Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 476
Filtrar
1.
J Toxicol Environ Health A ; 87(11): 457-470, 2024 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-38576186

RESUMEN

Glutamate is one of the predominant excitatory neurotransmitters released from the central nervous system; however, at high concentrations, this substance may induce excitotoxicity. This phenomenon is involved in numerous neuropathologies. At present, clinically available pharmacotherapeutic agents to counteract glutamatergic excitotoxicity are not completely effective; therefore, research to develop novel compounds is necessary. In this study, the main objective was to determine the pharmacotherapeutic potential of the hydroalcoholic extract of Psidium guajava (PG) in a model of oxidative stress-induced by exposure to glutamate utilizing Danio rerio larvae (zebrafish) as a model. Data showed that treatment with glutamate produced a significant increase in oxidative stress, chromatin damage, apoptosis, and locomotor dysfunction. All these effects were attenuated by pre-treatment with the classical antioxidant N-acetylcysteine (NAC). Treatment with PG inhibited oxidative stress responsible for cellular damage induced by glutamate. However, exposure to PG failed to prevent glutamate-initiated locomotor damage. Our findings suggest that under conditions of oxidative stress, PG can be considered as a promising candidate for treatment of glutamatergic excitotoxicity and consequent neurodegenerative diseases.


Asunto(s)
Psidium , Pez Cebra , Animales , Glutamatos/toxicidad , Estrés Oxidativo , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Hojas de la Planta
2.
Nat Commun ; 12(1): 847, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33558503

RESUMEN

A large G4C2-repeat expansion in C9orf72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Neuronal degeneration associated with this expansion arises from a loss of C9orf72 protein, the accumulation of RNA foci, the expression of dipeptide repeat (DPR) proteins, or all these factors. We report the discovery of a new targeting sequence that is common to all C9orf72 transcripts but enables preferential knockdown of repeat-containing transcripts in multiple cellular models and C9BAC transgenic mice. We optimize stereopure oligonucleotides that act through this site, and we demonstrate that their preferential activity depends on both backbone stereochemistry and asymmetric wing design. In mice, stereopure oligonucleotides produce durable depletion of pathogenic signatures without disrupting protein expression. These oligonucleotides selectively protect motor neurons harboring C9orf72-expansion mutation from glutamate-induced toxicity. We hypothesize that targeting C9orf72 with stereopure oligonucleotides may be a viable therapeutic approach for the treatment of C9orf72-associated neurodegenerative disorders.


Asunto(s)
Proteína C9orf72/genética , Expansión de las Repeticiones de ADN/genética , Mutación/genética , Oligonucleótidos/química , Oligonucleótidos/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proteína C9orf72/química , Exones/genética , Glutamatos/toxicidad , Intrones/genética , Ratones , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estereoisomerismo
3.
Toxicol In Vitro ; 65: 104780, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31978514

RESUMEN

Food additives are approved chemicals used for various purposes in foods; to provide nutritional safety, increase flavor, extend shelf life, reduce nutrient losses etc. In this study, the in vitro genotoxic effects of flavor enhancers, Monopotassium glutamate (MPG) and Magnesium diglutamate (MDG) were investigated in human peripheral blood lymphocytes by using chromosome aberrations (CAs), sister chromatid exchanges (SCEs), cytokinesis-block micronucleus cytome (CBMN-Cyt), and comet assays. Four concentrations of MPG (125, 250, 500, and 1000 µg/mL) and MDG (93.75, 187.5, 375, and 750 µg/mL) were used. Both food additives significantly reduced mitotic index and increased the frequency of CAs at high concentrations. MPG and MDG (except 93.75 µg/mL) significantly increased SCEs/Cell in concentration-dependent manner. In the CBMN-Cyt test, both MPG and MDG increased the formation of micronucleus, nuclear buds, and nucleoplasmic bridges compared to control in a concentration-dependent manner. However, these increases were statistically significant at higher concentrations. MPG (at 500 and 1000 µg/mL) and MDG (except 93.75 µg/mL) significantly increased DNA damages observed by comet assay. It is concluded from these results that MPG and MDG have clastogenic, mutagenic, aneugenic, and cytotoxic effects, particularly at high concentrations in human lymphocytes in vitro.


Asunto(s)
Aditivos Alimentarios/toxicidad , Glutamatos/toxicidad , Mutágenos/toxicidad , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Linfocitos/efectos de los fármacos , Pruebas de Mutagenicidad
4.
Transplantation ; 103(6): 1140-1151, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30801551

RESUMEN

BACKGROUND: Storage of donor hearts in cardioplegic solutions supplemented with conditioning agents activating endogenous mitochondrial protective signaling enhanced their postreperfusion recovery. The present study investigates the role of timing and duration of cardiac exposure to cyclosporine A (CsA), another putative mitochondrial protectant, on cardiac functional recovery and potential mechanisms of CsA action in an isolated working rat heart model of donor heart retrieval and storage. METHODS: After measurement of baseline function, hearts were arrested and stored for 6 hours at 4°C in either Celsior alone or Celsior + CsA (0.2 µM), then reperfused for 45 minutes in Krebs solution, when functional recovery was assessed. Two additional groups of Celsior-alone stored hearts were exposed to 0.2 µM CsA for the initial 15 minutes (nonworking period) or the full 45-minute period of reperfusion. Coronary effluent was collected pre- and poststorage for assessment of lactate dehydrogenase release. Tissue samples were collected at the end of each study for immunoblotting and histological studies. RESULTS: CsA supplementation during cold storage or the first 15-minute reperfusion significantly improved functional recovery and significantly increased phospho-AMPKαThr172 and phospho-ULK-1Ser757. Hearts exposed to CsA for 45 minutes at reperfusion recovered poorly with no phospho-AMP-activated protein kinase α activation, decreased phospho-eNOSSer633, and decreased mitochondrial cytochrome c content with increased lactate dehydrogenase release. CONCLUSIONS: Inclusion of CsA during cold storage is cardioprotective. Effects of CsA addition to the perfusate during reperfusion were time dependent, with benefits at 15 minutes but not 45 minutes of reperfusion. The toxic effect with the presence of CsA for the full 45-minute reperfusion is associated with impaired mitochondrial integrity and decreased eNOS phosphorylation.


Asunto(s)
Soluciones Cardiopléjicas/farmacología , Ciclosporina/farmacología , Trasplante de Corazón , Corazón/efectos de los fármacos , Soluciones Preservantes de Órganos/farmacología , Preservación de Órganos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Soluciones Cardiopléjicas/toxicidad , Isquemia Fría , Ciclosporina/toxicidad , Disacáridos/farmacología , Disacáridos/toxicidad , Electrólitos/farmacología , Electrólitos/toxicidad , Glutamatos/farmacología , Glutamatos/toxicidad , Glutatión/farmacología , Glutatión/toxicidad , Corazón/fisiopatología , Trasplante de Corazón/efectos adversos , Histidina/farmacología , Histidina/toxicidad , Preparación de Corazón Aislado , Masculino , Manitol/farmacología , Manitol/toxicidad , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Soluciones Preservantes de Órganos/toxicidad , Fosforilación , Ratas Wistar , Recuperación de la Función , Factores de Tiempo
5.
Mol Neurobiol ; 55(11): 8563-8585, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29564811

RESUMEN

Astrocytes play metabolic and structural support roles and contribute to the integrity of the blood-brain barrier (BBB), linking communication between neurons and the endothelium. Cyclin-dependent kinase 5 (CDK5) likely exerts a dual effect on the endothelium and astrocytes due to its involvement in migration and angiogenesis; the overactivation of CDK5 is associated with dysfunction in glutamate recapture and hypoxia. Recently, we proposed that CDK5-targeted astrocytes facilitate the recovery of neurological and motor function in transplanted ischemic rats. In the current study, we treated cerebral ischemic rats and endothelial cells exposed to glutamate toxicity with CDK5 knock-down (CDK5-KD) astrocytes to determine the role of CDK5 in neurovascular integrity. We found that the effects of CDK5-KD were sustained for 4 months, preventing neuronal and astrocyte loss, facilitating the recovery of the BBB via the production of BDNF by endogenous astrocytes (GFP-) surrounding vessels in the motor cortex and the corpus callosum of global ischemic rats, and improving neurological performance. These findings were supported by the in vitro findings of increased transendothelial resistance, p120-ctn+ adhesion and reduced intercellular gaps induced by a CDK5 inhibitor (roscovitine) in bEnd.3 cells in a glutamate-toxicity model. Additionally, CDK5-KD astrocytes in co-culture protected the endothelial cell viability, increased BDNF release from astrocytes, increased BDNF immunoreactivity in neighboring astrocytes and endothelial cells and enhanced cell adhesion in a glutamate-toxicity model. Altogether, these findings suggest that a CDK5 reduction in astrocytes protects the endothelium, which promotes BDNF release, endothelial adhesion, and the recovery of neurovascular unit integrity and brain function in ischemic rats.


Asunto(s)
Astrocitos/trasplante , Isquemia Encefálica/enzimología , Isquemia Encefálica/terapia , Encéfalo/irrigación sanguínea , Quinasa 5 Dependiente de la Ciclina/metabolismo , Técnicas de Silenciamiento del Gen , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Adhesión Celular , Línea Celular , Técnicas de Cocultivo , Cuerpo Calloso/metabolismo , Modelos Animales de Enfermedad , Impedancia Eléctrica , Células Endoteliales/metabolismo , Glutamatos/toxicidad , Masculino , Ratones , Actividad Motora , Corteza Motora/metabolismo , Corteza Motora/patología , Neuronas/metabolismo , Ratas Wistar , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología
6.
Cell Death Dis ; 9(2): 88, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29367744

RESUMEN

Glaucoma is a leading cause of irreversible blindness and characterized by progressive damage of retinal ganglion cells (RGCs). Growing evidences have linked impaired mitophagy with neurodegenerative diseases, while the E3 ubiquitin ligase parkin may play a key role. However, the pathophysiological relationship between parkin and glaucoma remains largely unknown. Using chronic hypertensive glaucoma rats induced by translimbal laser photocoagulation, we show here that the protein level of parkin and its downstream optineurin proteins were increased in hypertensive retinas. The ratio of LC3-II to LC3-I, the number of mitophagosomes, and unhealthy mitochondria were increased in hypertensive optic nerves. Overexpression of parkin by viral vectors increased RGC survival in glaucomatous rats in vivo and under excitotoxicity in vitro. It also promoted optineurin expression and improved mitochondrial health. In parkin-overexpressed glaucomatous rats, the ratio of LC3-II to LC3-I, LAMP1 level, and the number of mitophagosomes in optic nerve were decreased at 3 days, yet increased at 2 weeks following intraocular pressure (IOP) elevation. These findings demonstrate that dysfunction of mitophagy exist in RGCs of glaucomatous rats. Overexpression of parkin exerted a significant protective effect on RGCs and partially restored dysfunction of mitophagy in response to cumulative IOP elevation.


Asunto(s)
Glaucoma/metabolismo , Glaucoma/patología , Neuroprotección , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Supervivencia Celular , Enfermedad Crónica , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Glaucoma/fisiopatología , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamatos/toxicidad , Hipertensión/fisiopatología , Presión Intraocular , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Mitofagia , Neurotoxinas/toxicidad , Nervio Óptico/metabolismo , Ratas Sprague-Dawley , Células Ganglionares de la Retina/ultraestructura , Factor de Transcripción TFIIIA/metabolismo
7.
J Toxicol Sci ; 41(2): 311-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26961616

RESUMEN

Citrinin, a natural mycotoxin that is found in fermented foods, is known as a cytotoxin and nephrotoxin. Exposure to high doses of citrinin result in apoptosis; however, the effects of low doses are not fully understood. Glutamate excitotoxicity is responsible for neuronal death in acute neurological disorders including stroke, trauma and other neurodegenerative diseases. Here, we show the neuroprotective effect of low doses of citrinin against glutamate-induced excitotoxicity. We examined the effect of citrinin exposure on glutamate-induced cell death in cultured rat cortical neurons under two conditions: simultaneous treatment with citrinin 0.1 to 1,000 nM and glutamate (30 µM) for 1, 3 hr; the same simultaneous treatment for 3 hr after pretreatment with citrinin for 21 hr. Both the MTT and immunocytochemical assay showed significant neuroprotective effects at several doses and exposure times tested. All concentrations of citrinin tested showed no remarkable cell death following 14-day exposure, and no marked alterations to synapses. These data suggest that low doses of citrinin can be used as a neuroprotective agent against glutamate-induced excitotoxicity without additional harmful cellular alterations.


Asunto(s)
Muerte Celular/efectos de los fármacos , Corteza Cerebral/citología , Citrinina/farmacología , Glutamatos/toxicidad , Micotoxinas/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ratas Wistar , Factores de Tiempo
8.
Food Funct ; 7(2): 964-74, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26732548

RESUMEN

Numerous factors can induce oxidative stress in animal production and lead to growth retardation, disease, and even death. Arginine and N-carbamylglutamate can alleviate the effects of oxidative stress. However, the systematic changes in metabolic biochemistry linked to oxidative stress and arginine and N-carbamylglutamate treatment remain largely unknown. This study aims to examine the effects of arginine and N-carbamylglutamate on rat metabolism under oxidative stress. Thirty rats were randomly divided into three dietary groups (n = 10 each). The rats were fed a basal diet supplemented with 0 (control), 1% arginine, or 0.1% N-carbamylglutamate for 30 days. On day 28, the rats in each treatment were intraperitoneally injected with diquat at 12 mg per kg body weight or sterile solution. Urine and plasma samples were analyzed by metabolomics. Compared with the diquat group, the arginine + diquat group had significantly lower levels of acetamide, alanine, lysine, pyruvate, tyrosine, α-glucose, and ß-glucose in plasma; N-carbamylglutamate + diquat had higher levels of 3-hydroxybutyrate, 3-methylhistidine, acetone, allantoin, asparagine, citrate, phenylalanine, trimethylamine-N-oxide, and tyrosine, and lower levels of low density lipoprotein, lipid, lysine, threonine, unsaturated lipid, urea, and very low density lipoprotein (P < 0.05) in plasma. Compared with the diquat group, the arginine + diquat group had significantly higher levels of citrate, creatinine, homogentisate, and α-ketoglutarate while lower levels of acetamide, citrulline, ethanol, glycine, isobutyrate, lactate, malonate, methymalonate, N-acetylglutamate, N-methylnicotinamide, propionate, and ß-glucose (P < 0.05) in urine. Compared with the diquat group, the N-carbamylglutamate + diquat group had significantly higher levels of allantoin, citrate, homogentisate, phenylacetylglycine, α-ketoglutarate, and ß-glucose while lower levels of acetamide, acetate, acetone, benzoate, citrulline, ethanol, hippurate, lactate, N-acetylglutamate, nicotinamide, ornithine, and trigonelline (P < 0.05) in urine. Overall, these results suggest that arginine and N-carbamylglutamate can alter the metabolome associated with energy metabolism, amino acid metabolism, and gut microbiota metabolism under oxidative stress.


Asunto(s)
Arginina/toxicidad , Diquat/toxicidad , Glutamatos/toxicidad , Metaboloma/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Alanina Transaminasa/sangre , Aminoácidos/sangre , Animales , Aspartato Aminotransferasas/sangre , Metabolismo Energético , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Análisis Multivariante , Ratas , Ratas Sprague-Dawley
9.
ACS Nano ; 9(11): 10801-16, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26440073

RESUMEN

Alzheimer's disease (AD) exerts a heavy health burden for modern society and has a complicated pathological background. The accumulation of extracellular ß-amyloid (Aß) is crucial in AD pathogenesis, and Aß-initiated secondary pathological processes could independently lead to neuronal degeneration and pathogenesis in AD. Thus, the development of combination therapeutics that can not only accelerate Aß clearance but also simultaneously protect neurons or inhibit other subsequent pathological cascade represents a promising strategy for AD intervention. Here, we designed a nanostructure, monosialotetrahexosylganglioside (GM1)-modified reconstituted high density lipoprotein (GM1-rHDL), that possesses antibody-like high binding affinity to Aß, facilitates Aß degradation by microglia, and Aß efflux across the blood-brain barrier (BBB), displays high brain biodistribution efficiency following intranasal administration, and simultaneously allows the efficient loading of a neuroprotective peptide, NAP, as a nanoparticulate drug delivery system for the combination therapy of AD. The resulting multifunctional nanostructure, αNAP-GM1-rHDL, was found to be able to protect neurons from Aß(1-42) oligomer/glutamic acid-induced cell toxicity better than GM1-rHDL in vitro and reduced Aß deposition, ameliorated neurologic changes, and rescued memory loss more efficiently than both αNAP solution and GM1-rHDL in AD model mice following intranasal administration with no observable cytotoxicity noted. Taken together, this work presents direct experimental evidence of the rational design of a biomimetic nanostructure to serve as a safe and efficient multifunctional nanoplatform for the combination therapy of AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Gangliósido G(M1)/metabolismo , Lipoproteínas HDL/metabolismo , Nanopartículas/química , Administración Intranasal , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteínas E/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Terapia Combinada , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Gangliósido G(M1)/administración & dosificación , Gangliósido G(M1)/farmacocinética , Glutamatos/toxicidad , Lipoproteínas HDL/administración & dosificación , Lipoproteínas HDL/farmacocinética , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/patología , Ratones Endogámicos ICR , Microglía/efectos de los fármacos , Microglía/metabolismo , Datos de Secuencia Molecular , Nanopartículas/ultraestructura , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Tamaño de la Partícula , Fragmentos de Péptidos/metabolismo , Péptidos/química , Ratas Sprague-Dawley , Electricidad Estática , Distribución Tisular/efectos de los fármacos
10.
Drug Des Devel Ther ; 9: 4105-13, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26251576

RESUMEN

Celecoxib, a selective cyclooxygenase-2 inhibitor, is potentially useful for the treatment of colonic diseases such as colorectal cancer and colitis. However, the cardiovascular toxicity of celecoxib limits its routine use in the clinic. Generally, colon-specific delivery of a drug both increases the therapeutic availability in the large intestine and decreases the systemic absorption of the drug, most likely resulting in enhanced therapeutic effects against colonic diseases such as colitis and reduced systemic side effects. To develop a colon-specific prodrug of celecoxib that could reduce its cardiovascular toxicity and improve its therapeutic activity, dextran-glutamic acid-celecoxib conjugate (glutam-1-yl celecoxib-dextran ester [G1CD]) was prepared and evaluated. While stable in pH 1.2 and 6.8 buffer solutions and small-intestinal contents, G1CD efficiently released celecoxib in cecal contents. Oral administration of G1CD to rats delivered a larger amount of celecoxib to the large intestine than free celecoxib. G1CD prevented the systemic absorption of celecoxib and did not decrease the serum level of 6-ketoprostaglandin F1α, an inverse indicator of cardiovascular toxicity of celecoxib. Collectively, G1CD may be a polymeric colon-specific celecoxib prodrug with therapeutic and toxicological advantages.


Asunto(s)
Celecoxib/metabolismo , Colon/metabolismo , Inhibidores de la Ciclooxigenasa 2/metabolismo , Dextranos/metabolismo , Portadores de Fármacos , Glutamatos/metabolismo , Absorción Intestinal , Profármacos/metabolismo , 6-Cetoprostaglandina F1 alfa/sangre , Administración Oral , Animales , Tampones (Química) , Ciego/metabolismo , Celecoxib/administración & dosificación , Celecoxib/análogos & derivados , Celecoxib/síntesis química , Celecoxib/toxicidad , Química Farmacéutica , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Inhibidores de la Ciclooxigenasa 2/síntesis química , Inhibidores de la Ciclooxigenasa 2/toxicidad , Dextranos/administración & dosificación , Dextranos/síntesis química , Dextranos/toxicidad , Estabilidad de Medicamentos , Glutamatos/administración & dosificación , Glutamatos/síntesis química , Glutamatos/toxicidad , Concentración de Iones de Hidrógeno , Intestino Delgado/metabolismo , Masculino , Profármacos/administración & dosificación , Profármacos/síntesis química , Profármacos/toxicidad , Ratas Sprague-Dawley , Tecnología Farmacéutica/métodos
11.
Rinsho Shinkeigaku ; 54(12): 1066-8, 2014.
Artículo en Japonés | MEDLINE | ID: mdl-25519963

RESUMEN

The precise mechanisms of cortical damage in multiple sclerosis (MS) remain unknown. Microglia, the resident immune cells in the central nervous system (CNS), are involved in the chronic neuroinflammation in MS cortical lesions. Microglia produce various inflammatory cytokines such as IFN-γ and IL-ß, reactive oxygen species, and glutamate. IL-ß secretion is induced by NLRP3. ROS is induced by GM-CSF-producing Th17 cells. Glutamate is released via gap junctions. These molecules exert neurotoxicity. Meanwhile, damaged neurons produce fractalkine and FGF-2, which suppress microglial activation and enhance microglial neuroprotection through anti-inflammatory and anti-oxidant effect. Fractalkine accelerates microglial clearance of neuronal debris via inducing the release of MFG-E8. FGF-2 induces microglial migration through the FGFR3-Wnt-ERK signaling pathway. These molecules suppress microglial neuroinflammation, and enhance neuroprotection, which may give us clues for future therapeutic strategy cortical damage in MS.


Asunto(s)
Corteza Cerebral/patología , Microglía , Esclerosis Múltiple/patología , Antiinflamatorios , Antígenos de Superficie/metabolismo , Antioxidantes/farmacología , Proteínas Portadoras/fisiología , Movimiento Celular/efectos de los fármacos , Quimiocina CX3CL1/metabolismo , Quimiocina CX3CL1/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Uniones Comunicantes/metabolismo , Glutamatos/metabolismo , Glutamatos/toxicidad , Humanos , Interferón gamma/metabolismo , Interferón gamma/toxicidad , Interleucina-1beta/metabolismo , Interleucina-1beta/toxicidad , Sistema de Señalización de MAP Quinasas , Microglía/metabolismo , Microglía/patología , Proteínas de la Leche/metabolismo , Terapia Molecular Dirigida , Esclerosis Múltiple/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Fármacos Neuroprotectores , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/toxicidad , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/fisiología , Células Th17/fisiología , Vía de Señalización Wnt
12.
Oncotarget ; 5(18): 8528-43, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25138052

RESUMEN

The molecularly targeted agents, including anti-VEGF or anti-EGFR monoclonal antibody and some inhibitors of EGFR tyrosine kinase, are effective in the treatment of non-small-cell lung cancer (NSCLC) to a certain extent, but the benefit for a proportion of patients is still limited. Hence, it is necessary and urgent to develop more selective and effective molecular targeted agents against lung cancer. Here, we have synthesized novel theanine derivatives, methyl coumarin-3-carboxylyl L-theanine (TMC), ethyl coumarin-3-carboxylyl L-theanine (TEC), ethyl 6-fluorocoumarin- 3-carboxylyl L-theanine (TFC), and ethyl 6-nitrocoumarin-3-carboxylyl L-theanine (TNC), which are fluorescent small molecules, based on their parental compound theanine and studied their anticancer activities in vitro, ex vivo and in vivo models of human and mouse cancers. Our results show that the four theanine derivatives significantly inhibit the lung cancer cell migration and the growth of lung cancer and leukemia cell lines. TFC and TNC display enhanced effects with anticancer drugs cytarabine vincristine, andmethotrexate on inhibition of lung cancer cell growth and no toxicity to the normal human embryonic lung fibroblast and peripheral blood lymphocytes. TFC and TNC exhibit strong suppression of the highly metastatic Lewis lung cancer (LLC) and A549 tumor growth in tumor-bearing mice without toxicity to mice. TFC and TNC can effectively suppress the growth of lung cancer cells in vitro, ex vivo and in vivo by targeting EGFR/VEGFR-Akt/NF-κB pathways. Our study has suggested that TFC and TNC may have the therapeutic and/or adjuvant therapeutic applications in the treatment of lung cancers and other cancer.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Glutamatos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/patología , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Receptores ErbB/metabolismo , Femenino , Glutamatos/síntesis química , Glutamatos/toxicidad , Humanos , Células K562 , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Leucemia/patología , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Terapia Molecular Dirigida , FN-kappa B/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Mol Brain ; 7: 52, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25047048

RESUMEN

BACKGROUND: TRPM4 channels are Ca2+-activated nonselective cation channels which are deeply involved in physiological and pathological conditions. However, their trafficking mechanism and binding partners are still elusive. RESULTS: We have found the 14-3-3γ as a binding partner for TRPM4b using its N-terminal fragment from the yeast-two hybrid screening. Ser88 at the N-terminus of TRPM4b is critical for 14-3-3γ binding by showing GST pull-down and co-immunoprecipitation. Heterologous overexpression of 14-3-3γ in HEK293T cells increased TRPM4b expression on the plasma membrane which was measured by whole-cell recordings and cell surface biotinylation experiment. Surface expression of TRPM4b was greatly reduced by short hairpin RNA (shRNA) against 14-3-3γ. Next, endogenous TRPM4b-mediated currents were electrophysiologically characterized by application of glutamate and 9-phenanthrol, a TRPM4b specific antagonist in HT-22 cells which originated from mouse hippocampal neurons. Glutamate-induced TRPM4b currents were significantly attenuated by shRNAs against 14-3-3γ or TRPM4b in these cells. Finally, glutamate-induced cell death was greatly prevented by treatment of 9-phenanthrol or 14-3-3γ shRNA. CONCLUSION: These results showed that the cell surface expression of TRPM4 channels is mediated by 14-3-3γ binding, and the specific inhibition of this trafficking process can be a potential therapeutic target for glutamate-induced neuronal cell death.


Asunto(s)
Proteínas 14-3-3/deficiencia , Membrana Celular/metabolismo , Glutamatos/toxicidad , Neuronas/citología , Canales Catiónicos TRPM/metabolismo , Proteínas 14-3-3/metabolismo , Secuencia de Aminoácidos , Animales , Muerte Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HEK293 , Hipocampo/citología , Humanos , Activación del Canal Iónico/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Serina/metabolismo , Relación Estructura-Actividad , Canales Catiónicos TRPM/química
14.
BMC Cancer ; 14: 508, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25012241

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) patients with interstitial lung disease (ILD) need to be approached carefully given the high incidence of pulmonary toxicity. Pemetrexed (PEM) is the key drug for the treatment of NSCLC. However, its safety, especially with respect to the exacerbation of ILD, and efficacy in NSCLC patients with ILD have yet to be established. METHOD: We investigated the safety and efficacy of PEM monotherapy in NSCLC patients with or without idiopathic interstitial pneumonia (IIPs). The medical charts of these patients were retrospectively reviewed. RESULTS: Twenty-five patients diagnosed as having IIPs (IIPs group) and 88 patients without ILD (non-ILD group) were treated with PEM monotherapy at Juntendo University Hospital between 2009 and 2013. In the IIPs group, 12 patients were found to have usual interstitial pneumonitis (UIP) on chest computed tomography (CT) (UIP group) and the other 13 patients showed a non-UIP pattern on chest CT (non-UIP IIPs group). Three patients in the IIPs group (2 in the UIP group and 1 in the non-UIP IIPs group) and 1 in the non-ILD group developed pulmonary toxicity during treatment (3.5% overall, 12.0% in the IIPs group versus 1.1% in the non-ILD group). Moreover, all 3 patients in the IIPs group died of pulmonary toxicity. Overall survival tended to be longer in the non-ILD group than in the IIPs group (p = 0.08). Multivariate analyses demonstrated that IIPs was the only significant independent risk factor for PEM-related pulmonary toxicity. CONCLUSION: We found that the incidence of PEM-related pulmonary toxicity was significantly higher amongst NSCLC patients with IIPs than among those without IIPs. Particular care must be taken when administering PEM to treat NSCLC patients with IIPs.


Asunto(s)
Antineoplásicos/toxicidad , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Glutamatos/toxicidad , Guanina/análogos & derivados , Neumonías Intersticiales Idiopáticas/complicaciones , Neumonías Intersticiales Idiopáticas/mortalidad , Neoplasias Pulmonares/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Antineoplásicos/administración & dosificación , Femenino , Glutamatos/administración & dosificación , Guanina/administración & dosificación , Guanina/toxicidad , Humanos , Masculino , Persona de Mediana Edad , Pemetrexed , Análisis de Supervivencia
15.
Mol Med Rep ; 10(2): 931-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24840891

RESUMEN

Currently, chemotherapy and targeted therapies share the principal limitation of the emergence of drug resistance, which prevents these strategies from having lasting clinical benefits. The combination of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) with concurrent chemotherapy has been proposed as one strategy to overcome acquired resistance to EGFR-TKIs. The purpose of the present study was to investigate the combined effects of gefitinib and pemetrexed on EGFR-TKI-sensitive and EGFR­TKI­resistant human non-small cell lung cancer (NSCLC) cell lines. The antiproliferative effects of gefitinib and pemetrexed, alone and in combination, on the growth of NSCLC cell lines, were assessed using an MTT assay. The cytotoxic interaction between the two drugs was evaluated in vitro using the combination index (CI) method. Cell cycle distribution and apoptosis were analyzed by flow cytometry and alterations in signaling pathways were determined by western blot analysis. In the present study, it was identified that when cells were concurrently exposed to pemetrexed and gefitinib, cytotoxic synergism was present in the gefitinib-resistant PC9/GR human NSCLC cell line and antagonistic interactions were observed in the gefitinib-sensitive PC9 cell line. Synergism was associated with a combination of cell cycle effects of the different agents. In addition, the combination of pemetrexed and gefitinib decreased the levels of phosphorylated AKT, phosphorylated extracellular-signal-regulated kinase and B-cell lymphoma 2 as compared with those in the control. By contrast, antagonism was associated with gefitinib-induced G0/G1-phase blockade of gefitinib-sensitive cells, which interfered with the cell cycle-specific cytotoxicity of chemotherapy. The combination of pemetrexed and gefitinib generated synergistic effects in gefitinib-acquired resistant cells and antagonistic effects in gefitinib-sensitive cells, suggesting that EGFR-TKIs combined with pemetrexed may be a beneficial treatment strategy for NSCLC patients with acquired resistance to EGFR-TKIs.


Asunto(s)
Antineoplásicos/toxicidad , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/metabolismo , Glutamatos/toxicidad , Guanina/análogos & derivados , Quinazolinas/toxicidad , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Gefitinib , Guanina/toxicidad , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Pemetrexed , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
16.
Curr HIV Res ; 12(3): 174-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24862327

RESUMEN

Heme oxygenase-1 (HO-1) is a highly inducible and ubiquitous cellular enzyme that subserves cytoprotective responses to toxic insults, including inflammation and oxidative stress. In neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis, HO-1 expression is increased, presumably reflecting an endogenous neuroprotective response against ongoing cellular injury. In contrast, we have found that in human immunodeficiency virus (HIV) infection of the brain, which is also associated with inflammation, oxidative stress and neurodegeneration, HO-1 expression is decreased, likely reflecting a unique role for HO-1 deficiency in neurodegeneration pathways activated by HIV infection. We have also shown that HO-1 expression is significantly suppressed by HIV replication in cultured macrophages which represent the primary cellular reservoir for HIV in the brain. HO-1 deficiency is associated with release of neurotoxic levels of glutamate from both HIV-infected and immune-activated macrophages; this glutamate-mediated neurotoxicity is suppressed by pharmacological induction of HO-1 expression in the macrophages. Thus, HO-1 induction could be a therapeutic strategy for neuroprotection against HIV infection and other neuroinflammatory brain diseases. Here, we review various stimuli and signaling pathways regulating HO-1 expression in macrophages, which could promote neuronal survival through HO-1-modulation of endogenous antioxidant and immune modulatory pathways, thus limiting the oxidative stress that can promote HIV disease progression in the CNS. The use of pharmacological inducers of endogenous HO-1 expression as potential adjunctive neuroprotective therapeutics in HIV infection is also discussed.


Asunto(s)
Complejo SIDA Demencia/fisiopatología , Encéfalo/enzimología , Encéfalo/patología , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/biosíntesis , Regulación hacia Abajo , Glutamatos/metabolismo , Glutamatos/toxicidad , Humanos , Macrófagos/enzimología , Macrófagos/virología
17.
Neurochem Res ; 39(5): 922-31, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24706151

RESUMEN

Studies have suggested that mesenchymal stem cells (MSCs) can protect neuronal cells from excitotoxicity, but the underlying mechanisms are still remaining elusive. In the study, we show that microvesicles released by rat bone marrow-derived MSCs (rBMSC-MVs) protect rat pheochromocytoma PC12 cells from glutamate-induced excitotoxicity. BMSC-MVs upregulate Akt phosphorylation and Bcl-2 expression, downregulate Bax expression, and reduce the cleavage of caspase-3 in glutamate-treated PC12 cells. Such protective effects are partially abrogated by inhibiting PI3K, indicating that rBMSC-MVs act via the PI3K/Akt pathway. Transplantation of rBMSC-MVs may, therefore, be a promising strategy to treat cerebral injury or some other neuronal diseases involving excitotoxicity.


Asunto(s)
Glutamatos/toxicidad , Vesículas Transportadoras/fisiología , Animales , Células de la Médula Ósea/metabolismo , Masculino , Células Madre Mesenquimatosas , Células PC12 , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Neurosci Lett ; 564: 11-5, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24513236

RESUMEN

The perinatal hypoxic-ischemic insult frequently leads to mortality, morbidity and plays a key role in the later pathological consequences. The ischemic insult causes a massive release of glutamate and subsequent excitotoxic damage. The neuroactive steroid 3α5ß-pregnanolone glutamate (PG) is a NMDA receptor antagonist acting via use-dependent mechanism and can be used as a neuroprotective agent that may alleviate glutamatergic excitotoxicity in the brain. First, a possible neurotoxic effect of the PG, a novel use-dependent NMDA antagonist, was studied in immature rats. In addition, to compare this effect with a well-described non-competitive NMDA antagonist, the MK-801 (positive control) was used. Animals at postnatal day 12 (P12) were injected intraperitoneally with PG in a doses 1 or 10mg/kg or with MK-801 in a dose 1mg/kg. Effect of PG treatment on the immature brain was evaluated on Fluoro Jade B (FJB) stained sections. Second, a neuroprotective effect of the PG was studied in the model of focal cerebral ischemia in P12. Focal cerebral ischemia was induced by the infusion of the endothelin-1 (ET-1) into the right dorsal hippocampus. PG at the doses 1 or 10mg/kg was administrated intraperitoneally 5min after the end of ET-1 infusion. To evaluate the neuroprotective effect after the PG treatment FJB staining was used. Our results demonstrate a lack of the neurotoxicity of the PG in intact P12. In the second part of the study in the model of the focal ischemia we detected significantly lower occurrence of FJB-positive cells in the afflicted hippocampus in PG treated groups, while animals without PG treatment exhibited massive neurodegeneration. The neuroprotective potential of the PG can serve in the development of therapeutic strategies for brain damage induced by the glutamate excitotoxicity.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Glutamatos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Pregnanolona/análogos & derivados , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Glutamatos/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Pregnanolona/uso terapéutico , Pregnanolona/toxicidad , Ratas , Ratas Wistar
19.
Enzyme Microb Technol ; 55: 31-9, 2014 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-24411443

RESUMEN

Two biosurfactants, surfactin and fatty acyl-glutamate, were produced from genetically-modified strains of Bacillus subtilis on 2% glucose and mineral salts media in shake-flasks and bioreactors. Biosurfactant synthesis ceased when the main carbohydrate source was completely depleted. Surfactin titers were ∼30-fold higher than fatty acyl-glutamate in the same medium. When bacteria were grown in large aerated bioreactors, biosurfactants mostly partitioned to the foam fraction, which was recovered. Dispersion effectiveness of surfactin and fatty acyl-glutamate was evaluated by measuring the critical micelle concentration (CMC) and dispersant-to-oil ratio (DOR). The CMC values for surfactin and fatty acyl-glutamate in double deionized distilled water were 0.015 and 0.10 g/L, respectively. However, CMC values were higher, 0.02 and 0.4 g/L for surfactin and fatty acyl-glutamate, respectively, in 12 parts per thousand Instant Ocean®[corrected].sea salt, which has been partly attributed to saline-induced conformational changes in the solvated ionic species of the biosurfactants. The DORs for surfactin and fatty acyl-glutamate were 1:96 and 1:12, respectively, in water. In Instant Ocean® solutions containing 12 ppt sea salt, these decreased to 1:30 and 1:4, respectively, suggesting reduction in oil dispersing efficiency of both surfactants in saline. Surfactant toxicities were assessed using the Gulf killifish, Fundulus grandis, which is common in estuarine habitats of the Gulf of Mexico. Surfactin was 10-fold more toxic than fatty acyl-glutamate. A commercial surfactant, sodium laurel sulfate, had intermediate toxicity. Raising the salinity from 5 to 25 ppt increased the toxicity of all three surfactants; however, the increase was the lowest for fatty acyl-glutamate.


Asunto(s)
Glutamatos/aislamiento & purificación , Lipopéptidos/aislamiento & purificación , Péptidos Cíclicos/aislamiento & purificación , Contaminación por Petróleo , Tensoactivos/aislamiento & purificación , Contaminantes Químicos del Agua , Animales , Bacillus subtilis/metabolismo , Biodegradación Ambiental , Reactores Biológicos , Fermentación , Fundulidae/crecimiento & desarrollo , Glutamatos/biosíntesis , Glutamatos/farmacología , Glutamatos/toxicidad , Larva/efectos de los fármacos , Lipopéptidos/biosíntesis , Lipopéptidos/farmacología , Lipopéptidos/toxicidad , Micelas , Péptidos Cíclicos/biosíntesis , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/toxicidad , Salinidad , Tensión Superficial , Tensoactivos/metabolismo , Tensoactivos/farmacología , Tensoactivos/toxicidad
20.
PLoS One ; 8(5): e64423, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23691214

RESUMEN

Glutamate receptor (GluR)-mediated neurotoxicity is implicated in a variety of disorders ranging from ischemia to neural degeneration. Under conditions of elevated glutamate, the excessive activation of GluRs causes internalization of pathologic levels of Ca(2+), culminating in bioenergetic failure, organelle degradation, and cell death. Efforts to characterize cellular and molecular aspects of excitotoxicity and conduct therapeutic screening for pharmacologic inhibitors of excitogenic progression have been hindered by limitations associated with primary neuron culture. To address this, we evaluated glutamate-induced neurotoxicity in highly enriched glutamatergic neurons (ESNs) derived from murine embryonic stem cells. As of 18 days in vitro (DIV 18), ESNs were synaptically coupled, exhibited spontaneous network activity with neurotypic mEPSCs and expressed NMDARs and AMPARs with physiological current:voltage behaviors. Addition of 0.78-200 µM glutamate evoked reproducible time- and dose-dependent metabolic failure in 6 h, with a calculated EC50 value of 0.44 µM at 24 h. Using a combination of cell viability assays and electrophysiology, we determined that glutamate-induced toxicity was specifically mediated by NMDARs and could be inhibited by addition of NMDAR antagonists, increased extracellular Mg(2+) or substitution of Ba(2+) for Ca(2+). Glutamate treatment evoked neurite fragmentation and focal swelling by both immunocytochemistry and scanning electron microscopy. Presentation of morphological markers of cell death was dose-dependent, with 0.78-200 µM glutamate resulting in apoptosis and 3000 µM glutamate generating a mixture of necrosis and apoptosis. Addition of neuroprotective small molecules reduced glutamate-induced neurotoxicity in a dose-dependent fashion. These data indicate that ESNs replicate many of the excitogenic mechanisms observed in primary neuron culture, offering a moderate-throughput model of excitotoxicity that combines the verisimilitude of primary neurons with the flexibility and scalability of cultured cells. ESNs therefore offer a physiologically relevant platform that exhibits characteristic NMDAR responses, and appears suitable to evaluate molecular mechanisms of glutamate-induced excitotoxicity and screen for candidate therapeutics.


Asunto(s)
Neuronas/citología , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Células Madre/citología , Animales , Calcio/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Fenómenos Electrofisiológicos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glutamatos/toxicidad , Humanos , Ratones , Neuronas/metabolismo , Proteómica , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA