Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Cell Biochem ; 117(10): 2241-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26910498

RESUMEN

The objective of this study was to develop an in silico screening model for characterization of potential novel ligands from commercial drug libraries able to functionally activate certain olfactory receptors (ORs), which are members of the class A rhodopsin-like family of G protein couple receptors (GPCRs), in the brain of murine models of concussion. We previously found that concussions may significantly influence expression of certain ORs, for example, OR4M1 in subjects with a history of concussion/traumatic brain injury (TBI). In this study, we built a 3-D OR4M1 model and used it in in silico screening of potential novel ligands from commercial drug libraries. We report that in vitro activation of OR4M1 with the commercially available ZINC library compound 10915775 led to a significant attenuation of abnormal tau phosphorylation in embryonic cortico-hippocampal neuronal cultures derived from NSE-OR4M1 transgenic mice, possibly through modulation of the JNK signaling pathway. The attenuation of abnormal tau phosphorylation was rather selective since ZINC10915775 significantly decreased tau phosphorylation on tau Ser202/T205 (AT8 epitope) and tau Thr212/Ser214 (AT100 epitope), but not on tau Ser396/404 (PHF-1 epitope). Moreover, no response of ZINC10915775 was found in control hippocampal neuronal cultures derived from wild type littermates. Our in silico model provides novel means to pharmacologically modulate select ubiquitously expressed ORs in the brain through high affinity ligand activation to prevent and eventually to treat concussion induced down regulation of ORs and subsequent cascade of tau pathology. J. Cell. Biochem. 117: 2241-2248, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Conmoción Encefálica/complicaciones , Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas/metabolismo , Receptores Odorantes/química , Receptores Odorantes/metabolismo , Tauopatías/tratamiento farmacológico , Proteínas tau/metabolismo , Animales , Células Cultivadas , Simulación por Computador , Epítopos , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación , Ratas , Ratas Long-Evans , Tauopatías/etiología , Tauopatías/patología
2.
J Alzheimers Dis ; 16(2): 433-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19221432

RESUMEN

Abnormal misfoldings of the microtubule-associated protein tau, leading to the aggregation of tau into paired helical filaments that are ultimately deposited as neurofibrillary tangles, is a key neuropathologic feature of a number of neurodegenerative disorders collectively referred to as tauopathies. We recently observed that a particular grape seed polyphenolic extract (GSPE), namely, Meganatural-Az may attenuate the generation and stability of misfolded proteins. We hypothesized that Meganatural-Az GSPE might also attenuate tau protein misfolding that leads to the generation of tau filamentary aggregates that are critical for the initiation and progression of neurodegeneration and/or cognitive dysfunctions in tauopathies. In this study, we used in vitro aggregations of synthetic Ac(306)VQIVYK(311) tau peptide as a model system to explore whether Meganatural-Az GSPE might modulate aggregations of tau protein. We demonstrate that this GSPE is capable of inhibiting tau peptide aggregations, as well as dissociating preformed tau peptide aggregates. Results from this study suggest that this GSPE might provide beneficial disease-modifying bioactivities in tau-associated neurodegenerative disorders by modulating tau-mediated neuropathologic mechanisms. Our observation, in conjunction with the demonstrated bioavailability, as well as safety and tolerability, of this GSPE, supports the development of Meganatural-Az GSPE for the prevention and/or treatment of tau-associated neurodegenerative disorders.


Asunto(s)
Antioxidantes/metabolismo , Flavonoides/metabolismo , Fenoles/metabolismo , Extractos Vegetales/química , Tauopatías/metabolismo , Antioxidantes/farmacología , Relación Dosis-Respuesta a Droga , Flavonoides/farmacología , Modelos Biológicos , Péptidos/metabolismo , Fenoles/farmacología , Polifenoles , Unión Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos
3.
Nat Commun ; 9(1): 477, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396460

RESUMEN

Major depressive disorder is associated with abnormalities in the brain and the immune system. Chronic stress in animals showed that epigenetic and inflammatory mechanisms play important roles in mediating resilience and susceptibility to depression. Here, through a high-throughput screening, we identify two phytochemicals, dihydrocaffeic acid (DHCA) and malvidin-3'-O-glucoside (Mal-gluc) that are effective in promoting resilience against stress by modulating brain synaptic plasticity and peripheral inflammation. DHCA/Mal-gluc also significantly reduces depression-like phenotypes in a mouse model of increased systemic inflammation induced by transplantation of hematopoietic progenitor cells from stress-susceptible mice. DHCA reduces pro-inflammatory interleukin 6 (IL-6) generations by inhibiting DNA methylation at the CpG-rich IL-6 sequences introns 1 and 3, while Mal-gluc modulates synaptic plasticity by increasing histone acetylation of the regulatory sequences of the Rac1 gene. Peripheral inflammation and synaptic maladaptation are in line with newly hypothesized clinical intervention targets for depression that are not addressed by currently available antidepressants.


Asunto(s)
Antocianinas/farmacología , Ácidos Cafeicos/farmacología , Epigénesis Genética , Glucósidos/farmacología , Inflamación/genética , Plasticidad Neuronal/genética , Estrés Psicológico/genética , Animales , Antocianinas/administración & dosificación , Ácidos Cafeicos/administración & dosificación , Islas de CpG/efectos de los fármacos , Depresión/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Glucósidos/administración & dosificación , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Antígenos Comunes de Leucocito/genética , Masculino , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Neuropéptidos/genética , Neuropéptidos/metabolismo , Polifenoles/farmacología , Conducta Social , Estrés Psicológico/tratamiento farmacológico , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
4.
Neuromolecular Med ; 18(2): 177-89, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26984114

RESUMEN

Caregiving for a dementia patient is associated with increased risk of psychological and physical health problems. We investigated whether a mindfulness-based stress reduction (MBSR) training course for caregivers that closely models the MBSR curriculum originally established by the Center of Mindfulness at the University of Massachusetts may improve the psychological resilience of non-professional caregivers of Alzheimer's disease patients. Twenty adult non-professional caregivers of dementia patients participated in an 8-week MBSR training course. Caregiver stress, depression, burden, grief, and gene expression profiles of blood mononuclear cells were assessed at baseline and following MBSR. MBSR training significantly improved the psychological resilience of some of the caregivers. We identified predictive biomarkers whose expression is associated with the likelihood of caregivers to benefit from MBSR, and biomarkers whose expression is associated with MBSR psychological benefits. Our biomarker studies provide insight into the mechanisms of health benefits of MBSR and a basis for developing a personalized medicine approach for applying MBSR for promoting psychological and cognitive resilience in caregivers of dementia patients.


Asunto(s)
Biomarcadores/sangre , Cuidadores/educación , Cuidadores/psicología , Atención Plena , Estrés Psicológico/sangre , Estrés Psicológico/genética , Regulación de la Expresión Génica , Humanos , Atención Plena/educación , Atención Plena/normas , Estrés Psicológico/diagnóstico , Transcriptoma
5.
J Alzheimers Dis ; 54(2): 477-96, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27567821

RESUMEN

The release of paired helical filaments (PHFs) from neurons into the extracellular space may contribute to the propagation of tau pathology across brain regions in Alzheimer's disease (AD) and other tauopathies. The majority of available mechanistic studies exploring the pathologic role of extracellular PHFs are conducted in proliferating cell lines. Here, we compare how extracellular PHFs induce tauopathy in mitotic cells and in post-mitotic brain neurons. In a mitotic cell line (HEK 293T), extracellular exposure to AD PHFs leads to an intracellular "aggresomal" type deposition of tau, coincidental with redistribution of dynein, a retrograde motor protein. We also observed that PHFs impaired proteasome degradation, but not autophagy. Exposure of cells to proteasome inhibitors was sufficient to induce intracellular tau aggregate formation as well as reorganization of dynein and the intermediate filament protein, vimentin. Thus, in mitotic cells, extracellular PHFs promote cellular tau aggregation, in part, by interfering with cellular proteasome degradation processes. In contrast with our observations with proliferating cells, exposure of post-mitotic primary neuronal cultures to AD PHFs did not promote "aggresomal" tau deposition, but instead resulted in a widespread accumulation of phosphorylated tau-immunoreactive swellings in neuritic processes, characterized by disturbed cytoskeletal organization of dynein and vimentin. Collectively, our observations suggest that extracellular PHFs may contribute to the propagation of tau pathology by independent mechanisms in post-mitotic and mitotic brain cells. These outcomes indicate that in addition to post-mitotic brain neurons, mitotic brain cells should also be considered as targets for therapeutic interventions to attenuate propagation of tauopathy.


Asunto(s)
Encéfalo/metabolismo , Líquido Extracelular/metabolismo , Mitosis/fisiología , Ovillos Neurofibrilares/metabolismo , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/patología , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ovillos Neurofibrilares/patología
6.
Neurochem Int ; 89: 191-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26235983

RESUMEN

Sleep deprivation produces deficits in hippocampal synaptic plasticity and hippocampal-dependent memory storage. Recent evidence suggests that sleep deprivation disrupts memory consolidation through multiple mechanisms, including the down-regulation of the cAMP-response element-binding protein (CREB) and of mammalian target of rapamycin (mTOR) signaling. In this study, we tested the effects of a Bioactive Dietary Polyphenol Preparation (BDPP), comprised of grape seed polyphenol extract, Concord grape juice, and resveratrol, on the attenuation of sleep deprivation-induced cognitive impairment. We found that BDPP significantly improves sleep deprivation-induced contextual memory deficits, possibly through the activation of CREB and mTOR signaling pathways. We also identified brain-available polyphenol metabolites from BDPP, among which quercetin-3-O-glucuronide activates CREB signaling and malvidin-3-O-glucoside activates mTOR signaling. In combination, quercetin and malvidin-glucoside significantly attenuated sleep deprivation-induced cognitive impairment in -a mouse model of acute sleep deprivation. Our data suggests the feasibility of using select brain-targeting polyphenol compounds derived from BDPP as potential therapeutic agents in promoting resilience against sleep deprivation-induced cognitive dysfunction.


Asunto(s)
Encéfalo/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Extracto de Semillas de Uva/administración & dosificación , Polifenoles/administración & dosificación , Privación de Sueño/tratamiento farmacológico , Estilbenos/administración & dosificación , Animales , Encéfalo/metabolismo , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/metabolismo , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Extracto de Semillas de Uva/metabolismo , Ratones , Ratones Endogámicos C57BL , Polifenoles/metabolismo , Ratas , Ratas Sprague-Dawley , Resveratrol , Privación de Sueño/complicaciones , Privación de Sueño/metabolismo , Estilbenos/metabolismo
7.
Arch Neurol ; 60(3): 369-76, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12633148

RESUMEN

BACKGROUND: Caspase gene expression has previously been reported in terminal Alzheimer disease (AD) brain, but, currently, little is known about the temporal pattern of caspase gene expression relative to the onset and clinical progression of AD. OBJECTIVE: To derive a profile of caspase gene expression and proapoptotic indexes as a function of the clinical and neuropathologic progression of AD dementia. SETTING AND PATIENTS: Postmortem survey of nursing home patients characterized clinically by Clinical Dementia Rating (CDR) and neuropathologically by Consortium to Establish a Registry for Alzheimer's Disease criteria. DESIGN AND OUTCOME MEASURES: To assess messenger RNA expression of caspase-1, -2L, -2S, -3, -5, -6, -7, -8, and -9; apoptotic cell death by TUNEL assay; and poly (ADP-ribose) polymerase cleavage in postmortem brain tissue samples from cognitively normal (CDR 0), high risk of developing AD dementia (CDR 0.5), and severe dementia (CDR 5) cases. RESULTS: Compared with CDR 0 cases, elevated messenger RNA expression of caspase-1 and caspase-7 in the entorhinal cortex of CDR 0.5 cases coincided with increased poly (ADP-ribose) polymerase cleavage but not apoptotic cell injury. In the entorhinal cortex of CDR 5 cases, we found elevation of caspase-1, -2L, -3, -5, -6, -7, -8, and -9 and a greater than 4-fold increase in TUNEL-positive cells. Caspase messenger RNA expression was closely associated with neurofibrillary tangle and, to a lesser extent, neuritic plaque density. CONCLUSIONS: Proapoptotic mechanisms may be at play early in the onset of AD (before overt signs of apoptosis) and may be a conditional factor for later apoptotic cell injury or death. These data have relevance to potential therapeutic interventions for AD using selective caspase inhibitors.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Encéfalo/enzimología , Encéfalo/patología , Caspasas/genética , Anciano , Anciano de 80 o más Años , Apoptosis , Biomarcadores , Cognición , Progresión de la Enfermedad , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Mensajero/análisis
8.
Gene Expr ; 10(5-6): 271-8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12450219

RESUMEN

Several epidemiologic studies have reported that cyclooxygenase (COX) inhibitors prevent/delay the onset of Alzheimer's disease (AD). Recent experimental studies suggest that these compounds can also diminish amyloid-beta (Abeta) neuropathology in rodent models of AD. To explore the relationship of COX expression to Abeta neuropathology, we crossed mice expressing both mutant amyloid precursor protein [K670N/M671L (APP(swe)] and mutant PS1 (A246E) with mice expressing human COX-2 selectively in neurons. We show here that human COX-2 expression in APP(swe)/PS1/COX-2 mice induces potentiation of brain parenchymal amyloid plaque formation and a greater than twofold increase in prostaglandin E2 production, at 24 months of age. This increased amyloid plaque formation coincided with a preferential elevation of Abeta1-40 and Abeta1-42 with no change in total amyloid precursor protein (APP) expression/content in the brain. Collectively these data suggest that COX-2 influences APP processing and promotes amyloidosis in the brain.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Isoenzimas/fisiología , Prostaglandina-Endoperóxido Sintasas/fisiología , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Ciclooxigenasa 2 , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Humanos , Procesamiento de Imagen Asistido por Computador , Inflamación , Isoenzimas/metabolismo , Espectrometría de Masas , Proteínas de la Membrana , Ratones , Ratones Transgénicos , Mutación , Prostaglandina-Endoperóxido Sintasas/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo
9.
J Alzheimers Dis ; 34(2): 417-429, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23241557

RESUMEN

Traumatic brain injury (TBI) is a leading cause of death and disability among children and young adults in the United States. In this study, we explored whether changes in the gene expression profile of peripheral blood mononuclear cells (PBMC) may provide a clinically assessable "window" into the brain, reflecting molecular alterations following TBI that might contribute to the onset and progression of TBI clinical complications. We identified three olfactory receptor (OR) TBI biomarkers that are aberrantly down-regulated in PBMC specimens from TBI subjects. Down-regulation of these OR biomarkers in PBMC was correlated with the severity of brain injury and TBI-specific symptoms. A two- biomarker panel comprised of OR11H1 and OR4M1 provided the best criterion for segregating the TBI and control cases with 90% accuracy, 83.3% sensitivity, and 100% specificity. We found that the OR biomarkers are ectopically expressed in multiple brain regions, including the entorhinal-hippocampus system known to play an important role in memory formation and consolidation. Activation of OR4M1 led to attenuation of abnormal tau phosphorylation, possibly through JNK signaling pathway. Our results suggested that addition of the two-OR biomarker model to current diagnostic criteria may lead to improved TBI detection for clinical trials, and decreased expression of OR TBI biomarkers might be associated with TBI-induced tauopathy. Future studies exploring the physiological relevance of OR TBI biomarkers in the normal brain and in the brain following TBI will provide a better understanding of the biological mechanisms underlying TBI and insights into novel therapeutic targets for TBI.


Asunto(s)
Lesiones Encefálicas/sangre , Lesiones Encefálicas/epidemiología , Regulación hacia Abajo/fisiología , Leucocitos Mononucleares/metabolismo , Receptores Odorantes/antagonistas & inhibidores , Tauopatías/sangre , Tauopatías/epidemiología , Adulto , Biomarcadores/sangre , Lesiones Encefálicas/diagnóstico , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores Odorantes/biosíntesis , Receptores Odorantes/sangre , Tauopatías/diagnóstico , Adulto Joven
10.
Mol Immunol ; 56(4): 619-29, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23911420

RESUMEN

BACKGROUND: Complement component C5-derived C5a locally generated in the brain has been shown to protect against glutamate-induced neuronal apoptosis and beta-amyloid (Aß) toxicity, but the mechanism is not clear. In this study, we tested the hypothesis that C5a influences upstream signal transduction pathways associated with cAMP-response element-binding protein (CREB) activation, in which alterations of CREB levels are associated with cognitive deterioration in AD. METHODS: CREB signaling pathway, synaptic plasticity and cognitive function were studied in C5a receptor knockout mice (C5aR(-/-)), C5a over expressing mice (C5a/GFAP) and in Tg2576 mice, an AD mouse model. RESULTS: (1) Cognitive function is severely impaired in C5aR(-/-) mice, coincident with the down-regulated CREB/CEBP pathway in brain. (2) Either the application of recombinant-human-C5a (hrC5a) or exogenous expression of C5a in the brain of a mouse model (C5a/GFAP) enhances this pathway. (3) Application of hrC5a in brain slices from Tg2576 mice significantly improves deficits in long-term potentiation (LTP), while this effect is blocked by a specific AMPA receptor antagonist. (4) Searching for a pharmacological approach to locally mediate C5a responses in the brain, we found that low-dose human intravenous immunoglobulin (IVIG) treatment improves synaptic plasticity and cognitive function through C5a-mediated induction of the CREB/CEBP pathway, while the levels of Aß in the brain are not significantly affected. CONCLUSION: This study for the first time provides novel evidence suggesting that C5a may beneficially influence cognitive function in AD through an up-regulation of AMPA-CREB signaling pathway. IVIG may systematically improve cognitive function in AD brain by passing Aß toxicity.


Asunto(s)
Enfermedad de Alzheimer/terapia , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Complemento C5a/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Inmunoglobulinas Intravenosas/uso terapéutico , Receptores AMPA/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Complemento C5a/genética , Femenino , Humanos , Inmunoterapia/métodos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores AMPA/agonistas , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Transducción de Señal/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
11.
Int J Alzheimers Dis ; 2012: 321280, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666619

RESUMEN

We investigated the contents of the insulin receptor-beta subunit (IRß) and [Tyr1162/1163]-phosphorylated IRß as surrogate indices of total IR content and IR activation in postmortem hippocampal formation brain specimens from nondiabetic sporadic Alzheimer's disease (AD) cases. We found no significant changes in the brain contents of total IRß or [Tyr1162/1163]-phosphorylated IRß, suggesting normal IR content and activation in the brains of nondiabetic sporadic AD cases. Moreover, total IRß and [Tyr1162/1163]-phosphorylated IRß levels in the hippocampal formation are not correlated with the severity of amyloid or tau-neuropathology. Exploring the regulation of glycogen synthase kinase 3 (GSK3) α/ß, key IR-signaling components, we observed significantly lower levels of total GSK3 α/ß in brain specimens from nondiabetic AD cases, suggesting that impaired IR signaling mechanisms might contribute to the onset and/or progression of AD dementia. Outcomes from our study support the development of insulin-sensitizing therapeutic strategies to stimulate downstream IR signaling in nondiabetic AD cases.

12.
J Alzheimers Dis ; 31(2): 301-13, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22543850

RESUMEN

We explored whether changes in the expression profile of peripheral blood plasma proteins may provide a clinical, readily accessible "window" into the brain, reflecting molecular alterations following traumatic brain injury (TBI) that might contribute to TBI complications. We recruited fourteen TBI and ten control civilian participants for the study, and also analyzed banked plasma specimens from 20 veterans with TBI and 20 control cases. Using antibody arrays and ELISA assays, we explored differentially-regulated protein species in the plasma of TBI compared to healthy controls from the two independent cohorts. We found three protein biomarker species, monocyte chemotactic protein-1 (MCP-1), insulin-like growth factor-binding protein-3, and epidermal growth factor receptor, that are differentially regulated in plasma specimens of the TBI cases. A three-biomarker panel using all three proteins provides the best potential criterion for separating TBI and control cases. Plasma MCP-1 contents are correlated with the severity of TBI and the index of compromised axonal fiber integrity in the frontal cortex. Based on these findings, we evaluated postmortem brain specimens from 7 mild cognitive impairment (MCI) and 7 neurologically normal cases. We found elevated MCP-1 expression in the frontal cortex of MCI cases that are at high risk for developing Alzheimer's disease. Our findings suggest that additional application of the three-biomarker panel to current diagnostic criteria may lead to improved TBI detection and more sensitive outcome measures for clinical trials. Induction of MCP-1 in response to TBI might be a potential predisposing factor that may increase the risk for development of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/epidemiología , Lesiones Encefálicas/epidemiología , Quimiocina CCL2/biosíntesis , Predisposición Genética a la Enfermedad , Adulto , Anciano de 80 o más Años , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/genética , Biomarcadores/sangre , Lesiones Encefálicas/sangre , Lesiones Encefálicas/genética , Quimiocina CCL2/sangre , Quimiocina CCL2/genética , Femenino , Lóbulo Frontal/metabolismo , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
13.
Mol Neurodegener ; 6(1): 51, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21771318

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that affects spinal cord and cortical motor neurons. An increasing amount of evidence suggests that mitochondrial dysfunction contributes to motor neuron death in ALS. Peroxisome proliferator-activated receptor gamma co-activator-1α (PGC-1α) is a principal regulator of mitochondrial biogenesis and oxidative metabolism. RESULTS: In this study, we examined whether PGC-1α plays a protective role in ALS by using a double transgenic mouse model where PGC-1α is over-expressed in an SOD1 transgenic mouse (TgSOD1-G93A/PGC-1α). Our results indicate that PGC-1α significantly improves motor function and survival of SOD1-G93A mice. The behavioral improvements were accompanied by reduced blood glucose level and by protection of motor neuron loss, restoration of mitochondrial electron transport chain activities and inhibition of stress signaling in the spinal cord. CONCLUSION: Our results demonstrate that PGC-1α plays a beneficial role in a mouse model of ALS, suggesting that PGC-1α may be a potential therapeutic target for ALS therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA