Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 41(3): 605-613, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32410106

ABSTRACT

We have recently demonstrated that extracellular vesicles (EVs) derived from the human teeth stem cells improve motor symptoms and normalize tyrosine hydroxylase (TH) expression in the nigrostriatal structures of Parkinson's disease (PD) model rats obtained by 6-hydroxydopamine (6-OHDA) unilateral injection into the medial forebrain bundle (MFB). The aim of this study was to clarify: (1) how long therapeutic effects persist after discontinuation of 17-day intranasal administration of EVs in 6-OHDA rats; (2) may EVs reverse cognitive (learning/memory) dysfunction in these PD model rats; (3) whether and how the behavioral improvement may be related to the expression of TH and Nissl bodies count in the nigrostriatal structures. Our results demonstrated that in 6-OHDA rats, gait was normalized even ten days after discontinuation of EVs administration. EVs successfully reversed 6-OHDA-induced impairment in spatial learning/memory performance; however, the beneficial effect was shorter (up to post-treatment day 6) than that revealed for gait improvement. The shorter effect of EVs coincided with both full normalization of TH expression and Nissl bodies count in the nigrostriatal structures, while slight but significant increase in the 6-OHDA-decreased Nissl count persisted in the substantia nigra even on the post-treatment day 20, supposedly due to the continuation of protein synthesis in the living cells. The obtained data indicate the usefulness of further studies to find the optimal administration regimen which could be translated into clinical trials on PD patients, as well as to clarify other-apart from dopaminergic-neuromodulatory pathways involved in the EVs mechanism of action.


Subject(s)
Extracellular Vesicles/metabolism , Gait , Memory , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Administration, Intranasal , Animals , Behavior, Animal , Child , Corpus Striatum/pathology , Disease Models, Animal , Extracellular Vesicles/ultrastructure , Female , Humans , Male , Nissl Bodies/metabolism , Oxidopamine , Parkinson Disease/pathology , Rats, Wistar , Substantia Nigra/pathology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
2.
Eur J Pharmacol ; 881: 173290, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32580040

ABSTRACT

The earliest hallmarks of sporadic Alzheimer's disease (sAD) are impaired glucose metabolism, chronic neuroinflammation, diminished synaptic plasticity and subsequent cognitive decline. The safest antidiabetic drug metformin has shown both glucose metabolism-improving and cognition-enhancing action in type 2 diabetes patients and diabetic model animals. However, metformin has not been previously studied in intracerebroventricular streptozocin (STZ)-induced model of sAD. Therefore, our aim was to assess the preventive action of metformin in sAD model-rats. Firstly, the actions of metformin (75 and 100 mg/kg) on cognitive functions and sociability were examined. Secondly, we wanted to identify whether behavioral effects of metformin were provided via its action on brain glucose transport, neuronal/glial uptake and metabolism. Thirdly, the effects of metformin on neuroinflammation, acetylcholine esterase density and activity, as well as on synaptic plasticity were determined. Our results showed that metformin reversed STZ-induced impairments in spatial learning/memory performance and sociability, coinciding with normalization of brain glucose transport, uptake and metabolism. Microgliosis and astrogliosis were ameliorated by metformin in sAD model rats. Metformin also preserved hippocampal synaptic plasticity and normalized acetylcholine cleavage in the cortical and hippocampal tissues, as well as inhibited acetylcholine esterase activity in vitro. These data indicate the promise of further research of metformin in early brain pathologies to stop neurodegenerative before severe cognitive decline occurs.


Subject(s)
Alzheimer Disease/prevention & control , Behavior, Animal/drug effects , Brain/drug effects , Cognition/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Neuroprotective Agents/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/chemically induced , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , GPI-Linked Proteins/metabolism , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/metabolism , Glycogen Synthase Kinase 3/metabolism , Hypoglycemic Agents/administration & dosage , Injections, Intraventricular , Male , Metformin/administration & dosage , Morris Water Maze Test/drug effects , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Neuroprotective Agents/administration & dosage , Rats, Wistar , Social Behavior , Streptozocin
3.
Medicina (Kaunas) ; 55(7)2019 Jul 21.
Article in English | MEDLINE | ID: mdl-31330913

ABSTRACT

Background and Objectives: Previously we have shown that synthetic lunasin, a 43 amino acid residue-containing peptide, after its central (intracisternal) administration in mice demonstrated antagonism against dopaminergic drug behavioural effects, indicating a putative antipsychotic/anti-schizophrenic profile of lunasin. The aims of the present studies were: to test whether lunasin would show an influence on the dopaminergic system after intranasal administration, and to examine the effect(s) of lunasin on serotonin and glutamatergic systems, which could play an essential role in antipsychotic action. Materials and Methods: Lunasin was administered intra-nasally at doses 0.1 and 1 nmol/mouse in ICR mice (n = 7-8) and tested in an open field on hyperlocomotion caused by amphetamine; serotonin 5-HT 2A/2C receptor agonist 1-(2,5-dimethoxy-4-iodophenyl)- 2-aminopropane (DOI); and glutamate NMDA receptor antagonist phencyclidine. Following behavioural testing, the contents of neurotransmitters and their metabolites in brain hemispheres (n = 6-8) were assessed by ultra-high-performance liquid chromatography-time of flight mas-spectrometry (UHPLC-TOF-MS) method. Also, lunasin binding to serotonin receptors was assessed. Results: Lunasin intra-nasally fully normalized hyper-locomotion and brain monoamine levels in amphetamine- and DOI-treated mice brains. Phencyclidine behavioural effects were not influenced. In vitro receptor binding data demonstrated a low affinity of lunasin (at µM concentrations) compared with DOI (nM concentrations) for the 5-HT2A and 5-HT2C receptors. Conclusions: These results demonstrated, for the first time, that the intranasal administration of oligopeptide lunasin normalized mice behaviour and brain monoamine levels in experimental psychosis mice models. Its neuro-regulatory effects indicated a usefulness of this peptide molecule for the design of novel psychotropic agents.


Subject(s)
Antipsychotic Agents/analysis , Oligopeptides/therapeutic use , Administration, Intranasal , Amphetamines/administration & dosage , Amphetamines/adverse effects , Amphetamines/therapeutic use , Animals , Disease Models, Animal , Mice , Mice, Inbred ICR/metabolism , Motor Activity/drug effects , Oligopeptides/administration & dosage , Oligopeptides/pharmacology
4.
J Neurosci Res ; 97(6): 708-726, 2019 06.
Article in English | MEDLINE | ID: mdl-30742328

ABSTRACT

Neuroinflammation, oxidative stress, decreased glucose/energy metabolism, and disrupted neurotransmission are changes that occur early in sporadic Alzheimer's disease (AD), manifesting as mild cognitive impairment. Recently, the imbalanced function of the gamma-aminobutyric acid (GABA) system was identified as a critical factor in AD progression. Thus, maintaining balance among neurotransmitter systems, particularly the GABA system, can be considered a beneficial strategy to slow AD progression. The present study investigated the effects of the compound gammapyrone, a molecule containing three GABA moieties: "free" moiety attached to the position 4 of the 1,4-dihydropyridine (DHP) ring, and two "crypto" moieties as part of the DHP scaffold. The "free" and "crypto" GABA moieties are linked by a peptide bond (-CONH-), resulting in a peptide-mimicking structure. In a nontransgenic male rat AD model generated by intracerebroventricular (icv) streptozocin (STZ) administration, gammapyrone (0.1 and 0.5 mg/kg ip) mitigated the impairment of spatial learning and memory, prevented astroglial and microglial neuroinflammation, and normalized acetylcholine breakdown and GABA biosynthesis. In PC12 cells, gammapyrone protected against oxidative stress, mitochondrial dysfunction and apoptosis caused by the mitochondrial toxin di-2-ethylhexyl phthalate (DEHP). Gammapyrone did not bind to GABA-A and GABA-B receptors in vitro; therefore, we cannot attribute its neuroprotective action to a specific interaction with GABA receptors. Nevertheless, we suggest that the peptide-like regulatory mechanisms of gammapyrone or its allosteric modulatory properties are essential for the observed effects. Since, the icv STZ model resembles the early stages of AD, gammapyrone, and/or its congeners could be useful in the design of anti-dementia drugs.


Subject(s)
Alzheimer Disease/metabolism , Brain/drug effects , Brain/metabolism , Mitochondria/drug effects , Neuroprotective Agents/administration & dosage , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/metabolism , Acetylcholinesterase/metabolism , Animals , Astrocytes/drug effects , Cells, Cultured , Disease Models, Animal , Encephalitis/metabolism , Glutamate Decarboxylase/metabolism , Male , Maze Learning/drug effects , Memory/drug effects , Microglia/drug effects , Mitochondria/metabolism , Rats, Wistar , Receptors, GABA/metabolism , gamma-Aminobutyric Acid/administration & dosage
5.
Stem Cells Transl Med ; 8(5): 490-499, 2019 05.
Article in English | MEDLINE | ID: mdl-30706999

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder affecting millions of people worldwide. At present, there is no effective cure for PD; treatments are symptomatic and do not halt progression of neurodegeneration. Extracellular vesicles (EVs) can cross the blood-brain barrier and represent promising alternative to the classical treatment strategies. In the present study, we examined therapeutic effects of intranasal administration of EVs derived from human exfoliated deciduous teeth stem cells (SHEDs) on unilateral 6-hydroxydopamine (6-OHDA) medial forebrain bundle (MFB) rat model of PD. CatWalk gait tests revealed that EVs effectively suppressed 6-OHDA-induced gait impairments. All tested gait parameters (stand, stride length, step cycle, and duty cycle) were significantly improved in EV-treated animals when compared with 6-OHDA-lesion group rats. Furthermore, EVs slowed down numbers of 6-OHDA-induced contralateral rotations in apomorphine test. Improvements in motor function correlated with normalization of tyrosine hydroxylase expression in the striatum and substantia nigra. In conclusion, we demonstrated, for the first time, the therapeutic efficacy of intranasal administration of EVs derived from SHEDs in a rat model of PD induced by 6-OHDA intra-MFB lesion. Our findings could be potentially exploited for the development of new treatment strategies against PD.


Subject(s)
Administration, Intranasal/methods , Extracellular Vesicles/metabolism , Microscopy, Electron, Transmission/methods , Oxidopamine/therapeutic use , Parkinson Disease/drug therapy , Stem Cells/metabolism , Tooth/physiopathology , Tyrosine 3-Monooxygenase/metabolism , Aged , Animals , Corpus Striatum/pathology , Disease Models, Animal , Humans , Male , Oxidopamine/pharmacology , Parkinson Disease/pathology , Rats , Rats, Wistar , Substantia Nigra/pathology
6.
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
7.
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
8.
Pharmacol Res ; 113(Pt B): 781-787, 2016 11.
Article in English | MEDLINE | ID: mdl-27345857

ABSTRACT

The prevalence of Alzheimer's disease (AD) is higher in females than in males, and causes more severe cognitive, memory and behavioral impairments. Previously, in male transgenic (Tg) APPSweDI mice, we reported that the novel lipophilic 1,4-dihydropyridine (DHP) derivative AP-12 crossed the blood-brain barrier, blocked neuronal and vascular calcium channels, changed brain protein expression and improved behavior. In this study, we used female Tg APPSweDI mice to assess the effects of AP-12 on behavior, and brain protein expression, with a particular focus on those of the GABAergic system. The results showed that in female Tg mice, similar to male Tg mice, AP-12 improved spatial learning/memory performance in the water maze test and demonstrated anxiolytic effect in the elevated zero maze (after single administration of AP-12) and elevated plus maze (after chronic injections of AP-12). In addition, we demonstrated upregulated expression of glutamate decarboxylase 67 (GAD67) and vesicular GABA transporter (VGAT) in the cingulate cortex and hippocampus, pointing to the role of the GABAergic system as one of the neural networks dysregulated in AD. In both female and male mice, AP-12 did not change the expression of hippocampal Homer-1, a protein which is involved in synaptic plasticity. However, in cingulate cortex, the staining density of Homer-1 was significantly increased in female mice. Further, female mice (similar to male mice) did not show changes in brain AChE expression and in the amyloid beta load in the hippocampus and cingulate cortex. In conclusion, the memory enhancing, anxiolytic and protein expression effects of AP-12 did not show sex specificity in APPSweDI mice. Considering the ability of AP-12 to block brain calcium channels and improve memory by enhancing the GABAergic and synaptic plasticity processes, AP-12 is a promising compound which merits further pre-clinical studies to investigate its usefulness in the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Calcium Channel Blockers/pharmacology , Calcium/metabolism , Gyrus Cinguli/drug effects , Hippocampus/drug effects , Memory/drug effects , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Blood-Brain Barrier/metabolism , Dihydropyridines/pharmacology , Disease Models, Animal , Female , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Glutamate Decarboxylase/metabolism , Gyrus Cinguli/metabolism , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Neuronal Plasticity/drug effects , Up-Regulation/drug effects , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
9.
Pharmacol Res ; 113(Pt B): 754-759, 2016 11.
Article in English | MEDLINE | ID: mdl-27234899

ABSTRACT

This mini review is devoted to the design and pharmacological studies of novel atypical 1,4-dihydropyridine (DHP) derivatives which differ to a great extent from the traditional DHPs either by lack of neuronal calcium channel blocking activity and/or inability to protect mitochondrial processes. About 100 new DHP derivatives were screened and the mostly active were selected for detailed studies. The compounds of the series of the amino acid ("free" plus "crypto")-containing DHPs and lipophilic di-cyclic DHPs demonstrated long-lasting neuroprotective and/or memory-enhancing action, particularly at low doses (0.005-0.05mg/kg) in different neurodeficiency rat or mice models, and exerted neurotransmitter-modulating effects. The studies have shown an ability of these atypical DHPs to normalize the expression of neuronal proteins, which participate in the regulation of neurotransmission (particularly of the GABAergic system) and synaptic plasticity that has been impaired in animal models, including Alzheimer's disease transgenic mice. The obtained results indicate that the tested DHP compounds can be considered as candidate molecules either for their further chemical modifications or for the more detailed studies to identify cell targets essential for neuroprotection and memory enhancing.


Subject(s)
Dihydropyridines/pharmacology , Dihydropyridines/therapeutic use , Memory/drug effects , Neuroprotection/drug effects , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Humans , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Neurotransmitter Agents/metabolism
10.
PLoS One ; 10(6): e0127686, 2015.
Article in English | MEDLINE | ID: mdl-26042808

ABSTRACT

Ca2+ blockers, particularly those capable of crossing the blood-brain barrier (BBB), have been suggested as a possible treatment or disease modifying agents for neurodegenerative disorders, e.g., Alzheimer's disease. The present study investigated the effects of a novel 4-(N-dodecyl) pyridinium group-containing 1,4-dihydropyridine derivative (AP-12) on cognition and synaptic protein expression in the brain. Treatment of AP-12 was investigated in wild type C57BL/6J mice and transgenic Alzheimer's disease model mice (Tg APPSweDI) using behavioral tests and immunohistochemistry, as well as mass spectrometry to assess the blood-brain barrier (BBB) penetration. The data demonstrated the ability of AP-12 to cross the BBB, improve spatial learning and memory in both mice strains, induce anxiolytic action in transgenic mice, and increase expression of hippocampal and cortical proteins (GAD67, Homer-1) related to synaptic plasticity. The compound AP-12 can be seen as a prototype molecule for use in the design of novel drugs useful to halt progression of clinical symptoms (more specifically, anxiety and decline in memory) of neurodegenerative diseases, particularly Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/genetics , Brain/metabolism , Carrier Proteins/metabolism , Dihydropyridines/pharmacology , Glutamate Decarboxylase/metabolism , Memory/drug effects , Spatial Learning/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/pathology , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Homer Scaffolding Proteins , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Time Factors
11.
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
12.
J Neurosci Res ; 92(3): 338-46, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24273007

ABSTRACT

Mildronate, a carnitine congener drug, previously has been shown to provide neuroprotection in an azidothymidine-induced mouse model of neurotoxicity and in a Parkinson's disease rat model. The aim of this study was to investigate the effects of mildronate treatment on cognition and pathology in Alzheimer's disease (AD) model mice (APP(SweDI)). Mildronate was administered i.p. daily at 50 or 100 mg/kg for 28 days. At the end of treatment, the animals were behaviorally and cognitively tested, and brains were assessed for AD-related pathology, inflammation, synaptic markers, and acetylcholinesterase (AChE). The data show that mildronate treatment significantly improved animal performance in water maze and social recognition tests, lowered amyloid-ß deposition in the hippocampus, increased expression of the microglia marker Iba-1, and decreased AChE staining, although it did not alter expression of proteins involved in synaptic plasticity (GAP-43, synaptophysin, and GAD67). Taken together, these findings indicate mildronate's ability to improve cognition and reduce amyloid-ß pathology in a mouse model of AD and its possible therapeutic utility as a disease-modifying drug in AD patients.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cognition Disorders/drug therapy , Methylhydrazines/therapeutic use , Acetylcholinesterase/metabolism , Adjuvants, Immunologic/pharmacology , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cognition Disorders/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamate Decarboxylase/metabolism , Humans , Locomotion/drug effects , Locomotion/genetics , Methylhydrazines/pharmacology , Mice , Mice, Transgenic , Social Behavior
13.
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
14.
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
15.
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
16.
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
17.
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
18.
Medicina (Kaunas) ; 47(10): 552-9, 2011.
Article in English | MEDLINE | ID: mdl-22186119

ABSTRACT

BACKGROUND: Mildronate (3-[2,2,2-trimethylhydrazinium] propionate dihydrate) traditionally is a well-known cardioprotective drug. However, our recent studies convincingly demonstrated its neuroprotective properties. The aim of the present study was to evaluate the influence of mildronate on the expression of proteins that are involved in the differentiation and survival of the nigrostriatal dopaminergic neurons in the rat model of Parkinson's disease (PD). The following biomarkers were used: heat shock protein 70 (Hsp70, a molecular chaperone), glial cell line-derived nerve growth factor (GDNF, a growth factor promoting neuronal differentiation, regeneration, and survival), and neural cell adhesion molecule (NCAM). MATERIAL AND METHODS: PD was modeled by 6-hydroxydopamine (6-OHDA) unilateral intrastriatal injection in rats. Mildronate was administered at doses of 10, 20, and 50 mg/kg for 2 weeks intraperitoneally before 6-OHDA injection. Rat brains were dissected on day 28 after discontinuation of mildronate injections. The expression of biomarkers was assessed immunohistochemically and by western blot assay. RESULTS: 6-OHDA decreased the expression of Hsp70 and GDNF in the lesioned striatum and substantia nigra, whereas in mildronate-pretreated (20 and 50 mg/kg) rats, the expression of Hsp70 and GDNF was close to the control group values. NCAM expression also was decreased by 6-OHDA in the striatum and it was totally protected by mildronate at a dose of 50 mg/kg. In contrast, in the substantia nigra, 6-OHDA increased the expression of NCAM, while mildronate pretreatment (20 and 50 mg/kg) reversed the 6-OHDA-induced overexpression of NCAM close to the control values. CONCLUSION: The obtained data showed that mildronate was capable to regulate the expression of proteins that play a role in the homeostasis of neuro-glial processes.


Subject(s)
Cardiovascular Agents/administration & dosage , Methylhydrazines/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinson Disease, Secondary/drug therapy , Protein Biosynthesis/drug effects , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , Male , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/biosynthesis , Oxidopamine/antagonists & inhibitors , Oxidopamine/pharmacology , Parkinson Disease, Secondary/metabolism , Rats , Rats, Wistar , Substantia Nigra/drug effects , Substantia Nigra/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...