Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 26(7): 1376-1390, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28158580

RESUMEN

Gamma glutamyl cysteine ligase (GCL) is the rate-limiting enzyme for intracellular glutathione (GSH) synthesis. The GSH concentration and GCL activity are declining with age in the central nervous system (CNS), and is accompanied by elevated reactive oxygen species (ROS). To study the biological effects of low GSH levels, we disrupted its synthesis both at birth by breeding a Gclc loxP mouse with a thy1-cre mouse (NEGSKO mouse) and at a later age by breeding with a CaMKII-ERT2-Cre (FIGSKO mouse). NEGSKO mice with deficiency of the Gclc in their entire CNS neuronal cells develop at 4 weeks: progressive motor neuron loss, gait problems, muscle denervation and atrophy, paralysis, and have diminished life expectancy. The observed neurodegeneration in Gclc deficiency is of more chronic rather than acute nature as demonstrated by Gclc targeted single-neuron labeling from the inducible Cre-mediated knockout (SLICK) mice. FIGSKO mice with inducible Gclc deficiency in the forebrain at 23 weeks after tamoxifen induction demonstrate profound brain atrophy, elevated astrogliosis and neurodegeneration, particularly in the hippocampus region. FIGSKO mice also develop cognitive abnormalities, i.e. learning impairment and nesting behaviors based on passive avoidance, T-Maze, and nesting behavior tests. Mechanistic studies show that impaired mitochondrial glutathione homeostasis and subsequent mitochondrial dysfunction are responsible for neuronal cell loss. This was confirmed by mitochondrial electron transporter chain activity analysis and transmission electron microscopy that demonstrate remarkable impairment of state 3 respiratory activity, impaired complex IV function, and mitochondrial swollen morphology in the hippocampus and cerebral cortex. These mouse genetic tools of oxidative stress open new insights into potential pharmacological control of apoptotic signaling pathways triggered by mitochondrial dysfunction.


Asunto(s)
Corteza Cerebral/metabolismo , Glutamato-Cisteína Ligasa/genética , Glutatión/metabolismo , Mitocondrias/genética , Degeneración Nerviosa/genética , Animales , Apoptosis/genética , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Corteza Cerebral/ultraestructura , Glutamato-Cisteína Ligasa/deficiencia , Glutatión/biosíntesis , Humanos , Ratones , Ratones Noqueados , Mitocondrias/patología , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo
2.
J Cell Sci ; 129(9): 1815-1830, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28157684

RESUMEN

Deregulated Cdk5 causes neurotoxic amyloid beta peptide (Aß) processing and cell death, two hallmarks of Alzheimer's disease, through the Foxo3 transcriptional factor in hippocampal cells, primary neurons and an Alzheimer's disease mouse model. Using an innovative chemical genetic screen, we identified Foxo3 as a direct substrate of Cdk5 in brain lysates. Cdk5 directly phosphorylates Foxo3, which increased its levels and nuclear translocation. Nuclear Foxo3 initially rescued cells from ensuing oxidative stress by upregulating MnSOD (also known as SOD2). However, following prolonged exposure, Foxo3 upregulated Bim (also known as BCL2L11) and FasL (also known as FASLG) causing cell death. Active Foxo3 also increased Aß(1-42) levels in a phosphorylation-dependent manner. These events were completely inhibited either by expressing phosphorylation-resistant Foxo3 or by depleting Cdk5 or Foxo3, highlighting a key role for Cdk5 in regulating Foxo3. These results were confirmed in an Alzheimer's disease mouse model, which exhibited increased levels and nuclear localization of Foxo3 in hippocampal neurons, which preceded neurodegeneration and Aß plaque formation, indicating this phenomenon is an early event in Alzheimer's disease pathogenesis. Collectively, these results show that Cdk5-mediated phospho-regulation of Foxo3 can activate several genes that promote neuronal death and aberrant Aß processing, thereby contributing to the progression of neurodegenerative pathologies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteína Forkhead Box O3/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Proteína 11 Similar a Bcl2/genética , Proteína 11 Similar a Bcl2/metabolismo , Quinasa 5 Dependiente de la Ciclina/genética , Modelos Animales de Enfermedad , Proteína Forkhead Box O3/genética , Células HEK293 , Hipocampo/patología , Humanos , Ratones , Neuronas/patología , Estrés Oxidativo/genética , Fragmentos de Péptidos/genética , Fosforilación/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
3.
J Mol Cell Cardiol ; 105: 99-109, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28232072

RESUMEN

RATIONALE: The contractile dysfunction that underlies heart failure involves perturbations in multiple biological processes ranging from metabolism to electrophysiology. Yet the epigenetic mechanisms that are altered in this disease state have not been elucidated. SWI/SNF chromatin-remodeling complexes are plausible candidates based on mouse knockout studies demonstrating a combined requirement for the BRG1 and BRM catalytic subunits in adult cardiomyocytes. Brg1/Brm double mutants exhibit metabolic and mitochondrial defects and are not viable although their cause of death has not been ascertained. OBJECTIVE: To determine the cause of death of Brg1/Brm double-mutant mice, to test the hypothesis that BRG1 and BRM are required for cardiac contractility, and to identify relevant downstream target genes. METHODS AND RESULTS: A tamoxifen-inducible gene-targeting strategy utilizing αMHC-Cre-ERT was implemented to delete both SWI/SNF catalytic subunits in adult cardiomyocytes. Brg1/Brm double-mutant mice were monitored by echocardiography and electrocardiography, and they underwent rapidly progressive ventricular dysfunction including conduction defects and arrhythmias that culminated in heart failure and death within 3weeks. Mechanistically, BRG1/BRM repressed c-Myc expression, and enforced expression of a DOX-inducible c-MYC trangene in mouse cardiomyocytes phenocopied the ventricular conduction defects observed in Brg1/Brm double mutants. BRG1/BRM and c-MYC had opposite effects on the expression of cardiac conduction genes, and the directionality was consistent with their respective loss- and gain-of-function phenotypes. To support the clinical relevance of this mechanism, BRG1/BRM occupancy was diminished at the same target genes in human heart failure cases compared to controls, and this correlated with increased c-MYC expression and decreased CX43 and SCN5A expression. CONCLUSION: BRG1/BRM and c-MYC have an antagonistic relationship regulating the expression of cardiac conduction genes that maintain contractility, which is reminiscent of their antagonistic roles as a tumor suppressor and oncogene in cancer.


Asunto(s)
ADN Helicasas/metabolismo , Sistema de Conducción Cardíaco , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/metabolismo , Animales , ADN Helicasas/genética , Electrocardiografía , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones , Ratones Transgénicos , Mutación , Contracción Miocárdica/genética , Proteínas Nucleares/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción/genética
4.
Biochem Soc Trans ; 45(5): 1053-1066, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28778984

RESUMEN

Unlike DNA, oxidative damage to RNA has received little attention presumably due to the assumed transient nature of RNA. However, RNAs including mRNA can persist for several hours to days in certain tissues and are demonstrated to sustain greater oxidative damage than DNA. Because neuronal cells in the brain are continuously exposed to reactive oxygen species due to a high oxygen consumption rate, it is not surprising that neuronal RNA oxidation is observed as a common feature at an early stage in a series of neurodegenerative disorders. A recent study on a well-defined bacterial translation system has revealed that mRNA containing 8-oxo-guanosine (8-oxoGuo) has little effect on fidelity despite the anticipated miscoding. Indeed, 8-oxoGuo-containing mRNA leads to ribosomal stalling with a reduced rate of peptide-bond formation by 3-4 orders of magnitude and is subject to no-go decay, a ribosome-based mRNA surveillance mechanism. Another study demonstrates that transfer RNA oxidation catalyzed by cytochrome c (cyt c) leads to its depurination and cross-linking, which may facilitate cyt c release from mitochondria and subsequently induce apoptosis. Even more importantly, a discovery of oxidized microRNA has been recently reported. The oxidized microRNA causes misrecognizing the target mRNAs and subsequent down-regulation in the protein synthesis. It is noteworthy that oxidative modification to RNA not only interferes with the translational machinery but also with regulatory mechanisms of noncoding RNAs that contribute toward the biological complexity of the mammalian brain. Oxidative RNA damage might be a promising therapeutic target potentially useful for an early intervention of diverse neuropsychiatric disorders.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Oxidación-Reducción , Biosíntesis de Proteínas , ARN/química , Animales , Guanosina/análogos & derivados , Guanosina/metabolismo , Humanos , MicroARNs/química , Neuronas/química , ARN Mensajero/química , ARN Mensajero/metabolismo , ARN de Transferencia/química , Ribosomas/genética , Ribosomas/metabolismo
5.
Biochim Biophys Acta ; 1842(8): 1240-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24189435

RESUMEN

Alzheimer's disease (AD) exhibits extensive oxidative stress throughout the body, being detected peripherally as well as associated with the vulnerable regions of the brain affected in disease. Abundant evidence not only demonstrates the full spectrum of oxidative damage to neuronal macromolecules, but also reveals the occurrence of oxidative events early in the course of the disease and prior to the formation of the pathology, which support an important role of oxidative stress in AD. As a disease of abnormal aging, AD demonstrates oxidative damage at levels that significantly surpass that of elderly controls, which suggests the involvement of additional factor(s). Structurally and functionally damaged mitochondria, which are more proficient at producing reactive oxygen species but less so in ATP, are also an early and prominent feature of the disease. Since mitochondria are also vulnerable to oxidative stress, it is likely that a vicious downward spiral involving the interactions between mitochondrial dysfunction and oxidative stress contributes to the initiation and/or amplification of reactive oxygen species that is critical to the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Mitocondrias/patología , Estrés Oxidativo , Animales , Humanos
6.
J Neurochem ; 130(1): 115-25, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24601954

RESUMEN

Development of Alzheimer's disease (AD) has been linked to the de-regulation of estrogen and gonadotropins such as luteinizing hormone (LH). In this study, we found increases in AD pathology in the hippocampi of aged female 3xTg AD mice after ovariectomy that were unable to be reduced by estrogen therapy or down-regulation of serum LH levels. Despite the lack of effect of these treatments on AD pathology, down-regulation of serum LH but not estrogen improved factors associated with neuronal plasticity such as spatial memory, inhibition of glycogen synthase kinase-3 beta, expression of beta-catenin, and brain-derived neurotrophic factor transcription. Contrasting previous studies in younger mice, estrogen replacement was not able to rescue behavioral deficits, reduced glycogen synthase kinase-3 beta inhibition and increased hippocampal phosphorylation of tau. Of critical importance, serum LH was negatively correlated with brain LH in regions associated with spatial memory, and increases in brain LH correlated with cognitive improvement. This paralleled changes in human female AD brains which showed a significant reduction in brain LH mRNA compared to healthy age- and PMI-matched controls. Taken together, these findings should promote further research into the LH-dependent mechanisms associated with AD cognitive deficits as well as the effects of estrogen within the aged brain. In the aged triple transgenic Alzheimer's disease (AD) mouse model (3xAD-Tg), estrogen replacement after ovariectomy does not improve cognitive function, increases phosphorylated Tau levels and decreases inhibition of GSK3 beta. Luprolide acetate rescues ovariectomy-dependent cognitive function, increases signaling events associated with synaptic plasticity including GSK3 beta inhibition, but does not alter AD pathology. In the human AD female brain, luteinizing hormone (LH) mRNA levels are reduced. In the 3XAD-tg model, brain LH protein levels are reduced by ovariectomy and normalized by leuprolide acetate treatment. These treatment-dependent normalization of LH positively correlates with markers of neuroplasticity and cognitive improvement.


Asunto(s)
Envejecimiento/sangre , Cognición/fisiología , Regulación hacia Abajo/fisiología , Terapia de Reemplazo de Estrógeno/métodos , Estrógenos/administración & dosificación , Hormona Luteinizante/sangre , Ovariectomía , Animales , Biomarcadores/sangre , Células Cultivadas , Femenino , Gonadotropinas/sangre , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley
7.
J Neurochem ; 128(1): 162-72, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23895348

RESUMEN

Leptin signaling has received considerable attention in the Alzheimer disease (AD) field. Within the past decade, the peptide hormone has been demonstrated to attenuate tau hyperphosphorylation in neuronal cells and to be modulated by amyloid-ß. Moreover, a role in neuroprotection and neurogenesis within the hippocampus has been shown in animal models. To further characterize the association between leptin signaling and vulnerable regions in AD, we assessed the profile of leptin and the leptin receptor in AD and control patients. We analyzed leptin levels in CSF, and the concentration and localization of leptin and leptin receptor in the hippocampus. Significant elevations in leptin levels in both CSF and hippocampal tissue of AD patients, compared with age-matched control cases, indicate a physiological up-regulation of leptin in AD. However, the level of leptin receptor mRNA decreased in AD brain and the leptin receptor protein was localized to neurofibrillary tangles, suggesting a severe discontinuity in the leptin signaling pathway. Collectively, our results suggest that leptin resistance in the hippocampus may play a role in the characteristic changes associated with the disease. These findings are the first to demonstrate such dysregulated leptin-signaling circuitry and provide novel insights into the possible role of aberrant leptin signaling in AD. In this study, increased leptin was found in CSF and hippocampus in Alzheimer disease indicating its physiological up-regulation, yet leptin receptor mRNA was decreased and leptin receptor protein was localized to neurofibrillary tangles, suggesting a discontinuity in the leptin signaling pathway. The lack of leptin signaling within degenerating neurons may represent a novel neuronal leptin resistance in Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Leptina/fisiología , Neuronas/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/patología , Regulación hacia Abajo/fisiología , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Leptina/líquido cefalorraquídeo , Leptina/metabolismo , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/patología , Unión Proteica/fisiología , Adulto Joven
8.
Hum Mol Genet ; 21(5): 1138-44, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22100763

RESUMEN

In Alzheimer disease (AD), amyloid-ß (Aß) oligomer is suggested to play a critical role in imitating neurodegeneration, although its pathogenic mechanism remains to be determined. Recently, the cellular prion protein (PrP(C)) has been reported to be an essential co-factor in mediating the neurotoxic effect of Aß oligomer. However, these previous studies focused on the synaptic plasticity in either the presence or the absence of PrP(C) and no study to date has reported whether PrP(C) is required for the neuronal cell death, the most critical element of neurodegeneration in AD. Here, we show that Prnp(-/-) mice are resistant to the neurotoxic effect of Aß oligomer in vivo and in vitro. Furthermore, application of an anti-PrP(C) antibody or PrP(C) peptide prevents Aß oligomer-induced neurotoxicity. These findings are the first to demonstrate that PrP(C) is required for Aß oligomer-induced neuronal cell death, the pathology essential to cognitive loss.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Muerte Celular , Neuronas/fisiología , Proteínas PrPC/metabolismo , Priones/metabolismo , Péptidos beta-Amiloides/química , Animales , Anticuerpos/inmunología , Ratones , Proteínas PrPC/genética , Proteínas PrPC/inmunología , Proteínas Priónicas , Priones/genética , Técnicas de Cultivo de Tejidos
9.
BMC Cancer ; 14: 928, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25491510

RESUMEN

BACKGROUND: Amyloid-ß precursor protein (APP) is a highly conserved single transmembrane protein that has been linked to Alzheimer disease. Recently, the increased expression of APP in multiple types of cancers has been reported where it has significant correlation with the cancer cell proliferation. However, the function of APP in the pathogenesis of breast cancer has not previously been determined. In this study, we studied the pathological role of APP in breast cancer and revealed its potential mechanism. METHODS: The expression level of APP in multiple breast cancer cell lines was measured by Western blot analysis and the breast cancer tissue microarray was utilized to analyze the expression pattern of APP in human patient specimens. To interrogate the functional role of APP in cell growth and apoptosis, the effect of APP knockdown in MDA-MB-231 cells were analyzed. Specifically, multiple signal transduction pathways and functional alterations linked to cell survival and motility were examined in in vivo animal model as well as in vitro cell culture with the manipulation of APP expression. RESULTS: We found that the expression of APP is increased in mouse and human breast cancer cell lines, especially in the cell line possessing higher metastatic potential. Moreover, the analysis of human breast cancer tissues revealed a significant correlation between the level of APP and tumor development. Knockdown of APP (APP-kd) in breast cancer cells caused the retardation of cell growth in vitro and in vivo, with both the induction of p27(kip1) and caspase-3-mediated apoptosis. APP-kd cells also had higher sensitivity to treatment of chemotherapeutic agents, TRAIL and 5-FU. Such anti-tumorigenic effects shown in the APP-kd cells partially came from reduced pro-survival AKT activation in response to IGF-1, leading to activation of key signaling regulators for cell growth, survival, and pro-apoptotic events such as GSK3-ß and FOXO1. Notably, knock-down of APP in metastatic breast cancer cells limited cell migration and invasion ability upon stimulation of IGF-1. CONCLUSION: The present data strongly suggest that the increase of APP expression is causally linked to tumorigenicity as well as invasion of aggressive breast cancer and, therefore, the targeting of APP may be an effective therapy for breast cancer.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/genética , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Supervivencia Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones , Estadificación de Neoplasias , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Neurosci ; 32(48): 17262-72, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23197718

RESUMEN

Serine-arginine protein kinases 2 (SRPK2) is a cell cycle-regulated kinase that phosphorylates serine/arginine domain-containing proteins and mediates pre-mRNA splicing with unclear function in neurons. Here, we show that SRPK2 phosphorylates tau on S214, suppresses tau-dependent microtubule polymerization, and inhibits axonal elongation in neurons. Depletion of SRPK2 in dentate gyrus inhibits tau phosphorylation in APP/PS1 mouse and alleviates the impaired cognitive behaviors. The defective LTP in APP/PS1 mice is also improved after SRPK2 depletion. Moreover, active SRPK2 is increased in the cortex of APP/PS1 mice and the pathological structures of human Alzheimer's disease (AD) brain. Therefore, our study suggests SRPK2 may contribute to the formation of hyperphosphorylated tau and the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Conducta Animal/fisiología , Encéfalo/metabolismo , Aprendizaje por Laberinto/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Neuritas/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas tau/genética
11.
J Biol Chem ; 286(17): 15095-105, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21393248

RESUMEN

The prion protein (PrP) is best known for its association with prion diseases. However, a controversial new role for PrP in Alzheimer disease (AD) has recently emerged. In vitro studies and mouse models of AD suggest that PrP may be involved in AD pathogenesis through a highly specific interaction with amyloid-ß (Aß42) oligomers. Immobilized recombinant human PrP (huPrP) also exhibited high affinity and specificity for Aß42 oligomers. Here we report the novel finding that aggregated forms of huPrP and Aß42 are co-purified from AD brain extracts. Moreover, an anti-PrP antibody and an agent that specifically binds to insoluble PrP (iPrP) co-precipitate insoluble Aß from human AD brain. Finally, using peptide membrane arrays of 99 13-mer peptides that span the entire sequence of mature huPrP, two distinct types of Aß binding sites on huPrP are identified in vitro. One specifically binds to Aß42 and the other binds to both Aß42 and Aß40. Notably, Aß42-specific binding sites are localized predominantly in the octapeptide repeat region, whereas sites that bind both Aß40 and Aß42 are mainly in the extreme N-terminal or C-terminal domains of PrP. Our study suggests that iPrP is the major PrP species that interacts with insoluble Aß42 in vivo. Although this work indicated the interaction of Aß42 with huPrP in the AD brain, the pathophysiological relevance of the iPrP/Aß42 interaction remains to be established.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Priones/metabolismo , Anciano , Anciano de 80 o más Años , Sitios de Unión , Encéfalo/metabolismo , Estudios de Casos y Controles , Humanos , Persona de Mediana Edad , Unión Proteica , Solubilidad
12.
J Neurochem ; 120(3): 419-29, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22077634

RESUMEN

Mitochondrial dysfunction is a prominent feature of Alzheimer's disease (AD) brain. Our prior studies demonstrated reduced mitochondrial number in susceptible hippocampal neurons in the brain from AD patients and in M17 cells over-expressing familial AD-causing amyloid precursor protein (APP) mutant (APPswe). In the current study, we investigated whether alterations in mitochondrial biogenesis contribute to mitochondrial abnormalities in AD. Mitochondrial biogenesis is regulated by the peroxisome proliferator activator receptor gamma-coactivator 1α (PGC-1α)-nuclear respiratory factor (NRF)-mitochondrial transcription factor A pathway. Expression levels of PGC-1α, NRF 1, NRF 2, and mitochondrial transcription factor A were significantly decreased in both AD hippocampal tissues and APPswe M17 cells, suggesting a reduced mitochondrial biogenesis. Indeed, APPswe M17 cells demonstrated decreased mitochondrial DNA/nuclear DNA ratio, correlated with reduced ATP content, and decreased cytochrome C oxidase activity. Importantly, over-expression of PGC-1α could completely rescue while knockdown of PGC-1α could exacerbate impaired mitochondrial biogenesis and mitochondrial deficits in APPswe M17 cells, suggesting reduced mitochondrial biogenesis is likely involved in APPswe-induced mitochondrial deficits. We further demonstrated that reduced expression of p-CREB and PGC-1α in APPswe M17 cells could be rescued by cAMP in a dose-dependent manner, which could be inhibited by PKA inhibitor H89, suggesting that the PKA/CREB pathway plays a critical role in the regulation of PGC-1α expression in APPswe M17 cells. Overall, this study demonstrated that impaired mitochondrial biogenesis likely contributes to mitochondrial dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Hipocampo/ultraestructura , Mitocondrias/metabolismo , Biogénesis de Organelos , Adenosina Trifosfato/metabolismo , Anciano , Anciano de 80 o más Años , Precursor de Proteína beta-Amiloide/metabolismo , Proteína de Unión a CREB/metabolismo , Línea Celular Tumoral , ADN Mitocondrial/metabolismo , Relación Dosis-Respuesta a Droga , Complejo IV de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neuroblastoma/patología , Neuroblastoma/ultraestructura , Factores Nucleares de Respiración/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Interferencia de ARN/fisiología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transfección/métodos
13.
Biochim Biophys Acta ; 1802(1): 212-20, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19853657

RESUMEN

Mitochondrial dysfunction has long been associated with neurodegenerative disease. Therefore, mitochondrial protective agents represent a unique direction for the development of drug candidates that can modify the pathogenesis of neurodegeneration. This review discusses evidence showing that mitochondrial dysfunction has a central role in the pathogenesis of Alzheimer's, Parkinson's and Huntington's diseases and amyotrophic lateral sclerosis. We also debate the potential therapeutic efficacy of metabolic antioxidants, mitochondria-directed antioxidants and Szeto-Schiller (SS) peptides. Since these compounds preferentially target mitochondria, a major source of oxidative damage, they are promising therapeutic candidates for neurodegenerative diseases. Furthermore, we will briefly discuss the novel action of the antihistamine drug Dimebon on mitochondria.


Asunto(s)
Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Antioxidantes/metabolismo , Antioxidantes/uso terapéutico , Humanos , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo
14.
Lab Invest ; 91(12): 1777-86, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21968813

RESUMEN

The phosphorylated ribosomal protein S6 (pS6) is associated with the 40S ribosomal subunit in eukaryotes and is thought to have a role in RNA storage, degradation, and re-entry into translation. In this study, we found pS6 localized to granulovacuolar degeneration (GVD) within the pyramidal neurons. Immunohistochemical analysis found that nearly 20-fold more neurons contain pS6-positive granules in Alzheimer's disease (AD) hippocampus compared with age-matched controls. Further, pS6-positive granules were more common in neurons not containing neurofibrillary tangles (NFTs), were never associated with extracellular NFTs or in apoptotic neurons, and contained less RNA than neighboring pyramidal neurons not containing pS6-positive granules. In model systems, pS6 is a specific marker for stress granules, and another stress granule protein, p54/Rck, was also found to be a component of GVD in the current study. Stress granules are transient, intracellular, dense aggregations of proteins and RNAs that accumulate as a stress response, protecting cells from apoptosis and inappropriate transcriptional activity, often described as a form of 'molecular triage.' The RNA oxidation modification 8-hydroxyguanosine (8OHG) is strikingly increased in AD, yet this study reports that those neurons with pS6 granules display reduced RNA oxidation demonstrated by lower levels of 8OHG. Since chronic oxidative stress is central to AD pathogenesis, and RNA is a specific oxidative stress target and is intimately associated with stress granule biogenesis in model systems, we suggest that GVD in human brain parallel stress granules, and may in fact be more representative of early disease pathogenesis than traditionally believed. This proposed origin for GVD as a neuroprotective response, may represent a morphologic checkpoint between cell death and reversible cellular stress that proceeds in the absence of other inclusions.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/patología , Células Piramidales/patología , Proteína S6 Ribosómica/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Estudios de Casos y Controles , Femenino , Humanos , Inmunohistoquímica , Masculino , Ovillos Neurofibrilares , Oxidación-Reducción , Estrés Oxidativo , Células Piramidales/metabolismo , ARN Ribosómico/metabolismo , Adulto Joven
15.
J Neural Transm (Vienna) ; 118(1): 23-6, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20972807

RESUMEN

CD3ζ is a subunit of the CD3 molecule that, until recently, appeared restricted to T cells and natural killer cells. However, experimental studies have demonstrated a role of CD3ζ in dendritic outgrowth in the visual system as well as in synaptic plasticity. Given the increasing evidence for uncharacteristic recapitulation of neurodevelopmental processes in neurodegenerative diseases, in this study, we evaluated brains from subjects with Parkinson's disease and Lewy body dementia for evidence of aberrant CD3 expression. Our data shows marked CD3ζ in association with the α-synuclein containing pathological lesions, i.e., Lewy bodies and Lewy neurites, in the brains of subjects with Parkinson's disease and Lewy body dementia. This finding raises the novel concept of CD3 dysregulation in these disorders as a pathogenic factor and also furthers the increasing evidence that the recall of aberrant neurodevelopmental processes underlies the pathogenesis of neurodegenerative diseases.


Asunto(s)
Complejo CD3/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Femenino , Humanos , Inmunohistoquímica , Cuerpos de Lewy/patología , Cuerpos de Lewy/ultraestructura , Masculino , Persona de Mediana Edad , alfa-Sinucleína/metabolismo
16.
Cell Mol Biol Lett ; 16(3): 359-72, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21461981

RESUMEN

Post-mitotic neurons are typically terminally differentiated and in a quiescent status. However, in Alzheimer disease (AD), many neurons display ectopic re-expression of cell cycle-related proteins. Cyclin-dependent kinase 11 (CDK11) mRNA produces a 110-kDa protein (CDK11(p110)) throughout the cell cycle, a 58-kDa protein (CDK11(p58)) that is specifically translated from an internal ribosome entry site and expressed only in the G(2)/M phase of the cell cycle, and a 46-kDa protein (CDK11(p46)) that is considered to be apoptosis specific. CDK11 is required for sister chromatid cohesion and the completion of mitosis. In this study, we found that the expression patterns of CDK11 vary such that cytoplasmic CDK11 is increased in AD cellular processes, compared to a pronounced nuclear expression pattern in most controls. We also investigated the effect of amyloid precursor protein (APP) on CDK11 expression in vitro by using M17 cells overexpressing wild-type APP and APP Swedish mutant phenotype and found increased CDK11 expression compared to empty vector. In addition, amyloid-ß(25-35) resulted in increased CDK11 in M17 cells. These data suggest that CDK11 may play a vital role in cell cycle re-entry in AD neurons in an APP-dependent manner, thus presenting an intriguing novel function of the APP signaling pathway in AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , División Celular , Quinasas Ciclina-Dependientes/metabolismo , Fase G2 , Adulto , Anciano , Anciano de 80 o más Años , Precursor de Proteína beta-Amiloide/metabolismo , Western Blotting , Línea Celular , Secciones por Congelación , Hipocampo/patología , Humanos , Persona de Mediana Edad , Neuronas/enzimología , Neuronas/patología , Transporte de Proteínas , Adulto Joven
17.
Mol Neurodegener ; 16(1): 23, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33849621

RESUMEN

BACKGROUND: Recently, we and other researchers reported that brain metabolic disorders are implicated in Alzheimer's disease (AD), a progressive, devastating and incurable neurodegenerative disease. Hence, novel therapeutic approaches are urgently needed to explore potential and novel therapeutic targets/agents for the treatment of AD. The neuronal adiponectin receptor 1 (AdipoR1) is an emerging potential target for intervention in metabolic-associated AD. We aimed to validate this hypothesis and explore in-depth the therapeutic effects of an osmotin-derived adiponectin-mimetic novel nonapeptide (Os-pep) on metabolic-associated AD. METHODS: We used an Os-pep dosage regimen (5 µg/g, i.p., on alternating days for 45 days) for APP/PS1 in amyloid ß oligomer-injected, transgenic adiponectin knockout (Adipo-/-) and AdipoR1 knockdown mice. After behavioral studies, brain tissues were subjected to biochemical and immunohistochemical analyses. In separate cohorts of mice, electrophysiolocal and Golgi staining experiments were performed. To validate the in vivo studies, we used human APP Swedish (swe)/Indiana (ind)-overexpressing neuroblastoma SH-SY5Y cells, which were subjected to knockdown of AdipoR1 and APMK with siRNAs, treated with Os-pep and other conditions as per the mechanistic approach, and we proceeded to perform further biochemical analyses. RESULTS: Our in vitro and in vivo results show that Os-pep has good safety and neuroprotection profiles and crosses the blood-brain barrier. We found reduced levels of neuronal AdipoR1 in human AD brain tissue. Os-pep stimulates AdipoR1 and its downstream target, AMP-activated protein kinase (AMPK) signaling, in AD and Adipo-/- mice. Mechanistically, in all of the in vivo and in vitro studies, Os-pep rescued aberrant neuronal metabolism by reducing neuronal insulin resistance and activated downstream insulin signaling through regulation of AdipoR1/AMPK signaling to consequently improve the memory functions of the AD and Adipo-/- mice, which was associated with improved synaptic function and long-term potentiation via an AdipoR1-dependent mechanism. CONCLUSION: Our findings show that Os-pep activates AdipoR1/AMPK signaling and regulates neuronal insulin resistance and insulin signaling, which subsequently rescues memory deficits in AD and adiponectin-deficient models. Taken together, the results indicate that Os-pep, as an adiponectin-mimetic novel nonapeptide, is a valuable and promising potential therapeutic candidate to treat aberrant brain metabolism associated with AD and other neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Trastornos de la Memoria/prevención & control , Fármacos Neuroprotectores/farmacología , Receptores de Adiponectina/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/deficiencia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/genética , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Humanos , Resistencia a la Insulina , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Presenilina-1/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Adiponectina/genética , Transducción de Señal
18.
J Neurosci ; 29(28): 9090-103, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19605646

RESUMEN

Mitochondrial dysfunction is a prominent feature of Alzheimer's disease (AD) neurons. In this study, we explored the involvement of an abnormal mitochondrial dynamics by investigating the changes in the expression of mitochondrial fission and fusion proteins in AD brain and the potential cause and consequence of these changes in neuronal cells. We found that mitochondria were redistributed away from axons in the pyramidal neurons of AD brain. Immunoblot analysis revealed that levels of DLP1 (also referred to as Drp1), OPA1, Mfn1, and Mfn2 were significantly reduced whereas levels of Fis1 were significantly increased in AD. Despite their differential effects on mitochondrial morphology, manipulations of these mitochondrial fission and fusion proteins in neuronal cells to mimic their expressional changes in AD caused a similar abnormal mitochondrial distribution pattern, such that mitochondrial density was reduced in the cell periphery of M17 cells or neuronal process of primary neurons and correlated with reduced spine density in the neurite. Interestingly, oligomeric amyloid-beta-derived diffusible ligands (ADDLs) caused mitochondrial fragmentation and reduced mitochondrial density in neuronal processes. More importantly, ADDL-induced synaptic change (i.e., loss of dendritic spine and postsynaptic density protein 95 puncta) correlated with abnormal mitochondrial distribution. DLP1 overexpression, likely through repopulation of neuronal processes with mitochondria, prevented ADDL-induced synaptic loss, suggesting that abnormal mitochondrial dynamics plays an important role in ADDL-induced synaptic abnormalities. Based on these findings, we suggest that an altered balance in mitochondrial fission and fusion is likely an important mechanism leading to mitochondrial and neuronal dysfunction in AD brain.


Asunto(s)
Enfermedad de Alzheimer/patología , Regulación de la Expresión Génica/fisiología , Mitocondrias/patología , Proteínas Mitocondriales/metabolismo , Neuronas/ultraestructura , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Biotina/metabolismo , Células Cultivadas , Ciclooxigenasa 1/metabolismo , Dinaminas , Embrión de Mamíferos , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Proteínas Fluorescentes Verdes/genética , Hipocampo/patología , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Microscopía Inmunoelectrónica/métodos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Mitocondriales/genética , Mutación/genética , Neuroblastoma/patología , Neuroblastoma/ultraestructura , Cambios Post Mortem , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Transfección/métodos
19.
J Neurochem ; 112(4): 870-81, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19943850

RESUMEN

Declining levels of estrogen in women result in increases in gonadotropins such as luteinizing hormone (LH) through loss of feedback inhibition. LH, like estrogen, is modulated by hormone replacement therapy. However, the role of post-menopausal gonadotropin increases on cognition has not been evaluated. Here, we demonstrate that the down-regulation of ovariectomy-driven LH elevations using the gonadotropin releasing hormone super-analogue, leuprolide acetate, improves cognitive function in the Morris water maze and Y-maze tests in the absence of E2. Furthermore, our data suggest that these effects are independent of the modulation of estrogen receptors alpha and beta, or activation of CYP19 and StAR, associated with the production of endogenous E2. Importantly, pathways associated with improved cognition such as CaMKII and GluR1-Ser831 are up-regulated by leuprolide treatment but not by chronic long-term E2 replacement suggesting independent cognition-modulating properties. Our findings suggest that down-regulation of gonadotropins is as effective as E2 in modulating cognition but likely acts through different molecular mechanisms. These findings provide a potential novel protective strategy to treat menopause/age-related cognitive decline and/or prevent the development of AD.


Asunto(s)
Trastornos del Conocimiento/terapia , Regulación hacia Abajo/fisiología , Terapia de Reemplazo de Estrógeno/métodos , Gonadotropinas/sangre , Menopausia , Animales , Aromatasa/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Fármacos para la Fertilidad Femenina/uso terapéutico , Gonadotropinas/genética , Leuprolida/farmacología , Leuprolida/uso terapéutico , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ovariectomía/métodos , ARN Mensajero/metabolismo , Tiempo de Reacción/efectos de los fármacos , Receptores AMPA/metabolismo , Receptores de Estrógenos/química , Serina/genética
20.
J Neurochem ; 113(5): 1221-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20345761

RESUMEN

Cyclin-dependent kinase (Cdk) 5 and p38 activities are significantly increased in Alzheimer's Disease (AD). Both p38 and Cdk5 promote neurodegeneration upon deregulation. However, to date the mechanistic link between Cdk5 and p38 remains unclear. This study presents the first mechanism showing Cdk5 as a major regulator of p38 cascade in neurons and in transgenic mouse model of AD. Using beta-amyloid and glutamate as the neurotoxic stimuli, our results show that deregulated Cdk5 induces p38 activation by increasing reactive oxygen species (ROS) in neuronal cells and in primary cortical neurons. Elimination of ROS inhibits p38 activation, revealing ROS as major stimuli of the p38 cascade. Importantly, Cdk5-mediated p38 activation increases c-Jun expression, thereby revealing a mechanistic link between deregulated Cdk5 and c-Jun level in AD brains. c-Jun is over-expressed in AD, and is believed to contribute significantly to neurodegeneration. Based on the proposed mechanism, Cdk5 inhibition is more neuroprotective relative to p38 and c-Jun, suggesting that Cdk5 is an upstream regulator of neurodegenerative pathways triggered by p38 and a preferable therapeutic target for AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Quinasa 5 Dependiente de la Ciclina/fisiología , Neuronas/patología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Enfermedad de Alzheimer/enzimología , Péptidos beta-Amiloides/farmacología , Animales , Western Blotting , Calpaína/fisiología , Colorantes , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Ácido Glutámico/farmacología , Humanos , Inmunohistoquímica , MAP Quinasa Quinasa 6/metabolismo , Ratones , Ratones Transgénicos , Neuronas/enzimología , Fosforilación , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Sales de Tetrazolio , Tiazoles , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA