Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters










Publication year range
1.
Prog Neuropsychopharmacol Biol Psychiatry ; 84(Pt A): 102-114, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29407532

ABSTRACT

Drug withdrawal-associated aversive memories trigger relapse to drug-seeking behavior. Corticotrophin-releasing factor (CRF) is an important mediator of the reinforcing properties of drugs of abuse. However, the involvement of CRF1 receptor (CRF1R) in aversive memory induced by opiate withdrawal has yet to be elucidated. We used the conditioned-place aversion (CPA) paradigm to evaluate the role of CRF1R on opiate withdrawal memory acquisition, along with plasticity-related processes that occur after CPA within the basolateral amygdala (BLA) and dentate gyrus (DG). Male mice were rendered dependent on morphine and injected acutely with naloxone before paired to confinement in a naloxone-associated compartment. The CPA scores as well as the number of TH-positive neurons (in the NTS-A2 noradrenergic cell group), and the expression of the transcription factors Arc and pCREB (in the BLA and DG) were measured with and without CRF1R blockade. Mice subjected to conditioned naloxone-induced morphine withdrawal robustly expressed CPA. Pre-treatment with the selective CRF1R antagonist CP-154,526 before naloxone conditioning session impaired morphine withdrawal-induced aversive memory acquisition. CP-154,526 also antagonized the enhanced number of TH-positive neurons in the NTS-A2 that was seen after CPA. Increased Arc expression and Arc-pCREB co-localization were seen in the BLA after CPA, which was not modified by CP-154,526. In the DG, CPA was accompanied by a decrease of Arc expression and no changes in Arc-pCREB co-localization, whereas pre-treatment with CP-154,526 induced an increase in both parameters. These results indicate that CRF-CRF1R pathway could be a critical factor governing opiate withdrawal memory storage and retrieval and might suggest a role for TH-NA pathway in the effects of withdrawal on memory. Our results might indicate that the blockade of CRF1R could represent a promising pharmacological treatment strategy approach for the attenuation of the relapse to drug-seeking/taking behavior triggered by opiate withdrawal-associated aversive memories.


Subject(s)
Avoidance Learning/drug effects , Basolateral Nuclear Complex/drug effects , Conditioning, Psychological/drug effects , Dentate Gyrus/metabolism , Morphine Dependence/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Avoidance Learning/physiology , Basolateral Nuclear Complex/metabolism , Conditioning, Psychological/physiology , Corticotropin-Releasing Hormone/metabolism , Male , Memory/drug effects , Memory/physiology , Mice , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Random Allocation , Spatial Behavior/drug effects , Spatial Behavior/physiology
2.
Toxicol Appl Pharmacol ; 275(1): 28-35, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24398105

ABSTRACT

Opioid addiction is associated with cardiovascular disease. However, mechanisms linking opioid addiction and cardiovascular disease remain unclear. This study investigated the role of corticotropin-releasing factor (CRF) 1 receptor in mediating somatic signs and the behavioural states produced during withdrawal from morphine dependence. Furthermore, it studied the efficacy of CRF1 receptor antagonist, CP-154,526 to prevent the cardiac sympathetic activity induced by morphine withdrawal. In addition, tyrosine hydroxylase (TH) phosphorylation pathways were evaluated. Like stress, morphine withdrawal induced an increase in the hypothalamic-pituitary-adrenal (HPA) axis activity and an enhancement of noradrenaline (NA) turnover. Pre-treatment with CRF1 receptor antagonist significantly reduced morphine withdrawal-induced increases in plasma adrenocorticotropic hormone (ACTH) levels, NA turnover and TH phosphorylation at Ser31 in the right ventricle. In addition, CP-154,526 reduced the phosphorylation of extracellular signal-regulated kinase (ERK) after naloxone-precipitated morphine withdrawal. In addition, CP-154,526 attenuated the increases in body weight loss during morphine treatment and suppressed some of morphine withdrawal signs. Altogether, these results support the idea that cardiac sympathetic pathways are activated in response to naloxone-precipitated morphine withdrawal suggesting that treatment with a CRF1 receptor antagonist before morphine withdrawal would prevent the development of stress-induced behavioural and autonomic dysfunction in opioid addicts.


Subject(s)
Cardiotonic Agents/therapeutic use , Disease Models, Animal , Heart Ventricles/drug effects , Morphine Dependence/drug therapy , Narcotic Antagonists/adverse effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Substance Withdrawal Syndrome/prevention & control , Animals , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiopathology , MAP Kinase Signaling System/drug effects , Male , Morphine Dependence/physiopathology , Naloxone/adverse effects , Naloxone/therapeutic use , Narcotic Antagonists/therapeutic use , Phosphorylation/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiopathology , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Substance Withdrawal Syndrome/etiology , Tyrosine 3-Monooxygenase/metabolism , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/prevention & control , Weight Gain/drug effects
3.
Br J Pharmacol ; 168(8): 1889-901, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23215787

ABSTRACT

BACKGROUND AND PURPOSE: Alterations in transcription factors that regulate the development and maintenance of dopamine (DA) neurons (such as Nurr1 and Pitx3) play an important role in the pathogenesis of addiction diseases. We have examined the effects of acute and chronic morphine and morphine withdrawal on TH expression and activity as well as expression of Nurr1, Pitx3 and Ago2 in the ventral tegmental area (VTA) and nucleus accumbens (NAc) of the rat. EXPERIMENTAL APPROACH: Rats were injected acutely with morphine and decapitated 1 or 2 h later. Another set of rats were made dependent on morphine by implantation of two morphine pellets. Precipitated withdrawal was induced by injection of naloxone. Ago2, Pitx3, Nurr1, total TH (tTH), TH phosphorylated at Ser31 and at Ser40, and 3,4-Dihydroxyphenylacetic acid, and DA determination in the VTA and/or NAc were measured using immunoblotting, HPLC and immunofluorescence. KEY RESULTS: Acute morphine produced a marked increase in TH activity and DA turnover in the NAc, concomitantly with increased Nurr1 and Pitx3 expression in the VTA. In contrast, precipitated morphine withdrawal decreased TH activation, TH expression and did not increase DA turnover in the NAc. These effects paralleled decreases in Ago2 expression, which was accompanied by increased Nurr1 and Pitx3, TH activity and normalized TH protein levels in the VTA. CONCLUSIONS AND IMPLICATIONS: The combined decrease in Ago2 and increases in Nurr1 and Pitx3 might represent some of the mechanisms that served to protect against accumbal TH regulation observed in morphine withdrawn rats, which may be critical for DA bioavailability to influence behaviour.


Subject(s)
Argonaute Proteins/metabolism , Dopamine/metabolism , Mesencephalon/drug effects , Morphine/administration & dosage , Substance-Related Disorders/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Argonaute Proteins/genetics , Body Weight/drug effects , Drug Implants , Gene Expression Regulation/drug effects , Homeodomain Proteins/metabolism , Injections, Intraperitoneal , Male , Naloxone/administration & dosage , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Nucleus Accumbens/metabolism , Phosphorylation , Rats , Rats, Wistar , Substance-Related Disorders/pathology , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/metabolism
4.
J Pharmacol Exp Ther ; 342(3): 665-75, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22647273

ABSTRACT

The negative affective states of withdrawal involve the recruitment of brain and peripheral stress circuitry [e.g., noradrenergic activity, induction of the hypothalamo-pituitary-adrenocortical (HPA) axis, and the expression and activation of heat shock proteins (Hsps)]. The present study investigated the role of extracellular signal-regulated protein kinase (ERK) and ß-adrenoceptor on the response of stress systems to morphine withdrawal by the administration of [amino[(4-aminophenyl)thio]methylene]-2-(trifluoromethyl)benzeneacetonitrile (SL327), a selective inhibitor of ERK activation, or propranolol (a ß-adrenoceptor antagonist). Dependence on morphine was induced by a 7-day subcutaneous implantation of morphine pellets. Morphine withdrawal was precipitated on day 8 by the injection of naloxone (2 mg/kg s.c.). Plasma concentrations of adrenocorticotropin and corticosterone were determined by radioimmunoassay; noradrenaline (NA) turnover in left ventricle was determined by high-performance liquid chromatography; and catechol-O-methyl transferase (COMT) and Hsp27 expression and phosphorylation at Ser82 were determined by quantitative blot immunolabeling. Morphine-withdrawn rats showed an increase of NA turnover and COMT expression in parallel with an enhancement of adrenocorticotropin and plasma corticosterone concentrations. In addition, we observed an enhancement of Hsp27 expression and phosphorylation. Pretreatment with SL327 or propranolol significantly reduced morphine withdrawal-induced increases of plasma adrenocorticotropin and Hsp27 phosphorylation at Ser82 without any changes in plasma corticosterone levels. The present findings demonstrate that morphine withdrawal is capable of inducing the activation of HPA axis in parallel with an enhancement of Hsp27 expression and Hsp27 phosphorylation at Ser82 and suggest a role for ß-adrenoceptors and ERK pathways in mediating morphine-withdrawal activation of the HPA axis and cellular stress response.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , HSP27 Heat-Shock Proteins/metabolism , Heart/drug effects , Hypothalamo-Hypophyseal System/physiopathology , Morphine/adverse effects , Pituitary-Adrenal System/physiopathology , Substance Withdrawal Syndrome/physiopathology , Adrenergic beta-Antagonists/pharmacology , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/metabolism , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Catechol O-Methyltransferase/metabolism , Corticosterone/blood , Corticosterone/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Heart/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Male , Morphine Dependence/metabolism , Morphine Dependence/physiopathology , Naloxone/pharmacology , Norepinephrine/metabolism , Phosphorylation/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/metabolism , Substance Withdrawal Syndrome/metabolism
5.
Naunyn Schmiedebergs Arch Pharmacol ; 385(4): 397-409, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22207192

ABSTRACT

Tissue injury and/or opioids induce plastic changes in the spinal cord resulting in pain hypersensitivity; the contribution of the dorsal root ganglia (DRG) is poorly understood. We evaluated DRG phenotypic changes induced by surgery and/or remifentanil in a mice model of postoperative pain using as neuronal markers ERK1/2 and c-Fos; prodynorphin mRNA and dynorphin levels were also determined. We hypothesized that a correlation between nociception and DRG reactivity would occur. Surgery and/or remifentanil induced mechanical hypersensitivity, correlated with ERK1/2 phosphorylation and c-Fos expression in the DRG; changes were greater in the remifentanil + incision group and still present on day 14 (p < 0.01 vs. control). Intrathecal PD98059 (ERK1/2 inhibitor) partially reversed the mechanical hypersensitivity (44%, p < 0.05) observed in the remifentanil + incision group. In this group, significant increases in prodynorphin mRNA (at 2, 7, and 14 days, p < 0.01) roughly coincided with increases in dynorphin (days 2 and 14, p < 0.001) in the DRG. Remifentanil or incision (alone) also induced an up-regulation in prodynorphin mRNA expression on days 7 and 14 (p < 0.01, p < 0.05, respectively), partially correlating with dynorphin levels. On day 21, all molecular changes returned to control levels in all experimental conditions, concurring with the complete recovery of nociceptive thresholds. Surgery and/or remifentanil induce up-regulation of c-Fos and pERK in the DRG, approximately correlating with nociceptive behavior, also associated with an increased expression of prodynorphin/dynorphin. These changes support the role of the DRG in the development and maintenance of pain hypersensitivity after surgery. The findings could contribute to the development of new therapeutic agents focused on peripheral targets.


Subject(s)
Analgesics, Opioid/pharmacology , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Pain, Postoperative/metabolism , Piperidines/pharmacology , Animals , Behavior, Animal/drug effects , Dynorphins/metabolism , Enkephalins/genetics , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiopathology , Hyperalgesia/physiopathology , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pain, Postoperative/physiopathology , Protein Precursors/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Remifentanil
6.
J Mol Cell Cardiol ; 51(1): 129-39, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21530534

ABSTRACT

Heat shock protein 27 (Hsp27) is a well-known stress response protein that becomes phosphorylated through extracellular signal-regulated kinase (ERK). Different drugs of abuse, such as morphine and/or its withdrawal, induce severe stress situations. In this study, we investigated Hsp27 and phospho-Hsp27 expression during morphine dependence and withdrawal and evaluated the involvement of ERK in the phosphorylation of Hsp27 in the rat right ventricle. Dependence on morphine was induced by a 7-day s.c. implantation of morphine pellets. Morphine withdrawal was precipitated on day 8 by injection of naloxone (2 mg/kg, s.c.). ERK1/2, Hsp27 and phospho-Hsp27 at Ser15 were determined by quantitative blot immunolabeling using specific antibodies. Hsp27 expression was increased 30, 60, 90 and 120 min (144.5±14.2%, P<0.0001; 128.9±4.6%, P=0.04; 177.4±12.7, P<0.0001; and 136.2±11.0%, P=0.042, respectively) after saline injection to rats dependent on morphine. Naloxone-precipitated morphine withdrawal also increased the phosphorylation of Hsp27 at Ser15 at those time points (146.8±19.8%, P=0.034; 143.9±17.9%, P=0.032; 161.2±33.3%, P=0.029; and 152.2±25.5%, P=0.008, respectively). However, there were no changes in Hsp27 phosphorylation in the morphine dependent group injected with saline. In addition, there was an increase in the phosphorylation of ERK 60 min after naloxone injection in morphine dependent rats (pERK1: 116.3±4.2%, P=0.015 and pERK2: 117.2±1.5%, P=0.05). Pretreatment with SL327, an inhibitor of ERK phosphorylation, decreased activation (phosphorylation) of both ERK and Hsp27 (pERK1: 4.5±3.6%, P<0.0001; pERK2: 42.3±3.3%, P<0.0001; and pHsp27: 97.6±1.5%, P=0.008), suggesting that ERK activation triggers Hsp27 phosphorylation. The present findings demonstrate that morphine withdrawal is capable of inducing the activation of Hsp27 in the heart and suggest that phosphorylation of Hsp27 is closely linked to and also dependent on the ERK pathway.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , HSP27 Heat-Shock Proteins/metabolism , Morphine Dependence/metabolism , Morphine/adverse effects , Myocardium/metabolism , Naloxone/pharmacology , Substance Withdrawal Syndrome/metabolism , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , HSP27 Heat-Shock Proteins/genetics , Heart , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Immunoblotting , Myocardium/enzymology , Phosphorylation , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
7.
Naunyn Schmiedebergs Arch Pharmacol ; 382(5-6): 525-34, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20924561

ABSTRACT

Although dopamine (DA) has been extensively implicated in the morphine-induced conditioned place preference (CPP; a measure of reward), noradrenaline (NA) and other systems may play a larger role than previously suspected. The mesolimbic DA system, comprised of projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAc), receives noradrenergic innervations from the nucleus tractus solitaries (NTS)-A2 cell group and is modulated by NA. The purpose of the present study was to evaluate the turnover of DA and NA in the NAc and the site-specific phosphorylation of TH in the NAc, VTA, and NTS on the CPP mice conditioned by morphine. A dose-effect curve for morphine-induced CPP (0.5-8 mg/kg, s.c.) was obtained using 6-day conditioning sessions followed by a CPP test. TH phosphorylation was determined by quantitative blot immunolabeling and immunohistochemistry using phosphorylation state-specific antibodies; NA and DA turnover was evaluated by high-performance liquid chromatography. Morphine-induced CPP phosphorylates TH at serine (Ser)40 but not Ser31 in NAc, which is associated with an enhanced of DA and NA turnover. We also found that morphine-induced CPP increased levels of TH phosphorylated at Ser31 and Ser40 in the NTS. The present study demonstrates that morphine-induced CPP might stimulate TH activity and accelerate DA and NA turnover in the NAc via a mechanism involving phosphorylation of TH.


Subject(s)
Conditioning, Operant/drug effects , Morphine/pharmacology , Narcotics/pharmacology , Nucleus Accumbens/drug effects , Solitary Nucleus/drug effects , Tyrosine 3-Monooxygenase/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Dopamine/metabolism , Male , Methoxyhydroxyphenylglycol/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Norepinephrine/metabolism , Nucleus Accumbens/metabolism , Phosphorylation , Solitary Nucleus/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
8.
J Pharmacol Exp Ther ; 330(3): 771-82, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19567779

ABSTRACT

Our previous studies have shown that morphine withdrawal induced an increase in the expression of protein kinase (PK) A and mitogen-activated extracellular kinase (MAPK) pathways in the heart during morphine withdrawal. The purpose of the present study was to evaluate the interaction between PKA and extracellular signal-regulated kinase (ERK) signaling pathways mediating the cardiac adaptive changes observed after naloxone administration to morphine-dependent rats. Dependence on morphine was induced by a 7-day subcutaneous implantation of morphine pellets. Morphine withdrawal was precipitated on day 8 by an injection of naloxone (2 mg/kg). ERK1/2 and tyrosine hydroxylase (TH) phosphorylation was determined by quantitative blot immunolabeling using phosphorylation state-specific antibodies. Naloxone-induced morphine withdrawal activates ERK1/2 and phosphorylates TH at Ser31 in the right and left ventricle, with an increase in the mean arterial blood pressure and heart rate. When N-(2-guanidinoethyl)-5-isoquinolinesulfonamide (HA-1004), a PKA inhibitor, was infused, concomitantly with morphine, it diminished the expression of ERK1/2. In contrast, the infusion of calphostin C (a PKC inhibitor) did not modify the morphine withdrawal-induced activation of ERK1/2. The ability of morphine withdrawal to activate ERK that phosphorylates TH at Ser31 was reduced by HA-1004. The present findings demonstrate that the enhancement of ERK1/2 expression and the phosphorylation state of TH at Ser31 during morphine withdrawal are dependent on PKA and suggest cross-talk between PKA and ERK1/2 transduction pathway mediating morphine withdrawal-induced activation (phosphorylation) of TH.


Subject(s)
Analgesics, Opioid/adverse effects , Cyclic AMP-Dependent Protein Kinases/physiology , Heart/physiopathology , Mitogen-Activated Protein Kinases/physiology , Morphine/adverse effects , Signal Transduction/physiology , Substance Withdrawal Syndrome/physiopathology , Adaptation, Physiological/physiology , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Blotting, Western , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Hemodynamics/physiology , Isoquinolines/pharmacology , Male , Naloxone/pharmacology , Naphthalenes/pharmacology , Narcotic Antagonists/pharmacology , Phosphorylation , Protein Kinase C/biosynthesis , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tyrosine 3-Monooxygenase/physiology , Weight Gain/drug effects
9.
Br J Pharmacol ; 151(6): 787-97, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17549049

ABSTRACT

BACKGROUND AND PURPOSE: We have previously demonstrated that morphine withdrawal induced hyperactivity of the heart by activation of noradrenergic pathways innervating the left and right ventricle, as evaluated by noradrenaline turnover and c-Fos expression. The extracellular signal-regulated kinase (ERK) has been implicated in drug addiction, but its role in activation of the heart during morphine dependence remains poorly understood. Here, we have looked for activation of ERK during morphine withdrawal and if this activation induced gene expression. EXPERIMENTAL APPROACH: Dependence on morphine was induced by s.c. implantation of morphine pellets for 7 days. Morphine withdrawal was precipitated on day 8 by injection of naloxone (2 mg kg(-1), s.c.). ERK1/2, their phosphorylated forms and c-Fos were measured by western blotting and immunohistochemistry of cardiac tissue. KEY RESULTS: Naloxone-induced morphine withdrawal activated ERK1/2 and increased c-Fos expression in cardiac tissues. c-Fos expression was blocked by SL327, a drug that prevents ERK activation. CONCLUSIONS AND IMPLICATIONS: These results indicate that signalling through the ERKs is necessary for morphine withdrawal-induced hyperactivity of the heart and suggest that this pathway may also be involved in activation of immediate-early genes in both cytosolic and nuclear effector mechanisms that have the potential to bring about long-term changes in the heart.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Morphine/adverse effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/adverse effects , Substance Withdrawal Syndrome/metabolism , Aminoacetonitrile/analogs & derivatives , Animals , Blotting, Western , Gene Expression/drug effects , Genes, fos/drug effects , Heart Ventricles/drug effects , Immunohistochemistry , MAP Kinase Signaling System/drug effects , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Substance Withdrawal Syndrome/physiopathology
10.
J Neuroendocrinol ; 15(6): 586-91, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12716409

ABSTRACT

The content of corticotropin-releasing factor (CRF) and arginine vasopressin (AVP) in the hypothalamic paraventricular nucleus (PVN) increases during chronic morphine treatment. Because these experiments cannot distinguish between increased synthesis or reduced release, the present study measured changes in CRF and AVP mRNAs in the PVN by in situ hybridization. Concomitantly, changes in noradrenaline turnover in the PVN and changes in plasma corticosterone release were determined. Male rats were implanted with placebo (naive) or morphine pellets for 7 days. On day 7, groups of rats received an acute injection of either saline i.p. or morphine (30 mg/kg, i.p.). Acute morphine injection did not change the total size of the labelled area for CRF mRNA in the PVN of naive or morphine-pelleted rats, indicating that the number of CRF-containing neurones was unchanged. On the other hand, in rats chronically treated with morphine, the intensity of labelling for CRF mRNA was significantly reduced, suggesting a decrease in the synthesis of CRF. In placebo rats, injection of saline or morphine did not affect the surface hybridized for AVP mRNA. By contrast, in the morphine-group injected with saline, there was a significant reduction in the number of labelled neurones, measured by the size of labelled area. Similarly, there was a decrease in intensity of AVP mRNA expression in the parvocellular and magnocellular neurones of the PVN in the morphine-group injected with saline, suggesting a decreased synthesis of AVP in these neurones. In parallel with the decrease in the expression of CRF and AVP mRNAs in the PVN, there was a pronounced decrease in noradrenaline turnover and in the release of corticosterone in the morphine-pelleted rats. In conclusion, present results show that, in addition to modifications in corticosterone secretion and in noradrenaline turnover, chronic morphine administration produces a reduction in the synthesis of CRF and AVP.


Subject(s)
Arginine Vasopressin/metabolism , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/chemistry , Morphine/administration & dosage , RNA, Messenger/drug effects , Animals , Arginine Vasopressin/analysis , Body Weight/drug effects , Corticosterone/blood , Corticotropin-Releasing Hormone/analysis , Drug Administration Schedule , Gene Expression/drug effects , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , In Situ Hybridization , Male , Methoxyhydroxyphenylglycol/analysis , Methoxyhydroxyphenylglycol/metabolism , Narcotics/pharmacology , Norepinephrine/analysis , Norepinephrine/metabolism , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
11.
Eur J Anaesthesiol ; 20(3): 205-11, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12650491

ABSTRACT

BACKGROUND AND OBJECTIVE: This study was designed to compare the haemodynamic, electrophysiological and pharmacodynamic effects of three selective inhibitors of the different isoenzyme forms of phosphodiesterase (PDE) on ischaemia-induced dysrhythmias in the anaesthetized rat. The drugs used were pimobendan, a selective PDE III inhibitor, rolipram, a selective PDE IV inhibitor, and zaprinast, a selective PDE V inhibitor. METHODS: The coronary artery was occluded 15 min after commencing drug administration, and myocardial ischaemia was maintained for 30 min during which the heart rate and mean arterial pressure were recorded. cAMP and cGMP were determined by radioimmunoassay. RESULTS: Pretreatment with rolipram decreased the duration of ventricular tachycardia without any change in the incidences of dysrhythmias or the mortality rate. This drug did not modify ventricular content of adenosine 3',5'-cyclic monophosphate (cAMP) or guanosine 3',5'-cyclic monophosphate (cGMP). Pimobendan (1 mg kg(-1) + 0.1 mg kg(-1) min) decreased the duration of ventricular tachycardia. This dose of pimobendan and zaprinast (1 mg kg(-1) + 0.1 mg kg(-1) min(-1)) increased the incidence rate of ventricular fibrillation following coronary artery ligation and the mortality rate. Moreover, both drugs increased cGMP in the ventricle. CONCLUSIONS: The results demonstrated that pimobendan and zaprinast increased the incidence of dysrhythmias and the mortality rate, which was accompanied by an increase in the ventricular content of cGMP. Rolipram decreased the duration of ventricular tachycardia without a change in the cyclic nucleotide content or in the mortality rate.


Subject(s)
Anesthesia , Arrhythmias, Cardiac/drug therapy , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Phosphodiesterase Inhibitors/pharmacology , Purinones/pharmacology , Pyridazines/pharmacology , Rolipram/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases , Animals , Arrhythmias, Cardiac/mortality , Arrhythmias, Cardiac/physiopathology , Blood Pressure/drug effects , Coronary Vessels/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cyclic Nucleotide Phosphodiesterases, Type 5 , Heart Rate/drug effects , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Heart Ventricles/metabolism , Ligation , Male , Myocardial Ischemia/mortality , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley
12.
Neurosci Lett ; 334(1): 58-62, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12431775

ABSTRACT

Morphine withdrawal is characterized by an increase in the hypothalamus-pituitary-adrenocortical (HPA) axis activity. Here, by means of in situ hybridization, the changes in CRH and vasopressin (AVP) mRNAs have been analysed within the rat hypothalamic paraventricular nucleus (PVN) during morphine dependence and after naloxone-precipitated morphine withdrawal. CRH and AVP mRNA expression were analysed 30 min following administration of saline or naloxone to control groups and to morphine dependent rats. The data for in situ hybridization analysis of PVN neurons show that there were no changes in the total size of labelled area for CRH or AVP mRNA during morphine withdrawal, indicating that dependence on morphine does not involve alterations in the number of neurons expressing CRH or AVP mRNA. However, levels of mRNA encoding for CRH were decreased in the PVN during morphine dependence and withdrawal. By contrast, injection of saline or naloxone to morphine dependent rats did not affect the intensity of AVM mRNA expression. All these findings are discussed in term of cellular events that couple morphine dependence-increased HPA axis activity with changes in gene expression in selective neurons of the PVN.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Morphine Dependence/physiopathology , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/analysis , Substance Withdrawal Syndrome/physiopathology , Vasopressins/metabolism , Adrenocorticotropic Hormone/drug effects , Adrenocorticotropic Hormone/genetics , Animals , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , In Situ Hybridization , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Rats , Rats, Sprague-Dawley , Vasopressins/drug effects , Vasopressins/genetics
13.
Can J Physiol Pharmacol ; 79(10): 885-91, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11697748

ABSTRACT

The purpose of our study was to examine the effects of D1-and D2-dopamine receptors blockade on the changes in the ventricular content of catecholamines in rats withdrawn from morphine. Rats were given morphine by subcutaneous (s.c.) implantation of morphine pellets for 5 days. On the eighth day, morphine withdrawal was induced by s.c. administration of naloxone (1 mg/kg), and rats were killed 30 min later. Pretreatment with SCH 23390 (dopamine D1, D5 receptor antagonist) 15 min prior to naloxone administration suppressed some the behavioural signs of morphine withdrawal, whereas eticlopride (dopamine D2, D3, D4 receptor antagonist) did not. In addition, biochemical analysis indicate that SCH 23390 completely abolished the withdrawal-induced increase in noradrenaline and dopamine turnover in the right ventricle. By contrast, eticlopride did not block the hyperactivity of catecholaminergic neurons in the heart during morphine withdrawal. These data suggest that the hyperactivity of catecholaminergic neurons in the heart during morphine withdrawal is dependent upon D1 dopamine receptor activation. In addition, our results exclude the involvement of D2 dopamine receptors.


Subject(s)
Analgesics, Opioid , Catecholamines/physiology , Heart/innervation , Morphine , Neurons/physiology , Receptors, Dopamine/drug effects , Substance Withdrawal Syndrome/physiopathology , Animals , Benzazepines/pharmacology , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Heart/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Salicylamides/pharmacology , Ventricular Function
14.
Eur J Pharmacol ; 430(1): 59-68, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11698063

ABSTRACT

The transcription factor, Fos, is considered as a functional marker of activated neurons. We have shown previously that acute administration of morphine induces the expression of Fos in hypothalamic nuclei associated with control of the hypothalamus-pituitary-adrenocortex axis, such as the paraventricular nucleus and the supraoptic nucleus. In the current study, we examined the role of protein kinase A, protein kinase C and Ca2+ entry through L-type Ca2+ channels in naloxone-precipitated Fos expression in the paraventricular and supraoptic nuclei. After 7 days of morphine treatment, we did not observe any modification in Fos production. However, when opioid withdrawal was precipitated with naloxone a dramatic increase in Fos immunoreactivity was observed in the parvocellular division of the paraventricular nucleus and in the supraoptic nucleus. Chronic co-administration of chelerythrine (a selective protein kinase C inhibitor acting at its catalytic domain) with morphine did not affect the increase in Fos expression observed in nuclei from morphine withdrawn rats. In addition, infusion of calphostin C (another protein kinase C inhibitor, which interacts with its regulatory domain) did not modify the morphine withdrawal-induced expression of Fos. In contrast, when the selective protein kinase A inhibitor, N-(2'guanidinoethyl)-5-isoquinolinesulfonamide (HA-1004), was infused it greatly diminished the increased Fos production observed in morphine-withdrawn rats. Furthermore, chronic infusion of the selective L-type Ca2+ channel antagonist, nimodipine, significantly inhibited the enhancement of Fos induction in the paraventricular and supraoptic nuclei from morphine-withdrawn animals. Taken together, these data might indicate that protein kinase A activity is necessary for the expression of Fos during morphine withdrawal and that an up-regulated Ca2+ system might contribute to the activation of Fos. The present findings suggest that protein kinase A and Ca2+ influx through L-type Ca2+ channels might contribute to the activation of neuroendocrine cells in the paraventricular and supraoptic nuclei.


Subject(s)
Anterior Hypothalamic Nucleus/drug effects , Morphine/adverse effects , Proto-Oncogene Proteins c-fos/biosynthesis , Signal Transduction/physiology , Animals , Anterior Hypothalamic Nucleus/metabolism , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Hypothalamo-Hypophyseal System/drug effects , Male , Naloxone/administration & dosage , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/drug effects , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/metabolism , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism
15.
Naunyn Schmiedebergs Arch Pharmacol ; 364(3): 193-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11521160

ABSTRACT

Studies involving heart catecholaminergic systems in morphine-dependent rats have not established whether the adaptive changes observed in the heart during morphine withdrawal are mediated peripherally or centrally. In this study, naloxone (Nx), naloxone methiodide (NxM) and N-methyl levallorphan (NML), quaternary derivatives of Nx and levallorphan, respectively, that do not cross the blood-brain barrier, were administered to morphine-dependent rats and catecholamines and their metabolites determined in the right ventricle. Rats were made dependent on morphine by implantation of morphine pellets for 7 days. On day 8 animals received s.c. injections of saline, Nx (1 mg/kg), NxM (5 mg/kg) or NML (5 mg/kg) and were decapitated 30 min later. Noradrenaline (NA) and its metabolites normetanephrine (NMN) and 3-methoxy-4-hydroxyphenylethyleneglycol (MHPG) and dopamine (DA) and its metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) were determined by high-performance liquid chromatography with electrochemical detection. After NxM or NML administration to morphine-dependent rats there was a pronounced increase in NMN and DOPAC levels, as well as in NA and DA turnovers (as estimated by NMN/NA and DOPAC/DA ratios, respectively) in the right ventricle. Similarly, giving Nx to morphine-dependent rats increased NMN and DOPAC levels and NA and DA turnovers. In addition, in the paraventricular nucleus of the hypothalamus (PVN) NA and DA turnover, measured as the MHPG/NA or DOPAC/DA ratios, increased after Nx administration but not after NxM or NML These results suggest that the changes in cardiac sympathetic activity observed during morphine withdrawal are due to intrinsic mechanisms outside the central nervous system. These data may be important for understanding the adaptive changes induced in the heart in subjects dependent on opioids.


Subject(s)
Heart Ventricles/drug effects , Morphine/adverse effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Substance Withdrawal Syndrome/physiopathology , Animals , Chromatography, High Pressure Liquid , Heart Ventricles/metabolism , Male , Naloxone/analogs & derivatives , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley
16.
Can J Anaesth ; 48(5): 486-92, 2001 May.
Article in English | MEDLINE | ID: mdl-11394520

ABSTRACT

PURPOSE: The purpose of the present study was to evaluate the effects of GI104313, a chimeric molecule containing a phosphodiesterase inhibiting pyradazinone and a blocking phenoxpropanolamine, on ischemia-induced arrhythmias in anesthetized rats. METHOD: The coronary artery was occluded 15 min after commencing drug administration and myocardial ischemia was maintained for 30 min during which the heart rate and mean blood pressure were recorded. Cyclic AMP and GMP were determined by radio-immunoassay. RESULTS: GI104313 (0.1 micromol x kg(-1) plus 0.01 micromol x kg(-1) x min(-1) or 1 micromol x kg(-1) plus 0.1 micromol x kg(-1) x min(-1)) decreased the incidence of ventricular tachycardia (86% and 75%), ventricular fibrillation (28%, P <0.01 and 12%, P <0.001) and premature ventricular beats (164 +/- 27.0 and 114 +/- 28.5, P <0.05) following coronary artery ligation, resulting in a decrease in mortality (29% and 12%, P <0.05). Changes in cyclic nucleotide concentrations have been implicated in the genesis of ischemia-induced arrhythmias. However, in the present study GI104313 did not change the concentrations of adenosine 3':5'-cyclic monophosphate (cyclic AMP) (1.0 +/- 0.07 pmol x mg(-1), 1.0 +/- 0.05 pmol x mg(-1)) or guanosine 3':5'-cyclic monophosphate (cyclic GMP) (0.025 +/- 0.008 pmol x mg(-1) protein, 0.017 +/- 0.004 pmol x mg(-1) protein) in the left ventricle during ischemia-induced arrhythmias in anesthetized rats compared to saline (0.9 +/- 0.1 pmol x mg(-1) and 0.013 +/- 0.002 pmol x m(-1), respectively). CONCLUSION: Our results demonstrate that, in rats, GI104313 induced a decrease in both incidence of arrhythmias and mortality which was not associated with changes in ventricular cyclic nucleotide content.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/prevention & control , Myocardial Ischemia/complications , Nitriles/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Pyridazines/pharmacology , Animals , Hemodynamics/drug effects , Male , Milrinone/pharmacology , Myocardium/metabolism , Nucleotides, Cyclic/metabolism , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley
17.
Eur J Pharmacol ; 397(2-3): 311-8, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10844129

ABSTRACT

In the present study, we examined the effects of alpha(1)- and the alpha(2)-adrenoceptors blockade on the changes in the ventricular content of catecholamines in rats withdrawn from morphine. Rats were given morphine by s.c. implantation of morphine pellets for 5 days. On the seventh day, morphine withdrawal was induced by s.c. administration of naloxone (1 mg/kg), and rats were killed 30 min later. Pretreatment with yohimbine (alpha(2)-adrenoceptor) or prazosin (alpha(1)-adrenoceptor) 15 min prior to naloxone administration attenuated some of the behavioural signs of morphine withdrawal. In addition, biochemical analysis indicated that yohimbine completely abolished the withdrawal-induced increase in noradrenaline and dopamine turnover in the right ventricle. By contrast, prazosin did not block the hyperactivity of catecholaminergic neurons in the heart during withdrawal. These data suggest that the hyperactivity of catecholaminergic neurons in the heart during morphine withdrawal is dependent upon alpha(2)-adrenoceptor activation. In addition, the present results rule out the involvement of alpha(1)-adrenoceptors.


Subject(s)
Analgesics, Opioid/pharmacology , Catecholamines/metabolism , Heart Ventricles/drug effects , Morphine Dependence/metabolism , Morphine/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Dopamine/metabolism , Heart Ventricles/innervation , Heart Ventricles/metabolism , Male , Norepinephrine/metabolism , Normetanephrine/metabolism , Prazosin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Yohimbine/pharmacology
18.
J Pharmacol Exp Ther ; 293(2): 578-84, 2000 May.
Article in English | MEDLINE | ID: mdl-10773031

ABSTRACT

We have previously shown an enhanced activity of the pituitary-adrenal response in rats dependent on morphine, which occurs concomitantly with an increase in the activity of catecholaminergic terminals in the hypothalamic paraventricular nucleus (PVN). The present study examined the possible role of noradrenergic system in the regulation of opioid withdrawal-induced activation of the hypothalamus-pituitary-adrenocortical (HPA) axis activity. Rats were given morphine by s.c. implantation of morphine pellets for 7 days. On the seventh day, morphine withdrawal was induced by s.c. administration of naloxone (1 mg/kg), rats were sacrificed 30 min later, and changes in noradrenaline (NA) turnover (estimated by the 3-methoxy-4-hydroxyphenylethylen glycol/NA ratio) and in dopamine turnover (estimated by the 3,4-dihydroxyphenylacetic acid/dopamine ratio) in the PVN (HPLC with electrochemical detection) and in plasma corticosterone levels were determined. We found a parallelism between the behavioral signs of withdrawal, an increased activity of noradrenergic and dopaminergic terminals in the PVN, and the hypersecretion of the HPA axis. Pretreatment with alpha(1)- or alpha(2)-adrenoceptor antagonists prazosin or yohimbine, respectively, 15 min before naloxone administration significantly prevented the withdrawal-induced corticosterone hypersecretion and attenuated the behavioral signs of morphine withdrawal. In addition, biochemical analysis indicated that both prazosin and yohimbine completely abolished the withdrawal-induced increase in NA turnover in the PVN. In contrast, neither prazosin nor yohimbine modified the hyperactivity of dopaminergic terminals in the PVN during withdrawal. Collectively, these data suggest that the secretory activity in the HPA axis after morphine withdrawal results from an increase in noradrenergic activity that is dependent on alpha(1)- and alpha(2)-adrenoceptor activation. Activation of dopaminergic pathways might not contribute to the neuroendocrine response during withdrawal.


Subject(s)
Catecholamines/physiology , Morphine Dependence/physiopathology , Paraventricular Hypothalamic Nucleus/physiology , Pituitary-Adrenal System/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, alpha-2/physiology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Animals , Autonomic Pathways/drug effects , Autonomic Pathways/physiology , Corticosterone/blood , Dopamine/metabolism , Male , Morphine Dependence/metabolism , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Norepinephrine/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Prazosin/pharmacology , Rats , Rats, Sprague-Dawley , Yohimbine/pharmacology
19.
Endocrinology ; 141(4): 1366-76, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10746640

ABSTRACT

Administration of the preferential mu-opioid receptor agonist, morphine, and selective K-opioid receptor agonists elicits activation of the hypothalamus-pituitary-adrenocortical axis, although the site or the molecular mechanisms for these effects have not been determined. The expression ofFos, the protein product of the c-fos protooncogene, has been widely used as an anatomical marker of monitoring neuronal activity. In the present study we evaluated 1) the effects of the mu-opioid receptor agonist, morphine, and those of the selective K-opioid receptor agonist, trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)cyclohexyl-]benzeneacet amide methane sulfonate (U-50,488H), administration on the expression of Fos in hypothalamic nuclei; and 2) the possible modification of the activity of noradrenergic neurons known to send afferent projections to the paraventricular nucleus (PVN), the site of CRF neurons involved in initiating ACTH secretion. Using immunohistochemical staining of Fos, the present results indicate that acute treatment with either morphine or U-50,488H induces marked Fos immunoreactivity within the hypothalamus, including the medial parvicellular PVN and supraoptic and suprachiasmatic nuclei. Pretreatment with naloxone attenuated the effect of morphine, whereas nor-binaltorphimine, a selective kappa-opioid receptor antagonist, abolished the effect of U-50,488H on Fos induction. Correspondingly, morphine and U-50,488H injection increased the production of the cerebral noradrenaline metabolite 3-methoxy-4-hydroxyphenylethylene glycol as well as noradrenaline turnover in the PVN. These effects were antagonized by naloxone and nor-bin-altorphimine, respectively. All of these findings are discussed in terms of specific events that couple opioid-induced activation of the hypothalamus-pituitary-adrenocortical axis and noradrenergic activity with changes in gene expression in selective hypothalamic nuclei.


Subject(s)
Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists , Suprachiasmatic Nucleus/metabolism , Supraoptic Nucleus/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Male , Methoxyhydroxyphenylglycol/metabolism , Morphine/pharmacology , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Norepinephrine/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Suprachiasmatic Nucleus/drug effects , Supraoptic Nucleus/drug effects
20.
Pharmacol Toxicol ; 86(2): 68-70, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10728917

ABSTRACT

The effects of alpha,beta- and beta,gamma-methylene ATP on ventricular automaticity induced by local injury in the right ventricle of rats pretreated with reserpine, were investigated and compared with the effects of ATP and adenosine. Beta,gamma-methylene ATP but not alpha,beta-methylene ATP mimicked the inhibitory effect of both ATP and adenosine on the spontaneous automaticity In low concentrations, adenosine was more effective than ATP. Alpha,beta-methylene ATP caused little or no effect on ventricular automaticity. The results suggest that the effect of ATP might depend on its hydrolysis into AMP/adenosine.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Heart Rate/drug effects , Heart Ventricles/drug effects , Adenosine Triphosphate/pharmacology , Animals , Female , Heart Rate/physiology , Heart Ventricles/injuries , Heart Ventricles/physiopathology , Male , Molecular Mimicry , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...