Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell Mol Neurobiol ; 41(3): 605-613, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32410106

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares/metabolismo , Marcha , Memoria , Enfermedad de Parkinson/fisiopatología , Enfermedad de Parkinson/terapia , Administración Intranasal , Animales , Conducta Animal , Niño , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Vesículas Extracelulares/ultraestructura , Femenino , Humanos , Masculino , Cuerpos de Nissl/metabolismo , Oxidopamina , Enfermedad de Parkinson/patología , Ratas Wistar , Sustancia Negra/patología , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
2.
J Neurosci Res ; 97(6): 708-726, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30742328

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Ácido gamma-Aminobutírico/química , Ácido gamma-Aminobutírico/metabolismo , Acetilcolinesterasa/metabolismo , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Encefalitis/metabolismo , Glutamato Descarboxilasa/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Microglía/efectos de los fármacos , Mitocondrias/metabolismo , Ratas Wistar , Receptores de GABA/metabolismo , Ácido gamma-Aminobutírico/administración & dosificación
3.
Medicina (Kaunas) ; 55(7)2019 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-31330913

RESUMEN

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.


Asunto(s)
Antipsicóticos/análisis , Oligopéptidos/uso terapéutico , Administración Intranasal , Anfetaminas/administración & dosificación , Anfetaminas/efectos adversos , Anfetaminas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos ICR/metabolismo , Actividad Motora/efectos de los fármacos , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología
4.
Pharmacol Res ; 113(Pt B): 754-759, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27234899

RESUMEN

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.


Asunto(s)
Dihidropiridinas/farmacología , Dihidropiridinas/uso terapéutico , Memoria/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Animales , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Humanos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Neurotransmisores/metabolismo
5.
Pharmacol Res ; 113(Pt B): 781-787, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27345857

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Bloqueadores de los Canales de Calcio/farmacología , Calcio/metabolismo , Giro del Cíngulo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Memoria/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ansiolíticos/farmacología , Barrera Hematoencefálica/metabolismo , Dihidropiridinas/farmacología , Modelos Animales de Enfermedad , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Glutamato Descarboxilasa/metabolismo , Giro del Cíngulo/metabolismo , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
6.
J Neurosci Res ; 92(3): 338-46, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24273007

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Trastornos del Conocimiento/tratamiento farmacológico , Metilhidrazinas/uso terapéutico , Acetilcolinesterasa/metabolismo , Adyuvantes Inmunológicos/farmacología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato Descarboxilasa/metabolismo , Humanos , Locomoción/efectos de los fármacos , Locomoción/genética , Metilhidrazinas/farmacología , Ratones , Ratones Transgénicos , Conducta Social
7.
BMC Neurosci ; 14: 54, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23672601

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/metabolismo , Animales , Antidepresivos/farmacología , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/efectos de los fármacos , Embrión de Mamíferos , Femenino , Privación de Alimentos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas de Transporte de Neurotransmisores en la Membrana Plasmática/genética , Embarazo , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo
8.
Medicina (Kaunas) ; 49(7): 301-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24375241

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Aprendizaje/efectos de los fármacos , Metilhidrazinas/farmacología , Mitocondrias/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Mitocondrias/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Neuritis/metabolismo , Neuritis/patología , Neuronas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ratas
9.
Medicina (Kaunas) ; 48(10): 525-31, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23324249

RESUMEN

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.


Asunto(s)
Dihidropiridinas/uso terapéutico , Metilhidrazinas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/prevención & control , Animales , Dihidropiridinas/química , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Endotelina-1/farmacología , Masculino , Metilhidrazinas/química , Fármacos Neuroprotectores/química , Ratas , Ratas Wistar , Accidente Cerebrovascular/inducido químicamente
10.
Medicina (Kaunas) ; 47(10): 552-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22186119

RESUMEN

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.


Asunto(s)
Fármacos Cardiovasculares/administración & dosificación , Metilhidrazinas/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Factor Neurotrófico Derivado de la Línea Celular Glial/antagonistas & inhibidores , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/biosíntesis , Masculino , Moléculas de Adhesión de Célula Nerviosa/antagonistas & inhibidores , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Oxidopamina/antagonistas & inhibidores , Oxidopamina/farmacología , Enfermedad de Parkinson Secundaria/metabolismo , Ratas , Ratas Wistar , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo
11.
Int J Mol Sci ; 11(11): 4465-87, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-21151450

RESUMEN

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.


Asunto(s)
Metilhidrazinas/farmacología , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson Secundaria/metabolismo , Animales , Biomarcadores/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Metilhidrazinas/uso terapéutico , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oxidopamina/toxicidad , Ratas , Ratas Wistar , Receptor Notch3 , Receptores Notch/genética , Receptores Notch/metabolismo , Sustancia Negra/citología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
12.
Eur J Pharmacol ; 881: 173290, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32580040

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Hipoglucemiantes/farmacología , Metformina/farmacología , Fármacos Neuroprotectores/farmacología , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/psicología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipoglucemiantes/administración & dosificación , Inyecciones Intraventriculares , Masculino , Metformina/administración & dosificación , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuroglía/patología , Fármacos Neuroprotectores/administración & dosificación , Ratas Wistar , Conducta Social , Estreptozocina
13.
Pharmacol Biochem Behav ; 92(2): 267-71, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19135472

RESUMEN

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.


Asunto(s)
Ansiedad/inducido químicamente , Etanol/efectos adversos , Aprendizaje por Laberinto , Hormonas Estimuladoras de los Melanocitos/farmacología , Síndrome de Abstinencia a Sustancias/etiología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos ICR
14.
Neuropharmacology ; 144: 319-326, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30408486

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Diazepam/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Acetilcolina/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Gliosis/tratamiento farmacológico , Gliosis/metabolismo , Gliosis/patología , Glutamato Descarboxilasa/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Distribución Aleatoria , Ratas Wistar , Sinaptofisina/metabolismo , Ácido gamma-Aminobutírico/metabolismo
15.
Stem Cells Transl Med ; 8(5): 490-499, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30706999

RESUMEN

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.


Asunto(s)
Administración Intranasal/métodos , Vesículas Extracelulares/metabolismo , Microscopía Electrónica de Transmisión/métodos , Oxidopamina/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Células Madre/metabolismo , Diente/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo , Anciano , Animales , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Oxidopamina/farmacología , Enfermedad de Parkinson/patología , Ratas , Ratas Wistar , Sustancia Negra/patología
16.
J Mol Neurosci ; 35(2): 179-93, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18418736

RESUMEN

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.


Asunto(s)
Sistema de Transporte de Aminoácidos A/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos/genética , Evolución Molecular , Proteínas del Tejido Nervioso/genética , Filogenia , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Animales , Anticonvulsivantes/farmacología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiología , Diazepam/farmacología , Dopaminérgicos/farmacología , Sistema Endocrino/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Hibridación in Situ , Levodopa/farmacología , Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos , Músculo Esquelético/fisiología , Ratas , Ratas Wistar
17.
Neurotoxicology ; 29(1): 101-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17981335

RESUMEN

N-Methyl-d-aspartate (NMDA) receptor stimulation promotes neuronal survival and differentiation under both in vitro and in vivo conditions. We studied the effects of various NMDA receptor antagonists acting at different NMDA receptor binding sites and non-NMDA receptor antagonists on the development and survival of cerebellar granule cell (CGC) culture. Only three of the drugs tested induced neurotoxicity-MK-801 (non-competitive NMDA channel blocking antagonist), ifenprodil (an antagonist of the NR2B site and polyamine site of the NMDA receptor) and L-701.324 (full antagonist at glycine site), while CGP-37849 (a competitive NMDA antagonist), (+)-HA-966 (a partial agonist of the glycine site of the NMDA receptor), and NBQX (a competitively acting AMPA receptor antagonist) were not toxic at any concentration (1-100 microM) used. Among these drugs, only MK-801 was toxic for the immature CGC on second day in vitro (2DIV), and toxicity was diminished parallel to the neuronal maturation. In more mature neurons (7DIV), MK-801 demonstrated some neuroprotection, which diminished spontaneously occurring neuronal death in culture. Neither NMDA nor glutamate were able to prevent the neurotoxic effect of MK-801 at 2DIV. MK-801, ifenprodil and L-701.324 induced DNA fragmentation on 2DIV in CGC culture measured by the TUNEL method. The BOC-D-FMK, the universal caspase inhibitor, completely reversed MK-801-induced DNA fragmentation, suggesting an apoptotic pathway of MK-801-induced cell death. Neurite outgrowth as a characteristic feature of the development of CGC was diminished after treatment with MK-801, ifenprodil and L-701.324. In conclusion, the results of the present study demonstrate that only nonselective channel blocker MK-801 decreases cell viability, induces apoptosis and inhibits neurite outgrowth of CGC in a development-dependent manner.


Asunto(s)
Cerebelo/citología , Antagonistas de Aminoácidos Excitadores/farmacología , Neuronas/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/farmacología , Etiquetado Corte-Fin in Situ , N-Metilaspartato/farmacología , Ratas , Ratas Wistar , Sales de Tetrazolio , Tiazoles , Factores de Tiempo
18.
Pharmacol Biochem Behav ; 90(4): 712-6, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18582921

RESUMEN

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.


Asunto(s)
Anticonvulsivantes/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Receptores de GABA/metabolismo , Triterpenos/metabolismo , Triterpenos/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Bicuculina/farmacología , Flunitrazepam/metabolismo , Moduladores del GABA/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Tono Muscular/efectos de los fármacos , Triterpenos Pentacíclicos , Equilibrio Postural/efectos de los fármacos , Convulsiones/inducido químicamente , Convulsiones/psicología , Ácido gamma-Aminobutírico/metabolismo , Ácido Betulínico
19.
Cell Biochem Funct ; 26(5): 620-31, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18508390

RESUMEN

Previously mildronate, an aza-butyrobetaine derivative, was shown to be a cytoprotective drug, through its mechanism of action of inhibition of carnitine palmitoyltransferase-1, thus protecting mitochondria from long-chain fatty acid accumulation and subsequent damage. Recently in an azidothymidine (AZT)-induced cardiotoxicity model in vivo (in mice), we have found mildronate's ability of protecting heart tissue from nuclear factor kappaB abnormal expression. Preliminary data also demonstrate cerebro- and hepatoprotecting properties of mildronate in AZT-toxicity models. We suggest that mildronate may target its action predominantly to mitochondria. The present study in isolated rat liver mitochondria was designed to clarify mitochondrial targets for mildronate by using AZT as a model compound. The aim of this study was to investigate: (1) whether mildronate may protect mitochondria from AZT-induced toxicity; and (2) which is the most critical target in mitochondrial processes that is responsible for mildronate's regulatory action. The results showed that mildronate protected mitochondria from AZT-induced damage predominantly at the level of complex I, mainly by reducing hydrogen peroxide generation. Significant protection of AZT-caused inhibition of uncoupled respiration, ADP to oxygen ratio, and transmembrane potential were also observed. Mildronate per se had no effect on the bioenergetics, oxidative stress, or permeability transition of rat liver mitochondria. Since mitochondrial complex I is the first enzyme of the respiratory electron transport chain and its damage is considered to be responsible for different mitochondrial diseases, we may account for mildronate's effectiveness in the prevention of pathologies associated with mitochondrial dysfunctions.


Asunto(s)
Antimetabolitos/farmacología , Sistemas de Liberación de Medicamentos , Metilhidrazinas/farmacología , Mitocondrias Hepáticas/efectos de los fármacos , Enfermedades Mitocondriales/patología , Zidovudina/antagonistas & inhibidores , Zidovudina/toxicidad , Animales , Respiración de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Técnicas In Vitro , Masculino , Mitocondrias Hepáticas/patología , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/metabolismo , Permeabilidad/efectos de los fármacos , Ratas , Ratas Wistar
20.
Eur J Pharmacol ; 818: 381-399, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29133125

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Baclofeno/farmacología , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Muscimol/farmacología , Estreptozocina/efectos adversos , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratas , Ratas Wistar , Aprendizaje Espacial/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA