Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Neurobiol ; 59(7): 4124-4140, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35486224

RESUMEN

Alzheimer's disease (AD), currently the single leading cause of death still on the rise, almost always coexists alongside vascular cognitive impairment (VCI). In fact, the ischemic disease affects up to 90% of AD patients, with strokes and major infarctions representing over a third of vascular lesions. Studies also confirmed that amyloid plaques, typical of AD, are much more likely to cause dementia if strokes or cerebrovascular damage also exist, leading to the term "mixed pathology" cognitive impairment. Although its incidence is expected to grow, there are no satisfactory treatments. There is hence an urgent need for safe and effective therapies that preserve cognition, maintain function, and prevent the clinical deterioration that results from the progression of this irreversible, neurodegenerative disease. To our knowledge, this is the first study to investigate the effects of long-term treatment with C21, a novel angiotensin II type 2 receptor (AT2R) agonist, on the development of "mixed pathology" cognitive impairment. This was accomplished using a unique model that employs the fundamental elements of both AD and VCI. Treatment with C21/vehicle was started 1 h post-stroke and continued for 5 weeks in mice with concurrent AD pathology. Efficacy was established through a series of functional tests assessing various aspects of cognition, including spatial learning, short-term/working memory, long-term/reference memory, and cognitive flexibility, in addition to the molecular markers characteristic of AD. Our findings demonstrate that C21 treatment preserves cognitive function, maintains cerebral blood flow, and reduces Aß accumulation and toxic tau phosphorylation in AD animals post-stroke.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedades Neurodegenerativas , Accidente Cerebrovascular , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Animales , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Humanos , Imidazoles , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/complicaciones , Receptor de Angiotensina Tipo 2 , Accidente Cerebrovascular/complicaciones , Sulfonamidas , Tiofenos
2.
Neurochem Int ; 148: 105104, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34153352

RESUMEN

Although the exact etiology of Alzheimer's disease (AD) is poorly understood, experimental and clinical evidences suggest the contribution of neuroinflammation in the pathogenesis of AD. Pathologically, AD brain is characterized by an imbalance in redox status, elevated endoplasmic reticulum (ER) stress, synaptic dysfunction, inflammation, and progressive neurodegeneration. It has been noted that continuous accumulation of amyloid-beta (Aß) and intracellular neurofibrillary tangles (NFTs) in AD brain trigger ER stress, which contributes to neurodegeneration. Similarly, experimental evidences supports the hypothesis that thioredoxin-interacting protein (TXNIP), an endogenous regulator of redox regulator thioredoxin (TRX), is activated by ER stress and contributes to activation of NLRP3 (NOD-like receptor protein 3) inflammatory cascade in hippocampus of the AD brain. Hippocampus of postmortem human AD and aged matched non-AD controls were analyzed for the expression ER stress markers and TXNIP-NLRP3 inflammasome at cellular and molecular levels. We found higher expression of TXNIP at protein and transcript levels in close association with pathological markers of AD such as Aß and NFTs in AD hippocampus. In addition, our results demonstrated that TXNIP was co-localized in neurons and microglia. Moreover, expression of binding immunoglobulin protein (BiP), activated eukaryotic initiation factor-2α (eIf2α) and C/EBP homology protein (CHOP), proteins involved the development of ER stress, were elevated in AD hippocampus. Further, elevated expression of effector molecules of NLRP3 inflammasome activation such as apoptosis associated speck-like protein (ASC), cleaved caspase-1 and cleaved interleukin-1ß were observed in the AD hippocampus. The study suggests that TXNIP could be a link that connect ER stress with neuroinflammation. Thus, TXNIP can be a possible therapeutic target to mitigate the progression of neuroinflammation in the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas Portadoras/genética , Estrés del Retículo Endoplásmico , Hipocampo/patología , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Anciano , Anciano de 80 o más Años , Biomarcadores , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Factor 2 Eucariótico de Iniciación , Femenino , Humanos , Interleucina-1beta/metabolismo , Masculino , Persona de Mediana Edad , Factor de Transcripción CHOP/metabolismo
3.
Neurobiol Dis ; 156: 105399, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34029695

RESUMEN

Immune system hypersensitivity is believed to contribute to mental frailty in the elderly. Solid evidence indicates NOD-like receptor pyrin domain containing-3 (NLRP3)-inflammasome activation intimately connects aging-associated chronic inflammation (inflammaging) to senile cognitive decline. Thioredoxin interacting protein (TXNIP), an inducible protein involved in oxidative stress, is essential for NLRP3 inflammasome activity. This study aims to find whether TXNIP/NLRP3 inflammasome pathway is involved in senile dementia. According to our studies on sex-matched mice, TXNIP was significantly upregulated in aged animals, paralleled by the NLRP3-inflammasome over-activity leading to enhanced caspase-1 cleavage and IL-1ß maturation, in both sexes. This was closely associated with depletion of the anti-aging and cognition enhancing protein klotho, in aged males. Txnip knockout reversed age-related NLRP3-hyperactivity and enhanced thioredoxin (TRX) levels. Further, TXNIP inhibition along with verapamil replicated TXNIP/NLRP3-inflammasome downregulation in aged animals, with FOXO-1 and mTOR upregulation. These alterations concurred with substantial improvements in both cognitive and sensorimotor abilities. Together, these findings substantiate the pivotal role of TXNIP to drive inflammaging in parallel with klotho depletion and functional decline, and delineate thioredoxin system as a potential target to decelerate senile dementia.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/biosíntesis , Mediadores de Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/biosíntesis , Tiorredoxinas/biosíntesis , Envejecimiento/genética , Envejecimiento/patología , Animales , Encéfalo/patología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Femenino , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Estrés Oxidativo/fisiología , Tiorredoxinas/antagonistas & inhibidores , Tiorredoxinas/genética
4.
Cells ; 10(4)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916001

RESUMEN

Alzheimer's disease (AD), a progressive neurodegenerative disorder characterized by memory loss and cognitive decline, is a major cause of death and disability among the older population. Despite decades of scientific research, the underlying etiological triggers are unknown. Recent studies suggested that gut microbiota can influence AD progression; however, potential mechanisms linking the gut microbiota with AD pathogenesis remain obscure. In the present study, we provided a potential mechanistic link between dysbiotic gut microbiota and neuroinflammation associated with AD progression. Using a mouse model of AD, we discovered that unfavorable gut microbiota are correlated with abnormally elevated expression of gut NLRP3 and lead to peripheral inflammasome activation, which in turn exacerbates AD-associated neuroinflammation. To this end, we observe significantly altered gut microbiota compositions in young and old 5xFAD mice compared to age-matched non-transgenic mice. Moreover, 5xFAD mice demonstrated compromised gut barrier function as evident from the loss of tight junction and adherens junction proteins compared to non-transgenic mice. Concurrently, we observed increased expression of NLRP3 inflammasome and IL-1ß production in the 5xFAD gut. Consistent with our hypothesis, increased gut-microbial-inflammasome activation is positively correlated with enhanced astrogliosis and microglial activation, along with higher expression of NLRP3 inflammasome and IL-1ß production in the brains of 5xFAD mice. These data indicate that the elevated expression of gut-microbial-inflammasome components may be an important trigger for subsequent downstream activation of inflammatory and potentially cytotoxic mediators, and gastrointestinal NLRP3 may promote NLRP3 inflammasome-mediated neuroinflammation. Thus, modulation of the gut microbiota may be a potential strategy for the treatment of AD-related neurological disorders in genetically susceptible hosts.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/microbiología , Encéfalo/metabolismo , Microbioma Gastrointestinal , Inflamasomas/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/patología , Animales , Apoptosis , Encéfalo/patología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Tracto Gastrointestinal/patología , Inflamación/patología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/metabolismo
5.
Mol Neurobiol ; 58(1): 118-131, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32895786

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease that accounts for a majority of dementia cases. AD is characterized by progressive neuronal death associated with neuropathological lesions consisting of neurofibrillary tangles and senile plaques. While the pathogenesis of AD has been widely investigated, significant gaps in our knowledge remain about the cellular and molecular mechanisms promoting AD. Recent studies have highlighted the role of DNA damage, particularly DNA double-strand breaks (DSBs), in the progression of neuronal loss in a broad spectrum of neurodegenerative diseases. In the present study, we tested the hypothesis that accumulation of DNA DSB plays an important role in AD pathogenesis. To test our hypothesis, we examined DNA DSB expression and DNA repair function in the hippocampus of human AD and non-AD brains by immunohistochemistry, ELISA, and RT-qPCR. We observed increased DNA DSB accumulation and reduced DNA repair function in the hippocampus of AD brains compared to the non-AD control brains. Next, we found significantly increased levels of DNA DSB and altered levels of DNA repair proteins in the hippocampus of 5xFAD mice compared to non-transgenic mice. Interestingly, increased accumulation of DNA DSBs and altered DNA repair proteins were also observed in cellular models of AD. These findings provided compelling evidence that AD is associated with accumulation of DNA DSB and/or alteration in DSB repair proteins which may influence an important early part of the pathway toward neural damage and memory loss in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Roturas del ADN de Doble Cadena , Cambios Post Mortem , Animales , Células CHO , Células Cultivadas , Cricetulus , Reparación del ADN , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Ratones Transgénicos
6.
J Alzheimers Dis ; 68(1): 255-265, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30741672

RESUMEN

Alzheimer's disease (AD) is the most common form of age-associated dementia characterized by amyloid-ß plaques and neurofibrillary tangles. Recent studies have demonstrated that thioredoxin-interacting protein (TXNIP), an endogenous regulator of redox/glucose induced stress and inflammation, is now known to be upregulated in stroke, traumatic brain injury, diabetes and AD. We hypothesized that TXNIP overexpression sustains neurodegeneration through activation of the nucleotide binding and oligomerization domain-like receptor protein 3 in human AD brains. We analyzed TXNIP and the components of the NLRP3 inflammasome in the cortex of postmortem human brain samples by western blotting, real-time PCR, and immunohistochemical techniques in comparison with age-matched non-demented controls. Our results demonstrate that TXNIP protein as well as its mRNA levels in the cortex was significantly upregulated in AD compared to control brains. Moreover, using double immunofluorescence staining, TXNIP and interlukin-1ß (IL-1ß) were co-localized near Aß plaques and p-tau. These results suggest an association between TXNIP overexpression levels and AD pathogenesis. Further, a significant increased expression of cleaved caspase-1 and IL-1ß, the products of inflammasome activation, was detected in the cortex of AD brains. Together, these findings suggest that TXNIP, an upstream promising new therapeutic target, is a molecular link between inflammation and AD. The significant contribution of TXNIP to AD pathology suggests that strategies focusing on specific targeting of the TXNIP-NLRP3 inflammasome may lead to novel therapies for the management of AD and other age-related dementias.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Encéfalo/patología , Caspasa 1/metabolismo , Femenino , Humanos , Interleucina-1beta/metabolismo , Masculino , Persona de Mediana Edad , Placa Amiloide/metabolismo , Placa Amiloide/patología , Proteínas tau/metabolismo
7.
Neurosci Lett ; 692: 53-63, 2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30391320

RESUMEN

Converging evidence demonstrates an important role for gangliosides in brain function and neurodegenerative diseases. Exogenous GM1 is broadly neuroprotective, including in rodent, feline, and primate models of Parkinson's disease, and has shown positive effects in clinical trials. We and others have shown that inhibition of the ganglioside biosynthetic enzyme GD3 synthase (GD3S) increases endogenous levels GM1 ganglioside. We recently reported that targeted deletion of St8sia1, the gene that codes for GD3S, prevents motor impairments and significantly attenuates neurodegeneration induced by 1-methy-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The current study investigated the effects of GD3S inhibition on the neurotoxicity and parkinsonism induced by MPTP. Mice were injected intrastriatally with a lentiviral-vector-mediated shRNA construct targeting GD3S (shGD3S) or a scrambled-sequence control (scrRNA). An MPTP regimen of 18 mg/kg x 5 days reduced tyrosine-hydroxylase-positive neurons in the substantia nigra pars compacta of scrRNA-treated mice by nearly two-thirds. In mice treated with shGD3S the MPTP-induced lesion was approximately half that size. MPTP induced bradykinesia and deficits in fine motor skills in mice treated with scrRNA. These deficits were absent in shGD3S-treated mice. These results suggest that inhibition of GD3S protects against the nigrostriatal damage, bradykinesia, and fine-motor-skill deficits associated with MPTP administration.


Asunto(s)
Actividad Motora , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/terapia , Sialiltransferasas/genética , Animales , Cuerpo Estriado/enzimología , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/enzimología , Técnicas de Silenciamiento del Gen/métodos , Vectores Genéticos/fisiología , Lentivirus/fisiología , Masculino , Ratones Endogámicos C57BL , Trastornos Parkinsonianos/fisiopatología , Sialiltransferasas/metabolismo , Sustancia Negra/enzimología , Sustancia Negra/patología
8.
Environ Pollut ; 241: 279-288, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29843010

RESUMEN

Environmental exposure to air pollution has been linked to a number of health problems including organ rejection, lung damage and inflammation. While the deleterious effects of air pollution in adult animals are well documented, the long-term consequences of particulate matter (PM) exposure during animal development are uncertain. In this study we tested the hypothesis that environmental exposure to PM 2.5 µm in diameter in utero promotes long term inflammation and neurodegeneration. We evaluated the behavior of PM exposed animals using several tests and observed deficits in spatial memory without robust changes in anxiety-like behavior. We then examined how this affects the brains of adult animals by examining proteins implicated in neurodegeneration, synapse formation and inflammation by western blot, ELISA and immunohistochemistry. These tests revealed significantly increased levels of COX2 protein in PM2.5 exposed animal brains in addition to changes in synaptophysin and Arg1 proteins. Exposure to PM2.5 also increased the immunoreactivity for GFAP, a marker of activated astrocytes. Cytokine concentrations in the brain and spleen were also altered by PM2.5 exposure. These findings indicate that in utero exposure to particulate matter has long term consequences which may affect the development of both the brain and the immune system in addition to promoting inflammatory change in adult animals.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Sistema Nervioso/inmunología , Material Particulado/toxicidad , Pruebas de Toxicidad , Adulto , Contaminantes Atmosféricos/análisis , Contaminación del Aire/análisis , Animales , Ansiedad/inducido químicamente , Conducta Animal/efectos de los fármacos , Biomarcadores/análisis , Encéfalo/efectos de los fármacos , Exposición a Riesgos Ambientales/análisis , Humanos , Masculino , Ratones , Material Particulado/análisis , Fenotipo
9.
Behav Brain Res ; 298(Pt B): 192-201, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26393431

RESUMEN

In Parkinson's disease, cognitive deficits manifest as fronto-striatally-mediated executive dysfunction, with impaired attention, planning, judgment, and impulse control. We examined changes in executive function in mice lesioned with subchronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) using a 3-choice serial reaction-time (SRT) task, which included measures of sustained attention and impulse control. Each trial of the baseline SRT task comprised a pseudo-random pre-cue period ranging from 3 to 8 s, followed by a 1-s cue duration. MPTP impaired all measures of impulsive behavior acutely, but with additional training their performance normalized to saline control levels. When challenged with shorter cue durations, MPTP-lesioned mice had significantly slower reaction times than wild-type mice. When challenged with longer pre-cue times, the MPTP-lesioned mice exhibited a loss of impulse control at the longer durations. In lesioned mice, striatal dopamine was depleted by 54% and the number of tyrosine-hydroxylase-positive neurons in the substantia nigra pars compacta was reduced by 75%. Serotonin (5-HT) was unchanged in the striatum and prefrontal cortex (PFC), but the ratio of 5-hydroxyindolacetic acid (5-HIAA) to 5-HT was significantly reduced in the MPTP group in the PFC. In lesioned mice, prefrontal 5-HIAA/5-HT was significantly correlated with the executive impairments and striatal norepinephrine was associated with slower reaction times. None of the neurochemical measures was significantly associated with behavior in saline-treated controls. Taken together, these results show that prefrontal 5-HT turnover may play a pivotal role in MPTP-induced executive dysfunction.


Asunto(s)
Función Ejecutiva/fisiología , Intoxicación por MPTP/fisiopatología , Intoxicación por MPTP/psicología , Corteza Prefrontal/fisiopatología , Serotonina/metabolismo , Animales , Atención/fisiología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Ácido Hidroxiindolacético/metabolismo , Conducta Impulsiva/fisiología , Masculino , Ratones Endogámicos C57BL , Neuronas/fisiología , Pruebas Neuropsicológicas , Porción Compacta de la Sustancia Negra/fisiopatología , Tiempo de Reacción/fisiología , Tirosina 3-Monooxigenasa/metabolismo
10.
J Psychoactive Drugs ; 47(4): 267-75, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26266886

RESUMEN

Dextromethorphan (DXM) is a widely available antitussive that has, at elevated dose levels, euphoric and dissociative effects. This article presents the reported patterns and preferences of DXM use, and perceptions of DXM use among adult members of an online DXM community. Analyses were conducted of quantitative and qualitative responses from nine female and 43 male individuals, aged 18-63 years old. All respondents reported illegal and DXM drug use, beginning, on average, at 15.7 and 17.1 years of age, respectively. The majority of respondents first heard about DXM online or from a friend, preferred to use DXM alone, ingested substances concurrently with DXM to modify its effects, had not been to an emergency room or arrested because of their DXM use, and used DXM for its dissociative and mind-altering effects. DXM was perceived as safe and in no need of further regulation with only 14% of respondents mentioning DXM's addictive qualities. Findings from this sample of adult DXM users reveal a sophisticated subculture in which users report using DXM specifically to induce changes to their mental state and use a variety of substances to modify or enhance DXM's effects.


Asunto(s)
Antitusígenos , Dextrometorfano , Conocimientos, Actitudes y Práctica en Salud , Trastornos Relacionados con Sustancias , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Medios de Comunicación Sociales , Encuestas y Cuestionarios , Adulto Joven
11.
Mol Neurodegener ; 10: 24, 2015 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-26104027

RESUMEN

BACKGROUND: Cerebral infarction due to thrombosis leads to the most common type of stroke and a likely cause of age-related cognitive decline and dementia. Endothelial nitric oxide synthase (eNOS) generates NO, which plays a crucial role in maintaining vascular function and exerting an antithrombotic action. Reduced eNOS expression and eNOS polymorphisms have been associated with stroke and Alzheimer's disease (AD), the most common type of dementia associated with neurovascular dysfunction. However, direct proof of such association is lacking. Since there are no reports of complete eNOS deficiency in humans, we used heterozygous eNOS(+/-) mice to mimic partial deficiency of eNOS, and determine its impact on cerebrovascular pathology and perfusion of cerebral vessels. RESULTS: Combining cerebral angiography with immunohistochemistry, we found thrombotic cerebral infarctions in eNOS(+/-) mice as early as 3-6 months of age but not in eNOS(+/+) mice at any age. Remarkably, vascular occlusions in eNOS(+/-) mice were found almost exclusively in three areas: temporoparietal and retrosplenial granular cortexes, and hippocampus this distribution precisely matching the hypoperfused areas identified in preclinical AD patients. Moreover, progressive cerebral amyloid angiopaphy (CAA), blood brain barrier (BBB) breakdown, and cognitive impairment were also detected in aged eNOS(+/-) mice. CONCLUSIONS: These data provide for the first time the evidence that partial eNOS deficiency results in spontaneous thrombotic cerebral infarctions that increase with age, leading to progressive CAA and cognitive impairments. We thus conclude that eNOS(+/-) mouse may represent an ideal model of ischemic stroke to address early and progressive damage in spontaneously-evolving chronic cerebral ischemia and thus, study vascular mechanisms contributing to vascular dementia and AD.


Asunto(s)
Barrera Hematoencefálica/fisiología , Angiopatía Amiloide Cerebral/genética , Infarto Cerebral/genética , Trastornos del Conocimiento/genética , Óxido Nítrico Sintasa de Tipo III/fisiología , Envejecimiento , Animales , Angiopatía Amiloide Cerebral/patología , Infarto Cerebral/enzimología , Infarto Cerebral/patología , Infarto Cerebral/psicología , Circulación Cerebrovascular , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/patología , Progresión de la Enfermedad , Endotelio Vascular/fisiopatología , Regulación de la Expresión Génica , Heterocigoto , Hipocampo/irrigación sanguínea , Hipocampo/enzimología , Hipocampo/patología , Aprendizaje por Laberinto , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/genética , Trastornos de la Memoria/patología , Ratones , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Especificidad de Órganos , Lóbulo Parietal/irrigación sanguínea , Lóbulo Parietal/enzimología , Lóbulo Parietal/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Lóbulo Temporal/irrigación sanguínea , Lóbulo Temporal/enzimología , Lóbulo Temporal/patología
12.
Neurobiol Dis ; 78: 45-55, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25818006

RESUMEN

Physical exercise may provide protection against the cognitive decline and neuropathology associated with Alzheimer's disease, although the mechanisms are not clear. In the present study, APP/PSEN1 double-transgenic and wild-type mice were allowed unlimited voluntary exercise for 7months. Consistent with previous reports, wheel-running improved cognition in the double-transgenic mice. Interestingly, the average daily distance run was strongly correlated with spatial memory in the water maze in wild-type mice (r(2)=.959), but uncorrelated in transgenics (r(2)=.013). Proteomics analysis showed that sedentary transgenic mice differed significantly from sedentary wild-types with respect to proteins involved in synaptic transmission, cytoskeletal regulation, and neurogenesis. When given an opportunity to exercise, the transgenics' deficiencies in cytoskeletal regulation and neurogenesis largely normalized, but abnormal synaptic proteins did not change. In contrast, exercise enhanced proteins associated with cytoskeletal regulation, oxidative phosphorylation, and synaptic transmission in wild-type mice. Soluble and insoluble Aß40 and Aß42 levels were significantly decreased in both cortex and hippocampus of active transgenics, suggesting that this may have played a role in the cognitive improvement in APP/PSEN1 mice. ß-secretase was significantly reduced in active APP/PSEN1 mice compared to sedentary controls, suggesting a mechanism for reduced Aß. Taken together, these data illustrate that exercise improves memory in wild-type and APP-overexpressing mice in fundamentally different ways.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Actividad Motora , Proteómica , Memoria Espacial/fisiología , Enfermedad de Alzheimer/psicología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Conducta Animal , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Presenilina-1/genética , Presenilina-1/metabolismo
13.
Neurosci Res ; 96: 1-13, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25728560

RESUMEN

The structure of dendritic spines determines synaptic efficacy, a plastic process that mediates information processing in the vertebrate nervous system. Aberrant spine morphology, including alterations in shape, size, and number, are common in different brain diseases. Because of this, accurate and unbiased characterization of dendritic spine structure is vital to our ability to explore and understand their involvement in neuronal development, synaptic plasticity, and synaptic failure in neurological diseases. Investigators have attempted to elucidate the precise structure and function of dendritic spines for more than a hundred years, but their fundamental role in synaptic plasticity and neurological diseases remains elusive. Limitations and ambiguities in imaging techniques have exacerbated the challenges of acquiring accurate information about spines and spine features. However, recent advancements in molecular biology, protein engineering, immuno-labeling techniques, and the use of super-resolution nano-microscopy along with powerful image analysis software have provided a better understanding of dendritic spine architecture. Here we describe the pros and cons of the classical staining techniques used to study spine morphology, and the alteration of dendritic spines in various neuropathological conditions. Finally, we highlight recent advances in super-resolved nanoscale microscopy, and their potentials and pitfalls when used to explore dendritic spine dynamics.


Asunto(s)
Espinas Dendríticas/fisiología , Espinas Dendríticas/ultraestructura , Microscopía/métodos , Plasticidad Neuronal , Animales , Humanos , Inmunohistoquímica/métodos , Proteínas Luminiscentes , Ingeniería de Proteínas
14.
ASN Neuro ; 5(2): 141-8, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23565921

RESUMEN

In order to examine the potential involvement of gangliosides in AD (Alzheimer's disease), we compared the ganglioside compositions of the brains of a double-transgenic (Tg) mouse model [APP (amyloid precursor protein)/PSEN1 (presenilin)] of AD and a triple mutant mouse model with an additional deletion of the GD3S (GD3-synthase) gene (APP/PSEN1/GD3S(-/-)). These animals were chosen since it was previously reported that APP/PSEN1/GD3S(-/-) triple-mutant mice performed as well as WT (wild-type) control and GD3S(-/-) mice on a number of reference memory tasks. Cholinergic neuron-specific gangliosides, such as GT1aα and GQ1bα, were elevated in the brains of double-Tg mice (APP/PSEN1), as compared with those of WT mice. Remarkably, in the triple mutant mouse brains (APP/PSEN1/GD3S(-/-)), the concentration of GT1aα was elevated and as expected there was no expression of GQ1bα. On the other hand, the level of c-series gangliosides, including GT3, was significantly reduced in the double-Tg mouse brain as compared with the WT. Thus, the disruption of the gene of a specific ganglioside-synthase, GD3S, altered the expression of cholinergic neuron-specific gangliosides. Our data thus suggest the intriguing possibility that the elevated cholinergic-specific ganglioside, GT1aα, in the triple mutant mouse brains (APP/PSEN1/GD3S(-/-)) may contribute to the memory retention in these mice.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo/patología , Neuronas Colinérgicas/metabolismo , Gangliósidos/metabolismo , Regulación de la Expresión Génica/genética , Sialiltransferasas/deficiencia , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Antígenos de Superficie/metabolismo , Modelos Animales de Enfermedad , Humanos , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Presenilina-1/genética
15.
PLoS One ; 6(12): e29285, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22195039

RESUMEN

Converging evidence shows that GD3 ganglioside is a critical effector in a number of apoptotic pathways, and GM1 ganglioside has neuroprotective and noötropic properties. Targeted deletion of GD3 synthase (GD3S) eliminates GD3 and increases GM1 levels. Primary neurons from GD3S-/- mice are resistant to neurotoxicity induced by amyloid-ß or hyperhomocysteinemia, and when GD3S is eliminated in the APP/PSEN1 double-transgenic model of Alzheimer's disease the plaque-associated oxidative stress and inflammatory response are absent. To date, no small-molecule inhibitor of GD3S exists. In the present study we used sialidase from Vibrio cholerae (VCS) to produce a brain ganglioside profile that approximates that of GD3S deletion. VCS hydrolyzes GD1a and complex b-series gangliosides to GM1, and the apoptogenic GD3 is degraded. VCS was infused by osmotic minipump into the dorsal third ventricle in mice over a 4-week period. Sensorimotor behaviors, anxiety, and cognition were unaffected in VCS-treated mice. To determine whether VCS was neuroprotective in vivo, we injected kainic acid on the 25th day of infusion to induce status epilepticus. Kainic acid induced a robust lesion of the CA3 hippocampal subfield in aCSF-treated controls. In contrast, all hippocampal regions in VCS-treated mice were largely intact. VCS did not protect against seizures. These results demonstrate that strategic degradation of complex gangliosides and GD3 can be used to achieve neuroprotection without adversely affecting behavior.


Asunto(s)
Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/prevención & control , Neuraminidasa/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Cráneo/efectos de los fármacos , Cráneo/patología , Vibrio cholerae/enzimología , Animales , Ansiedad/complicaciones , Ansiedad/tratamiento farmacológico , Ansiedad/fisiopatología , Gangliósido G(M1)/análogos & derivados , Gangliósido G(M1)/metabolismo , Eliminación de Gen , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/patología , Inflamación/prevención & control , Ácido Kaínico , Memoria/efectos de los fármacos , Ratones , Actividad Motora/efectos de los fármacos , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/patología , Neuraminidasa/farmacología , Fármacos Neuroprotectores/farmacología , Neurotoxinas , Sialiltransferasas/metabolismo
16.
ASN Neuro ; 2(4): e00044, 2010 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-20930939

RESUMEN

The accumulation of Aß (amyloid ß-protein) is one of the major pathological hallmarks in AD (Alzheimer's disease). Gangliosides, sialic acid-containing glycosphingolipids enriched in the nervous system and frequently used as biomarkers associated with the biochemical pathology of neurological disorders, have been suggested to be involved in the initial aggregation of Aß. In the present study, we have examined ganglioside metabolism in the brain of a double-Tg (transgenic) mouse model of AD that co-expresses mouse/human chimaeric APP (amyloid precursor protein) with the Swedish mutation and human presenilin-1 with a deletion of exon 9. Although accumulation of Aß was confirmed in the double-Tg mouse brains and sera, no statistically significant change was detected in the concentration and composition of major ganglio-N-tetraosyl-series gangliosides in the double-Tg brain. Most interestingly, Chol-1α antigens (cholinergic neuron-specific gangliosides), such as GT1aα and GQ1bα, which are minor species in the brain, were found to be increased in the double-Tg mouse brain. We interpret that the occurrence of these gangliosides may represent evidence for generation of cholinergic neurons in the AD brain, as a result of compensatory neurogenesis activated by the presence of Aß.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Antígenos de Superficie/biosíntesis , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Gangliósidos/biosíntesis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Antígenos de Superficie/genética , Encéfalo/patología , Bovinos , Gangliósidos/genética , Humanos , Ratones , Ratones Transgénicos
17.
Neurobiol Aging ; 30(11): 1777-91, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18258340

RESUMEN

Gangliosides have been shown to be necessary for beta-amyloid (Abeta) binding and aggregation. GD3 synthase (GD3S) is responsible for biosynthesis of the b- and c-series gangliosides, including two of the four major brain gangliosides. We examined Abeta-ganglioside interactions in neural tissue from mice lacking the gene coding for GD3S (St8sia1), and in a double-transgenic (APP/PSEN1) mouse model of Alzheimer's disease cross-bred with GD3S-/- mice. In primary neurons and astrocytes lacking GD3S, Abeta-induced cell death and Abeta aggregation were inhibited. Like GD3S-/- and APP/PSEN1 double-transgenic mice, APP/PSEN1/GD3S-/- "triple-mutant" mice are indistinguishable from wild-type mice on casual examination. APP/PSEN1 double-transgenics exhibit robust impairments on a number of reference-memory tasks. In contrast, APP/PSEN1/GD3S-/- triple-mutant mice performed as well as wild-type control and GD3S-/- mice. Consistent with the behavioral improvements, both aggregated and unaggregated Abeta and associated neuropathology were almost completely eliminated in triple-mutant mice. These results suggest that GD3 synthase may be a novel therapeutic target to combat the cognitive deficits, amyloid plaque formation, and neurodegeneration that afflict Alzheimer's patients.


Asunto(s)
Amiloide/metabolismo , Memoria/fisiología , Placa Amiloide/patología , Sialiltransferasas/deficiencia , Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide/genética , Animales , Antígeno CD11b/metabolismo , Células Cultivadas , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Cromatografía de Gases y Espectrometría de Masas/métodos , Humanos , Peroxidación de Lípido/genética , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Ratones , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Placa Amiloide/genética , Presenilina-1/genética , Unión Proteica/genética
18.
J Neurosci ; 28(45): 11622-34, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-18987198

RESUMEN

Recent studies have revealed that disruption of vitamin A signaling observed in Alzheimer's disease (AD) leads to beta-amyloid (Abeta) accumulation and memory deficits in rodents. The aim of the present study was to evaluate the therapeutic effect of all-trans retinoic acid (ATRA), an active metabolite of vitamin A, on the neuropathology and deficits of spatial learning and memory in amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic mice, a well established AD mouse model. Here we report a robust decrease in brain Abeta deposition and tau phosphorylation in the blinded study of APP/PS1 transgenic mice treated intraperitoneally for 8 weeks with ATRA (20 mg/kg, three times weekly, initiated when the mice were 5 months old). This was accompanied by a significant decrease in the APP phosphorylation and processing. The activity of cyclin-dependent kinase 5, a major kinase involved in both APP and tau phosphorylation, was markedly downregulated by ATRA treatment. The ATRA-treated APP/PS1 mice showed decreased activation of microglia and astrocytes, attenuated neuronal degeneration, and improved spatial learning and memory compared with the vehicle-treated APP/PS1 mice. These results support ATRA as an effective therapeutic agent for the prevention and treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Antineoplásicos/uso terapéutico , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Tretinoina/uso terapéutico , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Astrocitos/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/patología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Presenilina-1/genética , Tiempo de Reacción/efectos de los fármacos
19.
J Neurochem ; 106(3): 1198-208, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18466336

RESUMEN

Oxidative stress is implicated in the cognitive deterioration associated with normal aging as well as neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. We investigated the effect of ascorbic acid (vitamin C) on oxidative stress, cognition, and motor abilities in mice null for gulono-gamma-lactone oxidase (Gulo). Gulo-/- mice are unable to synthesize ascorbic acid and depend on dietary ascorbic acid for survival. Gulo-/- mice were given supplements that provided them either with ascorbic acid levels equal to- or slightly higher than wild-type mice (Gulo-sufficient), or lower than physiological levels (Gulo-low) that were just enough to prevent scurvy. Ascorbic acid is a major anti-oxidant in mice and any reduction in ascorbic acid level is therefore likely to result in increased oxidative stress. Ascorbic acid levels in the brain and liver were higher in Gulo-sufficient mice than in Gulo-low mice. F(4)-neuroprostanes were elevated in cortex and cerebellum in Gulo-low mice and in the cortex of Gulo-sufficient mice. All Gulo-/- mice were cognitively normal but had a strength and agility deficit that was worse in Gulo-low mice. This suggests that low levels of ascorbic acid and elevated oxidative stress as measured by F(4)-neuroprostanes alone are insufficient to impair memory in the knockouts but may be responsible for the exacerbated motor deficits in Gulo-low mice, and ascorbic acid may have a vital role in maintaining motor abilities.


Asunto(s)
Deficiencia de Ácido Ascórbico/metabolismo , Cognición/fisiología , Trastornos de la Destreza Motora/metabolismo , Estrés Oxidativo/fisiología , Desempeño Psicomotor/fisiología , Animales , Ácido Ascórbico/biosíntesis , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Deficiencia de Ácido Ascórbico/tratamiento farmacológico , Deficiencia de Ácido Ascórbico/enzimología , Deficiencia de Ácido Ascórbico/genética , Cognición/efectos de los fármacos , Femenino , L-Gulonolactona Oxidasa/deficiencia , L-Gulonolactona Oxidasa/genética , L-Gulonolactona Oxidasa/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Actividad Motora/fisiología , Trastornos de la Destreza Motora/tratamiento farmacológico , Trastornos de la Destreza Motora/enzimología , Trastornos de la Destreza Motora/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Desempeño Psicomotor/efectos de los fármacos
20.
J Lipid Res ; 49(6): 1157-75, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18334715

RESUMEN

Gangliosides are expressed in the outer leaflet of the plasma membrane of the cells of all vertebrates and are particularly abundant in the nervous system. Ganglioside metabolism is closely associated with the pathology of Alzheimer's disease (AD). AD, the most common form of dementia, is a progressive degenerative disease of the brain characterized clinically by progressive loss of memory and cognitive function and eventually death. Neuropathologically, AD is characterized by amyloid deposits or "senile plaques," which consist mainly of aggregated variants of amyloid beta-protein (Abeta). Abeta undergoes a conformational transition from random coil to ordered structure rich in beta-sheets, especially after addition of lipid vesicles containing GM1 ganglioside. In AD brain, a complex of GM1 and Abeta, termed "GAbeta," has been found to accumulate. In recent years, Abeta and GM1 have been identified in microdomains or lipid rafts. The functional roles of these microdomains in cellular processes are now beginning to unfold. Several articles also have documented the involvement of these microdomains in the pathogenesis of certain neurodegenerative diseases, such as AD. A pivotal neuroprotective role of gangliosides has been reported in in vivo and in vitro models of neuronal injury, Parkinsonism, and related diseases. Here we describe the possible involvement of gangliosides in the development of AD and the therapeutic potentials of gangliosides in this disorder.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Gangliósidos/metabolismo , Conformación de Carbohidratos , Gangliósidos/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...