Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 285
Filter
1.
Acta Pharmacol Sin ; 41(3): 293-302, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31316182

ABSTRACT

Nav1.7 channels are mainly distributed in the peripheral nervous system. Blockade of Nav1.7 channels with small-molecule inhibitors in humans might provide pain relief without affecting the central nervous system. Based on the facts that many reported Nav1.7-selective inhibitors contain aryl sulfonamide fragments, as well as a tricyclic antidepressant, maprotiline, has been found to inhibit Nav1.7 channels, we designed and synthesized a series of compounds with ethanoanthracene and aryl sulfonamide moieties. Their inhibitory activity on sodium channels were detected with electrophysiological techniques. We found that compound 10o potently inhibited Nav1.7 channels stably expressed in HEK293 cells (IC50 = 0.64 Ā± 0.30 nmol/L) and displayed a high Nav1.7/Nav1.5 selectivity. In mouse small-sized dorsal root ganglion neurons, compound 10o (10, 100 nmol/L) dose-dependently decreased the sodium currents and dramatically suppressed depolarizing current-elicited neuronal discharge. Preliminary in vivo experiments showed that compound 10o possessed good analgesic activity: in a mouse visceral pain model, administration of compound 10o (30-100 mg/kg, i.p.) effectively and dose-dependently suppressed acetic acid-induced writhing.


Subject(s)
Analgesics/pharmacology , Drug Discovery , Maprotiline/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Sodium Channel Blockers/pharmacology , Sulfonamides/pharmacology , Acetic Acid , Analgesics/administration & dosage , Analgesics/chemistry , Animals , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Injections, Intraperitoneal , Male , Maprotiline/administration & dosage , Maprotiline/chemistry , Mice , Mice, Inbred ICR , Molecular Structure , Pain/chemically induced , Pain Measurement , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/chemistry , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/chemistry
2.
Bull Exp Biol Med ; 165(5): 597-601, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30225714

ABSTRACT

Single injection of muscarinic cholinoceptor blocker atropine (1 mg/kg) to outbred male rats reduced Ɵ-adrenergic responsiveness of erythrocytes (by 2.2 times) and the content of epinephrine granules on erythrocytes (by 1.5 times), significantly increased HR and rigidity of the heart rhythm, and manifold decreased the power of all spectral components of heart rhythm variability. Stimulation of the central neurotransmitter systems increased Ɵ-adrenergic responsiveness of erythrocytes (by 15-26%), decreased the number of epinephrine granules on erythrocytes (by 25-40%), and increased HR and cardiac rhythm intensity. These changes were most pronounced after stimulation of the serotoninergic system. Administration of atropine against the background of activation of central neurotransmitter systems did not decrease Ɵ-adrenergic responsiveness of erythrocytes (this parameter remained at a stably high level and even increased during stimulation of the dopaminergic system), but decreased the number of epinephrine granules on erythrocytes, increased HR, and dramatically decreased the power of all components of heart rhythm variability spectrum. The response to atropine was maximum against the background of noradrenergic system activation and less pronounced during stimulation of the serotoninergic system. Thus, substances that are complementary to cholinergic receptors modulated adrenergic effect on the properties of red blood cells, which, in turn, can modulate the adrenergic influences on the heart rhythm via the humoral channel of regulation. Stimulation of central neurotransmitter systems that potentiates the growth of visceral adrenergic responsiveness weakens the cholinergic modulation of the adrenergic influences, especially with respect to erythrocyte responsiveness. Hence, changes in the neurotransmitter metabolism in the body can lead to coupled modulation of reception and reactivity to adrenergic- and choline-like regulatory factors at the level of erythrocyte membranes, which can be important for regulation of heart rhythm.


Subject(s)
Atropine/pharmacology , Erythrocytes/drug effects , Heart Rate/drug effects , Heart/drug effects , Muscarinic Antagonists/pharmacology , Receptors, Muscarinic/metabolism , Acetylcholine/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-Agonists/pharmacology , Animals , Animals, Outbred Strains , Cardiotonic Agents/pharmacology , Cholinergic Agonists/pharmacology , Dopamine/pharmacology , Epinephrine/pharmacology , Erythrocytes/metabolism , Heart/physiology , Heart Rate/physiology , Maprotiline/pharmacology , Norepinephrine/pharmacology , Rats
3.
Int J Mol Sci ; 18(9)2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28895911

ABSTRACT

Autophagy is an essential catabolic program that forms part of the stress response and enables cells to break down their own intracellular components within lysosomes for recycling. Accumulating evidence suggests that autophagy plays vital roles in determining pathological outcomes of immune responses and tumorigenesis. Autophagy regulates innate and adaptive immunity affecting the pathologies of infectious, inflammatory, and autoimmune diseases. In cancer, autophagy appears to play distinct roles depending on the context of the malignancy by either promoting or suppressing key determinants of cancer cell survival. This review covers recent developments in the understanding of autophagy and discusses potential therapeutic interventions that may alter the outcomes of certain diseases.


Subject(s)
Autophagy/immunology , Autophagy/physiology , Immune System Diseases/immunology , Infections/immunology , Neoplasms/immunology , Adaptive Immunity/immunology , Animals , Autoimmune Diseases/immunology , Autophagy/drug effects , Benzylisoquinolines/pharmacology , Cholecalciferol/pharmacology , Humans , Immunity, Innate/immunology , Indoles , Isoquinolines/pharmacology , Lysosomes/metabolism , Maprotiline/pharmacology , Metformin/pharmacology , Phenols/pharmacology , Pyrroles/pharmacology , Resveratrol , Sirolimus/pharmacology , Spermidine/pharmacology , Stilbenes/pharmacology , Tetrahydroisoquinolines/pharmacology , Trehalose/pharmacology
4.
Bull Exp Biol Med ; 163(3): 302-306, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28744642

ABSTRACT

The effects of intraperitoneal DSP-4 (N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine, a noradrenergic neurotoxin) and maprotiline (an inhibitor of norepinephrine reuptake in synapses) on spectral components of heart rhythm variability were examined in outbred male and female rats treated with these agents in daily doses of 10 mg/kg for 3 days. At rest, DSP-4 elevated LF and VLF spectral components in male and female rats. Maprotiline elevated LF and VLF components in males at rest, increased HR and reduced all spectral components in resting females. Stress against the background of DSP-4 treatment sharply increased heart rate and reduced the powers of all spectral components (especially LF and VLF components). In maprotiline-treated rats, stress increased the powers of LF and VLF components. Thus, the central noradrenergic system participates in the formation of LF and VLF spectral components of heart rate variability at rest and especially during stressful stimulation, which can determine the phasic character of changes in the heart rate variability observed in stressed organism.


Subject(s)
Benzylamines/pharmacology , Heart Rate/drug effects , Heart/drug effects , Maprotiline/pharmacology , Neurotoxins/pharmacology , Neurotransmitter Uptake Inhibitors/pharmacology , Adrenergic Neurons/cytology , Adrenergic Neurons/drug effects , Animals , Animals, Outbred Strains , Drug Administration Schedule , Electrocardiography , Female , Heart/physiopathology , Injections, Intraperitoneal , Male , Rats , Rest , Stress, Physiological
5.
Molecules ; 21(1): 61, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26742028

ABSTRACT

The synthesis of the tetracyclic compounds 1-(4,5-dichloro-9,10-dihydro-9,10-ethanoanthracen-11-yl)-N-methylmethanamine (5) and 1-(1,8-dichloro-9,10-dihydro-9,10-ethanoanthracen-11-yl)-N-methylmethanamine (6) as a homologue of the anxiolytic and antidepressant drugs benzoctamine and maprotiline were described. The key intermediate aldehydes (3) and (4) were successfully synthesized via a [4 + 2] cycloaddition between acrolein and 1,8-dichloroanthracene. The synthesized compounds were investigated for antidepressant activity using the forced swimming test. Compounds (5), (6) and (3) showed significant reduction in the mice immobility indicating significant antidepressant effects. These compounds significantly reduced the immobility times at a dose 80 mg/kg by 84.0%, 86.7% and 71.1% respectively.


Subject(s)
Anthracenes/chemical synthesis , Anti-Anxiety Agents/chemical synthesis , Antidepressive Agents/chemical synthesis , Hydrocarbons, Chlorinated/chemical synthesis , Animals , Anthracenes/chemistry , Anthracenes/pharmacology , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacology , Hydrocarbons, Chlorinated/chemistry , Hydrocarbons, Chlorinated/pharmacology , Immobility Response, Tonic/drug effects , Maprotiline/chemistry , Maprotiline/pharmacology , Mice , Molecular Structure , Swimming
6.
Mol Pain ; 11: 47, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26260446

ABSTRACT

BACKGROUND: Although alterations in not only the pain sensitivity but also the analgesic effects of opioids have been reported under conditions of stress, the influence of unpredictable chronic mild stress (UCMS) on the antinociceptive effects of opioid analgesics remains to be fully investigated. The present study examined the influence of UCMS on the thermal pain sensitivity and antinociceptive effects of two opioid analgesics, morphine (an agonist of opioid receptors) and tramadol (an agonist of Āµ-opioid receptor and an inhibitor of both noradrenaline and serotonin transporters). We also examined the effects of pretreatment with maprotiline (a noradrenaline reuptake inhibitor) and escitalopram (a serotonin reuptake inhibitor) on the antinociceptive action of morphine in mice under an UCMS condition. RESULTS: Unpredictable chronic mild stress did not affect the basal thermal pain sensitivity in a mouse hot-plate test. Although morphine dose-dependently induced thermal antinociceptive effects under both the UCMS and non-stress conditions, the thermal antinociceptive effect of 3 mg/kg morphine under the UCMS condition was significantly lower than under the non-stressed condition. Unlike the case with morphine, we observed no significant difference in the thermal antinociceptive effect of tramadol between the UCMS and non-stress conditions. Furthermore, the reduced thermal antinociceptive effect of 3 mg/kg morphine under the UCMS condition was significantly ameliorated by pretreatment with 10 mg/kg maprotiline but not 3 mg/kg escitalopram. Pretreatment with neither maprotiline nor escitalopram alone was associated with an antinociceptive effect under either condition. CONCLUSIONS: We demonstrated that the antinociceptive effect of morphine but not tramadol was reduced in mice that had experienced UCMS. The reduced antinociceptive effect of morphine under the UCMS condition was ameliorated by pretreatment with maprotiline but not escitalopram. These results suggest that the reduced antinociceptive effects of morphine under conditions of chronic stress may be ameliorated by activation of the noradrenergic but not the serotonergic system.


Subject(s)
Analgesics/pharmacology , Morphine/pharmacology , Norepinephrine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Physiological/drug effects , Animals , Chronic Disease , Citalopram/pharmacology , Disease Models, Animal , Male , Maprotiline/pharmacology , Mice, Inbred BALB C , Temperature , Tramadol/pharmacology
7.
Int J Neuropsychopharmacol ; 15(8): 1087-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21835087

ABSTRACT

There is increasing interest in the pathophysiology and neurochemistry of the prefrontal cortex (PFC) in depression. Blood flow and metabolism are decreased in the PFC of patients with depression compared to controls. Changes in long-chain polyunsaturated fatty acids (PUFAs) are also associated with depression. This study was conducted to elucidate a possible role of PFC activity of an enzyme involved in the release of docosahexaenoic acid (DHA), i.e. calcium-independent phospholipase A2 (iPLA2), in the effects of the norepinephrine reuptake inhibitor (NRI) antidepressant, maprotiline, in mice. Treatment of Balb/C mice with maprotiline for 4 wk resulted in reduction in the level of behavioural despair, as determined by decreased immobility and increased climbing during the forced swim test. In contrast, mice treated with maprotiline plus bilateral prefrontal cortical injections of antisense oligonucleotide to iPLA2, showed significantly increased immobility and decreased climbing, to levels comparable to saline-treated controls, indicating abolishment of the antidepressant-like effect of maprotiline. Lipidomic analyses showed significant decreases in phosphatidylcholine species containing long-chain PUFAs and increases in lysophosphatidylcholine after maprotiline treatment, indicating increased PLA2 activity and endogenous release of eicosapentaenoic acid (EPA) or DHA after maprotiline treatment. These changes in lipid profiles were absent in mice that received maprotiline and PFC injections of antisense oligonucleotide to iPLA2. Together, the results indicate that PFC iPLA2 activity plays an important role in the antidepressant-like effect of maprotiline, possibly through endogenous release of long-chain PUFAs.


Subject(s)
Maprotiline/pharmacology , Phospholipases A2, Calcium-Independent/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Animals , Antidepressive Agents, Second-Generation , Calcium/metabolism , Cell Death/drug effects , Chromatography, High Pressure Liquid , Freezing Reaction, Cataleptic/drug effects , In Situ Nick-End Labeling , Indoles , Lipid Metabolism/drug effects , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides, Antisense/pharmacology , Swimming/psychology
8.
Nat Commun ; 13(1): 6796, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36357388

ABSTRACT

When the protein or calcium homeostasis of the endoplasmic reticulum (ER) is adversely altered, cells experience ER stress that leads to various diseases including neurodegeneration. Genetic deletion of an ER stress downstream effector, CHOP, significantly protects neuron somata and axons. Here we report that three tricyclic compounds identified through a small-scale high throughput screening using a CHOP promoter-driven luciferase cell-based assay, effectively inhibit ER stress by antagonizing their common target, histamine receptor H1 (HRH1). We further demonstrated that systemic administration of one of these compounds, maprotiline, or CRISPR-mediated retinal ganglion cell (RGC)-specific HRH1 inhibition, delivers considerable neuroprotection of both RGC somata and axons and preservation of visual function in two mouse optic neuropathy models. Finally, we determine that maprotiline restores ER homeostasis by inhibiting HRH1-mediated Ca2+ release from ER. In this work we establish maprotiline as a candidate neuroprotectant and HRH1 as a potential therapeutic target for glaucoma.


Subject(s)
Maprotiline , Retinal Ganglion Cells , Mice , Animals , Retinal Ganglion Cells/metabolism , Maprotiline/metabolism , Maprotiline/pharmacology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Disease Models, Animal , Homeostasis , Receptors, Histamine/metabolism
9.
Exp Clin Endocrinol Diabetes ; 130(9): 596-603, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35320846

ABSTRACT

Maprotiline is an antidepressant that has been found to cause hypoglycemia. However, the effect of maprotiline on diabetic nephropathy (DN) has not been investigated. Here, we explored the effect of maprotiline on human renal glomerular endothelial cells (HRGECs) in response to high glucose (HG) stimulation. We found that maprotiline attenuated HG-induced oxidative stress in HRGECs with decreased reactive oxygen species production and increased superoxide dismutase activity. Maprotiline repressed the HG-induced expression of cyclooxygenases 2 at both mRNA and protein levels in HRGECs. The increased thromboxane B2 level and decreased 6-keto-prostaglandin F1α level induced by HG were significantly attenuated by maprotiline treatment. Maprotiline also prevented the HG-induced increase in the permeability of HRGECs and the decrease in the zonula occludens-1 expression and downregulated HG-induced increase in the expression of protein kinase C-α (PKC-α) in HRGECs. This protective effect of maprotiline on HG-induced HRGECs dysfunction was abolished by overexpression of PKC-α. In conclusion, maprotiline displayed a protective effect on HG-challenged HRGECs, which was mediated by the regulation of PKC-α. These findings provide further evidence for the potential use of maprotiline for the treatment of DN.


Subject(s)
Diabetic Nephropathies , Endothelial Cells , Cells, Cultured , Diabetic Nephropathies/metabolism , Endothelial Cells/metabolism , Glucose/pharmacology , Humans , Kidney Glomerulus/metabolism , Maprotiline/metabolism , Maprotiline/pharmacology , Oxidative Stress , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandin-Endoperoxide Synthases/pharmacology , Protein Kinase C-alpha/metabolism , Protein Kinase C-alpha/pharmacology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase/pharmacology , Thromboxane B2/metabolism , Thromboxane B2/pharmacology
10.
Int J Cancer ; 128(7): 1712-23, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-20503272

ABSTRACT

Resistance to chemotherapy is a major obstacle for the success of cancer therapy and is most commonly attributed to the inability of cancer cells to die by apoptosis, the archetypal programed cell death (PCD) response. The development of anticancer drugs that can overcome this resistance to apoptosis and induce other forms of cell death is therefore paramount for efficient cancer therapy. We report that the antidepressants maprotiline and fluoxetine induce autophagic PCD in the chemoresistant Burkitt's lymphoma (BL) cell line DG-75, which does not involve caspases, DNA fragmentation or PARP cleavage, but is associated with the development of cytoplasmic vacuoles, all consistent with an autophagic mode of PCD. Autophagic PCD was confirmed by transmission electron microscopy, upregulation of Beclin-I and the extent of PCD being reduced by the autophagic inhibitor 3-MA. In contrast, these compounds induced apoptotic PCD in the biopsy-like chemosensitive BL MUTU-I cell line. We provide evidence that the chemoresistant DG-75 cells do not express the proapoptotic Bcl-2 proteins Bax and Bak, show diminished levels of stored intracellular calcium and display shortened rod-like mitochondria, all of which are known to be associated with a defective "apoptotic" response in cancer cells. PCD in the two cell lines has different Ca(2+) responses to maprotiline and fluoxetine, which may also account for their differential PCD responses. Our study, therefore, supports a new mechanistic role for maprotiline and fluoxetine as novel proautophagic agents in the treatment of resistant BL, and thus an alternative therapeutic application for these compounds.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Apoptosis , Burkitt Lymphoma/drug therapy , Fluoxetine/pharmacology , Maprotiline/pharmacology , Autophagy/drug effects , Calcium/chemistry , Cell Death , Cell Line, Tumor , Cell Separation , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Microscopy, Confocal/methods , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
11.
Article in Japanese | MEDLINE | ID: mdl-21409840

ABSTRACT

Substantial evidence indicates that brain neurons containing and secreting norepinephrine (NE) and corticotrophin-releasing hormone (CRH) are activated during stress. The acoustic startle reflex (ASR) can be enhanced by CRH neuronal activity in the central nucleus of the amygdala. Our previous study demonstrates an augmentation of the footshock-induced ASR (f-ASR) 1 day after chronic variable stress (CVS) for 13 days. In this study, to evaluate a long-term neural plasticity in NE-CRH systems after CVS, we examined f-ASR 1, 8 or 15 days after CVS. The augmented magnitude of the f-ASR 15 day after CVS was potentiated and delayed compared with that 1 day after CVS. The delayed augmentation of f-ASR was inhibited by repeated treatment with desipramine, maprotiline or paroxetine for 14 days after CVS. A single treatment with any antidepressant agent had no influence the f-ASR while a marked inhibition by a single dose of alprazolam, CRH1-receptor antagonist, prazosin and propranolol was observed. The decreased tyrosine hydroxylase activity in the locus coeruleus and the beta-adrenoceptor down-regulation in the amygdaloid complex might be involved in the inhibiton of the delayed augmentation of f-ASR by repeated antidepressant treatment, leading to the possibility that the delayed sensitization of CRH response to stress after CVS might contribute to the biological mechanism underlying the formation of pathological states such as anxiety and depressive disorders.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Reflex, Acoustic/physiology , Reflex, Startle/physiology , Stress, Psychological/physiopathology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Desipramine/pharmacology , Male , Maprotiline/pharmacology , Norepinephrine/physiology , Paroxetine/pharmacology , Rats , Rats, Wistar , Receptors, Corticotropin/antagonists & inhibitors , Reflex, Acoustic/drug effects , Reflex, Startle/drug effects , Time Factors
12.
Bioengineered ; 12(2): 12332-12344, 2021 12.
Article in English | MEDLINE | ID: mdl-34895041

ABSTRACT

Isoflurane-induced neurotoxicity has attracted much interest. Recent studies suggest that isoflurane causes microglial activation, resulting in an inflammatory response and microglial insult. Maprotiline is a novel drug that has been licensed as an antidepressant with considerable anti-inflammatory activity. However, it is still unknown whether maprotiline possesses a protective effect against isoflurane-induced microglial insult. Here, we found that maprotiline ameliorated isoflurane-caused reduction in BV2 microglial cell viability and lactate dehydrogenase (LDH) release. Maprotiline mitigated isoflurane-induced oxidative stress by inhibiting reactive oxygen species (ROS) production and increasing superoxide dismutase (SOD) activity. Isoflurane-induced expression and production of inflammatory markers including tumor necrosis factor (TNF-α), interleukin (IL)-1Ɵ, cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2) were decreased in maprotiline-treated cells. Maprotiline inhibited the mRNA and protein levels of Iba1, a marker of microglial activation, in isoflurane-induced BV2 cells. Maprotiline treatment restored isoflurane-induced reduction of TREM2 in BV2 microglial cells. In addition, the knockdown of TREM2 abolished the beneficial effects of maprotiline against isoflurane. Collectively, maprotiline exerted protective effects against isoflurane-caused oxidative stress, inflammatory response, and cell injury via regulating TREM2. These findings show that maprotiline prevented the isoflurane-induced microglial activation, indicating that maprotiline might be used as an optimal therapeutic agent for preventing the isoflurane-caused neurotoxicity.


Subject(s)
Isoflurane/pharmacology , Maprotiline/pharmacology , Membrane Glycoproteins/metabolism , Microglia/drug effects , Myeloid Cells/drug effects , Receptors, Immunologic/metabolism , Animals , Biomarkers/metabolism , Cell Survival/drug effects , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Inflammation/metabolism , L-Lactate Dehydrogenase/metabolism , Mice , Microglia/metabolism , Myeloid Cells/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism
13.
BMC Neurosci ; 11: 10, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20105322

ABSTRACT

BACKGROUND: Recent studies demonstrate that diverse antidepressant agents increase the cellular production of the nucleolipid CDP-diacylglycerol and its synthetic derivative, phosphatidylinositol, in depression-relevant brain regions. Pharmacological blockade of downstream phosphatidylinositide signaling disrupted the behavioral antidepressant effects in rats. However, the nucleolipid responses were resistant to inhibition by serotonin receptor antagonists, even though antidepressant-facilitated inositol phosphate accumulation was blocked. Could the neurochemical effects be additional to the known effects of the drugs on monoamine transmitter transporters? To examine this question, we tested selected agents in serotonin-depleted brain tissues, in PC12 cells devoid of serotonin transporters, and on the enzymatic activity of brain CDP-diacylglycerol synthase - the enzyme that catalyzes the physiological synthesis of CDP-diacylglycerol. RESULTS: Imipramine, paroxetine, and maprotiline concentration-dependently increased the levels of CDP-diacylglycerol and phosphatidylinositides in PC12 cells. Rat forebrain tissues depleted of serotonin by pretreatment with p-chlorophenylalanine showed responses to imipramine or maprotiline that were comparable to respective responses from saline-injected controls. With fluoxetine, nucleolipid responses in the serotonin-depleted cortex or hippocampus were significantly reduced, but not abolished. Each drug significantly increased the enzymatic activity of CDP-diacylglycerol synthase following incubations with cortical or hippocampal brain tissues. CONCLUSION: Antidepressants probably induce the activity of CDP-diacylglycerol synthase leading to increased production of CDP-diacylglycerol and facilitation of downstream phosphatidylinositol synthesis. Phosphatidylinositol-dependent signaling cascades exert diverse salutary effects in neural cells, including facilitation of BDNF signaling and neurogenesis. Hence, the present findings should strengthen the notion that modulation of brain phosphatidylinositide signaling probably contributes to the molecular mechanism of diverse antidepressant medications.


Subject(s)
Antidepressive Agents/pharmacology , Biogenic Monoamines/antagonists & inhibitors , Cytidine Diphosphate Diglycerides/biosynthesis , Imipramine/pharmacology , Maprotiline/pharmacology , Paroxetine/pharmacology , Animals , Antidepressive Agents/administration & dosage , Biogenic Monoamines/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cytidine Diphosphate Diglycerides/metabolism , Diacylglycerol Cholinephosphotransferase/metabolism , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Imipramine/administration & dosage , Male , Maprotiline/administration & dosage , Mice , Mice, Inbred C57BL , PC12 Cells , Paroxetine/administration & dosage , Rats , Rats, Sprague-Dawley , Serotonin/deficiency , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/deficiency , Serotonin Plasma Membrane Transport Proteins/metabolism
14.
Article in English | MEDLINE | ID: mdl-20160205

ABSTRACT

This manuscript reviews the preclinical in vitro, ex vivo, and nonhuman in vivo effects of psychopharmacological agents in clinical use on cell physiology with a view toward identifying agents with neuroprotective properties in neurodegenerative disease. These agents are routinely used in the symptomatic treatment of neurodegenerative disease. Each agent is reviewed in terms of its effects on pathogenic proteins, proteasomal function, mitochondrial viability, mitochondrial function and metabolism, mitochondrial permeability transition pore development, cellular viability, and apoptosis. Effects on the metabolism of the neurodegenerative disease pathogenic proteins alpha-synuclein, beta-amyloid, and tau, including tau phosphorylation, are particularly addressed, with application to Alzheimer's and Parkinson's diseases. Limitations of the current data are detailed and predictive criteria for translational clinical neuroprotection are proposed and discussed. Drugs that warrant further study for neuroprotection in neurodegenerative disease include pramipexole, thioridazine, risperidone, olanzapine, quetiapine, lithium, valproate, desipramine, maprotiline, fluoxetine, buspirone, clonazepam, diphenhydramine, and melatonin. Those with multiple neuroprotective mechanisms include pramipexole, thioridazine, olanzapine, quetiapine, lithium, valproate, desipramine, maprotiline, clonazepam, and melatonin. Those best viewed circumspectly in neurodegenerative disease until clinical disease course outcomes data become available, include several antipsychotics, lithium, oxcarbazepine, valproate, several tricyclic antidepressants, certain SSRIs, diazepam, and possibly diphenhydramine. A search for clinical studies of neuroprotection revealed only a single study demonstrating putatively positive results for ropinirole. An agenda for research on potentially neuroprotective agent is provided.


Subject(s)
Brain/drug effects , Brain/physiopathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Antioxidants/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Clonazepam/pharmacology , Clonazepam/therapeutic use , Desipramine/pharmacology , Desipramine/therapeutic use , Dibenzothiazepines/pharmacology , Dibenzothiazepines/therapeutic use , Dopamine Agonists/pharmacology , Dopamine Agonists/therapeutic use , Humans , Lithium Carbonate/pharmacology , Lithium Carbonate/therapeutic use , Maprotiline/pharmacology , Maprotiline/therapeutic use , Melatonin/therapeutic use , Neurodegenerative Diseases/metabolism , Olanzapine , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Pramipexole , Quetiapine Fumarate , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
15.
Int J Neuropsychopharmacol ; 12(7): 953-64, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19203412

ABSTRACT

Recently, there has been considerable interest in a possible link between changes in brain polyunsaturated fatty acids, neural membrane phospholipid degradation, serotonergic neurotransmission, and depression. The present study aims to examine effects of antidepressants on lipids in different regions of the brain at individual molecular species level, using the novel technique of lipidomics. Balb/C mice received daily intraperitoneal (i.p.) injections of 10 mg/kg of the antidepressants maprotiline, fluoxetine and paroxetine for 4 wk. The prefrontal cortex, hippocampus, striatum and cerebellum were harvested, and lipid profiles compared to those of saline-injected mice. Treatment with maprotiline and paroxetine, but not fluoxetine, resulted in significant decreases in phosphatidylcholine (PC) species, PC36:1, PC38:3, PC40:2p, PC40:6, PC40:5, PC42:7p, PC42:6p and PC42:5p in the prefrontal neocortex. The decreases in phospholipids were accompanied by increases in lysophospholipid species, lysoPC16:0, lysoPC18:2 and lysoPC18:0 in the prefrontal cortex, indicating increase in phospholipase A2 activity and possible release of long-chain fatty acids. Maprotiline and paroxetine treatment also resulted in decreases in sphingomyelin and increases in several ceramide species in the prefrontal cortex. It is postulated that endogenous release of long-chain fatty acids may be related to the mechanism of action of maprotiline and paroxetine.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Fatty Acids/metabolism , Fluoxetine/pharmacology , Maprotiline/pharmacology , Paroxetine/pharmacology , Animals , Antidepressive Agents/administration & dosage , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Brain/metabolism , Ceramides/metabolism , Cerebellum/drug effects , Cerebellum/metabolism , Chromatography, High Pressure Liquid , Fluoxetine/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Injections, Intraperitoneal , Lysophospholipids/metabolism , Male , Maprotiline/administration & dosage , Mice , Mice, Inbred BALB C , Paroxetine/administration & dosage , Phosphatidylcholines/metabolism , Phospholipases A2/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Spectrometry, Mass, Electrospray Ionization , Sphingomyelins/metabolism , Time Factors
16.
J Psychopharmacol ; 23(2): 143-56, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18515452

ABSTRACT

The clinical syndromes of anxiety and depression are now thought to exist along a temporal continuum and this construct has been modelled in a preclinical setting in chicks separated from conspecifics. This research sought to further the validity of the chick anxiety-depression continuum model. Dose-response studies using two classes of anxiolytics (chlordiazepoxide: 2.5, 5.0, 10.0, 15.0 mg/kg, and clonidine: 0.1, 0.15, 0.2, 0.25 mg/kg) and three classes of antidepressants (imipramine: 1.0, 3.0, 10.0, 15.0 mg/kg, maprotoline: 2.5, 5.0, 10.0, 20.0 mg/kg and fluoxetine: 1.0, 5.0, 10.0, 20.0 mg/kg) showed an ability to detect anxiolytic activity of chlordiazepoxide, clonidine, imipramine and maprotoline in the anxiety-like phase of the model and to detect antidepressant effects of imipramine, maprotoline and fluoxetine in the depression-like phase of the model. In addition, blood plasma interleukin-6, a biomarker of stress, was found to be elevated in response to social-separation stress. Collectively, these findings further characterize the model as a simulation of the anxiety-depression continuum and begin to establish the paradigm as a high-utility adjuvant to rodent screening assays for putative anxiolytic and antidepressant compounds.


Subject(s)
Anxiety/drug therapy , Anxiety/psychology , Depression/drug therapy , Depression/psychology , Animals , Animals, Newborn , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Chickens , Chlordiazepoxide/pharmacology , Clonidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Fluoxetine/pharmacology , Imipramine/pharmacology , Interleukin-6/blood , Maprotiline/pharmacology , Social Isolation/psychology , Stress, Psychological/drug therapy , Stress, Psychological/psychology
17.
Behav Brain Res ; 193(1): 140-3, 2008 Nov 03.
Article in English | MEDLINE | ID: mdl-18565601

ABSTRACT

There have been few comparisons of strains and antidepressants in the unpredictable chronic mild stress (UCMS) paradigm in mice. This study was undertaken to determine the influence of such factors using four antidepressants drugs including the tricyclics imipramine (20 mg/(kgday)) and desipramine (10 mg/(kgday)), the tetracyclic maprotiline (20 mg/(kgday)) and the selective serotonin reuptake inhibitor (SSRI) fluoxetine (10mg/(kgday)) in both Swiss and BALB/c mice. A 6-week UCMS regimen induced deterioration of the coat state and decreased grooming behaviours in the splash test in BALB/c mice but not Swiss mice. The four antidepressants reversed the UCMS-induced effects in BALB/c mice in both measures. However, imipramine and fluoxetine reached significance in the splash test while desipramine and maprotiline displayed only a trend. In conclusion, these results emphasize that BALB/c mice are more sensitive than Swiss mice for studying the effects of the UCMS model as well as for testing antidepressant-like properties.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Depression/drug therapy , Stress, Psychological/physiopathology , Animals , Animals, Outbred Strains , Antidepressive Agents/administration & dosage , Antidepressive Agents, Second-Generation/administration & dosage , Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Tricyclic/administration & dosage , Antidepressive Agents, Tricyclic/pharmacology , Anxiety/etiology , Anxiety/psychology , Chronic Disease , Depression/complications , Desipramine/administration & dosage , Desipramine/pharmacology , Fluoxetine/administration & dosage , Fluoxetine/pharmacology , Grooming/drug effects , Imipramine/administration & dosage , Imipramine/pharmacology , Injections, Intraperitoneal , Male , Maprotiline/administration & dosage , Maprotiline/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/pharmacology , Species Specificity , Stress, Psychological/etiology , Time Factors
18.
Prog Neuropsychopharmacol Biol Psychiatry ; 32(1): 135-9, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17825464

ABSTRACT

Agents such as clozapine, olanzapine and mirtazapine frequently trigger an increase in body weight. Though the mechanisms have not been thoroughly clarified, recent studies indicate a role for ghrelin in regulation of appetite and weight gain. We investigated the relation of maprotiline induced weight gain to serum ghrelin and adiponectin levels, as well as insulin resistance in lean subjects with depressive disorder. A total of 40 male lean subjects with depressive disorder were treated with maprotiline (150 mg/day) for 30-days. Clinical data, fasting plasma glucose, lipids, insulin levels, serum ghrelin and adiponectin concentrations were determined before and after treatment. Insulin resistance was estimated using the homeostasis model assessment (HOMA) formula. After 30 days of treatment with maprotiline, mean body mass index increased significantly. Blood ghrelin and insulin levels and HOMA indexes increased, and adiponectin concentration decreased (p<0.001, for all) after the treatment period. Changes in ghrelin levels correlated neither of the parameters tested; whereas decrease in plasma adiponectin was associated with an increase in BMI (r=-0.671, p<0.001). In conclusion, the results indicate that treatment of lean patients with depressive disorder with maprotiline results in an increase in serum ghrelin and reduction in adiponectin levels. Weight gain due to maprotiline treatment may be related to its negative effects on the metabolic variables.


Subject(s)
Adiponectin/blood , Antidepressive Agents, Second-Generation/pharmacology , Body Weight/drug effects , Depressive Disorder , Ghrelin/blood , Insulin Resistance/physiology , Maprotiline/pharmacology , Adult , Body Mass Index , Depressive Disorder/blood , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Humans , Insulin/blood , Male , Radioimmunoassay , Time Factors
19.
Neuro Endocrinol Lett ; 28(5): 614-20, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17984940

ABSTRACT

OBJECTIVE: To estimate the influence of long-term treatment with noradrenergic and serotonergic reuptake inhibitors on activity of pituitary-adrenocortical and sympatho-adrenomedullar systems in animals, we compared the effects of maprotiline (a selective inhibitor of noradrenaline reuptake) and fluxilan (a selective inhibitor of serotonin reuptake) on plasma noradrenaline (NA), adrenaline (A), adrenocorticotropic hormone (ACTH) and corticosterone (CORT) levels in unstressed control and rats exposed to chronic unpredictable mild stress (CUMS). METHODS: Plasma NA and A were assayed by a radioenzymatic method. Plasma CORT was measured using RIA kits and plasma ACTH by a chemiluminescent method. RESULTS: CUMS did not affect blood plasma NA, A and ACTH content, but elevated plasma CORT level. Maprotiline elevated plasma NA content both in unstressed control and CUMS group, whereas plasma A remained unchanged. Fluxilan acted significantly increasing plasma NA and A concentrations both in control and CUMS rats. Neither maprotiline nor fluxilan affected plasma ACTH level both in unstressed control and CUMS animals. Plasma CORT level in unstressed control rats remained unchanged after maprotiline and fluxilan treatment, while being significantly decreased in CUMS rats. CONCLUSION: Chronic treatment of adult rat males with maprotiline, a noradrenaline reuptake inhibitor activated sympathoneural system. On the other hand, fluxilan, a serotonin reuptake inhibitor activated both sympathoneural and adrenomedullar system, whereas both antidepressants desensitized HPA axis. The findings described here suggest that elevated plasma catecholamines my contribute to adverse effects of these drugs on cardiovascular parameters during antidepressant therapy.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Epinephrine/blood , Fluoxetine/pharmacology , Maprotiline/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/blood , Adrenal Medulla/drug effects , Adrenal Medulla/physiology , Adrenocorticotropic Hormone/blood , Analysis of Variance , Animals , Corticosterone/blood , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Male , Norepinephrine/blood , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Random Allocation , Rats , Rats, Wistar , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology
20.
Arch Pharm Res ; 30(4): 453-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17489361

ABSTRACT

Maprotiline, an atypical antidepressant, can induce prolonged QT and torsades de pointes. We studied the effects of maprotiline on human ether-a-go-go-related gene (HERG) channels expressed in Xenopus oocytes and HEK293 cells. Maprotiline induced a concentration-dependent decrease in current amplitudes at the end of the voltage steps and tail currents of HERG. The V1/2 values in the absence and presence of 1-20 microM maprotiline were not significantly different, while the values decreased according to the concentrations of the drug at 50-300 microM. The IC50 for a maprotiline block of HERG current in Xenopus oocytes did not change according to depolarization; 39.5 +/- 3.2 microM at -40 mV and 43.6 +/- 2.8 microM at +40 mV. The block of HERG by maprotiline was examined after treatment of trinitrobenzene sulfonic acid (TNBS), an amino-group reagent that neutralizes the positively charged amino-groups of peptide N-terminal and lysine residues. TNBS inhibited the change of V1/2 values induced by 50-300 mM maprotiline, and aggravated the drug-induced gmax decrease. The IC50 for the maprotiline-induced blockade of HERG currents in HEK293 cells at 36 degrees C was 0.13 microM at +20 mV. Our findings suggest that the arrhythmogenic side effects of maprotiline are caused by a blockade of HERG and possibly by a blockade of delayed rectifier K+ channel.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Maprotiline/pharmacology , Potassium Channel Blockers/pharmacology , Animals , ERG1 Potassium Channel , Female , Humans , Trinitrobenzenesulfonic Acid/pharmacology , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL