Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Dermatol Sci ; 113(3): 138-147, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38429137

RESUMEN

BACKGROUND: Postherpetic pain (PHP) is difficult to control. Although Neurotropin® (NTP) and methylcobalamin (MCB) are often prescribed to treat the pain, the efficacy of combined treatment for PHP remains imcompletely understood. OBJECTIVE: In this study, we investigate the combined effects of NTP and MCB on PHP in mice. METHODS: NTP and MCB were administered from day 10-29 after herpes simplex virus type-1 (HSV-1) infection. The pain-related responses were evaluated using a paint brush. The expression of neuropathy-related factor (ATF3) and nerve repair factors (GAP-43 and SPRR1A) in the dorsal root ganglion (DRG) and neurons in the skin were evaluated by immunohistochemical staining. Nerve growth factor (NGF) and neurotrophin-3 (NT3) mRNA expression levels were evaluated using real-time PCR. RESULTS: Repeated treatment with NTP and MCB after the acute phase inhibited PHP. Combined treatment with these drugs inhibited PHP at an earlier stage than either treatment alone. In the DRG of HSV-1-infected mice, MCB, but not NTP, decreased the number of cells expressing ATF3 and increased the number of cells expressing GAP-43- and SPRR1A. In addition, MCB, but not NTP, also increased and recovered non-myelinated neurons decreased in the lesional skin. NTP increased the mRNA levels of NTF3 in keratinocytes, while MCB increased that of NGF in Schwann cells. CONCLUSION: These results suggest that combined treatment with NTP and MCB is useful for the treatment of PHP. The combined effect may be attributed to the different analgesic mechanisms of these drugs.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Neuralgia Posherpética , Polisacáridos , Vitamina B 12/análogos & derivados , Ratones , Animales , Neuralgia Posherpética/tratamiento farmacológico , Factor de Crecimiento Nervioso/metabolismo , Proteína GAP-43/farmacología , Herpes Simple/complicaciones , Herpes Simple/tratamiento farmacológico , ARN Mensajero
2.
Environ Sci Pollut Res Int ; 30(38): 88685-88703, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37442924

RESUMEN

This study investigated the possible beneficial role of the bee venom (BV, Apis mellifera L.) against zinc oxide nanoparticles (ZNPs)-induced neurobehavioral and neurotoxic impacts in rats. Fifty male Sprague Dawley rats were alienated into five groups. Three groups were intraperitoneally injected distilled water (C 28D group), ZNPs (100 mg/kg b.wt) (ZNPs group), or ZNPs (100 mg/kg.wt) and BV (1 mg/ kg.bwt) (ZNPs + BV group) for 28 days. One group was intraperitoneally injected with 1 mL of distilled water for 56 days (C 56D group). The last group was intraperitoneally injected with ZNPs for 28 days, then BV for another 28 days at the same earlier doses and duration (ZNPs/BV group). Depression, anxiety, locomotor activity, spatial learning, and memory were evaluated using the forced swimming test, elevated plus maze, open field test, and Morris water maze test, respectively. The brain contents of dopamine, serotonin, total antioxidant capacity (TAC), malondialdehyde (MDA), and Zn were estimated. The histopathological changes and immunoexpressions of neurofilament and GAP-43 protein in the brain tissues were followed. The results displayed that BV significantly decreased the ZNPs-induced depression, anxiety, memory impairment, and spatial learning disorders. Moreover, the ZNPs-induced increment in serotonin and dopamine levels and Zn content was significantly suppressed by BV. Besides, BV significantly restored the depleted TAC but minimized the augmented MDA brain content associated with ZNPs exposure. Likewise, the neurodegenerative changes induced by ZNPs were significantly abolished by BV. Also, the increased neurofilament and GAP-43 immunoexpression due to ZNPs exposure were alleviated with BV. Of note, BV achieved better results in the ZNPs + BV group than in the ZNPs/BV group. Conclusively, these results demonstrated that BV could be employed as a biologically effective therapy to mitigate the neurotoxic and neurobehavioral effects of ZNPs, particularly when used during ZNPs exposure.


Asunto(s)
Venenos de Abeja , Nanopartículas , Síndromes de Neurotoxicidad , Óxido de Zinc , Ratas , Animales , Masculino , Abejas , Ratas Sprague-Dawley , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacología , Óxido de Zinc/metabolismo , Venenos de Abeja/farmacología , Venenos de Abeja/toxicidad , Dopamina/metabolismo , Dopamina/farmacología , Serotonina/metabolismo , Filamentos Intermedios/metabolismo , Antioxidantes/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Encéfalo
3.
J Ethnopharmacol ; 311: 116400, 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37003402

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The combination of Polygoni Multiflori Radix Praeparata (PMRP) and Acori Tatarinowii Rhizoma (ATR) is often used in traditional Chinese medicine to prevent and treat Alzheimer's disease (AD). However, it is not clear whether the effects and mechanisms of the decoction prepared by traditional decocting method (PA) is different from that prepared by modern decocting method (P + A). AIM OF THE STUDY: The present study aimed to investigate the differences in the protective effects of PA and P + A on scopolamine induced cognitive impairment, and to explore its potential mechanism. MATERIALS AND METHODS: To assess the protective effect of PA and P + A on cognitive dysfunction, the mice were orally administrated with PA (1.56, 6.24 g kg-1•day-1) and P + A (1.56, 6.24 g kg-1•day-1) for 26 days before co-treatment with scopolamine (4 mg kg-1•day-1, i.p.). The learning and memory abilities of mice were examined by Morris water maze test, and the expressions of proteins related to cholinergic system and synaptic function were detected by the methods of ELISA, real-time PCR and Western blotting. Then, molecular docking technique was used to verify the effect of active compounds in plasma after PA administration on Acetylcholinesterase (AChE) protein. Finally, the Ellman method was used to evaluate the effects of different concentrations of PA, P + A (1 µg/mL-100 mg/mL) and the compounds (1-100 µM) on AChE activity in vitro. RESULTS: On one hand, in the scopolamine-induced cognitive impairment mouse model, both of PA and P + A could improve the cognitive impairment, while the effect of PA on cognitive amelioration was better than that of P + A. Moreover, PA regulated the cholinergic and synaptic functions by enhancing the concentration of acetylcholine (ACh), the mRNA levels of CHT1, Syn, GAP-43 and PSD-95, and the related proteins (CHT1, VACHT, Syn, GAP-43 and PSD-95), and significantly inhibiting the expression of AChE protein. Meanwhile, P + A only up-regulated the mRNA levels of GAP-43 and PSD-95, increased the expressions of CHT1, VACHT, Syn, GAP-43 and PSD-95 proteins, and inhibited the expression of AChE protein. On the other hand, the in vitro study showed that some compounds including emodin-8-o-ß-d-Glucopyranoside, THSG and α-Asarone inhibited AChE protein activity with the IC50 values 3.65 µM, 5.42 µM and 9.43 µM, respectively. CONCLUSIONS: These findings demonstrate that both of PA and P + A can ameliorate the cognitive deficits by enhancing cholinergic and synaptic related proteins, while PA has the stronger improvement effect on the cholinergic function, which may be attributed to the compounds including THSG, emodin, emodin-8-O-ß-D-glucopyranoside and α-asarone. The present study indicated that PA has more therapeutic potential in the treatment of neurodegenerative diseases such as AD. The results provide the experimental basis for the clinical use of PA.


Asunto(s)
Disfunción Cognitiva , Emodina , Ratones , Animales , Escopolamina/farmacología , Acetilcolinesterasa/metabolismo , Emodina/farmacología , Simulación del Acoplamiento Molecular , Proteína GAP-43/farmacología , Colinérgicos/farmacología , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Aprendizaje por Laberinto
4.
Ecotoxicol Environ Saf ; 252: 114576, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36736231

RESUMEN

Nonylphenol (NP) is a typical environmental endocrine disruptor with estrogenic effects. It serves as an emulsifier and as the main ingredient of detergents and has become an increasingly common pollutant in both fresh and salt water, vegetables, and fruits. This study aimed to clarify whether NP exposure could lead to cognitive dysfunction and synaptic plasticity impairment, and also explore the mechanism of microRNA (miR)- 219a-5p regulation of N-methyl-D-aspartate receptor (NMDAR) in NP-induced synaptic plasticity impairment in vivo and in vitro. In vivo, 30 male Sprague-Dawley rats were randomly divided into 2 groups: blank control group (pure corn oil) and NP-exposed group [NP 80 mg/(kg·d)], with 15 rats in each group. In vitro, the extracted hippocampal neurons were divided into 6 groups: blank control group, mimics NC group, miR-219 mimics group, NP group (70 µmol/L NP), NP + mimics NC group, and NP + miR-219 mimics group. In vivo, the content of NP in hippocampal tissues after 90 days of NP exposure was significantly higher in the NP-stained group than in the blank control group. NP exposure could lead to a decrease in the ability to learn and memory, ability to remember, and space spatial memory ability in rats. The dendrites in the NP-stained group were disordered, with few dendritic spines and significantly decreased dendritic spine density. The postsynaptic densities were loosely arranged, the thickness and length of the postsynaptic densities shortened, and the length and width of the synaptic gap increased. Glutamine (Glu) and γ-aminobutyric acid (GABA) contents in hippocampal tissues decreased in the NP-stained group. The expression of miR-219a-5p mRNA decreased in the NP-stained group after 3 months of NP exposure. The expression of NMDAR1, NMDAR2A, NMDAR2B, nerve growth-associated protein (GAP-43), and Ca/calmodulin-dependent kinase II (CaMKII) mRNA/proteins decreased in the NP-stained group. In vitro, NMDAR protein expression decreased, while GAP-43 and CaMKII protein expression increased in the miR-219 mimics group compared with the control group. The expression levels of NMDAR and GAP-43 and CaMKII proteins were higher in the NP + miR-219 mimics group compared with the NP group. The levels of neurotransmitters Glu and GABA decreased in the NP and NP + mimics NC groups compared with the blank group. Shortened synaptic active band length, decreased thickness of postsynaptic densities, and shortened length of postsynaptic densities were observed in the NP, NP + mimics NC, and NP + miR-219 mimics groups compared with the blank control group. In vivo, NP exposure reduced learning memory capacity and neurotransmitter content in rats and caused a decrease in dendritic spine density and synaptic number density and a decrease in miR-219a-5p expression. In vitro, high expression of miR-219a-5p inhibited the expression of NMDAR, thus reducing the effect of NP on synaptic plasticity impairment in hippocampal neurons. Our study provided a scientific basis for the prevention of cognitive impairment owing to NP exposure and the development of targeted drug treatment strategies.


Asunto(s)
MicroARNs , Receptores de N-Metil-D-Aspartato , Ratas , Masculino , Animales , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Ratas Sprague-Dawley , MicroARNs/genética , MicroARNs/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacología , Plasticidad Neuronal/fisiología , ARN Mensajero
5.
Morphologie ; 107(356): 67-79, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35715368

RESUMEN

Adverse experiences during pregnancy have a negative impact on the neuronal structure and behavior of offspring, but the effects of a father's life events on the outcome of progeny are scarce. The present study is intended to investigate whether paternal stress affects the offspring brain structure, especially those regions concerned with learning and formation of memory, namely the hippocampus (HC) and prefrontal cortex (PFC), and also the expression of certain genes linked to learning and memory in the offspring. Induced stress to male rats by five stressors, one per day followed by allowing them to mate with the normal, unstressed female. Synaptophysin immunoreactivity was assessed in the tissue sections of the HC and PFC as well as expression of genes concerned with learning and memory was evaluated by RT-PCR in the progeny of stress-received males. The progeny of stressed rats had reduced antisynaptophysin immunoreactivity in the HC and PFC. The synaptic density in HC was less in the A-S (Offspring of male rats who received stress during adulthood) and PA-S (offspring of male rats who received stress during both adolescence and adulthood) than in P-S (offspring of male rats who received stress during adolescence) and C-C (offspring of control) groups. Similar results were observed even in the PFC. The results of post hoc tests proved that the HC and PFC of the progeny of stress-exposed rats exhibited considerably less synaptic density than control (P<0.05), and the levels of expression of GAP-43, GRIN1, M1, and SYP genes in HC and PFC were down-regulated. This study concludes that paternal adverse experiences can affect the offspring's synaptic plasticity and also the genes, which can regulate learning and formation of memory.


Asunto(s)
Hipocampo , Corteza Prefrontal , Embarazo , Ratas , Animales , Masculino , Femenino , Humanos , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacología , Hipocampo/metabolismo , Corteza Prefrontal/metabolismo , Aprendizaje , Padre , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacología , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinaptofisina/genética , Sinaptofisina/metabolismo , Sinaptofisina/farmacología
6.
Comput Math Methods Med ; 2022: 4949206, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35495894

RESUMEN

Optic neuritis (ON) is a common neurological disease, and the transplant of retinal ganglion cells (RGCs) has been thought as a promising strategy for improving the injury of the optic nerve system. Bone mesenchymal stem cells (BMSCs) have the potential to differentiate into neural cells. Several studies have indicated that GAP-43 is related with the regeneration of nerve cells, while the effect of GAP-43 on inducing BMSC differentiation remains unclear. In this study, the BMSCs were separated from the rats and identified with flow cytometry assay. The GAP-43 expressed vectors were transfected into the BMSCs, and the biomarkers of RGCs such as PAX6, LHX2, and ATOH7 were used to observe by qRT-PCR. Moreover, the effect of GAP-43-induced BMSCs (G-BMSCs) on ON improvement was also verified with rat models, and the activity of MAPK pathway was measured with western blot. Here, it was found that GAP-43 could obviously promote the differentiation of BMSCs, and increased PAX6, LHX2, ATOH7, BRN3A, and BRN3B were observed in the process of cell differentiation. Moreover, it was also found that G-BMSCs significantly increased the abundances of NFL and NFM in G-BMSCs, and GAP-43 could also enhance the activity of MAPK pathways in BMSCs. Therefore, this study suggested that GAP-43 could induce the differentiation of bone marrow-derived mesenchymal stem cells into retinal ganglial cells.


Asunto(s)
Células Madre Mesenquimatosas , Células Ganglionares de la Retina , Animales , Médula Ósea , Diferenciación Celular/fisiología , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacología , Humanos , Proteínas con Homeodominio LIM/metabolismo , Proteínas con Homeodominio LIM/farmacología , Ratas , Factores de Transcripción/metabolismo
7.
Metab Brain Dis ; 37(5): 1451-1463, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35348994

RESUMEN

1,4-butanediol (1,4-BD) is a known γ-hydroxybutyric acid (GHB) precursor which affects the nervous system after ingestion, leading to uncontrolled behavioral consequences. In the present study, we investigated whether 1,4-BD induces oxidative stress and inflammation in PC12 cells and evaluated the toxic effects of 1,4-BD associates with learning and memory. CCK-8 results revealed a dose-effect relationship between the cell viability of PC12 cells and 1,4-BD when the duration of action was 2 h or 4 h. Assay kits results showed that 1,4-BD decreased the levels of Glutathione (GSH), Glutathione peroxidase (GSH-px), Superoxide dismutase (SOD), Acetylcholine (Ach) and increased the levels of Malondialdehyde (MDA), Nitric oxide (NO) and Acetylcholinesterase (AchE). Elisa kits results indicated that 1,4-BD decreased the levels of synaptophysin I (SYN-1), Postsynaptic density protein-95 (PSD-95), Growth associated protein-43 (GAP-43) and increased the levels of Tumor necrosis factor alpha (TNF-α) and Interleukin- 6 (IL-6). RT-PCR results showed that the mRNA levels of PSD-95, SYN-1 and GAP-43 were significantly decreased. The expression of phosphorylation extracellular signal-regulated protein kinase 1/2 (p-ERK1/2), phosphorylation cAMP response element binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) proteins were significantly decreased in PC12 cells by protein blotting. Overall, these results suggest that 1,4-BD may affect synaptic plasticity via the ERK1/2-CREB-BDNF pathway, leading to Ach release reduction and ultimately to learning and memory impairment. Furthermore, oxidative stress and inflammation induced by 1,4-BD may also result in learning and memory deficits. These findings will enrich the toxicity data of 1.4-BD associated with learning and memory impairment.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Sistema de Señalización de MAP Quinasas , Acetilcolinesterasa/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Butileno Glicoles , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacología , Glutatión/metabolismo , Hipocampo/metabolismo , Inflamación/metabolismo , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/metabolismo , Células PC12 , Ratas , Transducción de Señal
8.
Curr Stem Cell Res Ther ; 17(8): 756-771, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34493197

RESUMEN

Neuronal damage or degeneration is the main feature of neurological diseases. Regulation of neurogenesis and neuronal differentiation is important in developing therapies to promote neuronal regeneration or synaptic network reconstruction. Neurogenesis is a multistage process in which neurons are generated and integrated into existing neuronal circuits. Neuronal differentiation is extremely complex because it can occur in different cell types and can be caused by a variety of inducers. Recently, natural compounds that induce neurogenesis and neuronal differentiation have attracted extensive attention. In this paper, the potential neural induction effects of medicinal plant-derived natural compounds on neural stem/progenitor cells (NS/PCs), the cultured neuronal cells, and mesenchymal stem cells (MSCs) are reviewed. The natural compounds that are efficacious in inducing neurogenesis and neuronal differentiation include phenolic acids, polyphenols, flavonoids, glucosides, alkaloids, terpenoids, quinones, coumarins, and others. They exert neural induction effects by regulating signal factors and cellspecific genes involved in the process of neurogenesis and neuronal differentiation, including specific proteins (ß-tubulin III, MAP-2, tau, nestin, neurofilaments, GFAP, GAP-43, NSE), related genes and proteins (STAT3, Hes1, Mash1, NeuroD1, notch, cyclin D1, SIRT1, Reggie-1), transcription factors (CREB, Nkx-2.5, Ngn1), neurotrophins (BDNF, NGF, NT-3), and signaling pathways (JAK/STAT, Wnt/ß-catenin, MAPK, PI3K/Akt, GSK-3ß/ß-catenin, Ca2+/CaMKII/ATF1, Nrf2/HO-1, BMP).The natural compounds with neural induction effects are of great value for neuronal regenerative medicine and provide promising prevention and treatment strategies for neurological diseases.


Asunto(s)
Ciclina D1 , beta Catenina , Factor Neurotrófico Derivado del Encéfalo/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/farmacología , Diferenciación Celular/fisiología , Cumarinas/farmacología , Ciclina D1/farmacología , Proteína GAP-43/farmacología , Glucósidos/farmacología , Glucógeno Sintasa Quinasa 3 beta/farmacología , Humanos , Factor 2 Relacionado con NF-E2/farmacología , Factor de Crecimiento Nervioso/farmacología , Nestina , Neurogénesis/fisiología , Fosfatidilinositol 3-Quinasas , Polifenoles/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/farmacología , Quinonas/farmacología , Sirtuina 1/farmacología , Terpenos/farmacología , Tubulina (Proteína) , beta Catenina/metabolismo
9.
J Mol Cell Cardiol ; 49(1): 79-87, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20138055

RESUMEN

Neuronal remodeling with increased sympathetic innervation density has been implicated in the pathogenesis of atrial fibrillation (AF). Recently, increased transcardiac nerve growth factor (NGF) levels were observed in a canine model of AF. Whether atrial myocytes or cardiac sympathetic neurons are the source of neurotrophins, and whether NGF is the main neurotrophic factor contributing to sympathetic nerve sprouting (SNS) in AF still remains unclear. Therefore, neonatal rat atrial myocytes were cultured under conditions of high frequency electrical field stimulation (HFES) to mimic rapid atrial depolarization. Likewise, sympathetic neurons from the superior cervical ganglia of neonatal rats were exposed to HFES to simulate the physiological effect of sympathetic stimulation. Real-time PCR, ELISA and Western blots were performed to analyze the expression pattern of NGF and neurotrophin-3 (NT-3). Baseline NGF and NT-3 content was 3-fold higher in sympathetic neurons than in atrial myocytes (relative NGF protein expression: 1+/-0.0 vs. 0.37+/-0.11, all n=5, p<0.05). HFES of sympathetic neurons induced a frequency dependent NGF and NT-3 gene and protein up-regulation (relative NGF protein expression: 0Hz=1+/-0.0 vs. 5Hz=1.13+/-0.19 vs. 50Hz=1.77+/-0.08, all n=5, 0Hz/5Hz vs. 50Hz p<0.05), with a subsequent increase of growth associated protein 43 (GAP-43) expression and morphological SNS. Moreover, HFES of sympathetic neurons increased the tyrosine kinase A (TrkA) receptor expression. HFES induced neurotrophic effects could be abolished by lidocaine, TrkA blockade or NGF neutralizing antibodies, while NT-3 neutralizing antibodies had no significant effect on SNS. In neonatal rat atrial myocytes, HFES resulted in myocyte hypertrophy accompanied by an increase in NT-3 and a decrease in NGF expression. In summary, this study provides evidence that high-rate electrical stimulation of sympathetic neurons mediates nerve sprouting by an increase in NGF expression that targets the TrkA receptor in an autocrine/paracrine manner.


Asunto(s)
Factor de Crecimiento Nervioso/farmacología , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes , Estimulación Eléctrica , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacología , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Neurotrofina 3/genética , Neurotrofina 3/metabolismo , Neurotrofina 3/farmacología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Ganglio Cervical Superior/efectos de los fármacos , Ganglio Cervical Superior/metabolismo , Sistema Nervioso Simpático/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
J Pharmacol Exp Ther ; 306(2): 752-62, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12730350

RESUMEN

Glutamate is the major excitatory transmitter in the brain. Recent developments in the molecular biology and pharmacology of the alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) subtype of glutamate receptors have led to the discovery of selective, potent, and systemically active AMPA receptor potentiators. These molecules enhance synaptic transmission and play important roles in plasticity and cognitive processes. In the present study, we first characterized a novel AMPA receptor potentiator, (R)-4'-[1-fluoro-1-methyl-2-(propane-2-sulfonylamino)-ethyl]-biphenyl-4-carboxylic acid methylamide (LY503430), on recombinant human GLUA1-4 and native preparations in vitro and then evaluated the potential neuroprotective effects of the molecule in rodent models of Parkinson's disease. Results indicated that submicromolar concentrations of LY503430 selectively enhanced glutamate-induced calcium influx into human embryonic kidney 293 cells transfected with human GLUA1, GLUA2, GLUA3, or GLUA4 AMPA receptors. The molecule also potentiated AMPA-mediated responses in native cortical, hippocampal, and substantia nigra neurons. We also report here that LY503430 provided dose-dependent functional and histological protection in animal models of Parkinson's disease. The neurotoxicity after unilateral infusion of 6-hydroxydopamine into either the substantia nigra or the striatum of rats and that after systemic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in mice were reduced. Interestingly, LY503430 also had neurotrophic actions on functional and histological outcomes when treatment was delayed until well after (6 or 14 days) the lesion was established. LY503430 also produced some increase in brain-derived neurotrophic factor in the substantia nigra and a dose-dependent increases in growth associated protein-43 (GAP-43) expression in the striatum. Therefore, we propose that AMPA receptor potentiators offer the potential of a new disease modifying therapy for Parkinson's disease.


Asunto(s)
Amidas/farmacología , Compuestos de Bifenilo/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/metabolismo , Receptores AMPA/agonistas , Sustancia Negra/citología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Células Cultivadas , Cuerpo Estriado/efectos de los fármacos , Dioxoles/farmacología , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/farmacología , Proteína GAP-43/farmacología , Hipocampo/citología , Humanos , Masculino , Neuronas/metabolismo , Oxidopamina/farmacología , Piperidinas/farmacología , Corteza Prefrontal/citología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
11.
Nat Neurosci ; 4(1): 38-43, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11135643

RESUMEN

In contrast to peripheral nerves, damaged axons in the mammalian brain and spinal cord rarely regenerate. Peripheral nerve injury stimulates neuronal expression of many genes that are not generally induced by CNS lesions, but it is not known which of these genes are required for regeneration. Here we show that co-expressing two major growth cone proteins, GAP-43 and CAP-23, can elicit long axon extension by adult dorsal root ganglion (DRG) neurons in vitro. Moreover, this expression triggers a 60-fold increase in regeneration of DRG axons in adult mice after spinal cord injury in vivo. Replacing key growth cone components, therefore, could be an effective way to stimulate regeneration of CNS axons.


Asunto(s)
Axones/metabolismo , Proteínas de Unión a Calmodulina , Conos de Crecimiento/metabolismo , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso , Neuronas/metabolismo , Médula Espinal/metabolismo , Animales , Axones/efectos de los fármacos , Axotomía , Separación Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/farmacología , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Proteína GAP-43/farmacología , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Expresión Génica , Conos de Crecimiento/efectos de los fármacos , Técnicas In Vitro , Ratones , Ratones Transgénicos , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/farmacología , Regeneración Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Médula Espinal/citología , Médula Espinal/efectos de los fármacos
12.
Can J Physiol Pharmacol ; 78(1): 81-5, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10741764

RESUMEN

The neuronal growth associated protein B-50/GAP-43 has been localized in synaptosomes both as an intact protein and as a partial proteolysis product (termed B-60) that has an N-terminal sequence SFRGHITR.... Because of the relationship of this amino acid sequence to those of the tethered ligand for the human proteinase activated receptors PAR1 (SFLLRN...) and PAR2 (SLIGKV...), we wished to determine whether the B-50/GAP-43-derived proteolytic fragment SFRGHITR (SFR(B60)) might function as a PAR-activating peptide (PAR-AP) to stimulate either PAR1 or PAR2. With the use of a newly developed PAR1/PAR2 receptor activation-desensitization assay, employing PAR1/PAR2-bearing cultured human embryonic kidney (HEK293) cells, we found that SFR(B60) could activate both PAR1 and PAR2 so as to elevate intracellular calcium with EC50 values of approximately 200 and 50 microM, respectively. We also showed that trypsin can rapidly degrade B-50 to smaller fragments that would include the sequence SFR(B60). Because PAR1 and PAR2 are present on neurones, our data raise the possibility that in certain circumstances in vivo, B-50/GAP-43 may play a signalling role by serving as a precursor for proteolytically generated PAR-activating peptides.


Asunto(s)
Proteína GAP-43/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Trombina/efectos de los fármacos , Secuencia de Aminoácidos , Señalización del Calcio , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Datos de Secuencia Molecular , Receptor PAR-1 , Receptor PAR-2 , Receptores de Trombina/metabolismo
13.
Mol Neurobiol ; 22(1-3): 99-113, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11414283

RESUMEN

Neuromodulin (GAP-43), neurogranin (RC3), and PEP-19 are small acid-stable proteins that bind calcium-poor calmodulin through a loosely conserved IQ-motif. Even though these proteins have been known for many years, much about their function in cells is not understood. It has recently become appreciated that calmodulin activity in cells is tightly controlled and that pools of otherwise free calmodulin are sequestered so as to restrict its availability for activating calcium/calmodulin-dependent enzymes. Neuromodulin, neurogranin, and PEP-19 appear to be major participants in this type of regulation. One way in which they do this is by providing localized increases in the concentration of calmodulin in cells so that the maximal level of target activation is increased. Additionally, they can function as calmodulin antagonists by directly inhibiting the association of calcium/calmodulin with enzymes and other proteins. Although neuromodulin, neurogranin, and PEP-19 were early representatives of the small IQ-motif-containing protein family, newer examples have come to light that expand the number of cellular systems through which the IQ-peptide/calmodulin interaction could regulate biological processes including gene transcription. It is the purpose of this review to examine the behavior of neuromodulin, neurogranin, and PEP-19 in paradigms that include both in vitro and in situ systems in order to summarize possible biological consequences that are linked to the expression of this type of protein. The use of protein:protein interaction chromatography is also examined in the recovery of a new calmodulin-binding peptide, CAP-19 (ratMBF1). Consistent with earlier predictions, at least one function of small IQ-motif proteins appears to be that they lessen the extent to which calcium-calmodulin-dependent enzymes become or stay activated. It also appears that these polypeptides can function to selectively inhibit activation of intracellular targets by some agonists while simultaneously permitting activation of these same targets by other agonists. Much of the mechanism for how this occurs is unknown, and possible explanations are examined. One of the biological consequences for a cell that expresses a calmodulin-regulatory protein could be an increased resistance to calcium-mediated toxicity. This possibility is examined for cells expressing PEP-19 and both anatomical and cell-biological data is described. The study of IQ-motif-containing small proteins has stimulated considerable thought as to how calcium signaling is refined in neurons. Current evidence suggests that signaling through calmodulin is not a fulminating and homogenous process but a spatially limited and highly regulated one. Data from studies on neuromodulin, neurogranin, and PEP-19 suggest that they play an important role in establishing some of the processes by which this regulation is accomplished.


Asunto(s)
Señalización del Calcio/fisiología , Proteínas de Unión a Calmodulina/fisiología , Calmodulina/fisiología , Proteína GAP-43/fisiología , Proteínas del Tejido Nervioso/fisiología , Enfermedad de Alzheimer/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Química Encefálica , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calmodulina/química , Proteínas de Unión a Calmodulina/química , Proteínas de Unión a Calmodulina/farmacología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Activación Enzimática/efectos de los fármacos , Proteína GAP-43/química , Proteína GAP-43/farmacología , Homeostasis , Humanos , Enfermedad de Huntington/metabolismo , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Degeneración Nerviosa , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/farmacología , Neurogranina , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I , Células PC12/efectos de los fármacos , Células PC12/enzimología , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...