Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuroreport ; 32(6): 518-524, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33788819

RESUMEN

Achyranthes bidentata polypeptide k (ABPPk), a powerful active component from a traditional Chinese medicinal herb-Achyranthes bidentata Bl., has exhibited promising neuroprotective activity due to its multiple-targeting capability. However, the effect of ABPPk on the survival, growth and axonal regeneration of spinal cord motor neurons remains unclear. Here, a modified method, which is more optimized for embryonic cells in ambient carbon dioxide levels, was used for acquisition of rat embryonic spinal cord motor neurons with high survival and purity. ABPPk concentration-dependently enhanced the neuronal viability and promoted the neurite outgrowth. Co-culture of motor neurons and skeletal myocytes model indicated that ABPPk enhanced the neuromuscular junction development and maturation. A microfluidic axotomy model was further established for the axonal disconnection, and ABPPk significantly accelerated the axonal regeneration of motor neurons. Furthermore, we demonstrated that the upregulation of three neurofilament protein subunits in motor neurons might be relevant to the mechanisms of the growth-promoting effect of ABPPk. Our findings provide an experimental and theoretical basis for the development of ABPPk as a potential application in the development of treatment strategy for nerve injury diseases.


Asunto(s)
Achyranthes , Axones/efectos de los fármacos , Neuronas Motoras/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Unión Neuromuscular/efectos de los fármacos , Proyección Neuronal/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Axotomía , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Técnicas In Vitro , Proteínas de Neurofilamentos/efectos de los fármacos , Proteínas de Neurofilamentos/metabolismo , Péptidos/farmacología , Traumatismos de los Nervios Periféricos , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología
2.
Med Sci Monit ; 25: 4627-4638, 2019 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-31266934

RESUMEN

BACKGROUND Subclinical epileptiform discharges (SEDs) are defined as epileptiform electroencephalographic (EEG) discharges without clinical signs of seizure in patients. The subthreshold convulsant discharge (SCD) is a frequently used model for SEDs. This study aimed to investigate the effect of levetiracetam (LEV), an anti-convulsant drug, on cognitive impairment of SCD model rats and to assess the associated mechanisms. MATERIAL AND METHODS A SCD rat model was established. Rats were divided into an SCD group, an SCD+ sodium valproate (VPA) group, and an SCD+ levetiracetam (LEV) group. The Morris water maze was used to evaluate the capacity of positioning navigation and space exploration. The field excitatory post-synaptic potentials (fEPSPs) were evaluated using a bipolar stimulation electrode. NCAM, GAP43, PS95, and CaMK II levels were detected using Western blot and RT-PCR, respectively. PKC activity was examined by a non-radioactive method. RESULTS LEV shortens the latency of platform seeking in SCD rats in positioning navigation. fEPSP slopes were significantly lower in the SCD group, and LEV treatment significantly enhanced the fEPSP slopes compared to the SCD group (P<0.05). The NCAM and GAP-43 levels were increased and PSD-95 levels were increased in SCD rats (P<0.05), which were improved by LEV treatment. The PKC activity and CaMK II levels were decreased in SCD rats and LEV treatment significantly enhanced PKC activity and increased CaMK II levels. CONCLUSIONS Cognitive impairment in of SCD model rats may be caused by decreased PKC activity, low expression of CaMK II, and inhibition of LTP formation. LEV can improve cognitive function by activating the PKC-GAP-43-CaMK signal transduction pathway.


Asunto(s)
Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/prevención & control , Levetiracetam/farmacología , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Modelos Animales de Enfermedad , Electroencefalografía , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Levetiracetam/metabolismo , Masculino , Fosforilación , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Convulsiones/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Ácido Valproico/uso terapéutico
3.
Behav Brain Res ; 362: 208-212, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30654123

RESUMEN

Anodal Direct Current Stimulation (DC) over prefrontal cortex improves working memory. This study investigated the influence of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) on prefrontal anodal DC-induced effects on spatial working memory and GAP-43 expression. Male Wistar rats well-trained in radial maze procedures received five sessions of anodal epidural DC stimulation (eDCS: 400 µA, 13 min, one daily session) over the left mPFC, or a respective sham procedure, and afterwards they received a single dose (1 mg/kg) of perampanel (PRP), an AMPARs antagonist, or vehicle 30 min before the performance of 4-h delayed task. The prefrontal cortex (PFC) and hippocampus (HPC) were removed 24-h later and GAP-43 (growth-associated protein) expression was measured by Western blot analysis. Repetitive eDCS decreased the number of errors in the 4-h post-delay performance (p < 0.05) and increased the expression of GAP-43 (p < 0.01) in the PFC when compared to sham stimulation. These behavioral and prefrontal molecular changes induced by the repetitive eDCS seem to involve AMPAR activity, because they were abolished when AMPARs were blocked by PRP (p < 0.01 and 0.05, respectively). Besides, in the HPC, changes of GAP-43 expression induced by eDCS was only seen when AMPARs were blocked by PRP. Therefore, the neuronal plasticity involving AMPARs may underlie, at least in part, the improving of spatial working memory and GAP-43 expression induced by the repetitive anodal prefrontal DC stimulation.


Asunto(s)
Proteína GAP-43/efectos de los fármacos , Memoria a Largo Plazo/efectos de los fármacos , Piridonas/farmacología , Receptores AMPA/antagonistas & inhibidores , Animales , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Nitrilos , Corteza Prefrontal/efectos de los fármacos , Ratas Wistar , Estimulación Transcraneal de Corriente Directa/métodos
4.
Neurochem Res ; 43(10): 1905-1913, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30088237

RESUMEN

Luteolin, a flavonoid isolated from Cirsium japonicum, has antioxidant, anti-inflammatory and neuroprotective activities. Our previous studies brought a prospect that luteolin benefited diabetic rats with cognitive impairments. In this study, we examined whether luteolin could suppress the inflammatory cytokines, thus increasing synapse-associated proteins in streptozotocin (STZ)-induced diabetes in rat models. The model rats underwent luteolin treatment for 8 consecutive weeks, followed by assessment of cognitive performances with MWM test. Nissl staining was employed to assess the neuropathological changes in the hippocampus and the effects of luteolin on diabetic rats. With animals sacrificed, expressions of inflammatory cytokines including interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) and synapse-associated proteins including growth-associated protein-43 (GAP-43) and synaptophysin (SYN) were determined. The results affirmed improvement of behavioral performances in the MWM test, downexpression of glycation end products (AGEs) in the plasma and the receptor for advanced glycation end products in the hippocampus, inhibition of IL-1ß and TNF-α in both the hippocampus and plasma in diabetic rats. Furthermore, luteolin treatment upregulated the expressions of GAP-43 and SYN in the hippocampus. Thus, luteolin could ameliorate the cognitive dysfunctions in STZ-induced diabetic rat model.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Proteína GAP-43/efectos de los fármacos , Luteolina/farmacología , Sinaptofisina/metabolismo , Animales , Disfunción Cognitiva/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Proteína GAP-43/metabolismo , Inflamación/tratamiento farmacológico , Masculino , Ratas Sprague-Dawley , Estreptozocina/farmacología , Sinaptofisina/efectos de los fármacos
5.
Tsitologiia ; 57(2): 144-52, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26035972

RESUMEN

We have studied the steady-state transmembrane current induced by amyloid and amyloid-like peptides in lipid bilayers in the presence of dipole modifiers. It has been shown that the addition of dipole modifier, phloretin, to the membrane bathing solutions leads to an increase in the multichannel activity of amyloid beta-peptide fragment 25-35, [Gly35]-amyloid beta-peptide fragment 25--35, prion protein fragment 106-126 and amyloid-like peptides myr-BASP1 (1--13), myr-BASP1(1--19) and GAP-43(1--40). We have found that the effect of phloretin is not the result of dipole potential changes due to adsorption of this modifier on the membrane. Using the various fragments of amyloid beta-peptide, presenilin, prion protein and neuronal proteins BASP1 and GAP-43 allowes to conclude that the steady-state peptide-induced transmembrane current in the case of addition of phloretin is due to the electrostatic interaction between the positively charged channel-forming agents and negatively charged dipole modifier. The results obtained by electron microscopy have demonstrated that this interaction increases degree of peptide oligomerization.


Asunto(s)
Péptidos beta-Amiloides/ultraestructura , Proteína GAP-43/ultraestructura , Proteínas de la Membrana/ultraestructura , Proteínas del Tejido Nervioso/ultraestructura , Fragmentos de Péptidos/ultraestructura , Priones/ultraestructura , Proteínas Represoras/ultraestructura , Péptidos beta-Amiloides/efectos de los fármacos , Proteína GAP-43/efectos de los fármacos , Humanos , Membrana Dobles de Lípidos/síntesis química , Membrana Dobles de Lípidos/metabolismo , Potenciales de la Membrana , Proteínas de la Membrana/efectos de los fármacos , Microscopía Electrónica , Proteínas del Tejido Nervioso/efectos de los fármacos , Fragmentos de Péptidos/efectos de los fármacos , Floretina/farmacología , Priones/efectos de los fármacos , Proteínas Represoras/efectos de los fármacos , Electricidad Estática
6.
Eur J Pain ; 18(10): 1480-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24756904

RESUMEN

BACKGROUND: Botulinum toxin A (Onabot/A) has been shown to have an antinociceptive effect. This might be due to an impairment of sensory nerves not only in the peripheral but also in the central nervous system. In this work, we analysed both systems by studying the effect of intrathecal (i.t.) administration of botulinum toxin A in an animal model of bladder pain and hyperactivity induced by cyclophosphamide (CYP). METHODS: Rats were implanted with an i.t. catheter at the L6 segment. Bladder pain was induced by intraperitoneal (i.p.) injection of CYP. Five experimental groups were created: (1) Saline i.p. + i.t.; (2) Onabot/A i.t.; (3) CYP i.p. + saline i.t.; (4) CYP i.p. + Onabot/A i.t. 48 h after CYP; and (5) Onabot/A i.t. 30 days. Mechanical sensitivity was assessed in the abdomen and hindpaws. Motor activity was observed in an open-field arena. Bladder reflex activity was evaluated by cystometry. At the end, bladders and spinal cord were immunoreacted (IR) against cleaved SNAP-25 (cSNAP-25), c-Fos, p-ERK, calcitonin gene-related peptide (CGRP) and GAP43. RESULTS: The toxin reduced pain symptoms, bladder hyperactivity, expression of neuronal activation markers and CGRP, typically up-regulated in this inflammatory model. The presence of cSNAP-25 was detected in the spinal cord and bladder fibres from animals treated with Onabot/A. No somatic or visceral motor impairments were observed. CONCLUSIONS: Our findings suggest that i.t. Onabot/A has a strong analgesic effect in a model of severe bladder pain. This route of administration can be further explored to treat intractable forms of pain.


Asunto(s)
Inhibidores de la Liberación de Acetilcolina/farmacología , Toxinas Botulínicas Tipo A/farmacología , Cistitis Intersticial/fisiopatología , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/fisiopatología , Médula Espinal/efectos de los fármacos , Vejiga Urinaria/efectos de los fármacos , Inhibidores de la Liberación de Acetilcolina/administración & dosificación , Animales , Toxinas Botulínicas Tipo A/administración & dosificación , Péptido Relacionado con Gen de Calcitonina/efectos de los fármacos , Péptido Relacionado con Gen de Calcitonina/metabolismo , Ciclofosfamida/envenenamiento , Cistitis Intersticial/inducido químicamente , Modelos Animales de Enfermedad , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Inmunohistoquímica , Inyecciones Espinales , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Médula Espinal/metabolismo , Proteína 25 Asociada a Sinaptosomas/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas/metabolismo , Vejiga Urinaria/metabolismo , Vejiga Urinaria/fisiopatología
7.
Muscle Nerve ; 47(6): 909-15, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23666783

RESUMEN

INTRODUCTION: Both target skeletal muscle (SKM) cells and neurotrophins (NTs) are essential for the maintenance of neuronal function and nerve-muscle communication. The effects of different NTs and SKM cells on growth-associated protein-43 (GAP-43) expression in dorsal root ganglion (DRG) neurons have not been clarified. METHODS: The morphological relationship between DRG neurons and SKM cells in neuromuscular cocultures was observed by scanning electron microscopy. The levels of GAP-43 and its mRNA were determined after administration of different NTs. RESULTS: DRG neurons demonstrated dense neurite outgrowth in the presence of NTs. Distinct NTs promoted GAP-43 and its mRNA expression in neuromuscular cocultures of DRG neurons and SKM cells. CONCLUSIONS: These results offer new clues for a better understanding of the effects of distinct NTs on GAP-43 expression in DRG sensory neurons in the presence of target SKM cells and implicate NTs and target SKM cells in DRG neuronal regeneration.


Asunto(s)
Proteína GAP-43/metabolismo , Ganglios Espinales/citología , Fibras Musculares Esqueléticas/fisiología , Factores de Crecimiento Nervioso/farmacología , Células Receptoras Sensoriales/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células Cultivadas , Técnicas de Cocultivo , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/fisiología , Microscopía Electrónica de Rastreo , Factor de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Neurotrofina 3/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/ultraestructura
8.
Neurochem Int ; 55(6): 397-405, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19409439

RESUMEN

Methamphetamine (METH) is a most commonly abused drug which damages nerve terminals by causing formation of reactive oxygen species (ROS), apoptosis, and finally neuronal damage. Fetal exposure to neurotoxic METH causes significant behavioral effects. The developing fetus is substantially deficient in most antioxidative enzymes, and may therefore be at high risk from both endogenous and drug-enhanced oxidative stress. Little is known about the effects of METH on vesicular proteins such as synaptophysin and growth-associated protein 43 (GAP-43) in the immature brain. The present study attempted to investigate the effects of METH-induced neurotoxicity in the dopaminergic system of the neonatal rat brain. Neonatal rats were subcutaneously exposed to 5-10mg/kg METH daily from postnatal day 4-10 for 7 consecutive days. The results showed that tyrosine hydroxylase enzyme levels were significantly decreased in the dorsal striatum, prefrontal cortex, nucleus accumbens and substantia nigra, synaptophysin levels decreased in the striatum and prefrontal cortex and growth-associated protein-43 (GAP-43) levels significantly decreased in the nucleus accumbens of neonatal rats. Pretreatment with 2mg/kg melatonin 30 min prior to METH administration prevented METH-induced reduction in tyrosine hydroxylase, synaptophysin and growth-associated protein-43 protein levels in different brain regions. These results suggest that melatonin provides a protective effect against METH-induced nerve terminal degeneration in the immature rat brain probably via its antioxidant properties.


Asunto(s)
Química Encefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Melatonina/farmacología , Metanfetamina/antagonistas & inhibidores , Fármacos Neuroprotectores/farmacología , Trastornos Relacionados con Anfetaminas/tratamiento farmacológico , Trastornos Relacionados con Anfetaminas/metabolismo , Trastornos Relacionados con Anfetaminas/fisiopatología , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Encéfalo/metabolismo , Química Encefálica/fisiología , Estimulantes del Sistema Nervioso Central/efectos adversos , Dopamina/biosíntesis , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Interacciones Farmacológicas/fisiología , Femenino , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Metanfetamina/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Ratas , Ratas Wistar , Sinaptofisina/efectos de los fármacos , Sinaptofisina/metabolismo , Tirosina 3-Monooxigenasa/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo , Degeneración Walleriana/inducido químicamente , Degeneración Walleriana/tratamiento farmacológico , Degeneración Walleriana/prevención & control
9.
Arch Toxicol ; 83(4): 373-80, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18633600

RESUMEN

The aim of this study was to assess the neurotoxicity of diazinon oxon (DZO), a major in vivo metabolite of the phosphorothionate insecticide diazinon (DZ), on differentiating mouse N2a neuroblastoma cells. When used at concentrations of 1, 5 and 10 microM, DZO did not cause cell death but it impaired the outgrowth of axon-like processes after 24 h. Densitometric scanning of Western blots of lysates of N2a cells revealed that exposure to 5 or 10 microM DZO for 24 h increased the expression of phosphorylated neurofilament heavy chain (NFH) compared to controls, while there was no significant change in total NFH. By contrast, treatment of N2a cells with 1-10 microM DZO resulted in marked reductions in the expression of the axon growth-associated protein GAP-43. DZO-treated cells also showed an increased expression of the heat shock protein HSP-70 compared to controls. The above biochemical changes were not temporally related to inhibition of acetylcholinesterase (AChE). These data suggest that biologically relevant, subcytotoxic levels of DZO may exert neurotoxic effects on differentiating cells and that the mechanisms involved are different from those attributed to its parent compound.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diazinón/análogos & derivados , Diazinón/toxicidad , Insecticidas/toxicidad , Neuritas/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Proteínas HSP70 de Choque Térmico/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Neuroblastoma , Proteínas de Neurofilamentos/efectos de los fármacos , Proteínas de Neurofilamentos/metabolismo , Neuronas/metabolismo , Neuronas/patología
10.
J Neurochem ; 103(3): 871-81, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17683485

RESUMEN

Oleic acid synthesized by astrocytes behaves as a neurotrophic factor for neurons, up-regulating the molecular markers of axonal and dendritic outgrowth, growth-associated protein 43 and microtubule-associated protein 2. In this work, the nature of the receptor involved in this neurotrophic effect was investigated. As oleic acid has been reported to be a ligand and activator of the peroxisome proliferator-activated receptor (PPAR), we focus on this family of receptors. Our results show that PPARalpha, beta/delta, and gamma are expressed in neurons in culture. However, only the agonists of PPARalpha, Wy14643, GW7647 and oleoylethanolamide, promoted neuronal differentiation, while PPAR beta/delta and gamma agonists did not modify neuronal differentiation. Consequently, we investigated the involvement of PPARalpha (Nr1c1) in oleic acid-induced neuronal differentiation. Our results indicate that oleic acid activates PPARalpha in neurons. In addition, the effect of oleic acid on neuronal morphology, growth-associated protein 43 and microtubule-associated protein 2 expression decreases in neurons after PPARalpha has been silenced by small interfering RNA. Taken together, our results suggest that PPARalpha could be the receptor for oleic acid in neurons, further broadening the range of functions attributed to this family of transcription factors. Although several works have reported that PPARalpha could be involved in neuroprotection, the present work provides the first evidence suggesting a role of PPARalpha in neuronal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Ácido Oléico/metabolismo , PPAR alfa/metabolismo , Animales , Butiratos/farmacología , Diferenciación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Forma de la Célula/fisiología , Células Cultivadas , Sistema Nervioso Central/citología , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/metabolismo , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Factores de Crecimiento Nervioso/farmacología , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Ácido Oléico/farmacología , PPAR alfa/agonistas , PPAR alfa/genética , Compuestos de Fenilurea/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar
11.
Exp Neurol ; 206(2): 209-19, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17580085

RESUMEN

A 25-35% reduction of brain cytochrome oxidase (COx) activity found in Alzheimer's disease (AD) could contribute to neuronal dysfunction and cognitive impairment. The present study replicated the reduction in brain COx activity in rats by administering sodium azide (NaN(3)) for 4 weeks via Alzet minipumps at the rate of 1 mg/kg/h, and determined its effect on hippocampal cholinergic transmission, spatial and episodic memory. NaN(3) caused a selective reduction in choline acetyltransferase (ChAT) immunoreactivity in the diagonal band, a major source of cholinergic input to the hippocampus and cingulate cortex, without altering the number of cholinergic neurons. NaN(3) also induced a significant increase in vesicular acetylcholine transporter (VAChT)-immunoreactive varicosities, GAP-43 in the subgranular layer and of transferrin receptors (TfR) in the hilus of the dentate gyrus. These neurochemical changes were associated with impairment in spatial learning in the Morris water maze and in episodic memory in the object recognition test. Chronic treatment with ladostigil, a novel cholinesterase and monoamine oxidase inhibitor, prevented the decrease in ChAT in the diagonal band, the compensatory increase in synaptic plasticity and TfR and the memory deficits without restoring COx activity. Ladostigil had no significant effect on ChAT activity, synaptic plasticity or TfR in control rats. Ladostigil may have a beneficial effect on cognitive deficits in AD patients that have a reduction in cortical COx activity and cholinergic hypofunction.


Asunto(s)
Acetilcolina/metabolismo , Fibras Colinérgicas/efectos de los fármacos , Complejo IV de Transporte de Electrones/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Indanos/farmacología , Trastornos de la Memoria/enzimología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/fisiopatología , Animales , Fibras Colinérgicas/metabolismo , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Inhibidores Enzimáticos/toxicidad , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Hipocampo/fisiopatología , Indanos/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/metabolismo , Núcleos Septales/efectos de los fármacos , Núcleos Septales/enzimología , Núcleos Septales/fisiopatología , Azida Sódica/toxicidad , Resultado del Tratamiento , Proteínas de Transporte Vesicular de Acetilcolina/efectos de los fármacos , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
12.
Brain Res ; 1149: 200-9, 2007 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-17382306

RESUMEN

Therapeutic approaches that promote both neuroprotection and neuroregeneration would be valuable for spinal cord (SC) injury therapies. Cyclosporin-A (CsA) is an immunosuppressant that, due to its mechanism of action, could both protect and regenerate the neural tissue after injury. Previous studies have already demonstrated that intraperitoneal administration of CsA at a dose of 2.5 mg/kg/12 h during the first 2 days after SC contusion, followed by 5 mg/kg/12 h orally, diminishes tissue damage and improves motor recovery. In order to evaluate the effect of this CsA dosing regimen on axonal growth, we assessed motor recovery, presence of axons establishing functional connections and expression of GAP-43 in rats subjected to a complete SC transection. The Basso-Beattie-Bresnahan rating scale did not show difference in motor recovery of CsA or vehicle-treated rats. Moreover, somato-sensorial evoked potentials demonstrated no functional connections in the SC of these animals. Nevertheless, histological studies showed that: i) a significant number of CsA-treated rats presented growing axons, although they deviated perpendicularly at the edge of the stumps, surrounding them, ii) the expression of GAP-43 in animals treated with CsA was higher than that observed in the control group. Finally, anterograde tracing of the corticospinal tract of rats subjected to an incomplete SC transection showed no axonal fibers reaching the caudal stump. In summary, CsA administered at the dosing-regimen that promotes neuroprotection in SC contused rats induces both GAP-43 expression and axonal growth; however, it failed to generate functional connections in SC transected animals.


Asunto(s)
Axones/efectos de los fármacos , Ciclosporina/farmacología , Regeneración Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Tractos Piramidales/efectos de los fármacos , Animales , Western Blotting , Electrofisiología , Femenino , Proteína GAP-43/biosíntesis , Proteína GAP-43/efectos de los fármacos , Inmunohistoquímica , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Traumatismos de la Médula Espinal
13.
Brain Res ; 1123(1): 68-79, 2006 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-17078935

RESUMEN

Rehmannia, a traditional Chinese medical herb, has a long history in age-related disease therapy. Previous work has indicated that catalpol is a main active ingredient performing neuroprotective effect in rehmannia, while the mechanism underlying the effect remains poorly understood. In this study, we attempt to investigate the effect of catalpol on presynaptic proteins and explore a potential mechanism. The hippocampal levels of GAP-43 and synaptophysin in 3 groups of 4 months (young group), 22-24 months (aged group) and catalpol-treated 22-24 months (catalpol-treated group) rats were evaluated by western blotting. Results clearly showed a significant decrease in synaptophysin (46.6%) and GAP-43 (61.4%) levels in the aged group against the young animals and an increase (45.0% and 31.8% respectively) in the catalpol-treated aged rats in comparison with the untreated aged group. In particular, synaptophysin immunoreactivity (OD) in the dentate granule layer of the hippocampus was increased 0.0251 in the catalpol-treated group as compared with the aged group. The study also revealed a catalpol-associated increase of PKC and BDNF in the hippocampus of the catalpol-treated group in comparison with the aged rats and highly correlated with synaptophysin and GAP-43. Such positive correlations between presynaptic proteins and signaling molecules also existed in the young group. These results suggested that catalpol could increase presynaptic proteins and up-regulate relative signaling molecules in the hippocampus of the aged rats. Consequently, it seemed to indicate that catalpol might ameliorate age-related neuroplasticity loss by "normalizing" presynaptic proteins and their relative signaling pathways in the aged rats.


Asunto(s)
Envejecimiento/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Glucósidos/farmacología , Hipocampo/efectos de los fármacos , Iridoides/farmacología , Plasticidad Neuronal/efectos de los fármacos , Proteína Quinasa C/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Análisis de Varianza , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Medicamentos Herbarios Chinos/farmacología , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Glucósidos Iridoides , Masculino , Fármacos Neuroprotectores/farmacología , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Estadísticas no Paramétricas , Sinaptofisina/efectos de los fármacos , Sinaptofisina/metabolismo , Regulación hacia Arriba
14.
J Neurosci Res ; 83(7): 1179-89, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16521124

RESUMEN

The vitamin D receptor (VDR) is believed to mediate different biologic actions of vitamin D3, an active metabolite of vitamin D, through regulation of gene expression after binding to specific DNA-response element (VDRE) on target genes. To further understand roles of both vitamin D3 and VDR in the central nervous system, we examined VDRE binding in nuclear extracts prepared from discrete rat brain regions and cultured rat cortical neurons by electrophoretic mobility shift assay. The highest activity of VDRE binding was found in the cerebellum among other brain regions examined, but sequence specific by taking into consideration the efficient competition with excess unlabeled VDRE but not with mutated VDRE. On in situ hybridization analysis, cells stained for VDR mRNA were abundant in neuron-enriched areas of cerebral cortex, hippocampus and cerebellar cortex in the mouse brain. Chronic treatment of vitamin D3 increased the expression of microtubule-associated protein-2, growth-associated protein-43 and synapsin-1 in cultured rat cortical neurons, suggesting a trophic role of vitamin D3 in differentiation and maturation of neurons. Neuronal cell death by brief glutamate exposure was significantly protected in cultured cortical neurons chronically treated with vitamin D3. Parallel studies showed that VDR mRNA was significantly upregulated 12-24 hr after brief glutamate exposure in cultured neurons chronically treated with vitamin D3, but not in those with vehicle alone. Our results suggest that vitamin D3 may play a role in mechanisms relevant to protective properties against the neurotoxicity of glutamate through upregulation of VDR expression in cultured rat cortical neurons.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Colecalciferol/farmacología , Ácido Glutámico/toxicidad , Neurotoxinas/antagonistas & inhibidores , Receptores de Calcitriol/genética , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Colecalciferol/metabolismo , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Esquema de Medicación , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Fármacos Neuroprotectores/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Elementos de Respuesta/efectos de los fármacos , Elementos de Respuesta/genética , Sinapsinas/efectos de los fármacos , Sinapsinas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
15.
J Nutr Biochem ; 16(9): 538-46, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16115542

RESUMEN

Effects of docosahexaenoic acid (DHA) on survival and neurite outgrowth were investigated in primary cultures of rat cortical neurons. Cell cultures were prepared from cortex on embryonic day 18 (E-18) for treatment with a series of DHA concentrations (12.5, 25, 50, 75, 100 and 200 microM). Docosahexaenoic acid (25-50 microM) significantly enhanced neuronal viability, but lower concentration of DHA (12.5 microM) did not show an obvious effect. In contrast, higher concentrations of DHA (100-200 microM) exerted the significant opposite effects by decreasing neuronal viability. Furthermore, treatment with 25 microM DHA significantly prevented the neurons from death after different culture days in vitro (DIV). Moreover, measurements from the cultures exposed to 25 microM DHA immediately after plating showed significant increases in the percentage of cells with neurites, the mean number of neurite branches, the total neuritic length per cell and the length of the longest neurite in each cell after 24 and 48 h in vitro (HIV). The DHA-treated neurons had greater growth-associated protein-43 (GAP-43) immunoactivity and higher phosphatidylserine (PS) and phosphatidylethanolamine (PE) contents, but lower phosphatidylcholine (PC) content than control neurons. The significant increased DHA contents were also observed in both PE and PS in the treated neurons. These findings suggest that optimal DHA (25 microM) may have positive effects on the survival and the neurite outgrowth of the cultured fetal rat cortical neurons, and the effects probably are related to DHA-stimulating neuron-specific protein synthesis and its enhancing the discrete phospholipid (PL) content through enrichment of DHA in the PL species.


Asunto(s)
Corteza Cerebral/citología , Ácidos Docosahexaenoicos/farmacología , Neuronas/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/embriología , Ácidos Grasos/metabolismo , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Neuritas/efectos de los fármacos , Neuronas/citología , Neuronas/metabolismo , Fosfolípidos/metabolismo , Ratas , Ratas Sprague-Dawley
17.
Biol Psychiatry ; 53(6): 530-7, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12644358

RESUMEN

BACKGROUND: The importance of molecular and cellular changes in hippocampus in major depression and in the mechanism of action of antidepressants has become increasingly clear. Identification of novel targets for antidepressants in hippocampus is important to understanding their therapeutic effects. METHODS: We used cDNA microarray to measure the expression patterns of multiple genes in primary cultured rat hippocampal cells. In situ hybridization and Northern and immunoblotting analysis were used to determine brain regional distribution and mRNA and protein levels of target genes. RESULTS: After comparing hybridized signals between control and desipramine treated groups, we found that chronic treatment with desipramine increased the expression of six genes and decreased the expression of two genes. One of the upregulated genes is growth associated protein GAP-43. In situ hybridization revealed that desipramine increased GAP-43 gene expression in dentate gyrus but not other brain regions. Northern and immunoblotting analysis revealed that desipramine increased GAP-43 mRNA and protein levels. GAP-43 expression is also increased by another antidepressant, tranylcypromine, but not by lithium or haloperidol. CONCLUSIONS: Because GAP-43 regulates growth of axons and modulates the formation of new connections, our findings suggest that desipramine may have an effect on neuronal plasticity in the central nervous system.


Asunto(s)
Antidepresivos Tricíclicos/administración & dosificación , Antidepresivos Tricíclicos/farmacología , Desipramina/administración & dosificación , Desipramina/farmacología , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Hipocampo/citología , Hipocampo/metabolismo , Animales , Antidepresivos Tricíclicos/uso terapéutico , Northern Blotting , Células Cultivadas , ADN Complementario/genética , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/metabolismo , Desipramina/uso terapéutico , Modelos Animales de Enfermedad , Esquema de Medicación , Hibridación in Situ , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Ratas , Ratas Sprague-Dawley
18.
J Neurocytol ; 32(9): 1077-89, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15044840

RESUMEN

Brain derived neurotrophic factor (BDNF) when added to explant cultures of both embryonic and adult retinal ganglion cell (RGC) axons exerted a marked effect on their growth cone size and complexity and also on the intensity of GAP-43, beta-III tubulin and F-actin immunoreaction product in their axons. GAP-43 was distributed in axons, lamellipodia, and filopodia whereas beta-III tubulin was distributed along the length of developing and adult regenerating axons and also in the C-domain of their growth cones. BDNF-treated developing RGC growth cones were larger and displayed increased numbers of GAP-43 and microtubule-containing branches. Although filopodia and lamellipodia were lost from both developing and adult RGC growth cones following trkB-IgG treatment, the intensity of the immunoreaction product of all these molecules was reduced and trkB-IgGs had no effect on the axonal distribution of betas-III tubulin and GAP-43. BDNF-treated growth cones also displayed increased numbers of F-actin containing filopodia and axonal protrusions. This study demonstrates, for the first time, that trkB-IgG treatment causes the loss of F-actin in the P-domain of growth cone tips in developing and regenerating RGC axons. Although microtubules and F-actin domains normally remained distinct in cultured growth cones, beta-III tubulin and F-actin overlapped within the growth cone C-domain, and within axonal protrusions of adult RGC axons, under higher concentrations of BDNF. The collapse of RGC growth cones appeared to correlate with the loss of F-actin. In vitro, trkB signalling may therefore be involved in the maintenance and stabilisation of RGC axons, by influencing F-actin polymerisation, stabilisation and distribution.


Asunto(s)
Actinas/metabolismo , Axones/metabolismo , Proteína GAP-43/metabolismo , Conos de Crecimiento/metabolismo , Factores de Crecimiento Nervioso/farmacología , Tubulina (Proteína)/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Actinas/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/ultraestructura , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Feto , Proteína GAP-43/efectos de los fármacos , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/ultraestructura , Inmunoglobulina G/metabolismo , Inmunoglobulina G/farmacología , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Técnicas de Cultivo de Órganos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Seudópodos/ultraestructura , Ratas , Ratas Wistar , Receptor trkB/efectos de los fármacos , Receptor trkB/metabolismo , Retina/citología , Retina/embriología , Retina/crecimiento & desarrollo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Tubulina (Proteína)/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
19.
Eur J Neurosci ; 16(10): 1939-48, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12453058

RESUMEN

Expression of neurotrophins (NTs) and their receptors is elevated in the adult CNS under several neuropathological conditions. We have investigated the anatomical and electrophysiological consequences of chronic NT-3 or NT-4/5 treatment on established organotypic hippocampal slice cultures maintained in vitro for > 14 days. Both NT-3 and NT-4/5 increased spontaneous, action potential-dependent excitatory synaptic activity (sEPSCs), but only NT-3 increased inhibitory synaptic activity (sIPSCs) in CA3 pyramidal cells. Both NTs strongly promoted spontaneous synaptic bursting activity. Spontaneous bursts of EPSCs were observed after either NT treatment but only NT-3-treated cultures exhibited an increase in spontaneous bursts of IPSCs. In addition, sIPSC bursts were eliminated by blocking glutamatergic excitation. The frequency of miniature inhibitory postsynaptic currents, but not miniature excitatory postsynaptic currents, was also increased by both NT-3 and NT-4/5. Furthermore, NT-3 and NT-4/5 induced an up-regulation of the growth-associated protein GAP-43, suggesting that neurotrophins may be able to induce axonal reorganization in established neuronal networks. CA1 pyramidal cells exhibited slight alterations in dendritic branching after NT-4/5, but not NT-3 treatment. We conclude that chronic treatment with NT-3 or NT-4/5 can affect an established hippocampal network by elevating spontaneous inhibitory and excitatory synaptic activity and inducing coordinated pre- and postsynaptic structural changes.


Asunto(s)
Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Factores de Crecimiento Nervioso/farmacología , Células Piramidales/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Animales Recién Nacidos , Axones/efectos de los fármacos , Técnicas de Cultivo , Dendritas/efectos de los fármacos , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/metabolismo , Hipocampo/citología , Inmunohistoquímica , Factores de Crecimiento Nervioso/fisiología , Inhibición Neural/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Neurotrofina 3/farmacología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar
20.
Cell Mol Biol (Noisy-le-grand) ; 48(6): 717-23, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12396084

RESUMEN

Abstract - The adult olfactory receptor neurons (ORNs), located in the olfactory epithelium (OE) are permanently renewed thanks to neuronal progenitors present in the deep part of the OE, the globose basal cells (GBCs). Following the ablation of their synaptic target, the olfactory bulb (OB), ORNs degenerate by apoptosis and a wave of neurogenesis, including proliferation of GBCs and neuronal differentiation of their progeny, restores the olfactory function. The Ginkgo biloba extract (EGb 761) (Beaufour Ipsen, France) was administered to adult mice at the doses of 50 or 100 mg/kg, following bilateral bulbectomy and its effects on the expression of PCNA, reflecting the number of proliferating GBCs and on growth associated protein 43 (GAP-43), expressed by differentiating neurons were measured by Western blotting. PCNA expression peaked 9 days post-bulbectomy in untreated animals, but 7 days post-lesion in EGb 761-treated animals. A simultaneous reduction in GAP-43 expression suggested that EGb 761 may temporarily favor the proliferation of GBCs rather than their entry into the differentiation pathway. Probably as a consequence of the earlier onset of the neurogenetic response to bulbectomy, neuronal differentiation was enhanced in the OE, 3 weeks post-bulbectomy. These data suggest that EGb 761 may have beneficial effects upon neurogenesis in the OE through changing the balance between proliferation and differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Mucosa Olfatoria/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Proteína GAP-43/biosíntesis , Proteína GAP-43/efectos de los fármacos , Proteína GAP-43/genética , Ginkgo biloba , Ratones , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...