Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neurosci Lett ; 790: 136898, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36195298

RESUMEN

Noopept (NP) is a proline-containing dipeptide with nootropic and neuroprotective properties. We have previously shown that NP significantly increased the frequency of spontaneous IPSCs in hippocampal CA1 pyramidal cells mediated by the activation of inhibitory interneurons in stratum radiatum. The cholinergic system plays an important role in the performance of cognitive functions, furthermore multiple behavioral and clinical facts link NP with the cholinergic system. The present study was undertaken to reveal the possible interaction of NP with neuronal nicotinic acetylcholine receptors (nAChRs). Currents were recorded from rat hippocampal neurons using the whole-cell, patch-clamp technique. NP (5 µM) increased the action potential firing frequency recorded from GABAergic interneurons in the stratum radiatum (SR) of CA1 region. This effect was almost completely abolished by the application of the α7 nAChR-selective antagonists α-bungarotoxin (α-BGT; 6 nM) and methyllycaconitine (MLA; 20 nM). The increase in the frequency of spontaneous IPSCs in CA1 pyramidal cells induced by NP was also eliminated by α7 nAChRs antagonists. These results imply the involvement of α7 nAChRs in the modulation of hippocampal neuronal activity caused by NP and indicate that a7 nAChRs are an important site of action of NP.


Asunto(s)
Nootrópicos , Receptores Nicotínicos , Animales , Ratas , Bungarotoxinas , Dipéptidos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interneuronas/metabolismo , Antagonistas Nicotínicos/farmacología , Nootrópicos/farmacología , Prolina/farmacología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas Sprague-Dawley , Receptores Nicotínicos/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
2.
Clin Exp Pharmacol Physiol ; 49(10): 1116-1125, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35748804

RESUMEN

Signs of metabolic syndrome and prediabetes preceding type 2 diabetes are modelled in an experiment using a high-fat diet (HFD). The aim of this work was to study the effect of a low molecular weight systemically active nerve growth factor mimetic, compound GK-2 (hexamethylenediamide bis[N-monosuccinyl-L-glutamyl-L-lysine]), on indicators of abdominal obesity, basal blood glucose level, glucose tolerance, cholesterol and triglyceride blood levels, as well as the morphological structure of the liver in male Wistar rats fed a HFD. Rats were divided into three groups: one of them received standard food (control) and two others were fed a HFD containing 45% fat, 35% carbohydrates and 20% protein, with a total caloric value of 516 kcal/100 g, over 12 weeks. Starting from the ninth week, for the next 4 weeks, one of the HFD groups was treated orally with saline whilst the other group was treated orally with GK-2 at a dose of 5 mg/kg. GK-2 was found to reduce the basal glycaemia level and improve glucose tolerance, as well as to reduce the blood level of cholesterol by 30% and that of triglycerides by 28% in comparison with the saline-treated HFD animals. GK-2 reduced the degree of abdominal obesity to the level of the healthy animals and eliminated morphological abnormalities in the liver caused by the HFD. The results of the study determine the feasibility of further GK-2 research as a potential agent for prediabetes treatment.


Asunto(s)
Diabetes Mellitus Tipo 2 , Estado Prediabético , Animales , Glucemia/metabolismo , Colesterol , Dieta Alta en Grasa/efectos adversos , Glucosa , Metabolismo de los Lípidos , Masculino , Peso Molecular , Factor de Crecimiento Nervioso/metabolismo , Obesidad/tratamiento farmacológico , Obesidad Abdominal , Estado Prediabético/tratamiento farmacológico , Ratas , Ratas Wistar , Delgadez
3.
Artículo en Inglés | MEDLINE | ID: mdl-35240968

RESUMEN

BACKGROUND: Growing pieces of evidence demonstrate a close relationship between type 2 diabetes (T2D) and neurodegenerative disorders such as Alzheimer's disease. The similarity of physiological and pathological processes occurring in pancreatic ß-cells and neurons over the course of these pathologies allows raising the question of the practicability of studying neuroprotective substances for their potential antidiabetic activity. OBJECTIVE: This review analyzes studies of antidiabetic and cytoprotective action on pancreatic ß- cells of the neuroprotective compounds that can attenuate the oxidative stress and enhance the expression of neurotrophins: low-molecular-weight NGF mimetic compound GK-2, selective anxiolytic afobazole, antidepressants lithium chloride, and lithium carbonate on the rat streptozotocin model of T2D. RESULTS: It was found that all the above-listed neuroprotective substances have a pronounced antidiabetic activity. The decrease in the ß-cells number, the average area of the pancreatic islets, as well as the violation of their morphological structure caused by the streptozotocin was significantly weakened by the therapy with the investigated neuroprotective substances. The extent of these morphological changes clearly correlates with the antihyperglycemic effect of these compounds. CONCLUSION: The presented data indicate that the neuroprotective substances attenuating the damaging effect of oxidative stress and neurotrophins deficit cannot only protect neurons but also exert their cytoprotective effect towards pancreatic ß-cells. These data may provide a theoretical basis for the further study of neuroprotective drugs as potential therapeutic options for T2D prevention and treatment.


Asunto(s)
Diabetes Mellitus Tipo 2 , Fármacos Neuroprotectores , Animales , Hipoglucemiantes , Factores de Crecimiento Nervioso , Ratas , Estreptozocina
4.
Curr Pharm Des ; 24(26): 3020-3027, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30295186

RESUMEN

The article is an overview of author's data obtained in the framework of the project "The Creation of dipeptide preparations" at the V.V. Zakusov Institute of Pharmacology, Moscow, Russia. Advantages of dipeptides over longer peptides consist in that they are orally active owing to higher stability and ability to penetrate biological barriers due to the presence of specific ATP-dependent transporters in enterocytes and blood-brain barrier. Two original approaches for dipeptide drugs design have been developed. Both of them are based on the idea of a leading role of central dipeptide fragment of the peptide chain beta-turn in the peptide-receptor interaction. The first approach, named "peptide drug-based design" represents the transformation of known nonpeptide drug into its dipeptide topological analog. The latter usually corresponds to a beta-turn of some regulatory peptide. The second approach represents the design of tripeptoide mimetic of the beta-turn of regulatory peptide or protein. The results of the studies, which led to the discovery of endogenous prototypes of the known non-peptide drugs piracetam and sulpiride, are presented herein. The paper discusses the process, based on the abovementioned principles, that was used in designing of nontoxic, orally available, highly effective dipeptide drugs: nootropic noopept, dipeptide analog of piracetam; antipsychotic dilept, neurotensin tripeptoid analog; selective anxiolytic GB-115, tripeptoid analog of CCK-4, and potential neuroprotector GK-2, homodimeric dipeptide analog of NGF.


Asunto(s)
Dipéptidos/síntesis química , Diseño de Fármacos , Administración Oral , Dipéptidos/administración & dosificación , Dipéptidos/química , Humanos , Estructura Molecular
5.
J Biomed Sci ; 21: 74, 2014 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-25096780

RESUMEN

BACKGROUND: Noopept (N-phenyl-acetyl-L-prolylglycine ethyl ester) was constructed as a dipeptide analog of the standard cognition enhancer, piracetam. Our previous experiments have demonstrated the cognition restoring effect of noopept in several animal models of Alzheimer disease (AD). Noopept was also shown to prevent ionic disbalance, excitotoxicity, free radicals and pro-inflammatory cytokines accumulation, and neurotrophine deficit typical for different kinds of brain damages, including AD. In this study, we investigated the neuroprotective action of noopept on cellular model of AD, Aß 25-35-induced toxicity in PC12 cells and revealed the underlying mechanisms. RESULTS: The neuroprotective effect of noopept (added to the medium at 10 µM concentration, 72 hours before Ðß 25-35) was studied on Ðß 25-35-induced injury (5 µM for 24 h) in PC12 cells. The ability of drug to protect the impairments of cell viability, calcium homeostasis, ROS level, mitochondrial function, tau phosphorylation and neurite outgrowth caused by Ðß 25-35 were evaluated. Following the exposure of PC12 cells to Ðß 25-35 an increase of the level of ROS, intracellular calcium, and tau phosphorylation at Ser396 were observed; these changes were accompanied by a decrease in cell viability and an increase of apoptosis. Noopept treatment before the amyloid-beta exposure improved PC12 cells viability, reduced the number of early and late apoptotic cells, the levels of intracellular reactive oxygen species and calcium and enhanced the mitochondrial membrane potential. In addition, pretreatment of PC12 cell with noopept significantly attenuated tau hyperphosphorylation at Ser396 and ameliorated the alterations of neurite outgrowth evoked by Аß25-35. CONCLUSIONS: Taken together, these data provide evidence that novel cognitive enhancer noopept protects PC12 cell against deleterious actions of Aß through inhibiting the oxidative damage and calcium overload as well as suppressing the mitochondrial apoptotic pathway. Moreover, neuroprotective properties of noopept likely include its ability to decrease tau phosphorylation and to restore the altered morphology of PC12 cells. Therefore, this nootropic dipeptide is able to positively affect not only common pathogenic pathways but also disease-specific mechanisms underlying Aß-related pathology.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis/efectos de los fármacos , Dipéptidos/farmacología , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Proteínas tau/metabolismo , Enfermedad de Alzheimer , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Células PC12 , Fosforilación , Ratas
6.
J Psychopharmacol ; 21(6): 611-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17092975

RESUMEN

The effects of the novel proline-containing nootropic and neuroprotective dipeptide, noopept (GVS-111, N-phenylacetyl-L-prolylglycine ethyl ester) were investigated in NMRI mice following olfactory bulbectomy. We have shown previously that these animals developed Alzheimer's disease (AD)-like behaviour, morphology and biochemistry including impairment of spatial memory, regional neuronal degeneration and elevated Abeta peptide brain levels. In the current investigation, spatial memory was assessed using the Morris water maze and serum antibodies to in vitro morphologically characterized amyloid structures of both Abeta((25-35)) peptide and equine lysozyme, as well as to neurotrophic glial factor S100b, were analyzed by enzyme-linked immunosorbent assay (ELISA). Noopept (administered at a dose of 0.01 mg/kg for a period of 21 days and during a further 5 days training) restored spatial memory and increased serum antibody levels to oligomers of Abeta((25-35)) peptide but not to equine lysozyme amyloid or S100b protein in bulbectomized animals. The positive immunotropic effect of noopept to Abeta((25-35)) peptide prefibrillar aggregates was more marked in sham-operated compared to the bulbectomized subjects which were characterized by an overall suppression of immunoreactivity. Enhancement of the immune response to Abeta((25-35)) peptide prefibrils caused by noopept may attenuate the neurotoxic consequences of amyloid fibrillization and also be associated with an improvement in spatial memory in bulbectomized mice. These actions of noopept, combined with its previously reported neuroprotective and cholinomimetic properties, suggests that this dipeptide may well be useful for improving cognitive deficits induced by neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/inmunología , Autoanticuerpos/sangre , Conducta Animal/efectos de los fármacos , Dipéptidos/farmacología , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Fragmentos de Péptidos/inmunología , Percepción Espacial/efectos de los fármacos , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/psicología , Animales , Dipéptidos/uso terapéutico , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Masculino , Ratones , Microscopía de Fuerza Atómica , Muramidasa/inmunología , Factores de Crecimiento Nervioso/inmunología , Fármacos Neuroprotectores/uso terapéutico , Nootrópicos/uso terapéutico , Bulbo Olfatorio/cirugía , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/inmunología , Factores de Tiempo
7.
Int J Dev Neurosci ; 21(3): 117-24, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12711349

RESUMEN

The neuroprotective activity of a novel N-acylprolyl-containing dipeptide analog of the nootropic 2-oxo-1-pyrrolidine acetamide (Piracetam) designated as GVS-111 (DVD-111/Noopept) was tested in two in vitro models of neuronal degeneration mediated by oxidative stress: normal human cortical neurons treated with H(2)O(2), and Down's syndrome (DS) cortical neurons. Incubation of normal cortical neurons with 50 microM H(2)O(2) for 1h resulted in morphological and structural changes consistent with neuronal apoptosis and in the degeneration of more than 60% of the neurons present in the culture. GVS-111 significantly increased neuronal survival after H(2)O(2)-treatment displaying a dose-dependent neuroprotective activity from 10nM to 100 microM, and an IC(50) value of 1.21+/-0.07 microM. GVS-111 inhibited the accumulation of intracellular free radicals and lipid peroxidation damage in neurons treated with H(2)O(2) or FeSO(4), suggesting an antioxidant mechanism of action. GVS-111 exhibited significantly higher neuroprotection compared to the standard cognition enhancer Piracetam, or to the antioxidants Vitamin E, propyl gallate and N-tert-butyl-2-sulpho-phenylnitrone (s-PBN). In DS cortical cultures, chronic treatment with GVS-111 significantly reduced the appearance of degenerative changes and enhanced neuronal survival. The results suggest that the neuroprotective effect of GVS-111 against oxidative damage and its potential nootropic activity may present a valuable therapeutic combination for the treatment of mental retardation and chronic neurodegenerative disorders.


Asunto(s)
Corteza Cerebral/patología , Dipéptidos/farmacología , Síndrome de Down/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Feto Abortado/efectos de los fármacos , Feto Abortado/patología , Feto Abortado/fisiopatología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Corteza Cerebral/fisiopatología , Síndrome de Down/embriología , Síndrome de Down/fisiopatología , Femenino , Compuestos Ferrosos/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Embarazo , Valores de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA