Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 501
Filter
1.
Toxicol Mech Methods ; 34(9): 1035-1044, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39021086

ABSTRACT

Chronic exposure to manganese compounds leads to accumulation of the manganese in the basal ganglia and hippocampus. High levels of manganese in these structures lead to oxidative stress, neuroinflammation, imbalance of brain neurotransmitters, and hyperactivation of calpains mediating neurotoxicity and causing motor and cognitive impairment. The purpose of this work was to study the effect of excess manganese chloride intake on rats' spatial memory and on dopamine-ß-hydroxylase (DßH) activity under conditions of calpain activity suppression. Rats were divided into 3 groups of 10 animals each. Group 1 received MnCl2 (30 days, 5 mg/kg/day, intranasally), group 2 received MnCl2 (30 days, 5 mg/kg/day, intranasally) and calpain inhibitor Cast (184-210) (30 days, 5 µg/kg/day, intranasally), and group 3 received sterile saline (30 days in a volume of 20 µl, intranasally). The spatial working memory was assessed using Morris water maze test. DßH activity was determined by HPLC. We have shown that in response to excessive intake of MnCl2, there was a development of cognitive impairments in rats, which was accompanied by a decrease in DßH activity in the hippocampus. The severity of cognitive impairment was reduced by inhibiting the activity of m-calpain. The protective effect of calpain inhibitors was achieved not through an effect on DßH activity. Thus, the development of therapeutic regimens for the treatment of manganism using dopaminomimetics and/or by inhibiting calpains, must be performed taking into account the manganese-induced decrease of DßH activity and the inability to influence this process with calpain inhibitors.


Subject(s)
Calpain , Chlorides , Dopamine beta-Hydroxylase , Manganese Compounds , Memory Disorders , Rats, Wistar , Animals , Male , Chlorides/toxicity , Calpain/metabolism , Calpain/antagonists & inhibitors , Memory Disorders/chemically induced , Memory Disorders/enzymology , Dopamine beta-Hydroxylase/metabolism , Dopamine beta-Hydroxylase/antagonists & inhibitors , Spatial Memory/drug effects , Brain/drug effects , Brain/enzymology , Brain/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Rats , Maze Learning/drug effects
2.
Behav Brain Res ; 471: 115116, 2024 08 05.
Article in English | MEDLINE | ID: mdl-38897419

ABSTRACT

The neural mechanisms underlying paternal care in biparental mammals are not well understood. The California mouse (Peromyscus californicus) is a biparental rodent in which virtually all fathers are attracted to pups, while virgin males vary widely in their behavior toward unrelated infants, ranging from attacking to avoiding to huddling and grooming pups. We previously showed that pharmacologically inhibiting the synthesis of the neurotransmitter norepinephrine (NE) with the dopamine ß-hydroxylase inhibitor nepicastat reduced the propensity of virgin male and female California mice to interact with pups. The current study tested the hypothesis that nepicastat would reduce pup-induced c-Fos immunoreactivity, a cellular marker of neural activity, in the medial preoptic area (MPOA), medial amygdala (MeA), basolateral amygdala (BLA), and bed nucleus of the stria terminalis (BNST), brain regions implicated in the control of parental behavior and/or anxiety. Virgin males were injected with nepicastat (75 mg/kg, i.p.) or vehicle 2 hours prior to exposure to either an unrelated pup or novel object for 60 minutes (n = 4-6 mice per group). Immediately following the 60-minute stimulus exposure, mice were euthanized and their brains were collected for c-Fos immunohistochemistry. Nepicastat reduced c-Fos expression in the MeA and MPOA of pup-exposed virgin males compared to vehicle-injected controls. In contrast, nepicastat did not alter c-Fos expression in any of the above brain regions following exposure to a novel object. Overall, these results suggest that the noradrenergic system might influence MeA and MPOA function to promote behavioral interactions with pups in virgin males.


Subject(s)
Dopamine beta-Hydroxylase , Paternal Behavior , Peromyscus , Preoptic Area , Septal Nuclei , Animals , Male , Dopamine beta-Hydroxylase/metabolism , Dopamine beta-Hydroxylase/antagonists & inhibitors , Paternal Behavior/physiology , Paternal Behavior/drug effects , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Preoptic Area/metabolism , Preoptic Area/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Female , Enzyme Inhibitors/pharmacology , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/metabolism , Corticomedial Nuclear Complex/drug effects , Corticomedial Nuclear Complex/metabolism , Norepinephrine/metabolism , Imidazoles , Thiones
3.
Pharmacol Rep ; 73(6): 1694-1711, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34236605

ABSTRACT

BACKGROUND: Opioid use disorders are serious contributors to the harms associated with the drug use. Unfortunately, therapeutic interventions for opioid addicts after detoxification have been limited and not sufficiently effective. Recently, several studies have led to promising results with disulfiram (DSF), a dopamine ß-hydroxylase (DBH) inhibitor, showing that it is a potent agent against not only alcohol but also addiction to various drugs. MATERIALS AND METHODS: This study was designed to examine whether DSF and nepicastat (NEP; another DBH inhibitor) modify morphine intake and reinstatement of seeking-behavior using the rat model of intravenous morphine self-administration. Additionally, we intended to estimate the effects of both inhibitors on the locomotor activity as well as on extracellular dopamine and its metabolite levels in the nucleus accumbens using microdialysis in naive rats. RESULTS: We demonstrated that both DBH inhibitors reduced responding to morphine self-administration. Moreover, DSF and NEP administered acutely before reinstatement test sessions consistently attenuated the reinforcing effects of morphine and a morphine-associated conditioned cue. The observed effects for lower doses (6.25-25 mg/kg; ip) of both DBH inhibitors seem to be independent of locomotor activity reduction and dopamine level in the nucleus accumbens. Neither DSF nor NEP administered daily during morphine abstinence with extinction training sessions had any effect on active lever-responding and changed the reinstatement induced by morphine priming doses. Reinstatement of drug-seeking behavior induced by a conditioned cue previously associated with morphine delivery was attenuated following repeated administration of DSF or NEP during the abstinence period. CONCLUSION: These results seem to point to the significance  of DBH inhibition as a potential pharmacotherapy against morphine use disorders.


Subject(s)
Disulfiram/pharmacology , Dopamine beta-Hydroxylase/antagonists & inhibitors , Imidazoles/pharmacology , Morphine/administration & dosage , Thiones/pharmacology , Animals , Dopamine/metabolism , Drug-Seeking Behavior/drug effects , Enzyme Inhibitors/pharmacology , Extinction, Psychological/drug effects , Male , Nucleus Accumbens/metabolism , Opioid-Related Disorders/drug therapy , Rats , Rats, Wistar , Recurrence , Self Administration
4.
Article in English | MEDLINE | ID: mdl-33161095

ABSTRACT

Dopamine beta-hydroxylase (DßH) plays an essential role in the synthesis of catecholamines (CA) in neuroendocrine networks. In the razor clam, Sinonovacula constricta a novel gene for DßH (ScDßH-α) was identified that belongs to the copper type II ascorbate-dependent monooxygenase family. Expression analysis revealed ScDßH-α gene transcripts were abundant in the liver and expressed throughout development. Knock-down of ScDßH-α in adult clams using siRNA caused a reduction in the growth rate compared to control clams. Reduced growth was associated with strong down-regulation of gene transcripts for the growth-related factors, platelet derived growth factors A (PDGF-A) (P < 0.001) 24 h after ScDßH-α knock-down, vascular endothelial growth factor (VEGF1) (P < 0.001) and platelet derived growth factor B (PDGF-B-2) (P < 0.001) 24 h and 48 h after ScDßH-α knock-down and transforming growth factor beta (TGF-ß1) (P < 0.001) 48 h and 72 h after ScDßH-α knock-down. Taken together the results suggest that the novel ScDßH-α gene through its role in CA synthesis is involved in growth regulation in the razor clam and possibly other bivalves.


Subject(s)
Bivalvia/growth & development , Bivalvia/genetics , Amino Acid Sequence , Animals , Bivalvia/immunology , Bivalvia/metabolism , Cloning, Molecular/methods , DNA, Complementary/genetics , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Gene Knockdown Techniques , Immunity, Innate , Phylogeny , Proto-Oncogene Proteins c-sis/metabolism , RNA Interference , Sequence Homology , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Dokl Biochem Biophys ; 493(1): 201-204, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32894465

ABSTRACT

This work is aimed at studying the mechanisms of reciprocal humoral regulation of noradrenaline-producing organs in rats in the perinatal period of development. The activity of noradrenaline synthesis enzymes tyrosine hydroxylase and dopamine-beta-hydroxylase was measured in the brain and adrenal glands 48 and 72 h after the injection of immunotoxin (anti-dopamine-beta-hydroxylase-saporin) into the rat brain ventricles. It was shown that, 48 h after the immunotoxin injection into the brain, the activity of tyrosine hydroxylase in the brain decreased; however, 72 h after the injection it reached the control levels. This fact indicates that noradrenaline synthesis in the survived neurons increases. In the adrenal glands, 72 h after the immunotoxin injection into the brain, the activity of dopamine-beta-hydroxylase increased. This points to a compensatory increase in the rate of noradrenaline synthesis in the adrenal glands when the synthesis of noradrenaline in the brain is inhibited.


Subject(s)
Adrenal Glands/metabolism , Brain/metabolism , Dopamine beta-Hydroxylase/metabolism , Norepinephrine/biosynthesis , Saporins/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Adrenal Glands/drug effects , Animals , Animals, Newborn , Brain/drug effects , Dopamine beta-Hydroxylase/antagonists & inhibitors , Immunotoxins/pharmacology , Male , Rats , Rats, Wistar
6.
Expert Opin Investig Drugs ; 29(9): 1043-1057, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32658551

ABSTRACT

INTRODUCTION: Despite a large number of commercially available drugs, hypertension and related cardiovascular diseases remain a global problem. It is thus imperative that novel drugs and therapeutic strategies are regularly identified, and alternative targets explored. Dopamine ß hydroxylase (DBH), a key player in the catecholamine biosynthetic pathway, may provide a therapeutic opportunity and should be extensively explored as a target for potent anti-hypertensives. Inhibitors of DBH have been successful in combating hypertension, as evidenced by the outcome of clinical trials for etamicastat and zamicastat. AREAS COVERED: We shed light on the strategies employed to identify inhibitors of the enzyme and outline the advantages that the target might offer. Structural and functional details of the enzyme are described along with specific methodologies for drug discovery that were never utilized for the therapeutic target. EXPERT OPINION: Effective inhibitors of the enzyme may be identified with computer-aided structure-based design. Adoption of new methodologies and the assessment of newly designed inhibitors in DBH-specific animal models will provide new, safe, and cost-effective therapeutic opportunities.


Subject(s)
Antihypertensive Agents/pharmacology , Dopamine beta-Hydroxylase/antagonists & inhibitors , Hypertension/drug therapy , Animals , Benzopyrans/pharmacology , Dopamine beta-Hydroxylase/metabolism , Drug Design , Drug Discovery , Enzyme Inhibitors/pharmacology , Humans , Hypertension/enzymology , Hypertension/physiopathology , Imidazoles/pharmacology
7.
Curr Opin Cardiol ; 33(4): 408-415, 2018 07.
Article in English | MEDLINE | ID: mdl-29702500

ABSTRACT

PURPOSE OF REVIEW: Hypertension (HTN) is a widespread and growing disease, with medication intolerance and side-effect present among many. To address these obstacles novel pharmacotherapy is an active area of drug development. This review seeks to explore future drug therapy for HTN in the preclinical and clinical arenas. RECENT FINDINGS: The future of pharmacological therapy in HTN consists of revisiting old pathways to find new targets and exploring wholly new approaches to provide additional avenues of treatment. In this review, we discuss the current status of the most recent drug therapy in HTN. New developments in well trod areas include novel mineralocorticoid antagonists, aldosterone synthase inhibitors, aminopeptidase-A inhibitors, natriuretic peptide receptor agonists, or the counter-regulatory angiotensin converting enzyme 2/angiotensin (Ang) (1-7)/Mas receptor axis. Neprilysin inhibitors popularized for heart failure may also still hold HTN potential. Finally, we examine unique systems in development never before used in HTN such as Na/H exchange inhibitors, vasoactive intestinal peptide agonists, and dopamine beta hydroxylase inhibitors. SUMMARY: A concise review of future directions of HTN pharmacotherapy.


Subject(s)
Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Hypertension/drug therapy , Cytochrome P-450 CYP11B2/antagonists & inhibitors , Dopamine beta-Hydroxylase/antagonists & inhibitors , Glutamyl Aminopeptidase/antagonists & inhibitors , Humans , Mineralocorticoid Receptor Antagonists/therapeutic use , Neprilysin/antagonists & inhibitors , Receptors, Atrial Natriuretic Factor/agonists , Receptors, Vasoactive Intestinal Peptide/agonists , Renin-Angiotensin System/drug effects , Sodium-Hydrogen Exchanger 3/antagonists & inhibitors
8.
Am J Physiol Regul Integr Comp Physiol ; 314(6): R811-R823, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29384699

ABSTRACT

Hindbrain catecholamine neurons convey gut-derived metabolic signals to an interconnected neuronal network in the hypothalamus and adjacent forebrain. These neurons are critical for short-term glycemic control, glucocorticoid and glucoprivic feeding responses, and glucagon-like peptide 1 (GLP-1) signaling. Here we investigate whether these pathways also contribute to long-term energy homeostasis by controlling obesogenic sensitivity to a high-fat/high-sucrose choice (HFSC) diet. We ablated hindbrain-originating catecholaminergic projections by injecting anti-dopamine-ß-hydroxylase-conjugated saporin (DSAP) into the paraventricular nucleus of the hypothalamus (PVH) of male rats fed a chow diet for up to 12 wk or a HFSC diet for 8 wk. We measured the effects of DSAP lesions on food choices; visceral adiposity; plasma glucose, insulin, and leptin; and indicators of long-term ACTH and corticosterone secretion. We also determined lesion effects on the number of carbohydrate or fat calories required to increase visceral fat. Finally, we examined corticotropin-releasing hormone levels in the PVH and arcuate nucleus expression of neuropeptide Y ( Npy), agouti-related peptide ( Agrp), and proopiomelanocortin ( Pomc). DSAP-injected chow-fed rats slowly increase visceral adiposity but quickly develop mild insulin resistance and elevated blood glucose. DSAP-injected HFSC-fed rats, however, dramatically increase food intake, body weight, and visceral adiposity beyond the level in control HFSC-fed rats. These changes are concomitant with 1) a reduction in the number of carbohydrate calories required to generate visceral fat, 2) abnormal Npy, Agrp, and Pomc expression, and 3) aberrant control of insulin secretion and glucocorticoid negative feedback. Long-term metabolic adaptations to high-carbohydrate diets, therefore, require intact forebrain catecholamine projections. Without them, animals cannot alter forebrain mechanisms to restrain increased visceral adiposity.


Subject(s)
Catecholamines/metabolism , Nerve Net/physiopathology , Obesity/physiopathology , Prosencephalon/physiopathology , Animals , Blood Glucose/metabolism , Diet , Dopamine beta-Hydroxylase/antagonists & inhibitors , Energy Intake , Insulin/blood , Intra-Abdominal Fat/drug effects , Leptin/blood , Male , Neural Pathways/physiopathology , Paraventricular Hypothalamic Nucleus , Rats , Rats, Sprague-Dawley , Saporins/pharmacology
9.
Eur J Pharm Sci ; 117: 35-40, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29428540

ABSTRACT

Inhibition of the biosynthesis of noradrenaline is a currently explored strategy for the treatment of hypertension, congestive heart failure and pulmonary arterial hypertension. While some dopamine ß-hydroxylase (DBH) inhibitors cross the blood-brain barrier (BBB) and cause central as well as peripheral effects (nepicastat), others have limited access to the brain (etamicastat, zamicastat). In this context, peripheral selectivity is clinically advantageous, in order to prevent alterations of noradrenaline levels in the CNS and the occurrence of adverse central effects. A limited brain exposure results from the combination of several factors, such as a reduced passive permeability or affinity for efflux transporters, but efflux liabilities may also lead to unwanted drug-drug interactions (DDIs) in the presence of co-administered substrates or inhibitors. Thus, the purpose of the study herein presented was to explore the interaction of P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP), the two major efflux transporters of the BBB that hamper the entry of several drugs to the brain, with the DBH inhibitors, etamicastat, nepicastat and zamicastat. Madin-Darby canine kidney cells (MDCK II) and transfected lines with human MDR1 (MDCK-MDR1) and ABCG2 (MDCK-BCRP) genes were used as a BBB surrogate model. P-gp and BCRP substrates and/or inhibitors were identified through intracellular accumulation and bidirectional permeability assays. The obtained data revealed that zamicastat is a concentration-dependent dual P-gp and BCRP inhibitor with IC50 values of 73.8 ±â€¯7.2 µM and 17.0 ±â€¯2.7 µM, while etamicastat and nepicastat inhibited BCRP to greater extent than P-gp, with IC50 values of 47.7 ±â€¯1.8 µM and 59.2 ±â€¯9.4 µM, respectively. Additionally, etamicastat was identified as P-gp and BCRP dual substrate, as demonstrated by net flux ratios of 5.84 and 3.87 and decreased >50% by verapamil and Ko143. Conversely, nepicastat revealed to be a P-gp-only substrate, with a net flux ratio of 2.01, reduced to 0.92 in the presence of verapamil. Furthermore, nepicastat displayed a consistently higher apparent permeability (>8.49 × 10-6 cm s-1) than etamicastat (<0.58 × 10-6 cm s-1). The identification of etamicastat as a dual efflux substrate suggests that P-gp and BCRP may be partially responsible for the limited central exposure of this compound, in association with its low passive permeability. Moreover, the weak efflux inhibitory potencies of etamicastat and nepicastat revealed a low DDI risk, while the dual P-gp/BCRP inhibition of zamicastat could be studied in the future with synergically effluxed compounds, for which BBB penetration is severely impaired.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Benzopyrans/pharmacology , Dopamine beta-Hydroxylase/antagonists & inhibitors , Imidazoles/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Thiones/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Cell Survival/drug effects , Dogs , Drug Interactions , Madin Darby Canine Kidney Cells , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
10.
Arch Dermatol Res ; 308(7): 473-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27278925

ABSTRACT

1-Phenyl-3-(2-thiazolyl)-2-thiourea (PTTU) is a well-characterized dopamine ß-hydroxylase inhibitor that prevents 6-hydroxydopamine-induced degenerative neuronal disease. However, the effect of PTTU on melanogenesis has not been reported. In this study, we examined the effect of PTTU on melanogenesis and studied its mechanism of action. We found that PTTU decreased melanin biosynthesis in a dose-dependent manner in normal human epidermal melanocytes (NHEMs). PTTU also inhibited tyrosinase catalytic activity in NHEMs. Moreover, PTTU treatment led to reduced protein levels of tyrosinase in NHEMs, while the protein levels of tyrosinase-related protein-1, tyrosinase-related protein-2, and microphthalmia-associated transcription factor were not affected. However, PTTU treatment did not affect the mRNA expression of tyrosinase. We found that PTTU-accelerated tyrosinase degradation via the ubiquitin-dependent proteasome pathway. In summary, we found that PTTU decreased melanin biosynthesis by decreasing the enzymatic activity and stability of tyrosinase. Our results indicate that PTTU could be used as a depigmentation agent for hyperpigmentation disorder.


Subject(s)
Melanins/biosynthesis , Melanocytes/metabolism , Monophenol Monooxygenase/antagonists & inhibitors , Phenylthiazolylthiourea/pharmacology , Cells, Cultured , Dopamine beta-Hydroxylase/antagonists & inhibitors , Epidermis/metabolism , Humans , Hyperpigmentation/drug therapy , Skin Pigmentation/drug effects
11.
J Am Soc Hypertens ; 10(3): 207-16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26803288

ABSTRACT

The aim of the present study was to evaluate the influence of chronic inhibition of dopamine ß-hydroxylase by etamicastat on the development of hypertension in the spontaneously hypertensive rat (SHR) and the sustainability of effects on the systolic and diastolic blood pressure in the SHR and the normotensive Wistar-Kyoto rat (WKY). WKY and SHR received etamicastat (10 mg/kg/d) from 5 weeks of age for 35 weeks in drinking water, and cardiovascular assessments were performed on a weekly basis. Etamicastat reduced systolic and diastolic blood pressure when SHRs reached the age of 16 weeks with mean decreases of 37 and 32 mm Hg, respectively, for the subsequent for 24 weeks of treatment, but did not prevent the increase in blood pressure (BP) aged between 5 and 11 week. The BP lowering effect of etamicastat in SHR was reversible on discontinuation and quickly resumed after reinstatement of therapy and was not accompanied by changes in heart rate. Etamicastat affected neither BP nor heart rate in WKY during 36 weeks of treatment. Etamicastat reduced urinary excretion of norepinephrine to a similar extent in WKY and SHR, accompanied by significant increases in urinary dopamine in SHR. Chronic administration of etamicastat did not adversely affected development of animals. Chronic dopamine ß-hydroxylase inhibition with etamicastat effectively decreases BP, although does not prevent the development of hypertension in the SHR.


Subject(s)
Antihypertensive Agents/therapeutic use , Benzopyrans/therapeutic use , Blood Pressure/drug effects , Dopamine beta-Hydroxylase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Hypertension/drug therapy , Imidazoles/therapeutic use , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/adverse effects , Benzopyrans/administration & dosage , Benzopyrans/adverse effects , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Heart Rate/drug effects , Humans , Hypertension/prevention & control , Imidazoles/administration & dosage , Imidazoles/adverse effects , Male , Norepinephrine/urine , Rats , Rats, Inbred SHR/urine , Rats, Inbred WKY/urine
12.
Addict Biol ; 21(1): 35-48, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25123018

ABSTRACT

Cocaine blocks plasma membrane monoamine transporters and increases extracellular levels of dopamine (DA), norepinephrine (NE) and serotonin (5-HT). The addictive properties of cocaine are mediated primarily by DA, while NE and 5-HT play modulatory roles. Chronic inhibition of dopamine ß-hydroxylase (DBH), which converts DA to NE, increases the aversive effects of cocaine and reduces cocaine use in humans, and produces behavioral hypersensitivity to cocaine and D2 agonism in rodents, but the underlying mechanism is unknown. We found a decrease in ß-arrestin2 (ßArr2) in the nucleus accumbens (NAc) following chronic genetic or pharmacological DBH inhibition, and overexpression of ßArr2 in the NAc normalized cocaine-induced locomotion in DBH knockout (Dbh -/-) mice. The D2/3 agonist quinpirole decreased excitability in NAc medium spiny neurons (MSNs) from control, but not Dbh -/- animals, where instead there was a trend for an excitatory effect. The Gαi inhibitor NF023 abolished the quinpirole-induced decrease in excitability in control MSNs, but had no effect in Dbh -/- MSNs, whereas the Gαs inhibitor NF449 restored the ability of quinpirole to decrease excitability in Dbh -/- MSNs, but had no effect in control MSNs. These results suggest that chronic loss of noradrenergic tone alters behavioral responses to cocaine via decreases in ßArr2 and cellular responses to D2/D3 activation, potentially via changes in D2-like receptor G-protein coupling in NAc MSNs.


Subject(s)
Arrestins/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Locomotion/drug effects , Neurons/drug effects , Nucleus Accumbens/drug effects , Receptors, Dopamine D2/metabolism , Animals , Arrestins/metabolism , Behavior, Animal/drug effects , Benzenesulfonates/pharmacology , Chromogranins , Dopamine Agonists/pharmacology , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine beta-Hydroxylase/genetics , GTP-Binding Protein alpha Subunits, Gs/antagonists & inhibitors , Mice , Mice, Knockout , Neurons/metabolism , Norepinephrine/metabolism , Nucleus Accumbens/metabolism , Quinpirole/pharmacology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D3/agonists , beta-Arrestins
13.
Addict Biol ; 21(1): 61-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25135633

ABSTRACT

Previous investigations indicate that the dopamine-ß-hydroxylase (DBH) inhibitors disulfiram and nepicastat suppress cocaine-primed reinstatement of cocaine self-administration behaviour. Moreover, both inhibitors increase dopamine release in the rat medial prefrontal cortex (mPFC) and markedly potentiate cocaine-induced dopamine release in this region. This study was aimed to clarify if the suppressant effect of DBH inhibitors on cocaine reinstatement was mediated by the high extracellular dopamine in the rat mPFC leading to a supra-maximal stimulation of D1 receptors in the dorsal division of mPFC, an area critical for reinstatement of cocaine-seeking behaviour. In line with previous microdialysis studies in drug-naïve animals, both DBH inhibitors potentiated cocaine-induced dopamine release in the mPFC, in the same animals in which they also suppressed reinstatement of cocaine seeking. Similar to the DBH inhibitors, L-DOPA potentiated cocaine-induced dopamine release in the mPFC and suppressed cocaine-induced reinstatement of cocaine-seeking behaviour. The bilateral microinfusion of the D1 receptor antagonist SCH 23390 into the dorsal mPFC not only prevented cocaine-induced reinstatement of cocaine seeking but also reverted both disulfiram- and L-DOPA-induced suppression of reinstatement. Moreover, the bilateral microinfusion of the D1 receptor agonist chloro-APB (SKF 82958) into the dorsal mPFC markedly attenuated cocaine-induced reinstatement of cocaine seeking. These results suggest that stimulation of D1 receptors in the dorsal mPFC plays a crucial role in cocaine-induced reinstatement of cocaine seeking, whereas the suppressant effect of DBH inhibitors and L-DOPA on drug-induced reinstatement is mediated by a supra-maximal stimulation of D1 receptors leading to their inactivation.


Subject(s)
Cocaine/administration & dosage , Disulfiram/pharmacology , Dopamine Agents/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Dopamine beta-Hydroxylase/antagonists & inhibitors , Drug-Seeking Behavior/drug effects , Imidazoles/pharmacology , Levodopa/pharmacology , Prefrontal Cortex/drug effects , Receptors, Dopamine D1/antagonists & inhibitors , Thiones/pharmacology , Animals , Behavior, Animal/drug effects , Benzazepines/pharmacology , Dopamine/metabolism , Extinction, Psychological , Male , Microdialysis , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/agonists , Self Administration
14.
Brain Behav ; 5(10): e00393, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26516613

ABSTRACT

INTRODUCTION: Disulfiram has been claimed to be useful in cocaine addiction therapy, its efficacy being attributed to dopamine-beta-hydroxylase (DBH) inhibition. Our previous results indicate that disulfiram and the selective DBH inhibitor nepicastat increase extracellular dopamine (DA) in the rat medial prefrontal cortex (mPFC), and markedly potentiated cocaine-induced increase. Concomitantly, in rats with cocaine self-administration history, cocaine-seeking behavior induced by drug priming was prevented, probably through overstimulation of D1 receptors due to the DA increase. The present research was aimed at studying the neurochemical mechanisms originating the enhanced DA release. METHODS: Noradrenergic system ablation was attained by intracerebroventricular (i.c.v.) administration of the neurotoxin anti-DBH-saporin (aDBH-sap). DA, noradrenaline (NA), and DOPAC were assessed by HPLC after ex vivo tissue extraction or in vivo microdialysis. Control and denervated rats were subjected to microdialysis in the mPFC and caudate nucleus to evaluate the effect of nepicastat-cocaine combination on extracellular DA levels and their regulation by α2-adrenoceptors. RESULTS: Fifteen days after neurotoxin or its vehicle administration, tissue and extracellular NA were reduced to less than 2% the control value, while extracellular DA was increased by approximately 100%. In control rats, nepicastat given alone and in combination with cocaine increased extracellular DA by about 250% and 1100%, respectively. In denervated rats, nepicastat slightly affected extracellular DA, while in combination with cocaine increased extracellular DA by 250%. No differences were found in the caudate nucleus. Clonidine almost totally reversed the extracellular DA elevation produced by nepicastat-cocaine combination, while it was ineffective in denervated rats. CONCLUSIONS: This research shows that the increase of extracellular DA produced by nepicastat alone or in combination with cocaine was prevented by noradrenergic denervation. The results indicate that nepicastat enhances DA release from noradrenergic terminals supposedly by removing NA from α2-autoreceptors. In addition to the inhibition of DA uptake, the latter mechanism may explain the synergistic effect of cocaine on nepicastat-induced DA release.


Subject(s)
Adrenergic Neurons/drug effects , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine/metabolism , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Prefrontal Cortex/drug effects , Thiones/pharmacology , Adrenergic Neurons/enzymology , Adrenergic Neurons/metabolism , Animals , Cocaine/administration & dosage , Dopamine beta-Hydroxylase/metabolism , Injections, Intraventricular , Male , Microdialysis , Norepinephrine/metabolism , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/enzymology , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Self Administration
15.
Am J Physiol Regul Integr Comp Physiol ; 309(7): R721-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26157062

ABSTRACT

Brainstem catecholamine neurons modulate sensory information and participate in control of cardiorespiratory function. These neurons have multiple projections, including to the paraventricular nucleus (PVN), which contributes to cardiorespiratory and neuroendocrine responses to hypoxia. We have shown that PVN-projecting catecholaminergic neurons are activated by hypoxia, but the function of these neurons is not known. To test the hypothesis that PVN-projecting catecholamine neurons participate in responses to respiratory challenges, we injected IgG saporin (control; n = 6) or anti-dopamine ß-hydroxylase saporin (DSAP; n = 6) into the PVN to retrogradely lesion catecholamine neurons projecting to the PVN. After 2 wk, respiratory measurements (plethysmography) were made in awake rats during normoxia, increasing intensities of hypoxia (12, 10, and 8% O2) and hypercapnia (5% CO2-95% O2). DSAP decreased the number of tyrosine hydroxylase-immunoreactive terminals in PVN and cells counted in ventrolateral medulla (VLM; -37%) and nucleus tractus solitarii (nTS; -36%). DSAP produced a small but significant decrease in respiratory rate at baseline (during normoxia) and at all intensities of hypoxia. Tidal volume and minute ventilation (VE) index also were impaired at higher hypoxic intensities (10-8% O2; e.g., VE at 8% O2: IgG = 181 ± 22, DSAP = 91 ± 4 arbitrary units). Depressed ventilation in DSAP rats was associated with significantly lower arterial O2 saturation at all hypoxic intensities. PVN DSAP also reduced ventilatory responses to 5% CO2 (VE: IgG = 176 ± 21 and DSAP = 84 ± 5 arbitrary units). Data indicate that catecholamine neurons projecting to the PVN are important for peripheral and central chemoreflex respiratory responses and for maintenance of arterial oxygen levels during hypoxic stimuli.


Subject(s)
Catecholamines/physiology , Hemodynamics/physiology , Hypoxia/physiopathology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Respiratory Mechanics/physiology , Animals , Blood Pressure/physiology , Brain Stem/cytology , Brain Stem/physiology , Dopamine beta-Hydroxylase/antagonists & inhibitors , Heart Rate/physiology , Hypercapnia/physiopathology , Immunohistochemistry , Male , Paraventricular Hypothalamic Nucleus/cytology , Rats , Rats, Sprague-Dawley , Reflex/physiology , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins , Telemetry
16.
Hypertens Res ; 38(9): 605-12, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25854989

ABSTRACT

Overactivity of the sympathetic nervous system has an important role in the development and progression of arterial hypertension. Catheter-based renal nerve ablation for the treatment of drug-resistant hypertension has recently been developed. An alternative strategy for the modulation of sympathetic nerve function is to reduce the biosynthesis of noradrenaline (NA) by inhibiting dopamine ß-hydroxylase (DßH), the enzyme that catalyzes the conversion of dopamine (DA) to NA in the sympathetic nerves. Renal denervation (RDN) surgery was performed in spontaneously hypertensive rats (SHR) to evaluate the effect of RDN on the DA and NA levels and on blood pressure over a 28-day period. The selective peripheral DßH inhibitor etamicastat (30 mg kg (-1)day(-1)) was administered to another cohort of SHR. RDN and etamicastat treatment had no effect on the renal function, as assessed by measuring the water balance response, renal function and urinary electrolyte levels. RDN significantly decreased the systolic blood pressure (SBP) and the diastolic blood pressure (DBP). A gradual return of the SBP and the DBP to the high baseline levels was observed over time. Conversely, treatment with etamicastat resulted in a significant decrease in the SBP and the DBP at all time points. On the last day of the assessment, NA levels in renal tissue were significantly decreased in both RDN and etamicastat-treated groups. In contrast, the NA levels in the left ventricle were decreased only in the etamicastat-treated group. Thus, RDN produces transitory decreases in blood pressure, whereas prolonged downregulation of sympathetic drive with the DßH inhibitor etamicastat results in a sustained decrease in the SBP and the DBP.


Subject(s)
Benzopyrans/pharmacology , Blood Pressure/drug effects , Dopamine beta-Hydroxylase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Kidney/innervation , Animals , Benzopyrans/pharmacokinetics , Denervation , Dopamine/metabolism , Dopamine beta-Hydroxylase/metabolism , Enzyme Inhibitors/pharmacokinetics , Hemodynamics/drug effects , Imidazoles/pharmacokinetics , Male , Norepinephrine/metabolism , Rats , Rats, Inbred SHR , Water-Electrolyte Balance/drug effects
17.
Xenobiotica ; 45(9): 828-39, 2015.
Article in English | MEDLINE | ID: mdl-25915108

ABSTRACT

1. This study explores the impact of permeability and P-glycoprotein (P-gp) efflux, upon brain exposure to etamicastat, a new dopamine-ß-hydroxylase (DBH) inhibitor and consequently brain levels of catecholamines. 2. Brain exposure to etamicastat (10 mg/kg), following intravenous administration to mice, was residual and upon oral administration of the same dose no compound was detected, concurring with the absence of effects upon brain catecholamines. The intravenous co-administration of elacridar (1.0 mg/kg), a known P-gp/BCRP dual modulator, significantly increased brain etamicastat exposure, but the levels attained were very low when compared to those of nepicastat, a centrally active DBH inhibitor. 3. In vitro permeability studies from apical-to-basal direction conducted in Caco-2 cells and MDCK-II cells showed that etamicastat apparent permeability was 1.2 × 10(-5) and 1.1 × 10(-6 )cm/s, respectively, 5- and 50-fold lower as compared to nepicastat. The secretory efflux ratio in MDCK-II cells overexpressing human P-gp showed an efflux ratio greater than 2, for both compounds, which was significantly decreased by elacridar. Despite its lower bioavailability and higher clearance, as compared to nepicastat, etamicastat showed preferential distribution to peripheral tissues and high plasma free fraction (15.5%), which may explain its effects upon peripheral DBH and catecholamine levels. 4. Though P-gp-mediated efflux may contribute to the limited brain penetration of etamicastat, the low permeability along with the pharmacokinetic properties of etamicastat may be perceived as the main contributors for its peripheral selectivity, which is advantageous for a cardiovascular drug candidate.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Benzopyrans/pharmacology , Brain/metabolism , Cell Membrane Permeability/drug effects , Imidazoles/pharmacology , Thiones/pharmacology , Acridines/administration & dosage , Acridines/pharmacology , Animals , Atenolol/pharmacology , Benzopyrans/blood , Benzopyrans/chemistry , Benzopyrans/pharmacokinetics , Biological Transport/drug effects , Blood Proteins/metabolism , Caco-2 Cells , Catecholamines/metabolism , Dogs , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine beta-Hydroxylase/metabolism , Humans , Imidazoles/blood , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Liver/drug effects , Liver/metabolism , Madin Darby Canine Kidney Cells , Male , Mice , Propranolol/pharmacology , Protein Binding/drug effects , Tetrahydroisoquinolines/administration & dosage , Tetrahydroisoquinolines/pharmacology , Thiones/blood , Thiones/chemistry , Thiones/pharmacokinetics , Tissue Distribution/drug effects
18.
Eur J Pharmacol ; 750: 98-107, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25641747

ABSTRACT

Etamicastat, a peripheral reversible dopamine-ß-hydroxylase inhibitor, blocked the hERG current amplitude with an IC50 value of 44.0µg/ml in HEK 293 cells. At 0.3 and 3µg/ml, etamicastat had no effects on the action potential (AP) in male dog Purkinje fibers. At 30µg/ml, etamicastat significantly affected resting membrane potential (+4%), AP amplitude (-4%), AP duration at 60% (-14%) and AP duration at 90% (+5%) repolarization, and AP triangulation (+79%). In the telemetered conscious male dog, etamicastat (up to 20mg/kg) had no effects on arterial blood pressure, heart rate and the PR interval. At 10 and 20mg/kg, the QTc interval was slightly prolonged (8-9% max, P<0.05). No arrhythmia or other changes in the morphology of the ECG were observed. The maximum observed plasma concentrations (Cmax) of etamicastat (i.e. 3h post-administration) were 1.4 and 3.7µg/ml at 10 and 20mg/kg, respectively. No deleterious effects, including ECG disturbance were observed in male and female dogs dosed by gavage with etamicastat (up to 20mg/kg/day) for 28 days. Mean plasma Cmax etamicastat levels ranged between 2.4 and 6.3µg/ml on Day 1 and Day 28 of treatment, respectively. It is concluded that the blockade of the delayed rectifier potassium channels by etamicastat together with the QTc interval prolongation observed in conscious dogs can be considered as modest with respect to the measured plasmatic concentrations. These findings suggest that etamicastat is not likely to prolong the QT interval at therapeutic doses (~0.2µg/ml).


Subject(s)
Benzopyrans/adverse effects , Dopamine beta-Hydroxylase/antagonists & inhibitors , Enzyme Inhibitors/adverse effects , Imidazoles/adverse effects , Purkinje Fibers/drug effects , Safety , Action Potentials/drug effects , Administration, Oral , Animals , Benzopyrans/administration & dosage , Benzopyrans/pharmacokinetics , Dogs , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Female , HEK293 Cells , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacokinetics , Male , Purkinje Fibers/physiology , Telemetry
19.
Eur J Pharmacol ; 751: 50-8, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25641750

ABSTRACT

The interaction of etamicastat, a novel peripherally acting dopamine-ß-hydroxylase (DBH) inhibitor, with the enzyme was studied using a classical kinetic approach and the pharmacodynamics effect of the compound upon administration to rats was also evaluated. SK-N-SH cell homogenates convert tyramine into octopamine with a Km value of 9 mM, and a Vmax of 1747 nmol/mg protein/h. The K(m) value for ascorbate was 3 mM. The inhibition of DBH by etamicastat and nepicastat, a known centrally acting DBH inhibitor, with IC50 values of 107 and 40 nM, respectively, was fully reversed by dilution. Non-linear fitting of the velocities, determined at various concentrations of substrate (tyramine) and co-substrate (ascorbic acid), and of etamicastat and nepicastat, indicated that the inhibition of DBH by both compounds follows a mixed-model inhibition mechanism, approaching competitive behavior with regards to the substrate tyramine, with K(i) values of 34 and 11 nM, respectively. Relatively to ascorbate, both compounds followed a mixed-model inhibition mechanism, approaching uncompetitive behavior. Oral administration of both compounds (at 30 mg/kg) inhibited adrenal DBH activity over time and significantly decreased noradrenaline levels in the heart. Nepicastat also decreased noradrenaline levels in the parietal cortex, but not etamicastat. Both compounds significantly decreased systolic and diastolic blood pressure in spontaneously hypertensive rats. In conclusion, etamicastat and nepicastat behave as multisubstrate DBH inhibitors, binding reversibly and preferentially to the reduced form of the enzyme, and simultaneously at the substrate and oxygen binding sites. Etamicastat, in contrast to nepicastat, offers the advantage of peripheral selectivity without central effects.


Subject(s)
Benzopyrans/metabolism , Benzopyrans/pharmacology , Dopamine beta-Hydroxylase/metabolism , Imidazoles/metabolism , Imidazoles/pharmacology , Thiones/metabolism , Adrenal Glands/drug effects , Adrenal Glands/enzymology , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Antihypertensive Agents/pharmacology , Benzopyrans/chemistry , Cell Line , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dopamine beta-Hydroxylase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/chemistry , Kinetics , Male , Models, Molecular , Protein Binding , Protein Conformation , Rats , Rats, Inbred SHR , Rats, Wistar
20.
Article in English | MEDLINE | ID: mdl-25602710

ABSTRACT

In the present study, we tested the hypothesis that the potent and selective dopamine-ß-hydroxylase (DßH) inhibitor nepicastat would have minimal effects on cardiovascular and pharmacokinetic parameters associated with cocaine administration and would reduce the positive subjective effects produced by cocaine. We conducted a double-blind, placebo-controlled, inpatient study of oral nepicastat (0, 80 and 160mg) concurrent with intravenous (IV) cocaine (0, 10, 20 and 40mg) in non-treatment seeking participants who metcriteria for cocaine use disorder. Safety analyses revealed that nepicastat was well-tolerated and there were no differences in adverse events observed after nepicastat plus cocaine vs. cocaine alone. In addition, the pharmacokinetic properties of cocaine administration were not altered by nepicastat treatment. Cocaine-induced cardiovascular and subjective effects were evaluated for completers in the cohort randomized to nepicastat (n=13) using a within-subjects statistical analysis strategy. Specifically, the cardiovascular and subjective effects of cocaine were assessed in the presence of placebo (0mg), 80mg of nepicastat or 160mg of nepicastat on study Days 4, 8 and 12, respectively. Analyses revealed a main effect of nepicastat to reduce several cocaine-induced positive subjective effects. Taken together, these data indicate that nepicastat is safe when co-administered with cocaine and may suppress its positive subjective effects, and may be viable as a pharmacotherapy for treatment of cocaine use disorder.


Subject(s)
Cocaine-Related Disorders/drug therapy , Dopamine beta-Hydroxylase/metabolism , Enzyme Inhibitors/therapeutic use , Imidazoles/therapeutic use , Thiones/therapeutic use , Adult , Analysis of Variance , Cardiovascular System/drug effects , Cocaine-Related Disorders/blood , Dopamine beta-Hydroxylase/antagonists & inhibitors , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Inhibitors/blood , Female , Follow-Up Studies , Humans , Imidazoles/blood , Male , Pain Measurement , Psychiatric Status Rating Scales , Reinforcement, Psychology , Thiones/blood
SELECTION OF CITATIONS
SEARCH DETAIL