Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 144: 319-326, 2019 01.
Article in English | MEDLINE | ID: mdl-30408486

ABSTRACT

Early manifestations of Alzheimer's disease (AD) include neuroinflammation, disrupted neurotransmission and cognitive deficits. Impairment of the GABAergic system is essentially involved in the pathogenesis of AD. Traditionally, agonists of GABAA receptors at doses above 1 mg/kg are known to possess memory impairing effects. However, we have previously found that GABAA receptor GABA site ligand muscimol at very low doses acted contrary - enhanced spatial learning/memory, as well as prevented neuroinflammation and augmented neurotransmission in AD model rats. Therefore, in the present study we focused on the assessment of the effects of non-sedative - very low (0.05 mg/kg) and moderate (1 mg/kg) - doses of diazepam, a positive allosteric modulator of benzodiazepine site of GABAA receptors. Its effects on spatial learning/memory and brain proteins related to neuroinflammation (GFAP and Iba-1), synaptic plasticity (SYP1), as well as acetylcholine breakdown and GABA biosynthesis were studied. Non-transgenic AD model rats (intracerebroventricular streptozocin injection) were used with the aim to mimic the pre-dementia stage of AD in humans. The obtained data showed that diazepam at both doses protected against streptozocin induced detrimental effects by enhancing spatial learning/memory, preventing neuroinflammation, preserving synaptic plasticity, as well as normalizing the hippocampal and cortical protein expression related to acetylcholine breakdown and GABA biosynthesis. One may suggest that at low and moderate doses diazepam is targeting non-specific, probably allosteric GABAA receptor sites, thus leading to stimulatory effects that can be beneficial for diazepam use in early pre-dementia stages of AD.


Subject(s)
Alzheimer Disease/drug therapy , Diazepam/administration & dosage , Neuroprotective Agents/administration & dosage , Acetylcholine/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gliosis/drug therapy , Gliosis/metabolism , Gliosis/pathology , Glutamate Decarboxylase/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Memory Disorders/drug therapy , Memory Disorders/metabolism , Memory Disorders/pathology , Random Allocation , Rats, Wistar , Synaptophysin/metabolism , gamma-Aminobutyric Acid/metabolism
2.
Eur J Pharmacol ; 818: 381-399, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29133125

ABSTRACT

Recent studies devoted to neuroprotection have focused on the role of the gamma-aminobutyric acid (GABA) system in regulating neuroinflammatory processes which play a key role in the neurodegenerative processes observed in Alzheimer's disease (AD) by inducing glial cell overactivation and impairing neurotransmission. Data on the efficacy of classical GABA-A and GABA-B receptor agonists (muscimol and baclofen, respectively) in animal models of AD are not available. Moreover, no published studies have examined the ability of optimal doses of these compounds to prevent neuroinflammation, the alterations in neurotransmission and cognitive deficits. In the present study, we used a non-transgenic rat model of AD obtained by intracerebroventricular streptozocin (STZ) injection and assessed the effects of muscimol and baclofen at very low doses (0.01-0.05mg/kg) on spatial memory and the expression of cortical and hippocampal proteins related to neuroinflammation, namely proteins involved in astroglial functions (glial fibrillary acidic protein, GFAP), GABA synthesis (GABA synthesizing enzyme, glutamic acid decarboxylase 67, GAD67) and acetylcholine degradation (acetylcholine esterase). The presented study demonstrated that in a rat model of STZ-induced AD both muscimol and baclofen at the tested doses exerted memory-enhancing and anti-inflammatory effects, as well as normalization of acetylcholine esterase and GABA expression. We suggested that the function of very low doses of GABA receptor agonists differs from typical GABA-related inhibition and may be mediated by the allosteric sites of GABA receptors or other non-specific cell regulatory pathways.


Subject(s)
Alzheimer Disease/physiopathology , Baclofen/pharmacology , Brain/drug effects , Cognition/drug effects , Gene Expression Regulation/drug effects , Muscimol/pharmacology , Streptozocin/adverse effects , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Male , Memory/drug effects , Rats , Rats, Wistar , Spatial Learning/drug effects
3.
Eur J Pharmacol ; 745: 76-83, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25446926

ABSTRACT

The present study investigates the efficacy of mildronate, a carnitine congener, to protect stress and haloperidol-induced impairment of memory in rats and the expression of brain protein biomarkers involved in synaptic plasticity, such as brain-derived neurotrophic factor (BDNF), acetylcholine esterase and glutamate decarboxylase 67 (GAD67). Two amnesia models were used: 2h immobilization stress and 3-week haloperidol treatment. Stress caused memory impairment in the passive avoidance test and induced a significant 2-fold BDNF elevation in hippocampal and striatal tissues that was completely inhibited by mildronate. Mildronate decreased the level of GAD67 (but not acetylcholine esterase) expression by stress. Haloperidol decrease by a third hippocampal BDNF and acetylcholine esterase (but not GAD67) expression, which was normalized by mildronate; it also reversed the haloperidol-induced memory impairment in Barnes test. The results suggest the usefulness of mildronate as protector against neuronal disturbances caused by stress or haloperidol.


Subject(s)
Brain/drug effects , Memory/drug effects , Methylhydrazines/pharmacology , Acetylcholinesterase/metabolism , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Biomarkers/metabolism , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Carnitine/analogs & derivatives , Carnitine/pharmacology , GPI-Linked Proteins/metabolism , Glutamate Decarboxylase/metabolism , Haloperidol/toxicity , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Stress, Physiological
4.
Medicina (Kaunas) ; 49(7): 301-9, 2013.
Article in English | MEDLINE | ID: mdl-24375241

ABSTRACT

This review for the first time summarizes the data obtained in the neuropharmacological studies of mildronate, a drug previously known as a cardioprotective agent. In different animal models of neurotoxicity and neurodegenerative diseases, we demonstrated its neuroprotecting activity. By the use of immunohistochemical methods and Western blot analysis, as well as some selected behavioral tests, the new mechanisms of mildronate have been demonstrated: a regulatory effect on mitochondrial processes and on the expression of nerve cell proteins, which are involved in cell survival, functioning, and inflammation processes. Particular attention is paid to the capability of mildronate to stimulate learning and memory and to the expression of neuronal proteins involved in synaptic plasticity and adult neurogenesis. These properties can be useful in neurological practice to protect and treat neurological disorders, particularly those associated with neurodegeneration and a decline in cognitive functions.


Subject(s)
Adjuvants, Immunologic/pharmacology , Learning/drug effects , Methylhydrazines/pharmacology , Mitochondria/drug effects , Nerve Tissue Proteins/biosynthesis , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Disease Models, Animal , Humans , Mice , Mitochondria/metabolism , Nerve Regeneration/drug effects , Neuritis/metabolism , Neuritis/pathology , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Rats
5.
Behav Brain Res ; 256: 5-9, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23933157

ABSTRACT

The present study for the first time is devoted to identify central effects of synthetic lunasin, a 43 amino acid peptide. A markedly expressed neuroleptic/cataleptic effect was observed at low (0.1-10 nmol/mouse) centrally administered doses in male C57Bl/6 mice. Lunasin considerably reduced the amphetamine hyperlocomotion but weakly apomorphine climbing behaviour. No influence on ketamine and bicuculline effects was observed. Binding assay studies demonstrated modest affinity of lunasin for the dopamine D1 receptor (Ki=60 ± 15 µM). In a functional assay of cAMP accumulation on live cells lunasin antagonised apomorphine effect on D1 receptor activation (pEC50=6.1 ± 0.3), but had no effect in cells expressing D2 receptors. The obtained data suggest that lunasin's action at least in part is provided via dopaminergic D1 receptor pathways. However, other non-identified mechanisms (probably intracellular) may play an important role in lunasin's central action. Nevertheless further studies of lunasin are promising, particularly taking into account a necessity for novel type of antipsychotic drugs.


Subject(s)
Brain/drug effects , Central Nervous System Agents/pharmacology , Motor Activity/drug effects , Receptors, Dopamine D1/metabolism , Soybean Proteins/pharmacology , Amphetamine/pharmacology , Animals , Apomorphine/pharmacology , Bicuculline/adverse effects , Brain/physiology , Catalepsy/chemically induced , Catalepsy/drug therapy , Cyclic AMP/metabolism , Dopamine Agents/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , HEK293 Cells , Humans , Ketamine/pharmacology , Male , Mice, Inbred C57BL , Motor Activity/physiology , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/metabolism , Seizures/chemically induced , Seizures/drug therapy , Seizures/physiopathology , Soybean Proteins/administration & dosage
6.
BMC Neurosci ; 14: 54, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23672601

ABSTRACT

BACKGROUND: The vesicular B0AT3 transporter (SLC6A17), one of the members of the SLC6 family, is a transporter for neutral amino acids and is exclusively expressed in brain. Here we provide a comprehensive expression profile of B0AT3 in mouse brain using in situ hybridization and immunohistochemistry. RESULTS: We confirmed previous expression data from rat brain and used a novel custom made antibody to obtain detailed co-labelling with several cell type specific markers. B0AT3 was highly expressed in both inhibitory and excitatory neurons. The B0AT3 expression was highly overlapping with those of vesicular glutamate transporter 2 (VGLUT2) and vesicular glutamate transporter 1 (VGLUT1). We also show here that Slc6a17mRNA is up-regulated in animals subjected to short term food deprivation as well as animals treated with the serotonin reuptake inhibitor fluoxetine and the dopamine/noradrenaline reuptake inhibitor bupropion. CONCLUSIONS: This suggests that the B0AT3 transporter have a role in regulation of monoaminergic as well as glutamatergic synapses.


Subject(s)
Central Nervous System/physiology , Gene Expression Regulation/physiology , Nerve Tissue Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Animals , Antidepressive Agents/pharmacology , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/drug effects , Embryo, Mammalian , Female , Food Deprivation , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Pregnancy , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism
7.
Pharmacol Biochem Behav ; 106: 68-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23537732

ABSTRACT

Previously we demonstrated that mildronate [3-(2,2,2-trimethylhydrazinium) propionate dihydrate], a representative of the aza-butyrobetaine class of compounds, protects mitochondrial metabolism under conditions such as ischemia. Mildronate also acted as a neuroprotective agent in an azidothymidine-induced mouse model of neurotoxicity, as well as in a rat model of Parkinson's disease. These observations suggest that mildronate may stimulate processes involved in cell survival and change expression of proteins involved in neurogenic processes. The present study investigated the influence of mildronate on learning and memory in the passive avoidance response (PAR) test and the active conditioned avoidance response (CAR) test in rats. The CAR test employed also bromodeoxyuridine (BrdU)-treated animals. Hippocampal cell BrdU incorporation was then immunohistochemically assessed in BrdU-treated, CAR-trained rats to identify proliferating cells. In addition, the expression of hippocampal proteins which could serve as memory enhancement biomarkers was evaluated and compared to non-trained animals' data. These biomarkers included glutamic acid decarboxylase 65/67 (GAD65/67), acetylcholine esterase (AChE), growth-associated protein-43 (GAP-43) and the transcription factor c-jun/activator protein-1 (AP-1). The results showed that mildronate enhanced learning/memory formation that coincided with the proliferation of neural progenitor cells, changing/regulating of the expression of biomarker proteins which are involved in the activation of glutamatergic and cholinergic pathways, transcription factors and adhesion molecule. The data from our study suggest that mildronate may be useful as a possible cognitive enhancer for the treatment of patients with neurodegenerative diseases with dementia.


Subject(s)
Hippocampus/drug effects , Learning/drug effects , Memory/drug effects , Methylhydrazines/pharmacology , Nerve Tissue Proteins/metabolism , Acetylcholinesterase/metabolism , Animals , Blotting, Western , Bromodeoxyuridine/metabolism , Glutamate Decarboxylase/metabolism , Hippocampus/metabolism , Male , Rats , Rats, Wistar
8.
Medicina (Kaunas) ; 48(10): 525-31, 2012.
Article in English | MEDLINE | ID: mdl-23324249

ABSTRACT

BACKGROUND AND OBJECTIVE: Ischemic stroke may initiate a reperfusion injury leading to brain damage cascades where inflammatory mechanisms play a major role. Therefore, the necessity for the novel stroke-protecting agents whose the mechanism of action is focused on their anti-inflammatory potency is still on the agenda for drug designers. Our previous studies demonstrated that cerebrocrast (a 1,4-dihydropyridine derivative) and mildronate (a representative of the aza-butyrobetaine class) possessed considerable anti-inflammatory and neuroprotective properties in different in vitro and in vivo model systems. The present study investigated their stroke-protecting ability in an endothelin-1 (ET-1)-induced ischemic stroke model in rats. MATERIAL AND METHODS: Male Wistar rats were pretreated (for 7 days, per os) with cerebrocrast (0.1 mg/kg), mildronate (100 mg/kg), or their combination, followed by the intracerebral injection of ET-1. Functional and behavioral tests were carried out up to 14 days after the ET-1 injection. Ex vivo, the number of degenerated neurons and the infarction size in the cerebral cortical tissue were assessed histologically. RESULTS: Cerebrocrast and mildronate effectively normalized ET-1-induced disturbances in neurological status, improved the muscle tone, and decreased the number of degenerated cortical cells. Both drugs also reduced the infarction size, and cerebrocrast showed at least a 2-fold higher activity than mildronate. The combination of both drugs did not cause a more pronounced effect in comparison with the action of drugs administered separately. CONCLUSIONS: The 1,4-dihydropyridine and aza-butyrobetaine structures may serve for the design of novel stroke-protecting agents to prevent severe neurological poststroke consequences.


Subject(s)
Dihydropyridines/therapeutic use , Methylhydrazines/therapeutic use , Neuroprotective Agents/therapeutic use , Stroke/prevention & control , Animals , Dihydropyridines/chemistry , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Therapy, Combination , Endothelin-1/pharmacology , Male , Methylhydrazines/chemistry , Neuroprotective Agents/chemistry , Rats , Rats, Wistar , Stroke/chemically induced
9.
J Pharm Pharmacol ; 63(2): 230-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21235587

ABSTRACT

OBJECTIVES: Taurine, a sulfur-containing amino acid, has high hydrophilicity and is poorly absorbed. Tauropyrone, a taurine-containing 1,4-dihydropyridine derivative, is suggested to have greater activity than taurine owing to improved physicochemical properties that facilitate delivery of the compound to target cells. The aim of this study was to determine whether the 1,4-dihydropyridine moiety in tauropyrone improves the pharmacological efficacy of taurine in vitro and in vivo. METHODS: The effects of taurine and tauropyrone, as well as of the 1,4-dihydropyridine moiety were compared in in-vitro experiments to determine the binding to GABA receptors and influence on mitochondrial processes (isolated rat liver mitochondria), and in in-vivo tests to assess the influence on behavioural effects caused by the GABA-A receptor ligands, bicuculline, diazepam and ethanol. KEY FINDINGS: Unlike taurine, tauropyrone did not display binding activity for the GABA-A receptor, and only taurine (but not tauropyrone) at low doses (0.1, 1.0 and 10 mg/kg) antagonised the bicuculline-induced convulsion effect. Taurine and tauropyrone had no effect on diazepam myorelaxing action, and they both exerted a comparable 'anti-ethanol' effect (shortening of the ethanol-sleeping time). Taurine and tauropyrone did not influence processes of mitochondrial bioenergetics. CONCLUSIONS: The action of tauropyrone at the level of the GABA-A receptor differs qualitatively from that of taurine, probably because of its 1,4-dihydropyridine moiety, which may hinder access to the GABA-A receptor GABA site. Tauropyrone does not show improved pharmacological efficacy in in-vitro and in-vivo studies in comparison with taurine.


Subject(s)
Behavior, Animal/drug effects , Mitochondria/drug effects , Receptors, GABA-A/metabolism , Taurine/analogs & derivatives , Taurine/pharmacology , Animals , Bicuculline/pharmacology , Diazepam/pharmacology , Dihydropyridines/pharmacology , Energy Metabolism/drug effects , Ethanol/pharmacology , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Inbred ICR , Mitochondria/metabolism , Motor Activity/drug effects , Muscle Tonus/drug effects , Protein Binding/drug effects , Rats , Rats, Wistar , Rotarod Performance Test/methods , Seizures/chemically induced , Structure-Activity Relationship
10.
Int J Mol Sci ; 11(11): 4465-87, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21151450

ABSTRACT

Previously, we have found that mildronate [3-(2,2,2-trimethylhydrazinium) propionate dihydrate], a small molecule with charged nitrogen and oxygen atoms, protects mitochondrial metabolism that is altered by inhibitors of complex I and has neuroprotective effects in an azidothymidine-neurotoxicity mouse model. In the present study, we investigated the effects of mildronate in a rat model of Parkinson's disease (PD) that was generated via a unilateral intrastriatal injection of the neurotoxin 6-hydroxydopamine (6-OHDA). We assessed the expression of cell biomarkers that are involved in signaling cascades and provide neural and glial integration: the neuronal marker TH (tyrosine hydroxylase); ubiquitin (a regulatory peptide involved in the ubiquitin-proteasome degradation system); Notch-3 (a marker of progenitor cells); IBA-1 (a marker of microglial cells); glial fibrillary acidic protein, GFAP (a marker of astrocytes); and inducible nitric oxide synthase, iNOS (a marker of inflammation). The data show that in the 6-OHDA-lesioned striatum, mildronate completely prevented the loss of TH, stimulated Notch-3 expression and decreased the expression of ubiquitin, GFAP and iNOS. These results provide evidence for the ability of mildronate to control the expression of an array of cellular proteins and, thus, impart multi-faceted homeostatic mechanisms in neurons and glial cells in a rat model of PD. We suggest that the use of mildronate provides a protective effect during the early stages of PD that can delay or halt the progression of this neurodegenerative disease.


Subject(s)
Methylhydrazines/pharmacology , Neuroglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/metabolism , Animals , Biomarkers/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Male , Methylhydrazines/therapeutic use , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidopamine/toxicity , Rats , Rats, Wistar , Receptor, Notch3 , Receptors, Notch/genetics , Receptors, Notch/metabolism , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
11.
Neurosci Lett ; 470(2): 100-5, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20036318

ABSTRACT

Mildronate, a representative of the aza-butyrobetaine class of drugs with proven cardioprotective efficacy, was recently found to prevent dysfunction of complex I in rat liver mitochondria. The present study demonstrates that mildronate also acts as a neuroprotective agent. In a mouse model of azidothymidine (anti-HIV drug) neurotoxicity, mildronate reduced the azidothymidine-induced alterations in mouse brain tissue: it normalized the increase in caspase-3, cellular apoptosis susceptibility protein (CAS) and iNOS expression assessed by quantitative and semi-quantitative analysis. Mildronate also normalized the changes in cytochrome c oxidase (COX) expression, reduced the expression of glial fibrillary acidic protein (GFAP) and cellular infiltration. The present results show that the neuroprotective action of mildronate results at least partially from anti-neurodegenerative (anti-apoptotic) and anti-inflammatory mechanisms. It might be suggested that the molecular conformation of mildronate can facilitate its easy binding to mitochondria, and regulate the expression of different signal molecules, hence maintaining cellular signaling and survival.


Subject(s)
Brain/drug effects , Brain/physiopathology , Methylhydrazines/pharmacology , Neuroprotective Agents/pharmacology , Animals , Anti-HIV Agents/toxicity , Caspase 3/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Disease Models, Animal , Electron Transport Complex IV/metabolism , Glial Fibrillary Acidic Protein/metabolism , Lymphocytes/drug effects , Lymphocytes/physiology , Male , Methylhydrazines/chemistry , Mice , Mice, Inbred Strains , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/physiopathology , Nitric Oxide Synthase Type II/metabolism , Zidovudine/toxicity
12.
Pharmacol Biochem Behav ; 92(2): 267-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19135472

ABSTRACT

Little is known about the endogenous functions of gamma1- and gamma2-melanocyte stimulating hormones (gamma1- and gamma2-MSH). Although gamma-MSHs bind to melanocortin receptor subtypes 3 and 4, we have previously shown that these peptides also influence non-melanocortinergic processes, such as dopaminergic and GABAergic. The aim of this study was to determine the effects of gamma1- and gamma2-MSH (at doses 0.3, 1 and 2 nmol/mouse/5 microl) on the anxiety levels in mice in elevated plus maze. Three experimental paradigms were performed to assess the effects of peptides on: a) ethanol withdrawal; b) acute ethanol-induced anxiolytic action; c) peptides per se. We used ethanol as the model substance, since its action involves either dopaminergic/GABAergic or melanocortinergic processes. gamma-MSHs were administered intracisternally in mice and behavioural responses were assessed in the elevated plus maze test. This study provides the first demonstration of an anxiogenic effect of gamma1- and gamma2-MSH, their synergistic/additive effect on ethanol withdrawal-induced anxiety behaviour, and an antagonism of peptides involved in the anxiolytic action of ethanol. Furthermore, results suggest that gamma-MSHs belong to an anxiogenic peptide family that may play an important role in anxiety disorders as well as in the development of alcohol dependence and/or alcohol withdrawal-induced behaviours.


Subject(s)
Anxiety/chemically induced , Ethanol/adverse effects , Maze Learning , Melanocyte-Stimulating Hormones/pharmacology , Substance Withdrawal Syndrome/etiology , Animals , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred ICR
13.
Basic Clin Pharmacol Toxicol ; 103(5): 401-6, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18801031

ABSTRACT

This study demonstrates the effective protection by compounds of atypical 1,4-dihydropyridine (DHP) series cerebrocrast, glutapyrone and tauropyrone against neuro- and cardiotoxicity caused by the model compound azidothymidine, a well-known mitochondria-compromising anti-HIV drug. In previous in vitro experiments, we have demonstrated distinct effects of these DHP compounds to influence mitochondrial functioning. In the present in vivo experiments, DHP compounds were administered intraperitoneally in mice daily for 2 weeks, per se and in combinations with azidothymidine at doses: azidothymidine 50 mg/kg; cerebrocrast 0.1 mg/kg; glutapyrone 1 mg/kg; and tauropyrone 1 mg/kg. At the end of the experiment, mice were killed, heart and brain tissues were removed and examined ex vivo histopathologically and immunohistochemically. NF-kappaBp65 and caspase-3 were used as the markers indicating inflammatory and apoptotic events, respectively. Cerebrocrast (dicyclic structure) was the most potent DHP, which effectively reduced azidothymidine-induced overexpression of NF-kappaBp65 and caspase-3 in mouse myocardium and brain cortex. Glutapyrone per se increased the number of caspase-3-positive cells in the brain, whereas it reduced NF-kappaBp65 and caspase-3 expression in cardiac tissue caused by azidothymidine. Tauropyrone showed dual action: per se it increased caspase-3 in the brain and NF-kappaBp65 expression in the heart, but it considerably reduced these activations in azidothymidine-treated mice. This study provides the first demonstration of a distinct pharmacological action for atypical DHP compounds in cardiac and brain tissues. The dicyclic structure of cerebrocrast is considered beneficial for neuro- and cardioprotection at least in part via mitochondrial targeting and consequent regulation of inflammatory and apoptotic processes.


Subject(s)
Dihydropyridines/pharmacology , Glutamates/pharmacology , Taurine/analogs & derivatives , Animals , Anti-HIV Agents/toxicity , Apoptosis/drug effects , Caspase 3/drug effects , Caspase 3/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Gene Expression Regulation/drug effects , Heart Diseases/chemically induced , Heart Diseases/drug therapy , Inflammation/drug therapy , Male , Mice , Mice, Inbred ICR , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Taurine/pharmacology , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , Zidovudine/toxicity
14.
Planta Med ; 74(5): 491-6, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18543146

ABSTRACT

Naturally occurring benzodiazepines have been identified in regular food such as wheat and potato, but there is still no evidence that potato extracts can affect CNS responses in vivo. Here we found that undiluted potato juice and potato juice diluted with saline 1 : 2 administered 10 min intracisternally ( I. C.) and 30 min per os before bicuculline exerted significant anticonvulsant activity in the bicuculline-induced seizure threshold test in mice. In vitro, potato juice from different harvests at dilution series from 10 % to 0.000001 %, diluted 100,000-fold, displaced 50 % of gamma-aminobutyric acid (GABA) receptor ligand [ (3)H]GABA and diluted 40-fold displaced 50 % of [(3)H]flunitrazepam from binding sites in mice forebrain membranes. The low content of diazepam (0.04 +/- 0.01 mg/kg) determined by HPLC and mass spectrometry in the potato extracts could not sustain the anticonvulsant activity of potato juice in vivo; therefore we hypothesized that potato juice might contain GABA (A) receptor GABA-site active compounds. The findings of this study suggest that potato juice as well as potato taken as food may have the capacity of influencing brain GABA-ergic activity.


Subject(s)
Anticonvulsants/analysis , Behavior, Animal/drug effects , Diazepam/analysis , Plant Extracts/pharmacology , Receptors, GABA/metabolism , Solanum tuberosum/chemistry , Animals , Anticonvulsants/pharmacology , Bicuculline , Chromatography, High Pressure Liquid , Diazepam/pharmacology , Male , Mass Spectrometry , Mice , Mice, Inbred ICR , Receptors, GABA/drug effects
15.
Pharmacol Biochem Behav ; 90(4): 712-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18582921

ABSTRACT

The lupane type pentacyclic triterpenes: lupeol, betulin, and betulinic acid are widely distributed natural compounds. Recently, pharmaceutical compositions from plant extracts (family Marcgraviaceae) containing betulinic acid, have been patented as anxiolytic remedies. To extend our knowledge of the CNS effects of the triterpenes, we suggest here that the chemically related lupeol, betulin and betulinic acid may interact with the brain neurotransmitter gamma-aminobutyric acid (GABA) receptors in vitro and in vivo. Using radioligand receptor-binding assay, we showed that only betulin bound to the GABA(A)-receptor sites in mice brain in vitro and antagonised the GABA(A)-receptor antagonist bicuculline-induced seizures in mice after intracisternal and intraperitoneal administration. Neither betulinic acid nor lupeol bound to GABA(A) receptor nor did they inhibit bicuculline-induced seizures in vivo. These findings demonstrate for the first time the CNS effects of betulin in vivo, and they also show distinct GABA(A)-receptor-related properties of lupane type triterpenes. These findings may open new avenues in understanding the central effects of betulin, and they also indicate possibilities for novel drug design on the basis of betulin structure.


Subject(s)
Anticonvulsants/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Receptors, GABA/metabolism , Triterpenes/metabolism , Triterpenes/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bicuculline/pharmacology , Flunitrazepam/metabolism , GABA Modulators/metabolism , Male , Mice , Mice, Inbred ICR , Muscle Tonus/drug effects , Pentacyclic Triterpenes , Postural Balance/drug effects , Seizures/chemically induced , Seizures/psychology , gamma-Aminobutyric Acid/metabolism , Betulinic Acid
16.
J Mol Neurosci ; 35(2): 179-93, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18418736

ABSTRACT

Members of the solute carrier families (SLC) 32, 36, and 38, together also designated the beta-group of SLCs, are known to transport neutral amino acids. In this paper, we show that these three families were present before the split of the animal lineage and that they are likely to share a common decent. We also show that the APF transporters found in plants are most likely homologous to the mammalian beta-group, suggesting that this type of transporters arouse early in the evolution of eukaryotes. We performed detailed tissue expression analysis of all the members of the beta-group in rat and found several examples of highly specific expression patterns, with SLC38A7 being exclusively found in liver, SLC38A5 in blood, and SLC38A4 in muscle and liver. Moreover, we found that SLC38A10 is expressed in several endocrine organs. We also found that SLC38A1 is highly up regulated in the cortex from rats treated with diazepam and that SLC38A2 is significantly down regulated in the same tissue. In addition, we performed a detailed expression analysis of SLC38A1 and SLC38A6 in mouse brain using in situ hybridization, which showed that both these transporters are widely expressed in the brain.


Subject(s)
Amino Acid Transport System A/genetics , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems/genetics , Evolution, Molecular , Nerve Tissue Proteins/genetics , Phylogeny , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Animals , Anticonvulsants/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Diazepam/pharmacology , Dopamine Agents/pharmacology , Endocrine System/physiology , Gene Expression/drug effects , Gene Expression/physiology , Humans , In Situ Hybridization , Levodopa/pharmacology , Liver/physiology , Male , Mice , Mice, Inbred Strains , Muscle, Skeletal/physiology , Rats , Rats, Wistar
17.
Basic Clin Pharmacol Toxicol ; 99(4): 323-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17040219

ABSTRACT

Azidothymidine, a nucleoside-analogue reverse transcriptase inhibitor (NRTI), is a commonly used antiretroviral drug in AIDS treatment, however its use is limited by severe toxic side effects due to its influence on mitochondria that result in myopathy, particularly affecting the cardiac muscle. We suggest that effective protection of azidothymidine-induced cardiopathology can be expected from drugs that are capable of targeting mitochondria. Therefore the present study in mice was carried out with mildronate, a cardioprotective drug of the aza-butyrobetaine class, which previously has been shown to act as a highly potent protector of mitochondrial processes. In our study, saline (control), azidothymidine (50 mg/kg), mildronate (50, 100 and 200 mg/kg), and azidothymidine + mildronate (at the doses mentioned) were injected intraperitoneally daily in separate groups of mice for two weeks. At the termination of the experiment, mice were sacrificed, the hearts were removed and cardiac tissue was examined morphologically and immunohistochemically. It was found that azidothymidine, compared to control and mildronate groups, induced major morphologic changes in cardiac tissue, which were manifestated as degeneration and inflammation. These changes were prevented when mildronate was co-administered with azidothymidine. Mildronate also reduced the azidothymidine-induced expression of nuclear factor kappaBp65 (NF-kappaBp65). The obtained data demonstrate a high ability of mildronate of preventing azidothymidine-induced cardiopathologic changes, and suggest mildronate's indirect action on azidothymidine-caused oxidative stress reactions leading to mitochondrial dysfunction. This offers a rational combination of mildronate with azidothymidine or other anti-HIV drugs for beneficial application in AIDS therapy.


Subject(s)
Cardiovascular Agents/therapeutic use , Heart Diseases/prevention & control , Methylhydrazines/therapeutic use , Mitochondria/drug effects , Zidovudine , Animals , Cardiovascular Agents/pharmacology , Disease Models, Animal , Heart Diseases/chemically induced , Methylhydrazines/pharmacology , Mice , Mice, Inbred ICR
SELECTION OF CITATIONS
SEARCH DETAIL
...