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










Base de datos
Intervalo de año de publicación
1.
Neurotox Res ; 35(1): 150-159, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30088187

RESUMEN

Organophosphorus (OPs) compounds have been widely used in agriculture, industry, and household, and the neurotoxicity induced by them is still a cause of concern. The main toxic mechanism of OPs is the inhibition of acetylcholinesterase (AChE); however, the delayed neuropathy induced by OPs (OPIDN) is mediated by other mechanisms such as the irreversible inhibition of 70% of NTE activity (neuropathy target esterase) that leads to axonal degeneration. Liraglutide is a long-lasting GLP-1 analog clinically used as antidiabetic. Its neurotrophic and neuroprotective effects have been demonstrated in vitro and in experimental models of neurodegenerative diseases. As in OPIDN, axonal degeneration also plays a role in neurodegenerative diseases. Therefore, this study investigated the protective potential of liraglutide against the neurotoxicity of OPs by using mipafox as a neuropathic agent (at a concentration able to inhibit and age 70% of NTE activity) and a neuronal model with SH-SY5Y neuroblastoma cells, which express both esterases. Liraglutide protected cells against the neurotoxicity of mipafox by increasing neuritogenesis, the uptake of glucose, the levels of cytoskeleton proteins, and synaptic-plasticity modulators, besides decreasing the pro-inflammatory cytokine interleukin 1ß and caspase-3 activity. This is the first study to suggest that liraglutide might induce beneficial effects against the delayed, non-cholinergic neurotoxicity of OPs.


Asunto(s)
Isoflurofato/análogos & derivados , Liraglutida/farmacología , Fármacos Neuroprotectores/farmacología , Plaguicidas/toxicidad , Línea Celular Tumoral , Glucosa/metabolismo , Humanos , Hipoglucemiantes/farmacología , Interleucina-1beta/metabolismo , Isoflurofato/toxicidad , Proyección Neuronal/efectos de los fármacos , Proyección Neuronal/fisiología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Neuroprotección/efectos de los fármacos , Neuroprotección/fisiología , Síndromes de Neurotoxicidad/tratamiento farmacológico
2.
Neurotox Res ; 34(1): 32-46, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29260495

RESUMEN

Cisplatin is a highly effective chemotherapeutic drug that is toxic to the peripheral nervous system. Findings suggest that axons are early targets of the neurotoxicity of cisplatin. Although many compounds have been reported as neuroprotective, there is no effective treatment against the neurotoxicity of cisplatin. Caffeic acid phenethyl ester (CAPE) is a propolis component with neuroprotective potential mainly attributed to antioxidant and anti-inflammatory mechanisms. We have recently demonstrated the neurotrophic potential of CAPE in a cellular model of neurotoxicity related to Parkinson's disease. Now, we have assessed the neurotrophic and neuroprotective effects of CAPE against cisplatin-induced neurotoxicity in PC12 cells. CAPE (10 µM) attenuated the inhibition of neuritogenesis and the downregulation of markers of neuroplasticity (GAP-43, synapsin I, synaptophysin, and 200-kD neurofilament) induced by cisplatin (5 µM). This concentration of cisplatin does not affect cell viability, and it was used in order to assess the early neurotoxic events triggered by cisplatin. When a lethal dose of cisplatin was used (IC50 = 32 µM), CAPE (10 µM) increased cell viability. The neurotrophic effect of CAPE is not dependent on NGF nor is it additive to the effect of NGF, but it might involve the activation of the NGF-high-affinity receptors (trkA). The involvement of other neurotrophin receptors such as trkB and trkC is unlikely. This is the first study to demonstrate the protective potential of CAPE against the neurotoxicity of cisplatin and to suggest the involvement of trkA receptors in the neuroprotective mechanism of CAPE. Based on these findings, the beneficial effect of CAPE on cisplatin-induced peripheral neuropathy should be further investigated.


Asunto(s)
Ácidos Cafeicos/farmacología , Cisplatino/farmacología , Factor de Crecimiento Nervioso/metabolismo , Fármacos Neuroprotectores/farmacología , Neurotoxinas/farmacología , Alcohol Feniletílico/análogos & derivados , Transducción de Señal/efectos de los fármacos , Análisis de Varianza , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Proteína GAP-43/metabolismo , Neuroblastoma/patología , Proteínas de Neurofilamentos/metabolismo , Proyección Neuronal/efectos de los fármacos , Células PC12/efectos de los fármacos , Alcohol Feniletílico/farmacología , Ratas , Sinapsinas/metabolismo , Sinaptofisina/metabolismo
3.
J Toxicol Environ Health A ; 80(19-21): 1086-1097, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28862523

RESUMEN

Some organophosphorus compounds (OP), including the pesticide mipafox, produce late onset distal axonal degeneration, known as organophosphorus-induced delayed neuropathy (OPIDN). The underlying mechanism involves irreversible inhibition of neuropathy target esterase (NTE) activity, elevated intracellular calcium levels, increased activity of calcium-activated proteases and impaired neuritogenesis. Voltage-gated calcium channels (VGCC) appear to play a role in several neurologic disorders, including OPIDN. Therefore, this study aimed to examine and compare the neuroprotective effects of T-type (amiloride) and L-type (nimodipine) VGCC blockers induced by the inhibitory actions of mipafox on neurite outgrowth and axonal proteins of retinoic-acid-stimulated SH-SY5Y human neuroblastoma cells, a neuronal model widely employed to determine the neurotoxicity attributed to OP. Both nimodipine and amiloride significantly blocked augmentation of intracellular calcium levels and activity of calpains, as well as decreased neurite length, number of differentiated cells, and lowered concentrations of growth-associated protein 43 (GAP-43) and synapsin induced by mipafox. Only nimodipine inhibited reduction of synaptophysin levels produced by mipafox. These findings demonstrate a role for calcium and VGCC in the impairment of neuronal plasticity mediated by mipafox. Data also demonstrated the neuroprotective potential of T-type and L-type VGCC blockers to inhibit OP-mediated actions, which may be beneficial to counteract cases of pesticide poisoning.


Asunto(s)
Amilorida/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Insecticidas/toxicidad , Isoflurofato/análogos & derivados , Neuritas/efectos de los fármacos , Nimodipina/farmacología , Axones/efectos de los fármacos , Línea Celular Tumoral , Humanos , Isoflurofato/toxicidad
4.
Toxicol In Vitro ; 39: 84-92, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27939611

RESUMEN

Some organophosphorus compounds (OPs) induce a neurodegenerative disorder known as organophosphate-induced delayed neuropathy (OPIDN), which is related to irreversible inhibition of neuropathy target esterase (NTE) and impairment of neurite outgrowth. The present study addresses the effects of trichlorfon, mipafox (neuropathic model) and paraoxon (non-neuropathic model) on neurite outgrowth and neuroplasticity-related proteins in retinoic-acid-stimulated SH-SY5Y cells, a cellular model widely used to study the neurotoxicity of OPs. Mipafox (20µM) decreased cellular differentiation and the expression of neurofilament 200 (NF-200), growth associated- (GAP-43) and synaptic proteins (synapsin I and synaptophysin); whereas paraoxon (300µM) induced no effect on cellular differentiation, but significant decrease of NF-200, GAP-43, synapsin I and synaptophysin as compared to controls. However, the effects of paraoxon on these proteins were significantly lower than the effects of mipafox. In conclusion, axonal cytoskeletal proteins, as well as axonal plasticity-related proteins are more effectively affected by neuropathic (mipafox) than by non-neuropathic (paraoxon) OPs, suggesting that they might play a role in the mechanism of OPIDN. At high concentration (1mM), trichlorfon induced effects similar to those of the neuropathic OP, mipafox (20µM), but also caused high inhibition of AChE. Therefore, these effects are unlikely to occur in humans at non-lethal doses of trichlorfon.


Asunto(s)
Axones/efectos de los fármacos , Inhibidores de la Colinesterasa/toxicidad , Insecticidas/toxicidad , Isoflurofato/análogos & derivados , Paraoxon/toxicidad , Triclorfón/toxicidad , Acetilcolinesterasa/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular Tumoral , Citoesqueleto/efectos de los fármacos , Proteína GAP-43/metabolismo , Humanos , Isoflurofato/toxicidad , L-Lactato Deshidrogenasa/metabolismo , Plasticidad Neuronal , Sinapsinas/metabolismo
5.
Chem Biol Interact ; 261: 86-95, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-27871898

RESUMEN

Beta-caryophyllene (BCP) is a phytocannabinoid whose neuroprotective activity has been mainly associated with selective activation of cannabinoid-type-2 (CB2) receptors, inhibition of microglial activation and decrease of inflammation. Here, we addressed the potential of BCP to induce neuritogenesis in PC12 cells, a model system for primary neuronal cells that express trkA receptors, respond to NGF and do not express CB2 receptors. We demonstrated that BCP increases the survival and activates the NGF-specific receptor trkA in NGF-deprived PC12 cells, without increasing the expression of NGF itself. The neuritogenic effect of BCP in PC12 cells was abolished by k252a, an inhibitor of the NGF-specific receptor trkA. Accordingly, BCP did not induce neuritogenesis in SH-SY5Y neuroblastoma cells, a neuronal model that does not express trkA receptors and do not respond to NGF. Additionally, we demonstrated that BCP increases the expression of axonal-plasticity-associated proteins (GAP-43, synapsin and synaptophysin) in PC12 cells. It is known that these proteins are up-regulated by NGF in neurons and neuron-like cells, such as PC12 cells. Altogether, these findings suggest that BCP activates trka receptors and induces neuritogenesis by a mechanism independent of NGF or cannabinoid receptors. This is the first study to show such effects of BCP and their beneficial role in neurodegenerative processes should be further investigated.


Asunto(s)
Cannabinoides/farmacología , Neuritas/metabolismo , Neurogénesis/efectos de los fármacos , Receptores de Cannabinoides/metabolismo , Sesquiterpenos/farmacología , Animales , Carbazoles/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Alcaloides Indólicos/farmacología , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuritas/efectos de los fármacos , Células PC12 , Sesquiterpenos Policíclicos , Ratas , Receptor trkA/antagonistas & inhibidores , Receptor trkA/metabolismo
6.
Neurochem Res ; 41(11): 2993-3003, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27473385

RESUMEN

Cisplatin is the most effective and neurotoxic platinum chemotherapeutic agent. It induces a peripheral neuropathy characterized by distal axonal degeneration that might progress to degeneration of cell bodies and apoptosis. Most symptoms occur nearby distal axonal branches and axonal degeneration might induce peripheral neuropathy regardless neuronal apoptosis. The toxic mechanism of cisplatin has been mainly associated with DNA damage, but cisplatin might also affect neurite outgrowth. Nevertheless, the neurotoxic mechanism of cisplatin remains unclear. We investigated the early effects of cisplatin on axonal plasticity by using non-cytotoxic concentrations of cisplatin and PC12 cells as a model of neurite outgrowth and differentiation. PC12 cells express NGF-receptors (trkA) and respond to NGF by forming neurites, branches and synaptic vesicles. For comparison, we used a neuronal model (SH-SY5Y cells) that does not express trkA nor responds to NGF. Cisplatin did not change NGF expression in PC12 cells and decreased neurite outgrowth in both models, suggesting a NGF/trkA independent mechanism. It also reduced axonal growth (GAP-43) and synaptic (synapsin I and synaptophysin) proteins in PC12 cells, without inducing mitochondrial damage or apoptosis. Therefore, cisplatin might affect axonal plasticity before DNA damage, NGF/trkA down-regulation, mitochondrial damage or neuronal apoptosis. This is the first study to show that neuroplasticity-related proteins might be early targets of the neurotoxic action of cisplatin and their role on cisplatin-induced peripheral neuropathy should be investigated in vivo.


Asunto(s)
Cisplatino/farmacología , Factor de Crecimiento Nervioso/metabolismo , Proyección Neuronal/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Proteína GAP-43/metabolismo , Neuritas/efectos de los fármacos , Neuritas/fisiología , Células PC12 , Ratas , Receptores de Factor de Crecimiento Nervioso/metabolismo
7.
Toxicol In Vitro ; 30(1 Pt B): 231-40, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26556726

RESUMEN

Cannabidiol (CBD) is a non-psychoactive constituent of Cannabis sativa with potential to treat neurodegenerative diseases. Its neuroprotection has been mainly associated with anti-inflammatory and antioxidant events; however, other mechanisms might be involved. We investigated the involvement of neuritogenesis, NGF receptors (trkA), NGF, and neuronal proteins in the mechanism of neuroprotection of CBD against MPP(+) toxicity in PC12 cells. CBD increased cell viability, differentiation, and the expression of axonal (GAP-43) and synaptic (synaptophysin and synapsin I) proteins. Its neuritogenic effect was not dependent or additive to NGF, but it was inhibited by K252a (trkA inhibitor). CBD did not increase the expression of NGF, but protected against its decrease induced by MPP(+), probably by an indirect mechanism. We also evaluated the neuritogenesis in SH-SY5Y cells, which do not express trkA receptors. CBD did not induce neuritogenesis in this cellular model, which supports the involvement of trkA receptors. This is the first study to report the involvement of neuronal proteins and trkA in the neuroprotection of CBD. Our findings suggest that CBD has a neurorestorative potential independent of NGF that might contribute to its neuroprotection against MPP(+), a neurotoxin relevant to Parkinson's disease.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Cannabidiol/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Neuritas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/prevención & control , Receptor trkA/fisiología , Animales , Axones/metabolismo , Humanos , Factor de Crecimiento Nervioso/fisiología , Neuritas/fisiología , Neuroblastoma/patología , Células PC12 , Ratas , Sinapsis/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...