Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Mol Sci ; 24(20)2023 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-37894744

RESUMEN

Alzheimer's disease (AD) is characterized by the accumulation of amyloid ß (Aß) plaques in the brain, leading to cognitive impairment and other clinical symptoms. The 5XFAD mouse model is commonly used in AD research because it expresses five human transgenes that result in the accumulation of Aß plaques and cognitive decline at a relatively early age. Behavioral experiments are frequently conducted using this model; however, the effect size has not yet been reported. In this study, we examined basic cognition and locomotion in 5XFAD mice with a C57BL6/J background (5XFAD-J) at 6 months of age, a period in which impairments of cognitive function and locomotion are commonly observed. We analyzed the effect sizes of cognitive and locomotive experiments in the 5XFAD mice compared with those in the wild-type mice. Our results suggest that for long-term memory analysis, the novel object recognition test (p = 0.013, effect size 1.24) required a sample size of at least 12 to obtain meaningful results. Moreover, analysis of general locomotion over total distance with the Laboratory Animal Behavior Observation, Registration and Analysis System (LABORAS) test during the dark phase (p = 0.007, effect size -1.37) needed a sample size of 10 for a statistical power (1-ß) of 0.8. In conclusion, we can conduct more ethical and scientifically rigorous animal experiments using 5XFAD mice based on the effect and sample sizes suggested in this study.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Ratones , Animales , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides , Ratones Transgénicos , Escala de Evaluación de la Conducta , Cognición , Modelos Animales de Enfermedad
2.
Int J Mol Sci ; 24(19)2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37834402

RESUMEN

Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases and a major contributor to dementia. Although the cause of this condition has been identified long ago as aberrant aggregations of amyloid and tau proteins, effective therapies for it remain elusive. The complexities of drug development for AD treatment are often compounded by the impermeable blood-brain barrier and low-yield brain delivery. In addition, the use of high drug concentrations to overcome this challenge may entail side effects. To address these challenges and enhance the precision of delivery into brain regions affected by amyloid aggregation, we proposed a transferrin-conjugated nanoparticle-based drug delivery system. The transferrin-conjugated melittin-loaded L-arginine-coated iron oxide nanoparticles (Tf-MeLioNs) developed in this study successfully mitigated melittin-induced cytotoxicity and hemolysis in the cell culture system. In the 5XFAD mouse brain, Tf-MeLioNs remarkably reduced amyloid plaque accumulation, particularly in the hippocampus. This study suggested Tf-LioNs as a potential drug delivery platform and Tf-MeLioNs as a candidate for therapeutic drug targeting of amyloid plaques in AD. These findings provide a foundation for further exploration and advancement in AD therapeutics.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Ratones , Animales , Péptidos beta-Amiloides/metabolismo , Meliteno/farmacología , Transferrina/metabolismo , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Amiloide/metabolismo , Nanopartículas Magnéticas de Óxido de Hierro , Ratones Transgénicos , Placa Amiloide/metabolismo , Modelos Animales de Enfermedad
3.
Int J Mol Sci ; 23(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36293147

RESUMEN

The typical pathological features of Alzheimer's disease (AD) are the accumulation of amyloid plaques in the brain and reactivity of glial cells such as astrocytes and microglia. Clinically, the development of AD and obesity are known to be correlated. In this study, we analyzed the changes in AD pathological characteristics in 5XFAD mice after obesity induction through a high-fat diet (HFD). Surprisingly, high-density lipoprotein and apolipoprotein AI (APOA-I) serum levels were increased without low-density lipoprotein alteration in both HFD groups. The reactivity of astrocytes and microglia in the dentate gyrus of the hippocampus and fornix of the hypothalamus in 5XFAD mice was decreased in the transgenic (TG)-HFD high group. Finally, the accumulation of amyloid plaques in the dentate gyrus region of the hippocampus was also significantly decreased in the TG-HFD high group. These results suggest that increased high-density lipoprotein level, especially with increased APOA-I serum level, alleviates the pathological features of AD and could be a new potential therapeutic strategy for AD treatment.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/patología , Placa Amiloide/patología , Dieta Alta en Grasa/efectos adversos , Péptidos beta-Amiloides , Apolipoproteína A-I , Lipoproteínas HDL/uso terapéutico , Ratones Transgénicos , Modelos Animales de Enfermedad , Obesidad/etiología , Lipoproteínas LDL
4.
Acta Neuropathol ; 132(6): 859-873, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27785573

RESUMEN

TDP-43 proteinopathy, initially associated with ALS and FTD, is also found in 30-60% of Alzheimer's disease (AD) cases and correlates with worsened cognition and neurodegeneration. A major component of this proteinopathy is depletion of this RNA-binding protein from the nucleus, which compromises repression of non-conserved cryptic exons in neurodegenerative diseases. To test whether nuclear depletion of TDP-43 may contribute to the pathogenesis of AD cases with TDP-43 proteinopathy, we examined the impact of depletion of TDP-43 in populations of neurons vulnerable in AD, and on neurodegeneration in an AD-linked context. Here, we show that some populations of pyramidal neurons that are selectively vulnerable in AD are also vulnerable to TDP-43 depletion in mice, while other forebrain neurons appear spared. Moreover, TDP-43 depletion in forebrain neurons of an AD mouse model exacerbates neurodegeneration, and correlates with increased prefibrillar oligomeric Aß and decreased Aß plaque burden. These findings support a role for nuclear depletion of TDP-43 in the pathogenesis of AD and provide strong rationale for developing novel therapeutics to alleviate the depletion of TDP-43 and functional antemortem biomarkers associated with its nuclear loss.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Proteínas de Unión al ADN/deficiencia , Enfermedades Neurodegenerativas/etiología , Placa Amiloide , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Antineoplásicos Hormonales/uso terapéutico , Autofagia/genética , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Placa Amiloide/etiología , Placa Amiloide/genética , Placa Amiloide/patología , Presenilina-1/genética , Presenilina-1/metabolismo , Prosencéfalo/patología , Tamoxifeno/uso terapéutico
5.
Neuron ; 112(4): 611-627.e8, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38086372

RESUMEN

Social animals compete for limited resources, resulting in a social hierarchy. Although different neuronal subpopulations in the medial prefrontal cortex (mPFC), which has been mechanistically implicated in social dominance behavior, encode distinct social competition behaviors, their identities and associated molecular underpinnings have not yet been identified. In this study, we found that mPFC neurons projecting to the nucleus accumbens (mPFC-NAc) encode social winning behavior, whereas mPFC neurons projecting to the ventral tegmental area (mPFC-VTA) encode social losing behavior. High-throughput single-cell transcriptomic analysis and projection-specific genetic manipulation revealed that the expression level of POU domain, class 3, transcription factor 1 (Pou3f1) in mPFC-VTA neurons controls social hierarchy. Optogenetic activation of mPFC-VTA neurons increases Pou3f1 expression and lowers social rank. Together, these data demonstrate that discrete activity and gene expression in separate mPFC projections oppositely orchestrate social competition and hierarchy.


Asunto(s)
Núcleo Accumbens , Área Tegmental Ventral , Animales , Área Tegmental Ventral/fisiología , Núcleo Accumbens/fisiología , Conducta Social , Corteza Prefrontal/fisiología , Neuronas
6.
Proc Natl Acad Sci U S A ; 107(37): 16320-4, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20660762

RESUMEN

Tat activating regulatory DNA-binding protein (Tardbp or TDP-43), a highly conserved metazoan DNA/RNA binding protein thought to be involved in RNA transcription and splicing, has been linked to the pathophysiology of amyotrophic lateral sclerosis and frontotemporal lobar degeneration and is essential for early embryonic development. However, neither the physiological role of TDP-43 in the adult nor its downstream targets are well defined. To address these questions, we developed conditional Tardbp-KO mice and embryonic stem (ES) cell models. Here, we show that postnatal deletion of Tardbp in mice caused dramatic loss of body fat followed by rapid death. Moreover, conditional Tardbp-KO ES cells failed to proliferate. Importantly, high-throughput DNA sequencing analysis on the transcriptome of ES cells lacking Tardbp revealed a set of downstream targets of TDP-43. We show that Tbc1d1, a gene known to mediate leanness and linked to obesity, is down-regulated in the absence of TDP-43. Collectively, our results establish that TDP-43 is critical for fat metabolism and ES cell survival.


Asunto(s)
Tejido Adiposo/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Eliminación de Gen , Proteínas Nucleares/metabolismo , Obesidad/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas Activadoras de GTPasa , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Proteínas Nucleares/genética , Obesidad/genética , Células Madre/citología , Células Madre/metabolismo
7.
J Neurochem ; 119(6): 1282-93, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21967036

RESUMEN

Various environmental factors are known to influence the onset and progression of Alzheimer's disease (AD). Environmental enrichment was reported to improve cognitive performance in various Alzheimer's transgenic mice via an amyloid-related or unrelated mechanism. However, stress has been found to accelerate amyloid deposition and cognitive deficits in many AD models. The aim of this study was to determine whether environmental enrichment compensates for the effects of stress on disease progression in the Tg2576 mice, an established AD model. We housed Tg2576 mice under environmental enrichment, enrichment plus stress, stress, or control conditions at 3 months of age. In this study, we first report that environmental enrichment counteracts the effects of stress in terms of cognitive deficits, tau phosphorylation, neurogenesis, and neuronal proliferation during AD-like disease progression. These results strongly implicate the importance of environmental factors as a major modulator for the disease progression of AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Progresión de la Enfermedad , Ambiente , Estrés Psicológico/fisiopatología , Factores de Edad , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Análisis de Varianza , Animales , Movimiento Celular/fisiología , Corticosterona/sangre , Quinasa 5 Dependiente de la Ciclina/metabolismo , Giro Dentado/citología , Giro Dentado/fisiología , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neurogénesis/fisiología , Neuropéptidos/metabolismo , Fragmentos de Péptidos/metabolismo , Fosfotransferasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas tau/metabolismo
8.
Front Psychiatry ; 12: 671722, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34744808

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has disrupted our everyday life. Along with the fear of getting infected or of having loved ones infected, the lifestyle changes and the socioeconomic consequences of the pandemic have profound impact on mental health of the general population. While numerous studies on immediate psychological responses to COVID-19 are being published, there is a lack of discussion on its possible long-term sequelae. In this study, we systematically reviewed and meta-analyzed longitudinal studies that examined mental health of the general population prior to and during the pandemic. Furthermore, we explored the long-term psychiatric implications of the pandemic with data from South Korea. Our analysis showed that the number of suicidal deaths during the pandemic was lower than the previous years in many countries, which is in contrast with the increased depression, anxiety, and psychological distress in the general population in South Korea as well as in other countries. To explain this phenomenon, we propose a possibility of delayed impacts. The post-traumatic stress, long-term consequences of social restrictions, and maladaptive response to the "new normal" are discussed in the paper. COVID-19 being an unprecedented global crisis, more research and international collaboration are needed to understand, to treat, and to prevent its long-term effects on our mental health.

9.
Mol Brain ; 14(1): 111, 2021 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-34246283

RESUMEN

The morphological dynamics of astrocytes are altered in the hippocampus during memory induction. Astrocyte-neuron interactions on synapses are called tripartite synapses. These control the synaptic function in the central nervous system. Astrocytes are activated in a reactive state by STAT3 phosphorylation in 5XFAD mice, an Alzheimer's disease (AD) animal model. However, changes in astrocyte-neuron interactions in reactive or resting-state astrocytes during memory induction remain to be defined. Here, we investigated the time-dependent changes in astrocyte morphology and the number of astrocyte-neuron interactions in the hippocampus over the course of long-term memory formation in 5XFAD mice. Hippocampal-dependent long-term memory was induced using a contextual fear conditioning test in 5XFAD mice. The number of astrocytic processes increased in both wild-type and 5XFAD mice during memory formation. To assess astrocyte-neuron interactions in the hippocampal dentate gyrus, we counted the colocalization of glial fibrillary acidic protein and postsynaptic density protein 95 via immunofluorescence. Both groups revealed an increase in astrocyte-neuron interactions after memory induction. At 24 h after memory formation, the number of tripartite synapses returned to baseline levels in both groups. However, the total number of astrocyte-neuron interactions was significantly decreased in 5XFAD mice. Administration of Stattic, a STAT3 phosphorylation inhibitor, rescued the number of astrocyte-neuron interactions in 5XFAD mice. In conclusion, we suggest that a decreased number of astrocyte-neuron interactions may underlie memory impairment in the early stages of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Astrocitos/patología , Comunicación Celular , Trastornos de la Memoria/patología , Neuronas/patología , Animales , Forma de la Célula , Giro Dentado/patología , Modelos Animales de Enfermedad , Ratones Transgénicos
10.
Autophagy ; 16(4): 672-682, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31242080

RESUMEN

A shared neuropathological hallmark in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is nuclear clearance and cytoplasmic aggregation of TARDBP/TDP-43 (TAR DNA binding protein). We previously showed that the ability of TARDBP to repress nonconserved cryptic exons was impaired in brains of patients with ALS and FTD, suggesting that its nuclear depletion contributes to neurodegeneration. However, the critical pathways impacted by the failure to repress cryptic exons that may contribute to neurodegeneration remain undefined. Here, we report that transcriptome analysis of TARDBP-deficient neurons revealed downregulation of ATG7, a critical gene required for macroautophagy/autophagy. Mouse and Drosophila models lacking TARDBP/TBPH in motor neurons exhibiting age-dependent neurodegeneration and motor deficits showed reduction of ATG7 and accumulation of SQSTM1/p62 inclusions. Importantly, genetic upregulation of the autophagy pathway improved motor function and survival in TBPH-deficient flies. Together with our observation that ATG7 is reduced in ALS-FTD brain tissues, these findings identify the autophagy pathway as one key effector of nuclear depletion of TARDBP that contributes to neurodegeneration. We thus suggest that the autophagy pathway is a therapeutic target for ALS-FTD and other disorders exhibiting TARDBP pathology.Abbreviations: ALS: amyotrophic lateral sclerosis; ANOVA: analysis of variance; ChAT: choline acetyltransferase; CTSD: cathepsin D; FTD: frontotemporal dementia; LAMP1: lysosomal associated membrane protein 1; NMJ: neuromuscular junction; RBFOX3/NeuN: RNA binding fox-1 homolog 3; SQSTM1: sequestosome 1; TARDBP/TDP-43: TAR DNA binding protein 43.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuronas Motoras/metabolismo , Animales , Autofagia/genética , Autofagia/fisiología , Encéfalo/metabolismo , Humanos , Ratones Transgénicos , Neuronas Motoras/patología , Regulación hacia Arriba
11.
Cells ; 9(9)2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32872335

RESUMEN

Monoamine oxidase (MAO) has been implicated in neuroinflammation, and therapies targeting MAO are of interest for neurodegenerative diseases. The small-molecule drug tranylcypromine, an inhibitor of MAO, is currently used as an antidepressant and in the treatment of cancer. However, whether tranylcypromine can regulate LPS- and/or Aß-induced neuroinflammation in the brain has not been well-studied. In the present study, we found that tranylcypromine selectively altered LPS-induced proinflammatory cytokine levels in BV2 microglial cells but not primary astrocytes. In addition, tranylcypromine modulated LPS-mediated TLR4/ERK/STAT3 signaling to alter neuroinflammatory responses in BV2 microglial cells. Importantly, tranylcypromine significantly reduced microglial activation as well as proinflammatory cytokine levels in LPS-injected wild-type mice. Moreover, injection of tranylcypromine in 5xFAD mice (a mouse model of AD) significantly decreased microglial activation but had smaller effects on astrocyte activation. Taken together, our results suggest that tranylcypromine can suppress LPS- and Aß-induced neuroinflammatory responses in vitro and in vivo.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Lipopolisacáridos/metabolismo , Inhibidores de la Monoaminooxidasa/uso terapéutico , Tranilcipromina/uso terapéutico , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Inhibidores de la Monoaminooxidasa/farmacología , Tranilcipromina/farmacología
12.
Biol Psychiatry ; 88(10): 746-757, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32622465

RESUMEN

BACKGROUND: Cholinergic interneurons (ChINs) in the nucleus accumbens (NAc) play critical roles in processing information related to reward. However, the contribution of ChINs to the emergence of addiction-like behaviors and its underlying molecular mechanisms remain elusive. METHODS: We employed cocaine self-administration to identify two mouse subpopulations: susceptible and resilient to cocaine seeking. We compared the subpopulations for physiological responses with single-unit recording of NAc ChINs, and for gene expression levels with RNA sequencing of ChINs sorted using fluorescence-activated cell sorting. To provide evidence for a causal relationship, we manipulated the expression level of dopamine D2 receptor (DRD2) in ChINs in a cell type-specific manner. Using optogenetic activation combined with a double whole-cell recording, the effect of ChIN-specific DRD2 manipulation on each synaptic input was assessed in NAc medium spiny neurons in a pathway-specific manner. RESULTS: Susceptible mice showed higher levels of nosepoke responses under a progressive ratio schedule, and impairment in extinction and punishment procedures. DRD2 was highly abundant in the NAc ChINs of susceptible mice. Elevated abundance of DRD2 in NAc ChINs was sufficient and necessary to express high cocaine motivation, putatively through reduction of ChIN activity during cocaine exposure. DRD2 overexpression in ChINs mimicked cocaine-induced effects on the dendritic spine density and the ratios of excitatory inputs between two distinct medium spiny neuron cell types, while DRD2 depletion precluded cocaine-induced synaptic plasticity. CONCLUSIONS: These findings provide a molecular mechanism for dopaminergic control of NAc ChINs that can control the susceptibility to cocaine-seeking behavior.


Asunto(s)
Trastornos Relacionados con Cocaína , Cocaína , Animales , Colinérgicos , Dopamina , Interneuronas/metabolismo , Ratones , Ratones Transgénicos , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo
13.
Mol Cell Biol ; 26(11): 4327-38, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16705182

RESUMEN

Amyloid precursor protein (APP) has eight potential phosphorylation sites in its cytoplasmic domain. Recently, it has demonstrated that the constitutive phosphorylation of APP at T668 (APP695 isoform numbering) was observed specifically in the brain. Neuron-specific phosphorylation of APP at T668 is thought to be important for neuronal functions of APP, although its exact physiological significance remains to be clarified. In this study, we show that the phosphorylation of the APP intracellular domain (AICD) at T668 is essential for its binding to Fe65 and its nuclear translocation and affects the resultant neurotoxicity, possibly mediated through the induction of glycogen synthase kinase 3beta and tau phosphorylation by enhancing the formation of a ternary complex with Fe65 and CP2 transcription factor. Taken together, these results suggest that the phosphorylation of AICD at T668 contributes to the neuronal degeneration in Alzheimer's disease (AD) by regulating its translocation into the nucleus and then affects neurodegeneration; therefore, the specific inhibitor of T668 phosphorylation might be the target of AD therapy.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Núcleo Celular/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Treonina/metabolismo , Transporte Activo de Núcleo Celular , Enfermedad de Alzheimer/patología , Animales , Encéfalo/citología , Encéfalo/patología , Muerte Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Proteínas Nucleares/metabolismo , Células PC12 , Fosforilación , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Proteínas tau/metabolismo
14.
Sci Rep ; 9(1): 4615, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30874581

RESUMEN

Animal studies using chronic social defeat stress (CSDS) in mice showed that brain-derived neurotrophic factor (BDNF) signaling in the mesolimbic dopamine (DA) circuit is important for the development of social aversion. However, the downstream molecular targets after BDNF release from ventral tegmental area (VTA) DA terminals are unknown. Here, we show that depressive-like behaviors induced by CSDS are mediated in part by Gadd45b downstream of BDNF signaling in the nucleus accumbens (NAc). We show that Gadd45b mRNA levels are increased in susceptible but not resilient mice. Intra-NAc infusion of BDNF or optical stimulation of VTA DA terminals in NAc enhanced Gadd45b expression levels in the NAc. Importantly, Gadd45b downregulation reversed social avoidance in susceptible mice. Together, these data suggest that Gadd45b in NAc contributes to susceptibility to social stress. In addition, we investigated the function of Gadd45b in demethylating CpG islands of representative gene targets, which have been associated with a depressive phenotype in humans and animal models. We found that Gadd45b downregulation changes DNA methylation levels in a phenotype-, gene-, and locus-specific fashion. Together, these results highlight the contribution of Gadd45b and changes in DNA methylation in mediating the effects of social stress in the mesolimbic DA circuit.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Desmetilación del ADN/efectos de los fármacos , Depresión/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , ADN/metabolismo , Dopamina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Núcleo Accumbens/metabolismo , Conducta Social , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/metabolismo
15.
Neuropsychopharmacology ; 32(11): 2393-404, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17406652

RESUMEN

Minocycline is a semi-synthetic tetracycline antibiotic that effectively crosses the blood-brain barrier. Minocycline has been reported to have significant neuroprotective effects in models of cerebral ischemia, traumatic brain injury, amyotrophic lateral sclerosis, and Huntington's and Parkinson's diseases. In this study, we demonstrate that minocycline has neuroprotective effects in in vitro and in vivo Alzheimer's disease models. Minocycline was found to attenuate the increases in the phosphorylation of double-stranded RNA-dependent serine/threonine protein kinase, eukaryotic translation initiation factor-2 alpha and caspase 12 activation induced by amyloid beta peptide1-42 treatment in NGF-differentiated PC 12 cells. In addition, increases in the phosphorylation of eukaryotic translation initiation factor-2 alpha were attenuated by administration of minocycline in Tg2576 mice, which harbor mutated human APP695 gene including the Swedish double mutation and amyloid beta peptide(1-42)-infused rats. We found that minocycline administration attenuated deficits in learning and memory in amyloid beta peptide(1-42)-infused rats. Increased phosphorylated state of eukaryotic translation initiation factor-2 alpha is observed in Alzheimer's disease patients' brains and may result in impairment of cognitive functions in Alzheimer's disease patients by decreasing the efficacy of de novo protein synthesis required for synaptic plasticity. On the basis of these results, minocycline may prove to be a good candidate as an effective therapeutic agent for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento , Minociclina/uso terapéutico , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Reacción de Prevención/efectos de los fármacos , Encéfalo/patología , Estudios de Casos y Controles , Muerte Celular/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Minociclina/farmacología , Factor de Crecimiento Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Células PC12/efectos de los fármacos , Fragmentos de Péptidos , Ratas , Ratas Wistar , Transfección
16.
FASEB J ; 20(6): 729-31, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16467370

RESUMEN

Although chronic stress is known to be linked with memory and other neurological disorders, little is known about the relationship between chronic stress and the onset or development of Alzheimer's disease (AD). In this study, we investigated the effects of long-term stress on the onset and severity of cognitive deficits and pathological changes in APPV717I-CT100 mice overexpressing human APP-CT100 containing the London mutation (V717I) after exposure to immobilization stress. We found that chronic immobilization stress accelerated cognitive impairments, as accessed by the Passive avoidance and the Social Transfer of Food Preference (STFP) tests. Moreover, the numbers and densities of vascular and extracellular deposits containing amyloid beta peptide (Abeta) and carboxyl-terminal fragments of amyloid precursor protein (APP-CTFs), which are pathologic markers of AD, were significantly elevated in stressed animals, especially in the hippocampus. Moreover, stressed animals, also showed highly elevated levels of neurodegeneration and tau phosphorylation and increased intraneuronal Abeta and APP-CTFs immunoreactivities in the hippocampus and in the entorhinal and piriform cortex. This study provides the first evidence that chronic stress accelerates the onset and severity of cognitive deficits and that these are highly correlated with pathological changes, which thus indicates that chronic stress may be an important contributor to the onset and development of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Discapacidades para el Aprendizaje/metabolismo , Trastornos de la Memoria/metabolismo , Estrés Psicológico/metabolismo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Corteza Cerebral/metabolismo , Enfermedad Crónica , Corticosterona/sangre , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Restricción Física/efectos adversos , Estrés Psicológico/genética
17.
Mol Neurodegener ; 12(1): 13, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28153034

RESUMEN

BACKGROUND: TDP-43 proteinopathy is a prominent pathological feature that occurs in a number of human diseases including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and inclusion body myositis (IBM). Our recent finding that TDP-43 represses nonconserved cryptic exons led us to ask whether cell type-specific cryptic exons could exist to impact unique molecular pathways in brain or muscle. METHODS: In the present work, we investigated TDP-43's function in various mouse tissues to model disease pathogenesis. We generated mice to conditionally delete TDP-43 in excitatory neurons or skeletal myocytes and identified the cell type-specific cryptic exons associated with TDP-43 loss of function. RESULTS: Comparative analysis of nonconserved cryptic exons in various mouse cell types revealed that only some cryptic exons were common amongst stem cells, neurons, and myocytes; the majority of these nonconserved cryptic exons were cell type-specific. CONCLUSIONS: Our results suggest that in human disease, TDP-43 loss of function may impair cell type-specific pathways.


Asunto(s)
Proteínas de Unión al ADN/genética , Exones/genética , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Neuronas/metabolismo , Animales , Modelos Animales de Enfermedad , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteinopatías TDP-43/genética
18.
CNS Neurol Disord Drug Targets ; 15(8): 935-944, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27577574

RESUMEN

We previously demonstrated that dehydroevodiamine•HCl (DHED), which was purified from Evodia rutaecarpa Bentham (Rutaceae), has beneficial effects on memory impairment and neuronal damage in three disease models. To investigate the preventive action of DHED in Alzheimer's disease (AD), a progressive neurodegenerative disorder characterized by memory decline, amyloid-ß (Aß) protein-containing neuritic plaques and neurofibrillary tangles, in this study, we proposed that DHED may be therapeutically effective against the memory impairment and disease-related neurochemical changes that occur in Tg2576 (Tg) mice. DHED (0.5 mg/kg) was intraperitoneally administered to 7-month-old Tg and wild type mice for 4 months. In passive avoidance and water maze tests, DHED improved memory impairment of Tg mice after 4 months of administration. DHED also reduced cortical levels of soluble Aß40, soluble Aß42 and total Aß peptides in the Tg mice. Additionally, we investigated whether DHED may be a ß-secretase inhibitor that affects the production of Aß related to the formation of neuritic plaques. DHED directly inhibited ß-secretase activity in a concentrationdependent manner. The concentration required for 50 % enzyme inhibition (IC50) was 40.96 µM, and DHED may act as a competitive inhibitor of ß-secretase. Moreover, DHED interacted strongly with BACE1 (ß-secretase 2QP8), as demonstrated in the analysis of the binding mode of DHED in the active site of human BACE1. In conclusion, DHED may exhibit therapeutic effects for AD as a ß-secretase inhibitor.


Asunto(s)
Alcaloides/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Reacción de Prevención/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/genética , Ratones , Ratones Transgénicos , Modelos Moleculares , Mutación/genética , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética
19.
Brain Res ; 1462: 26-39, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22608070

RESUMEN

Recently, missense mutations in the gene TARDBP encoding TDP-43 have been linked to familial ALS. The discovery of genes encoding these RNA binding proteins, such as TDP-43 and FUS/TLS, raised the notion that altered RNA metabolism is a major factor underlying the pathogenesis of ALS. To begin to unravel how mutations in TDP-43 cause dysfunction and death of motor neurons, investigators have employed both gain- and loss-of-function studies in rodent model systems. Here, we will summarize major findings from the initial sets of TDP-43 transgenic and knockout rodent models, identify their limitations, and point to future directions toward clarification of disease mechanism(s) and testing of therapeutic strategies that ultimately may lead to novel therapy for this devastating disease. This article is part of a Special Issue entitled RNA-Binding Proteins.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteinopatías TDP-43/genética , Proteinopatías TDP-43/patología , Adiposidad/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , ADN/genética , Modelos Animales de Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Datos de Secuencia Molecular , Neuronas Motoras/metabolismo , Neuronas Motoras/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA