schäfer M, et al. Developmental expression of δ-opioid receptors during maturation of the parasympathetic, sympathetic, and sensory innervations of the neonatal heart: early targets for opioid regulation of autonomic control. J Comp Neurol. 2011;519:957–71.PubMed
10.
go back to reference Mousa SA, Shaqura M, Schäper J, Huang W, Treskatsch S, Habazettl H, et al. Identification of mu- and kappa-opioid receptors as potential targets to regulate parasympathetic, sympathetic, and sensory neurons within rat intracardiac ganglia. J Comp Neurol. 2010;518:3836–47.PubMed Mousa SA, Shaqura M, Schäper J, Huang W, Treskatsch S, Habazettl H, et al. Identification of mu- and kappa-opioid receptors as potential targets to regulate parasympathetic, sympathetic, and sensory neurons within rat intracardiac ganglia. J Comp Neurol. 2010;518:3836–47.PubMed
11.
go back to reference Bolte C, Newman G, Schultz JEJ. Kappa and delta opioid receptor signaling is augmented in the failing heart. J Mol Cell Cardiol. 2009;47:493–503.PubMedPubMedCentral Bolte C, Newman G, Schultz JEJ. Kappa and delta opioid receptor signaling is augmented in the failing heart. J Mol Cell Cardiol. 2009;47:493–503.PubMedPubMedCentral
12.
go back to reference Imai N, Kashiki M, Woolf PD, Liang CS. Comparison of cardiovascular effects of mu- and delta-opioid receptor antagonists in dogs with congestive heart failure. Am J Phys. 1994;267:H912–7. Imai N, Kashiki M, Woolf PD, Liang CS. Comparison of cardiovascular effects of mu- and delta-opioid receptor antagonists in dogs with congestive heart failure. Am J Phys. 1994;267:H912–7.
13.
go back to reference Treskatsch S, Feldheiser A, Rosin AT, Sifringer M, Habazettl H, Mousa SA, et al. A modified approach to induce predictable congestive heart failure by volume overload in rats. PLoS One. 2014;9:e87531–7.PubMedPubMedCentral Treskatsch S, Feldheiser A, Rosin AT, Sifringer M, Habazettl H, Mousa SA, et al. A modified approach to induce predictable congestive heart failure by volume overload in rats. PLoS One. 2014;9:e87531–7.PubMedPubMedCentral
14.
go back to reference Dent MR, Tappia PS, Dhalla NS. Gender differences in apoptotic signaling in heart failure due to volume overload. Apoptosis. 2010;15:499–510.PubMed Dent MR, Tappia PS, Dhalla NS. Gender differences in apoptotic signaling in heart failure due to volume overload. Apoptosis. 2010;15:499–510.PubMed
15.
go back to reference Hill KG, Sable HJK, Ferraro FM, Kiefer SW. Chronic naltrexone treatment and ethanol responsivity in outbred rats. Alcohol Clin Exp Res. 2010;34:272–9.PubMed Hill KG, Sable HJK, Ferraro FM, Kiefer SW. Chronic naltrexone treatment and ethanol responsivity in outbred rats. Alcohol Clin Exp Res. 2010;34:272–9.PubMed
16.
go back to reference Rzasa Lynn R, Galinkin JL. Naloxone dosage for opioid reversal: current evidence and clinical implications. Ther Adv Drug Saf. 2018;9:63–88.PubMed Rzasa Lynn R, Galinkin JL. Naloxone dosage for opioid reversal: current evidence and clinical implications. Ther Adv Drug Saf. 2018;9:63–88.PubMed
17.
go back to reference Crabtree BL. Review of naltrexone, a long-acting opiate antagonist. Clin Pharm. 1984;3:273–80.PubMed Crabtree BL. Review of naltrexone, a long-acting opiate antagonist. Clin Pharm. 1984;3:273–80.PubMed
18.
go back to reference Saha DC, Saha AC, Malik G, Astiz ME, Rackow EC. Comparison of cardiovascular effects of tiletamine-zolazepam, pentobarbital, and ketamine-xylazine in male rats. J Am Assoc Lab Anim Sci. 2007;46:74–80.PubMed Saha DC, Saha AC, Malik G, Astiz ME, Rackow EC. Comparison of cardiovascular effects of tiletamine-zolazepam, pentobarbital, and ketamine-xylazine in male rats. J Am Assoc Lab Anim Sci. 2007;46:74–80.PubMed
19.
go back to reference Page IH. The mosaic theory of arterial hypertension - its interpretation. Perspect Biol Med. 1967;10:325–33.PubMed Page IH. The mosaic theory of arterial hypertension - its interpretation. Perspect Biol Med. 1967;10:325–33.PubMed
20.
go back to reference Heidrich FM, Zhang K, Estrada M, Huang Y, Giordano FJ, Ehrlich BE. Chromogranin B regulates calcium signaling, nuclear factor kappaB activity, and brain natriuretic peptide production in cardiomyocytes. Circ Res. 2008;102:1230–8.PubMedPubMedCentral Heidrich FM, Zhang K, Estrada M, Huang Y, Giordano FJ, Ehrlich BE. Chromogranin B regulates calcium signaling, nuclear factor kappaB activity, and brain natriuretic peptide production in cardiomyocytes. Circ Res. 2008;102:1230–8.PubMedPubMedCentral
21.
go back to reference Mansour A, Burke S, Pavlic RJ, Akil H, Watson SJ. Immunohistochemical localization of the cloned kappa 1 receptor in the rat CNS and pituitary. Neuroscience. 1996;71:671–90.PubMed Mansour A, Burke S, Pavlic RJ, Akil H, Watson SJ. Immunohistochemical localization of the cloned kappa 1 receptor in the rat CNS and pituitary. Neuroscience. 1996;71:671–90.PubMed
22.
go back to reference Morton ME, Froehner SC. The alpha 1 and alpha 2 polypeptides of the dihydropyridine-sensitive calcium channel differ in developmental expression and tissue distribution. Neuron. 1989;2:1499–506.PubMed Morton ME, Froehner SC. The alpha 1 and alpha 2 polypeptides of the dihydropyridine-sensitive calcium channel differ in developmental expression and tissue distribution. Neuron. 1989;2:1499–506.PubMed
23.
go back to reference Balijepalli RC, Foell JD, Hall DD, Hell JW, Kamp TJ. Localization of cardiac L-type Ca (2+) channels to a caveolar macromolecular signaling complex is required for beta (2)-adrenergic regulation. Proc Natl Acad Sci. 2006;103:7500–5.PubMedPubMedCentral Balijepalli RC, Foell JD, Hall DD, Hell JW, Kamp TJ. Localization of cardiac L-type Ca (2+) channels to a caveolar macromolecular signaling complex is required for beta (2)-adrenergic regulation. Proc Natl Acad Sci. 2006;103:7500–5.PubMedPubMedCentral
24.
go back to reference Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3:1101–8.PubMed Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3:1101–8.PubMed
25.
go back to reference Villemagne PSR, Dannals RF, Ravert HT, Frost JJ. PET imaging of human cardiac opioid receptors. Eur J Nucl Med Mol Imaging. 2002;29:1385–8.PubMed Villemagne PSR, Dannals RF, Ravert HT, Frost JJ. PET imaging of human cardiac opioid receptors. Eur J Nucl Med Mol Imaging. 2002;29:1385–8.PubMed
26.
go back to reference Treskatsch S, Shakibaei M, Feldheiser A, Shaqura M, Dehe L, Roepke TK, et al. Ultrastructural changes associated with myocardial apoptosis, in failing rat hearts induced by volume overload. Int J Cardiol. 2015;197:327–32.PubMed Treskatsch S, Shakibaei M, Feldheiser A, Shaqura M, Dehe L, Roepke TK, et al. Ultrastructural changes associated with myocardial apoptosis, in failing rat hearts induced by volume overload. Int J Cardiol. 2015;197:327–32.PubMed
27.
go back to reference Rosén A, Lundeberg T, Bytner B, Nylander I. Central changes in nociceptin dynorphin B and Met-enkephalin-Arg-Phe in different models of nociception. Brain Res. 2000;857:212–8.PubMed Rosén A, Lundeberg T, Bytner B, Nylander I. Central changes in nociceptin dynorphin B and Met-enkephalin-Arg-Phe in different models of nociception. Brain Res. 2000;857:212–8.PubMed
28.
go back to reference Feldman PD, Parveen N, Sezen S. Cardiovascular effects of Leu-enkephalin in the nucleus tractus solitarius of the rat. Brain Res. 1996;709:331–6.PubMed Feldman PD, Parveen N, Sezen S. Cardiovascular effects of Leu-enkephalin in the nucleus tractus solitarius of the rat. Brain Res. 1996;709:331–6.PubMed
29.
go back to reference Bovill JG, Sebel PS, Stanley TH. Opioid analgesics in anesthesia: with special reference to their use in cardiovascular anesthesia. Anesthesiology. 1984;61:731–55.PubMed Bovill JG, Sebel PS, Stanley TH. Opioid analgesics in anesthesia: with special reference to their use in cardiovascular anesthesia. Anesthesiology. 1984;61:731–55.PubMed
30.
go back to reference See Hoe L, Patel HH, Peart JN. Delta opioid receptors and cardioprotection. Handb Exp Pharmacol. 2018;247:301–34.PubMed See Hoe L, Patel HH, Peart JN. Delta opioid receptors and cardioprotection. Handb Exp Pharmacol. 2018;247:301–34.PubMed
31.
go back to reference Meine TJ, Roe MT, Chen AY, Patel MR, Washam JB, Ohman EM, et al. Association of intravenous morphine use and outcomes in acute coronary syndromes: results from the CRUSADE quality improvement initiative. Am Heart J. 2005;149:1043–9.PubMed Meine TJ, Roe MT, Chen AY, Patel MR, Washam JB, Ohman EM, et al. Association of intravenous morphine use and outcomes in acute coronary syndromes: results from the CRUSADE quality improvement initiative. Am Heart J. 2005;149:1043–9.PubMed
32.
go back to reference Cozzolino D, Sasso FC, Salvatore T, Torella M, Cittadini A, Gentile S, et al. Acute effects of β-endorphin on cardiovascular function in patients with mild to moderate chronic heart failure. Am Heart J. 2004;148:530–7. Cozzolino D, Sasso FC, Salvatore T, Torella M, Cittadini A, Gentile S, et al. Acute effects of β-endorphin on cardiovascular function in patients with mild to moderate chronic heart failure. Am Heart J. 2004;148:530–7.
33.
go back to reference Vargish T, Beamer KC, Daly T, Riggs TR. Morphine sulfate depression of cardiac function is attenuated by opiate receptor antagonism with naloxone. Circ Shock. 1987;23:189–95.PubMed Vargish T, Beamer KC, Daly T, Riggs TR. Morphine sulfate depression of cardiac function is attenuated by opiate receptor antagonism with naloxone. Circ Shock. 1987;23:189–95.PubMed
34.
go back to reference Yatani A, Imai N, Himura Y, Suematsu M, Liang CS. Chronic opiate-receptor inhibition in experimental congestive heart failure in dogs. Am J Phys. 1997;272:H478–84. Yatani A, Imai N, Himura Y, Suematsu M, Liang CS. Chronic opiate-receptor inhibition in experimental congestive heart failure in dogs. Am J Phys. 1997;272:H478–84.
35.
go back to reference Pepe S. Cross-talk of opioid peptide receptor and ß-adrenergic receptor signalling in the heart. Cardiovasc Res. 2004;63:414–22.PubMed Pepe S. Cross-talk of opioid peptide receptor and ß-adrenergic receptor signalling in the heart. Cardiovasc Res. 2004;63:414–22.PubMed
36.
go back to reference Yeganeh-Hajahmadi M, Najafipour H, Farzaneh F, Esmaeili-Mahani S, Joukar S. Effect of apelin on cardiac contractility in acute reno-vascular hypertension: the role of apelin receptor and kappa opioid receptor heterodimerization. Iran J Basic Med Sci. 2018;21:1305–15.PubMedPubMedCentral Yeganeh-Hajahmadi M, Najafipour H, Farzaneh F, Esmaeili-Mahani S, Joukar S. Effect of apelin on cardiac contractility in acute reno-vascular hypertension: the role of apelin receptor and kappa opioid receptor heterodimerization. Iran J Basic Med Sci. 2018;21:1305–15.PubMedPubMedCentral
37.
go back to reference Rostamzadeh F, Najafipour H, Yeganeh-Hajahmadi M, Esmaeili-mahani S, Joukar S, Iranpour M. Heterodimerization of apelin and opioid receptors and cardiac inotropic and lusitropic effects of apelin in 2K1C hypertension: role of pERK1/2 and PKC. Life Sci. 2017;191:24–33.PubMed Rostamzadeh F, Najafipour H, Yeganeh-Hajahmadi M, Esmaeili-mahani S, Joukar S, Iranpour M. Heterodimerization of apelin and opioid receptors and cardiac inotropic and lusitropic effects of apelin in 2K1C hypertension: role of pERK1/2 and PKC. Life Sci. 2017;191:24–33.PubMed
38.
go back to reference Rostamzadeh F, Najafipour H, Yeganeh-Hajahmadi M, Joukar S. Opioid receptors mediate inotropic and depressor effects of apelin in rats with 2K1C-induced chronic renovascular hypertension. Clin Exp Pharmacol Physiol. 2018;45:187–97.PubMed Rostamzadeh F, Najafipour H, Yeganeh-Hajahmadi M, Joukar S. Opioid receptors mediate inotropic and depressor effects of apelin in rats with 2K1C-induced chronic renovascular hypertension. Clin Exp Pharmacol Physiol. 2018;45:187–97.PubMed
39.
go back to reference Liang CS, Imai N, Stone CK, Woolf PD, Kawashima S, Tuttle RR. The role of endogenous opioids in congestive heart failure: effects of nalmefene on systemic and regional hemodynamics in dogs. Circulation. 1987;75:443–51.PubMed Liang CS, Imai N, Stone CK, Woolf PD, Kawashima S, Tuttle RR. The role of endogenous opioids in congestive heart failure: effects of nalmefene on systemic and regional hemodynamics in dogs. Circulation. 1987;75:443–51.PubMed
40.
go back to reference Wong TM, Shan J. Modulation of sympathetic actions on the heart by opioid receptor stimulation. J Biomed Sci. 2001;8:299–306.PubMed Wong TM, Shan J. Modulation of sympathetic actions on the heart by opioid receptor stimulation. J Biomed Sci. 2001;8:299–306.PubMed
41.
go back to reference Weir MR, Dzau VJ. The renin-angiotensin-aldosterone system: a specific target for hypertension management. Am J Hypertens. 1999;12:205S–13S.PubMed Weir MR, Dzau VJ. The renin-angiotensin-aldosterone system: a specific target for hypertension management. Am J Hypertens. 1999;12:205S–13S.PubMed
42.
go back to reference Schmittinger CA, Torgersen C, Luckner G, Schröder DCH, Lorenz I, Dünser MW. Adverse cardiac events during catecholamine vasopressor therapy: a prospective observational study. Intensive Care Med. 2012;38:950–8.PubMed Schmittinger CA, Torgersen C, Luckner G, Schröder DCH, Lorenz I, Dünser MW. Adverse cardiac events during catecholamine vasopressor therapy: a prospective observational study. Intensive Care Med. 2012;38:950–8.PubMed
43.
go back to reference Shahani R, Klein LV, Marshall JG, Nicholson S, Rubin BB, Walker PM, et al. Hemorrhage-induced alpha-adrenergic signaling results in myocardial TNF-alpha expression and contractile dysfunction. Am J Physiol-Heart C. 2001;281:H84–92. Shahani R, Klein LV, Marshall JG, Nicholson S, Rubin BB, Walker PM, et al. Hemorrhage-induced alpha-adrenergic signaling results in myocardial TNF-alpha expression and contractile dysfunction. Am J Physiol-Heart C. 2001;281:H84–92.
44.
go back to reference Shimizu M, Tanaka R, Uchida M, et al. Effect of angiotensin II type 1 receptor blocker on cardiac angiotensin-converting enzyme and chymase-like activities, and cardiac fibrosis in cardiomyopathic hamsters. J Vet Med Sci. 2006;68:227–33.PubMed Shimizu M, Tanaka R, Uchida M, et al. Effect of angiotensin II type 1 receptor blocker on cardiac angiotensin-converting enzyme and chymase-like activities, and cardiac fibrosis in cardiomyopathic hamsters. J Vet Med Sci. 2006;68:227–33.PubMed
45.
go back to reference Shaqura M, Mohamed DM, Aboryag NB, Bedewi L, Dehe L, Treskatsch S, et al. Pathological alterations in liver injury following congestive heart failure induced by volume overload in rats. PLoS One. 2017;12:e0184161.PubMedPubMedCentral Shaqura M, Mohamed DM, Aboryag NB, Bedewi L, Dehe L, Treskatsch S, et al. Pathological alterations in liver injury following congestive heart failure induced by volume overload in rats. PLoS One. 2017;12:e0184161.PubMedPubMedCentral
46.
go back to reference Aboryag NB, Mohamed DM, Dehe L, et al. Histopathological changes in the kidney following congestive heart failure by volume overload in rats. Oxidative Med Cell Longev. 2017;2017:6894040–10. Aboryag NB, Mohamed DM, Dehe L, et al. Histopathological changes in the kidney following congestive heart failure by volume overload in rats. Oxidative Med Cell Longev. 2017;2017:6894040–10.
47.
go back to reference Raynor K, Kong H, Mestek A, et al. Characterization of the cloned human mu opioid receptor. J Pharmacol Exp Ther. 1995;272:423–8.PubMed Raynor K, Kong H, Mestek A, et al. Characterization of the cloned human mu opioid receptor. J Pharmacol Exp Ther. 1995;272:423–8.PubMed
48.
go back to reference Niciu MJ, Arias AJ. Targeted opioid receptor antagonists in the treatment of alcohol use disorders. CNS Drugs. 2013;27:777–87.PubMedPubMedCentral Niciu MJ, Arias AJ. Targeted opioid receptor antagonists in the treatment of alcohol use disorders. CNS Drugs. 2013;27:777–87.PubMedPubMedCentral
49.
go back to reference Walker EA, Sterious SN. Opioid antagonists differ according to negative intrinsic efficacy in a mouse model of acute dependence. Br J Pharmacol J. 2005;145:975–83. Walker EA, Sterious SN. Opioid antagonists differ according to negative intrinsic efficacy in a mouse model of acute dependence. Br J Pharmacol J. 2005;145:975–83.
50.
go back to reference Emmerson PJ, Clark MJ, Mansour A, Akil H, Woods JH, Medzihradsky F. Characterization of opioid agonist efficacy in a C6 glioma cell line expressing the mu opioid receptor. J Pharmacol Exp Ther. 1996;278:1121–7.PubMed Emmerson PJ, Clark MJ, Mansour A, Akil H, Woods JH, Medzihradsky F. Characterization of opioid agonist efficacy in a C6 glioma cell line expressing the mu opioid receptor. J Pharmacol Exp Ther. 1996;278:1121–7.PubMed
Metadata
Title
Chronic Naltrexone Therapy Is Associated with Improved Cardiac Function in Volume Overloaded Rats
Authors
Lukas Dehe
Mohammed Shaqura
Michael Nordine
Helmut Habazettl
Petra von Kwiatkowski
Helena Schluchter
Mehdi Shakibaei
Shaaban A. Mousa
Michael Schäfer
Sascha Treskatsch
Publication date
01-08-2021
Publisher
Springer US
Published in
Cardiovascular Drugs and Therapy / Issue 4/2021
Print ISSN: 0920-3206
Electronic ISSN: 1573-7241
DOI
https://doi.org/10.1007/s10557-020-07132-4

Other articles of this Issue 4/2021

Cardiovascular Drugs and Therapy 4/2021 Go to the issue