Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nutr Neurosci ; 25(7): 1374-1391, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33345721

RESUMEN

OBJECTIVE: Parkinson's disease (PD) is a progressive motor disease of unknown etiology. Although neuroprotective ability of endogenous bile acid, tauroursodeoxycholic acid (TUDCA), shown in various diseases, including an acute model of PD,the potential therapeutic role of TUDCA in progressive models of PD that exhibit all aspects of PD has not been elucidated. In the present study, mice were assigned to one of four treatment groups: (1) Probenecid (PROB); (2) TUDCA, (3) MPTP + PROB (MPTPp); and (3) TUDCA + MPTPp. Methods: Markers for dopaminergic function, neuroinflammation, oxidative stress and autophagy were assessed using high performance liquid chromatography (HPLC), immunohistochemistry (IHC) and western blot (WB) methods. Locomotion was measured before and after treatments. Results: MPTPp decreased the expression of dopamine transporters (DAT) and tyrosine hydroxylase (TH), indicating dopaminergic damage, and induced microglial and astroglial activation as demonstrated by IHC analysis. MPTPp also decreased DA and its metabolites as demonstrated by HPLC analysis. Further, MPTPp-induced protein oxidation; increased LAMP-1 expression indicated autophagy and the promotion of alpha-synuclein (α-SYN) aggregation. Discussion: Pretreatment with TUDCA protected against dopaminergic neuronal damage, prevented the microglial and astroglial activation, as well as the DA and DOPAC reductions caused by MPTPp. TUDCA by itself did not produce any significant change, with data similar to the negative control group. Pretreatment with TUDCA prevented protein oxidation and autophagy, in addition to inhibiting α-SYN aggregation. Although TUDCA pretreatment did not significantly affect locomotion, only acute treatment effects were measured, indicating more extensive assessments may be necessary to reveal potential therapeutic effects on behavior. Together, these results suggest that autophagy may be involved in the progression of PD and that TUDCA may attenuate these effects. The efficacy of TUDCA as a novel therapy in patients with PD clearly warrants further study.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Animales , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas , Humanos , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/prevención & control , Ácido Tauroquenodesoxicólico/farmacología , Ácido Tauroquenodesoxicólico/uso terapéutico
2.
Cell Biol Int ; 38(2): 145-53, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24155099

RESUMEN

Several dual-specificity phosphatases (DUSPs) that play key roles in the direct or indirect inactivation of different MAP kinases (MAPKs) have been implicated in human cancers over the past decade. This has led to a growing interest in identifying DUSPs and their specific inhibitors for further testing and validation as therapeutic targets in human cancers. However, the lack of understanding of the complex regulatory mechanisms and cross-talks between MAPK signaling pathways, combined with the fact that DUSPs can act as a double-edged sword in cancer progression, calls for a more careful and thorough investigation. Among the various types of brain cancer, glioblastoma multiforme (GBM) is notorious for its aggressiveness and resistance to current treatment modalities. This has led to the search for new molecular targets, particularly those involving various signaling pathways. DUSPs appear to be a promising target, but much more information on DUSP targets and their effects on GBM is needed before potential therapies can be developed, tested, and validated. This review identifies and summarize the specific roles of DUSP1, DUSP4, DUSP6 and DUSP26 that have been implicated in GBM.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Fosfatasas de Especificidad Dual/antagonistas & inhibidores , Fosfatasas de Especificidad Dual/metabolismo , Inhibidores Enzimáticos/farmacología , Glioblastoma/tratamiento farmacológico , Terapia Molecular Dirigida , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Descubrimiento de Drogas , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Terapia Molecular Dirigida/métodos , Transducción de Señal/efectos de los fármacos
3.
Regul Toxicol Pharmacol ; 70(3): 641-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25265367

RESUMEN

MRI was utilized to probe T2 changes in living brain following exposure of rats to one of ten classical neurotoxicants. Brains were subsequently perfused for classical neuropathology examination. This approach was predicated on the assumption that the T2 changes represent loci of neurotoxicity encompassing those seen using neuropathology techniques. The traditional neurotoxicologic approach of selecting a few arbitrary brain sections is dramatically improved by MRI targeting that can indicate the location(s) at which to collect "smart sections" for subsequent workup. MRI scans can provide the equivalent of 64 coronal sections; the number estimated for full coverage of the rat brain if only traditional neuropathology is utilized. Use of MRI allows each animal to serve as its own control as well as longitudinal observations of the life cycle of the neurotoxic lesion(s) (inception, apex and regression). Optimization of time of sacrifice and selection of an appropriate stain based on MRI-identified brain areas could be greatly enhanced should this approach prove successful. The application of full brain MRI imaging that informs neuropathology offers the potential to dramatically improve detection of neurotoxicity produced by new drugs and facilitate new drug development, review and approval processes, and to qualify an imaging biomarker of neuropathology.


Asunto(s)
Encéfalo/efectos de los fármacos , Neurotoxinas/toxicidad , Animales , Encéfalo/patología , Encéfalo/fisiología , Mapeo Encefálico , Imagen por Resonancia Magnética , Masculino , Síndromes de Neurotoxicidad/patología , Ratas Sprague-Dawley
4.
Hum Mol Genet ; 20(4): 659-69, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21106706

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by expanded polyglutamine repeats in the huntingtin (Htt) protein. Mutant Htt may damage and kill striatal neurons by a mechanism involving reduced production of brain-derived neurotrophic factor (BDNF) and increased oxidative and metabolic stress. Because electroconvulsive shock (ECS) can stimulate the production of BDNF and protect neurons against stress, we determined whether ECS treatment would modify the disease process and provide a therapeutic benefit in a mouse model of HD. ECS (50 mA for 0.2 s) or sham treatment was administered once weekly to male N171-82Q Htt mutant mice beginning at 2 months of age. Endpoints measured included motor function, striatal and cortical pathology, and levels of protein chaperones and BDNF. ECS treatment delayed the onset of motor symptoms and body weight loss and extended the survival of HD mice. Striatal neurodegeneration was attenuated and levels of protein chaperones (Hsp70 and Hsp40) and BDNF were elevated in striatal neurons of ECS-treated compared with sham-treated HD mice. Our findings demonstrate that ECS can increase the resistance of neurons to mutant Htt resulting in improved functional outcome and extended survival. The potential of ECS as an intervention in subjects that inherit the mutant Htt gene merits further consideration.


Asunto(s)
Progresión de la Enfermedad , Electrochoque , Enfermedad de Huntington/patología , Enfermedad de Huntington/terapia , Mutación/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transducción de Señal , Análisis de Supervivencia
5.
J Neurosci Res ; 91(5): 671-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23404341

RESUMEN

Tumor necrosis factor-α (TNF) plays a prominent role in the brain damage and functional deficits that result from ischemic stroke. It was recently reported that the thalidomide analog 3,6'-dithiothalidomide (3,6'-DT) can selectively inhibit the synthesis of TNF in cultured cells. We therefore tested the therapeutic potential of 3,6'-DT in a mouse model of focal ischemic stroke. Administration of 3,6'-DT immediately prior to a stroke or within 3 hr after the stroke reduced infarct volume, neuronal death, and neurological deficits, whereas thalidomide was effective only when administered prior to stroke. Neuroprotection was accompanied by decreased inflammation; 3,6'-DT-treated mice exhibited reduced expression of TNF, interleukin-1ß, and inducible nitric oxide synthase; reduced numbers of activated microglia/macrophages, astrocytes, and neutrophils; and reduced expression of intercellular adhesion molecule-1 in the ischemic brain tissue. 3,6'-DT treatment attenuated stroke-induced disruption of the blood-brain barrier by a mechanism that appears to involve suppression of matrix metalloproteinase-9 and preservation of occludin. Treatment with 3,6'-DT did not reduce ischemic brain damage in mice lacking TNF receptors, consistent with a critical role for suppression of TNF production and TNF signaling in the therapeutic action of 3,6'-DT. These findings suggest that anti-inflammatory mechanisms underlie the therapeutic actions of 3,6-DT in an animal model of stroke.


Asunto(s)
Antiinflamatorios/uso terapéutico , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Talidomida/análogos & derivados , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Etiquetado Corte-Fin in Situ , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/genética , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Talidomida/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
Nat Med ; 12(6): 621-3, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16680150

RESUMEN

Mice transgenic for antisense Notch and normal mice treated with inhibitors of the Notch-activating enzyme gamma-secretase showed reduced damage to brain cells and improved functional outcome in a model of focal ischemic stroke. Notch endangers neurons by modulating pathways that increase their vulnerability to apoptosis, and by activating microglial cells and stimulating the infiltration of proinflammatory leukocytes. These findings suggest that Notch signaling may be a therapeutic target for treatment of stroke and related neurodegenerative conditions.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Endopeptidasas/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Accidente Cerebrovascular/patología , Secretasas de la Proteína Precursora del Amiloide , Animales , Apoptosis , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Células Cultivadas , Endopeptidasas/genética , Inhibidores Enzimáticos/metabolismo , Humanos , Leucocitos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Péptidos/genética , Péptidos/metabolismo , Ratas , Receptor Notch1/genética , Daño por Reperfusión , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia , Resultado del Tratamiento
7.
PLoS Biol ; 7(8): e1000176, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19688041

RESUMEN

During embryogenesis, the neural stem cells (NSC) of the developing cerebral cortex are located in the ventricular zone (VZ) lining the cerebral ventricles. They exhibit apical and basal processes that contact the ventricular surface and the pial basement membrane, respectively. This unique architecture is important for VZ physical integrity and fate determination of NSC daughter cells. In addition, the shorter apical process is critical for interkinetic nuclear migration (INM), which enables VZ cell mitoses at the ventricular surface. Despite their importance, the mechanisms required for NSC adhesion to the ventricle are poorly understood. We have shown previously that one class of candidate adhesion molecules, laminins, are present in the ventricular region and that their integrin receptors are expressed by NSC. However, prior studies only demonstrate a role for their interaction in the attachment of the basal process to the overlying pial basement membrane. Here we use antibody-blocking and genetic experiments to reveal an additional and novel requirement for laminin/integrin interactions in apical process adhesion and NSC regulation. Transient abrogation of integrin binding and signalling using blocking antibodies to specifically target the ventricular region in utero results in abnormal INM and alterations in the orientation of NSC divisions. We found that these defects were also observed in laminin alpha2 deficient mice. More detailed analyses using a multidisciplinary approach to analyse stem cell behaviour by expression of fluorescent transgenes and multiphoton time-lapse imaging revealed that the transient embryonic disruption of laminin/integrin signalling at the VZ surface resulted in apical process detachment from the ventricular surface, dystrophic radial glia fibers, and substantial layering defects in the postnatal neocortex. Collectively, these data reveal novel roles for the laminin/integrin interaction in anchoring embryonic NSCs to the ventricular surface and maintaining the physical integrity of the neocortical niche, with even transient perturbations resulting in long-lasting cortical defects.


Asunto(s)
Ventrículos Cerebrales , Regulación del Desarrollo de la Expresión Génica , Cadenas beta de Integrinas/metabolismo , Neocórtex/embriología , Transducción de Señal , Células Madre/citología , Animales , Adhesión Celular , Diferenciación Celular , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/embriología , Ventrículos Cerebrales/fisiología , Embrión de Mamíferos , Procesamiento de Imagen Asistido por Computador , Cadenas beta de Integrinas/genética , Laminina/genética , Laminina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Neocórtex/citología , Neocórtex/metabolismo , Neuronas/citología , Neuronas/metabolismo
8.
Acta Neurochir Suppl ; 113: 59-64, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22116425

RESUMEN

Acetazolamide (AZA), used in treatment of early or infantile hydrocephalus, is effective in some cases, while its effect on the choroid plexus (CP) remains ill-defined. The drug reversibly inhibits aquaporin-4 (AQP4), the most ubiquitous "water pore" in the brain, and perhaps modulation of AQP1 (located apically on CP cells) by AZA may reduce cerebrospinal fluid (CSF) production. We sought to elucidate the effect of AZA on AQP1 and fluid flow in CP cell cultures.CP tissue culture from 10-day Sprague-Dawley rats and a TRCSF-B cell line were grown on Transwell permeable supports and treated with 100 µM AZA. Fluid assays to assess direction and extent of fluid flow, and AQP1 expression patterns by immunoblot, Immuncytochemistry (ICC), and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) were performed.Immunoblots and ICC analyses showed a decrease in AQP1 protein shortly after AZA treatment (lowest at 12 h), with transient AQP1 reduction mediated by mRNA expression (lowest at 6 h). Transwell fluid assays indicated a fluid shift at 2 h, before significant changes in AQP1 mRNA or protein levels.Timing of AZA effect on AQP1 suggests the drug alters protein transcription, while affecting fluid flow by a concomitant method. It is plausible that other mechanisms account for these phenomena, as the processes may occur independently.


Asunto(s)
Acetazolamida/farmacología , Acuaporina 1/metabolismo , Inhibidores de Anhidrasa Carbónica/farmacología , Plexo Coroideo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Animales , Animales Recién Nacidos , Permeabilidad Capilar/efectos de los fármacos , Plexo Coroideo/metabolismo , Dextranos , Hidrodinámica , Técnicas de Cultivo de Órganos , ARN Mensajero/metabolismo , Ratas , Rodaminas , Factores de Tiempo
9.
Stroke ; 42(9): 2589-94, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21737799

RESUMEN

BACKGROUND AND PURPOSE: Activation of Notch worsens ischemic brain damage as antisense knockdown or pharmacological inhibition of the Notch pathway reduces the infarct size and improves the functional outcome in a mouse model of stroke. We sought to determine whether Notch activation contributes to postischemic inflammation by directly modulating the microglial innate response. METHODS: The microglial response and the attendant inflammatory reaction were evaluated in Notch1 antisense transgenic (Tg) and in nontransgenic (non-Tg) mice subjected to middle cerebral artery occlusion with or without treatment with a γ-secretase inhibitor (GSI). To investigate the impact of Notch on microglial effector functions, primary mouse microglia and murine BV-2 microglial cell line were exposed to oxygen glucose deprivation or lipopolysaccharide in the presence or absence of GSI. Immunofluorescence labeling, Western blotting, and reverse-transcription polymerase chain reaction were performed to measure microglial activation and production of inflammatory cytokines. The nuclear translocation of nuclear factor-κB in microglia was assessed by immunohistochemistry. The neurotoxic potential of microglia was determined in cocultures. RESULTS: Notch1 antisense mice exhibit significantly lower numbers of activated microglia and reduced proinflammatory cytokine expression in the ipsilateral ischemic cortices compared to non-Tg mice. Microglial activation also was attenuated in Notch1 antisense cultures and in non-Tg cultures treated with GSI. GSI significantly reduced nuclear factor-κB activation and expression of proinflammatory mediators and markedly attenuated the neurotoxic activity of microglia in cocultures. CONCLUSIONS: These findings establish a role for Notch signaling in modulating the microglia innate response and suggest that inhibition of Notch might represent a complementary therapeutic approach to prevent reactive gliosis in stroke and neuroinflammation-related degenerative disorders.


Asunto(s)
Isquemia Encefálica/metabolismo , Núcleo Celular/metabolismo , Gliosis/metabolismo , Microglía/metabolismo , Receptor Notch1/metabolismo , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/inmunología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/inmunología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Línea Celular , Núcleo Celular/genética , Núcleo Celular/inmunología , Técnicas de Cocultivo , Citocinas/biosíntesis , Citocinas/genética , Citocinas/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Gliosis/genética , Gliosis/inmunología , Gliosis/patología , Gliosis/terapia , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/terapia , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Ratones , Ratones Transgénicos , Microglía/inmunología , Microglía/patología , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Oligopéptidos/farmacología , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Receptor Notch1/inmunología
10.
J Gerontol A Biol Sci Med Sci ; 76(1): 23-31, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32154567

RESUMEN

The accumulation of amyloid-ß (Aß) is a characteristic event in the pathogenesis of Alzheimer's disease (AD). Aquaporin 1 (AQP1) is a membrane water channel protein belonging to the AQP family. AQP1 levels are elevated in the cerebral cortex during the early stages of AD, but the role of AQP1 in AD pathogenesis is unclear. We first determined the expression and distribution of AQP1 in brain tissue samples of AD patients and two AD mouse models (3xTg-AD and 5xFAD). AQP1 accumulation was observed in vulnerable neurons in the cerebral cortex of AD patients, and in neurons affected by the Aß or tau pathology in the 3xTg-AD and 5xFAD mice. AQP1 levels increased in neurons as aging progressed in the AD mouse models. Stress stimuli increased AQP1 in primary cortical neurons. In response to cellular stress, AQP1 appeared to translocate to endocytic compartments of ß- and γ-secretase activities. Ectopic expression of AQP1 in human neuroblastoma cells overexpressing amyloid precussir protein (APP) with the Swedish mutations reduced ß-secretase (BACE1)-mediated cleavage of APP and reduced Aß production without altering the nonamyloidogenic pathway. Conversely, knockdown of AQP1 enhanced BACE1 activity and Aß production. Immunoprecipitation experiments showed that AQP1 decreased the association of BACE1 with APP. Analysis of a human database showed that the amount of Aß decreases as the expression of AQP1 increases. These results suggest that the upregulation of AQP1 is an adaptive response of neurons to stress that reduces Aß production by inhibiting the binding between BACE1 and APP.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/fisiología , Precursor de Proteína beta-Amiloide/fisiología , Amiloide/biosíntesis , Acuaporina 1/fisiología , Enfermedad de Alzheimer/metabolismo , Animales , Acuaporina 1/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Neuronas/metabolismo
11.
J Biol Chem ; 284(27): 18323-33, 2009 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-19447887

RESUMEN

The endoplasmic reticulum (ER) is a key organelle regulating intracellular Ca(2+) homeostasis. Oxidants and mitochondria-derived free radicals can target ER-based Ca(2+) regulatory proteins and cause uncontrolled Ca(2+) release that may contribute to protracted ER stress and apoptosis. Several ER stress proteins have been suggested to counteract the deregulation of ER Ca(2+) homeostasis and ER stress. Here we showed that knockdown of Herp, an ubiquitin-like domain containing ER stress protein, renders PC12 and MN9D cells vulnerable to 1-methyl-4-phenylpyridinium-induced cytotoxic cell death by a mechanism involving up-regulation of CHOP expression and ER Ca(2+) depletion. Conversely, Herp overexpression confers protection by blocking 1-methyl-4-phenylpyridinium-induced CHOP up-regulation, ER Ca(2+) store depletion, and mitochondrial Ca(2+) accumulation in a manner dependent on a functional ubiquitin-proteasomal protein degradation pathway. Deletion of the ubiquitin-like domain of Herp or treatment with a proteasomal inhibitor abolished the central function of Herp in ER Ca(2+) homeostasis. Thus, elucidating the underlying molecular mechanism(s) whereby Herp counteracts Ca(2+) disturbances will provide insights into the molecular cascade of cell death in dopaminergic neurons and may uncover novel therapeutic strategies to prevent and ameliorate Parkinson disease progression.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Calcio/metabolismo , Intoxicación por MPTP/fisiopatología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neuronas/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Retículo Endoplásmico/metabolismo , Homeostasis/fisiología , Humanos , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/patología , Proteínas de la Membrana/química , Ratones , Neuronas/citología , Células PC12 , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño , Ratas , Estrés Fisiológico/fisiología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Transfección , Ubiquitina/metabolismo
12.
J Neurochem ; 114(1): 323-34, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20456017

RESUMEN

Because the olfactory system plays a major role in food consumption, and because 'food addiction' and associated morbidities have reached epidemic proportions, we tested the hypothesis that dietary energy restriction can modify adverse effects of cocaine on behavior and olfactory cellular and molecular plasticity. Mice maintained on an alternate day fasting (ADF) diet exhibited increased baseline locomotion and increased cocaine-sensitized locomotion during cocaine conditioning, despite no change in cocaine conditioned place preference, compared with mice fed ad libitum. Levels of dopamine and its metabolites in the olfactory bulb (OB) were suppressed in mice on the ADF diet compared with mice on the control diet, independent of acute or chronic cocaine treatment. The expression of several enzymes involved in dopamine metabolism including tyrosine hydroxylase, monoamine oxidases A and B, and catechol-O-methyltransferase were significantly reduced in OBs of mice on the ADF diet. Both acute and chronic administration of cocaine suppressed the production of new OB cells, and this effect of cocaine was attenuated in mice on the ADF diet. Cocaine administration to mice on the control diet resulted in up-regulation of OB genes involved in mitochondrial energy metabolism, synaptic plasticity, cellular stress responses, and calcium- and cAMP-mediated signaling, whereas multiple olfactory receptor genes were down-regulated by cocaine treatment. ADF abolished many of the effects of cocaine on OB gene expression. Our findings reveal that dietary energy intake modifies the neural substrates underlying some of the behavioral and physiological responses to repeated cocaine treatment, and also suggest novel roles for the olfactory system in addiction. The data further suggest that modification of dietary energy intake could provide a novel potential approach to addiction treatments.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cocaína/farmacología , Dieta , Ayuno , Bulbo Olfatorio/efectos de los fármacos , Animales , Proliferación Celular , Dopamina/metabolismo , Ingestión de Energía , Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos
13.
J Am Soc Nephrol ; 19(7): 1360-70, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18385426

RESUMEN

Although neurologic sequelae of acute kidney injury (AKI) are well described, the pathogenesis of acute uremic encephalopathy is poorly understood. This study examined the short-term effect of ischemic AKI on inflammatory and functional changes of the brain in mice by inducing bilateral renal ischemia for 60 min and studying the brains 24 h later. Compared with sham mice, mice with AKI had increased neuronal pyknosis and microgliosis in the brain. AKI also led to increased levels of the proinflammatory chemokines keratinocyte-derived chemoattractant and G-CSF in the cerebral cortex and hippocampus and increased expression of glial fibrillary acidic protein in astrocytes in the cortex and corpus callosum. In addition, extravasation of Evans blue dye into the brain suggested that the blood-brain barrier was disrupted in mice with AKI. Because liver failure also leads to encephalopathy, ischemic liver injury was induced in mice with normal renal function; neuronal pyknosis and glial fibrillary acidic protein expression were not increased, suggesting differential effects on the brain depending on the organ injured. For evaluation of the effects of AKI on brain function, locomotor activity was studied using an open field test. Mice subjected to renal ischemia or bilateral nephrectomy had moderate to severe declines in locomotor activity compared with sham-operated mice. These data demonstrate that severe ischemic AKI induces inflammation and functional changes in the brain. Targeting these pathways could reduce morbidity and mortality in critically ill patients with severe AKI.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Encéfalo/fisiopatología , Permeabilidad Capilar/fisiología , Encefalitis/etiología , Agua/metabolismo , Lesión Renal Aguda/sangre , Lesión Renal Aguda/complicaciones , Animales , Encéfalo/metabolismo , Proteína C-Reactiva/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Factor Estimulante de Colonias de Granulocitos/sangre , Etiquetado Corte-Fin in Situ , Isquemia/fisiopatología , Fallo Hepático Agudo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Actividad Motora/fisiología , Neuronas/fisiología , Proteínas/metabolismo
14.
Neuromolecular Med ; 20(1): 63-72, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29299869

RESUMEN

Acute ischemic stroke causes a high rate of deaths and permanent neurological deficits in survivors. Current interventional treatment, in the form of enzymatic thrombolysis, benefits only a small percentage of patients. Brain ischemia triggers mobilization of innate immunity, specifically the complement system and Toll-like receptors (TLRs), ultimately leading to an exaggerated inflammatory response. Here we demonstrate that intravenous immunoglobulin (IVIG), a scavenger of potentially harmful complement fragments, and C1-esterase inhibitor (C1-INH), an inhibitor of complement activation, exert a beneficial effect on the outcome of experimental brain ischemia (I) and reperfusion (R) injury induced by transient occlusion of middle cerebral artery in mice. Both IVIG and C1-INH significantly and in a dose-responsive manner reduced brain infarction size, neurological deficit and mortality when administered to male mice 30 min before ischemia or up to 6 h after the onset of reperfusion. When combined, suboptimal doses of IVIG and C1-INH potentiated each other's neuroprotective therapeutic effects. Complement C3 and TLR2 signals were colocalized and significantly greater in brain cells adjacent to infracted brain lesions when compared to the corresponding regions of the contralateral hemisphere and to control (sham) mice. Treatment with IVIG and C1-INH effectively reduced deposition of C3b and downregulated excessive TLR2 and p-JNK1 expression at the site of I/R injury. Taken together, these results provide a rationale for potential use of IVIG and C1-INH, alone or in combination with ischemic stroke and other neurological conditions that involve inappropriately activated components of the innate immune system.


Asunto(s)
Proteína Inhibidora del Complemento C1/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Trastornos Neurológicos de la Marcha/prevención & control , Inmunoglobulinas Intravenosas/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Activación de Complemento/efectos de los fármacos , Proteína Inhibidora del Complemento C1/administración & dosificación , Complemento C3b/análisis , Inactivadores del Complemento/administración & dosificación , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Femenino , Trastornos Neurológicos de la Marcha/etiología , Inmunoglobulinas Intravenosas/administración & dosificación , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Método Simple Ciego , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/genética , Regulación hacia Arriba
15.
Neurosci Lett ; 687: 1-9, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30025832

RESUMEN

Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, whose hallmark is the loss of dopamine terminals in the substantia nigra pars compacta (SNpc). PD is usually diagnosed after the appearance of motor symptoms, when about 70% of neurons in the SNpc have already been lost. Because of that, it is important to search for new methods that aid in the early diagnosis of PD. In recent years, microRNAs (miRs) have emerged as potential biomarkers for a variety of diseases and hold the potential to be used to aid in the diagnosis of PD. Therefore, the aim of this study was to characterize if specific miRs are differentially expressed in serum in a mouse model of PD. To induce PD-like damage, mice were subcutaneously injected with 25 mg/kg of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) by administering 10 doses over a period of 5 weeks, with 3.5 days between doses. Expression of 71 different microRNAs was quantified in serum separated from blood collected at day 35, using next-generation sequencing. Histological analysis and quantification of neurotransmitters were performed to confirm dopaminergic neurodegeneration. Chronic MPTP treatment induced loss of dopaminergic terminals in the SNpc and caudate putamen, confirmed by a decrease in the number of tyrosine hydroxylase and dopamine transporter positive cells. In addition, MPTP decreased the concentration of dopamine and its metabolites in the SNpc, simulating the damage observed in PD. From the 71 miRs analyzed, only 4 were differentially expressed after MPTP treatment. Serum levels of miR19b, miR124, miR126a and miR133b were significantly decreased in MPTP-treated mice compared to control. These data suggest that specific miRs are downregulated in a pre-clinical model of PD and hold the potential to be used as biomarkers to aid in the diagnosis of this disease. Further experiments need to be conducted to validate the use of these miRs as biomarkers of PD in additional pre-clinical models as well as in samples from patients diagnosed with PD.


Asunto(s)
Encéfalo/metabolismo , MicroARNs/sangre , Trastornos Parkinsonianos/sangre , Animales , Biomarcadores/sangre , Encéfalo/patología , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología
16.
Mol Endocrinol ; 20(8): 1894-911, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16574742

RESUMEN

The expression of human (h) calcitonin (CT) and its receptor (CTR) is localized to basal epithelium in benign prostates but is distributed in whole epithelium of malignant prostates. Moreover, the abundance of hCT and CTR mRNA in primary prostate tumors positively correlates with the tumor grade. We tested the hypothesis that the modulation of endogenous hCT expression of prostate cancer (PC) cell lines alters their oncogenicity. The effect of modulation of hCT expression on oncogenic characteristics was examined in LNCaP and PC-3M cell lines. The endogenous hCT expression was modulated using either constitutively active expression vector containing hCT cDNA or anti-hCT hammerhead ribozymes. The changes in the oncogenicity of cell sublines was assessed with cell proliferation assays, invasion assays, colony formation assays, and in vivo growth in athymic nude mice. Up-regulation of hCT in PC-3M cells and or enforced hCT expression in LNCaP cells dramatically enhanced their oncogenic characteristics. In contrast, the down-regulation of hCT in PC-3M cells led to a dramatic decline in their oncogenicity. These results, when combined with our other results, that the expression of hCT in primary PCs increase with tumor grade, suggest an important role for hCT in the progression of PC to a metastatic phenotype.


Asunto(s)
Calcitonina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Neoplasias de la Próstata/patología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Animales , Calcitonina/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Expresión Génica , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Células Madre Neoplásicas , Neoplasias de la Próstata/metabolismo , ARN Mensajero/metabolismo , Receptores de Superficie Celular/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Transfección , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 65(18): 8519-29, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16166333

RESUMEN

Although a strong correlation between neuroendocrine differentiation and angiogenesis of prostate cancer has been reported, no mechanistic link between the two events has been established. Because neuropeptide calcitonin is secreted by prostate tumors and endothelial cells are known to express calcitonin receptor-like receptor, we examined the potential action of calcitonin on endothelial cells. The presence of calcitonin receptor, calcitonin receptor-like receptor, and receptor activity-modifying proteins in human microvessel endothelial-1 cells was tested by reverse transcriptase-PCR (RT-PCR). The proangiogenic action of calcitonin was examined in several in vitro models of angiogenesis using HMEC-1 cells and also in vivo using dorsal skinfold assays. Calcitonin expression of PC-3M cells was modulated, and its effect on angiogenesis was examined in in vitro as well as in vivo models. The results of RT-PCR and radioligand receptor assays showed the presence of functional calcitonin receptor in HMEC-1 cells. Calcitonin stimulated all phases of angiogenesis through the calcitonin receptor, but its effect on tube morphogenesis by endothelial cells occurred at the concentration of the Kd of calcitonin receptor. Silencing of calcitonin receptor expression in HMEC-1 cells abolished calcitonin-induced tube formation. Vascular endothelial growth factor antibodies attenuated but did not abolish calcitonin-induced tube morphogenesis. PC-3M prostate cancer cells induced angiogenesis in in vivo and in vitro models. Overexpression of calcitonin in PC-3M cells increased their angiogenic activity, whereas the silencing of calcitonin expression abolished it. These results show that prostate tumor-derived calcitonin may play an important role in prostate tumor growth by regulating intratumoral vascularization.


Asunto(s)
Calcitonina/farmacología , Células Endoteliales/efectos de los fármacos , Animales , Sitios de Unión , Calcitonina/biosíntesis , Calcitonina/genética , Calcitonina/metabolismo , Proteína Similar al Receptor de Calcitonina , Procesos de Crecimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Colágeno , ADN Complementario/genética , Combinación de Medicamentos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Laminina , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Proteoglicanos , Receptores de Calcitonina/biosíntesis , Piel/irrigación sanguínea , Estimulación Química , Factor A de Crecimiento Endotelial Vascular/farmacología
18.
Cytokine Growth Factor Rev ; 31: 37-48, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27079372

RESUMEN

Adipose tissue is a highly vascularized endocrine organ, and its secretion profiles may vary with obesity. Adiponectin is secreted by adipocytes that make up adipose tissue. Worldwide, obesity has been designated a serious health problem among women and is associated with a variety of metabolic disorders and an increased risk of developing cancer of the cervix, ovaries, uterus (uterine/endometrial), and breast. In this review, the potential link between obesity and female-specific malignancies is comprehensively presented by discussing significant features of the intriguing and complex molecule, adiponectin, with a focus on recent findings highlighting its molecular mechanism of action in female-specific carcinogenesis.


Asunto(s)
Adiponectina/metabolismo , Carcinogénesis/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Femenino , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Receptores de Adiponectina/metabolismo
19.
Exp Neurol ; 273: 151-60, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26277686

RESUMEN

OBJECTIVE: Multiple sclerosis (MS) is a debilitating neurological disorder involving an autoimmune reaction to oligodendrocytes and degeneration of the axons they ensheath in the CNS. Because the damage to oligodendrocytes and axons involves local inflammation and associated oxidative stress, we tested the therapeutic efficacy of combined treatment with a potent anti-inflammatory thalidomide analog (lenalidomide) and novel synthetic anti-oxidant cerium oxide nanoparticles (nanoceria) in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS: C57BL/6 mice were randomly assigned to a control (no EAE) group, or one of the four myelin oligodendrocyte glycoprotein-induced EAE groups: vehicle, lenalidomide, nanoceria, or lenalidomide plus nanoceria. During a 23 day period, clinical EAE symptoms were evaluated daily, and MRI brain scans were performed at 11-13 days and 20-22 days. Histological and biochemical analyses of brain tissue samples were performed to quantify myelin loss and local inflammation. RESULTS: Lenalidomide treatment alone delayed symptom onset, while nanoceria treatment had no effect on symptom onset or severity, but did promote recovery; lenalidomide and nanoceria each significantly attenuated white matter pathology and associated inflammation. Combined treatment with lenalidomide and nanoceria resulted in a near elimination of EAE symptoms, and reduced white matter pathology and inflammatory cell responses to a much greater extent than either treatment alone. INTERPRETATION: By suppressing inflammation and oxidative stress, combined treatment with lenalidomide and nanoceria can reduce demyelination and associated neurological symptoms in EAE mice. Our preclinical data suggest a potential application of this combination therapy in MS.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Cerio/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Talidomida/análogos & derivados , Análisis de Varianza , Animales , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Lenalidomida , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , ARN Mensajero , Talidomida/uso terapéutico , Factores de Tiempo
20.
Int J Oncol ; 25(5): 1407-14, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15492832

RESUMEN

The expression of matrix metalloproteinases (MMPs), particularly MMP-9, is significantly increased during tumor progression and is thought to play a major role in mediating angiogenic process. Since microvasculature plays an important role in controlling tumor growth, we investigated the effects of MMP-9 inhibition on endothelial cell migration and tube formation, two determinants of angiogenesis. Adenoviral-mediated MMP-9 downregulation inhibited endothelial cell migration in cell wounding and spheroid migration assays. To determine the effects of MMP-9 reduction in glioblastoma/endothelial co-cultures, we used a three-dimensional co-culture assay of glioblastoma spheroids and endothelial spheroids. Untreated controls showed invasion of both cell populations into each other whereas treatment of the co-cultures with adenoviral antisense MMP-9 particles resulted in reduced invasion. Next, inhibition of MMP-9 by adenoviral vectors in endothelial cells was assessed for in vitro capillary-like structure formation either by co-culture with glioblastoma cells or exposure to glioblastoma-conditioned medium. Addition of conditioned medium from human glioblastoma cells to endothelial cells treated with antisense MMP-9 adenoviral vectors or co-cultures of glioblastoma cell lines with MMP-9-reduced endothelial cells resulted in reduced capillary-like tube formation demonstrating the key role of MMP-9 in endothelial cell network organization. Examination of in vitro capillary-like tube structure formation using Matrigel showed a significant decrease in MMP-9 downregulated endothelial cells as compared to controls. In conclusion, the inhibition of MMP-9 is required for inhibition of endothelial cell migration and tube formation and is likely to be of importance in cerebral angiogenesis for therapeutic targets.


Asunto(s)
Neoplasias Encefálicas/patología , Movimiento Celular/fisiología , Células Endoteliales/fisiología , Glioblastoma/patología , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/farmacología , Neovascularización Patológica , Adenoviridae , Regulación hacia Abajo , Células Endoteliales/patología , Vectores Genéticos , Humanos , Invasividad Neoplásica/patología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA