Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 19(1): 303, 2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36527099

RESUMEN

BACKGROUND: Considerable evidence indicates that a signaling crosstalk between the brain and periphery plays important roles in neurological disorders, and that both acute and chronic peripheral inflammation can produce brain changes leading to cognitive impairments. Recent clinical and epidemiological studies have revealed an increased risk of cognitive impairment and dementia in individuals with impaired pulmonary function. However, the mechanistic underpinnings of this association remain unknown. Exposure to SiO2 (silica) particles triggers lung inflammation, including infiltration by peripheral immune cells and upregulation of pro-inflammatory cytokines. We here utilized a mouse model of lung silicosis to investigate the crosstalk between lung inflammation and memory. METHODS: Silicosis was induced by intratracheal administration of a single dose of 2.5 mg SiO2/kg in mice. Molecular and behavioral measurements were conducted 24 h and 15 days after silica administration. Lung and hippocampal inflammation were investigated by histological analysis and by determination of pro-inflammatory cytokines. Hippocampal synapse damage, amyloid-ß (Aß) peptide content and phosphorylation of Akt, a proxy of hippocampal insulin signaling, were investigated by Western blotting and ELISA. Memory was assessed using the open field and novel object recognition tests. RESULTS: Administration of silica induced alveolar collapse, lung infiltration by polymorphonuclear (PMN) cells, and increased lung pro-inflammatory cytokines. Lung inflammation was followed by upregulation of hippocampal pro-inflammatory cytokines, synapse damage, accumulation of the Aß peptide, and memory impairment in mice. CONCLUSION: The current study identified a crosstalk between lung and brain inflammatory responses leading to hippocampal synapse damage and memory impairment after exposure to a single low dose of silica in mice.


Asunto(s)
Neumonía , Silicosis , Animales , Ratones , Dióxido de Silicio/toxicidad , Ratones Endogámicos C57BL , Silicosis/patología , Neumonía/inducido químicamente , Neumonía/patología , Inflamación/inducido químicamente , Inflamación/patología , Pulmón/patología , Sinapsis/patología , Péptidos beta-Amiloides , Hipocampo/patología , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/patología , Citocinas
3.
J Alzheimers Dis ; 82(3): 1067-1074, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34151795

RESUMEN

BACKGROUND: Alzheimer's disease (AD) and Lewy body disease (LBD) are complex neurodegenerative disorders that have been associated with brain inflammation and impaired neurotransmission. OBJECTIVE: We aimed to determine concentrations of multiple cytokines, chemokines, and neurotransmitters previously associated with brain inflammation and synapse function in cerebrospinal fluid (CSF) from AD and LBD patients. METHODS: We examined a panel of 50 analytes comprising neurotransmitters, cytokines, chemokines, and hormones in CSF in a cohort of patients diagnosed with mild cognitive impairment (MCI), AD, LBD, or non-demented controls (NDC). RESULTS: Among neurotransmitters, noradrenaline (NA) was increased in AD CSF, while homovanillic acid (HVA), a dopamine metabolite, was reduced in both AD and LBD CSF relative to NDC. Six cytokines/chemokines out of 30 investigated were reliably detected in CSF. CSF vascular endothelial growth factor (VEGF) was significantly reduced in LBD patients relative to NDC. CONCLUSIONS: CSF alterations in NA, HVA, and VEGF in AD and LBD may reflect pathogenic features of these disorders and provide tools for improved diagnosis. Future studies are warranted to replicate current findings in larger, multicenter cohorts.

4.
Cell Rep ; 30(7): 2180-2194.e8, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32075735

RESUMEN

Obesity has been associated with cognitive decline, atrophy of brain regions related to learning and memory, and higher risk of developing dementia. However, the molecular mechanisms underlying these neurological alterations are still largely unknown. Here, we investigate the effects of palmitate, a saturated fatty acid present at high amounts in fat-rich diets, in the brain. Palmitate is increased in the cerebrospinal fluid (CSF) of overweight and obese patients with amnestic mild cognitive impairment. In mice, intracerebroventricular infusion of palmitate impairs synaptic plasticity and memory. Palmitate induces astroglial and microglial activation in the mouse hippocampus, and its deleterious impact is mediated by microglia-derived tumor necrosis factor alpha (TNF-α) signaling. Our results establish that obesity is associated with increases in CSF palmitate. By defining a pro-inflammatory mechanism by which abnormal levels of palmitate in the brain impair memory, the results further suggest that anti-inflammatory strategies may attenuate memory impairment in obesity.


Asunto(s)
Trastornos de la Memoria/etiología , Obesidad/líquido cefalorraquídeo , Palmitatos/líquido cefalorraquídeo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Humanos , Trastornos de la Memoria/patología , Ratones , Obesidad/patología
5.
J Alzheimers Dis ; 73(2): 585-596, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31815695

RESUMEN

Familial hypercholesterolemia (FH) is a genetic disorder caused by dysfunction of low density lipoprotein receptors (LDLr), resulting in elevated plasma cholesterol levels. FH patients frequently exhibit cognitive impairment, a finding recapitulated in LDLr deficient mice (LDLr-/-), an animal model of FH. In addition, LDLr-/- mice are more vulnerable to the deleterious memory impact of amyloid-ß (Aß), a peptide linked to Alzheimer's disease. Here, we investigated whether the expression of proteins involved in Aß metabolism are altered in the brains of adult or middle-aged LDLr-/- mice. After spatial memory assessment, Aß levels and gene expression of LDLr related-protein 1, proteins involved in Aß synthesis, and apoptosis-related proteins were evaluated in prefrontal cortex and hippocampus. Moreover, the location and cell-specificity of apoptosis signals were evaluated. LDLr-/- mice presented memory impairment, which was more severe in middle-aged animals. Memory deficit in LDLr-/- mice was not associated with altered expression of proteins involved in Aß processing or changes in Aß levels in either hippocampus or prefrontal cortex. We further found that the expression of Bcl-2 was reduced while the expression of Bax was increased in both prefrontal cortex and hippocampus in 3- and 14-month-old LDLr-/-mice Finally, LDLr-/- mice presented increased immunoreactivity for activated caspase-3 in the prefrontal cortex and hippocampus. The activation of caspase 3 was predominantly associated with neurons in LDLr-/- mice. Cognitive impairment in LDLr-/- mice is thus accompanied by an exacerbation of neuronal apoptosis in brain regions related to memory formation, but not by changes in Aß processing or levels.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Apoptosis/genética , Química Encefálica/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética , Envejecimiento/metabolismo , Envejecimiento/psicología , Animales , Caspasa 3 , Colesterol/sangre , Expresión Génica , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Corteza Prefrontal/metabolismo
6.
Front Neurosci ; 13: 265, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30983955

RESUMEN

Western societies experienced drastic changes in eating habits during the past century. The modern nutritional profile, typically rich in saturated fats and refined sugars, is recognized as a major contributing factor, along with reduced physical activity, to the current epidemics of metabolic disorders, notably obesity and diabetes. Alongside these conditions, recent years have witnessed a gradual and significant increase in prevalence of brain diseases, particularly mood disorders. While substantial clinical/epidemiological evidence supports a correlation between metabolic and neuropsychiatric disorders, the mechanisms of pathogenesis in the latter are often multifactorial and causal links have been hard to establish. Neuroinflammation stands out as a hallmark feature of brain disorders that may be linked to peripheral metabolic dyshomeostasis caused by an unhealthy diet. Dietary fatty acids are of particular interest, as they may play a dual role, both as a component of high-calorie obesogenic diets and as signaling molecules involved in inflammatory responses. Here, we review current literature connecting diet-related nutritional imbalance and neuropsychiatric disorders, focusing on the role of dietary fatty acids as signaling molecules directly relevant to inflammatory processes and to neuronal function.

7.
Elife ; 82019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30694175

RESUMEN

Leptin regulates energy balance and also exhibits neurotrophic effects during critical developmental periods. However, the actual role of leptin during development is not yet fully understood. To uncover the importance of leptin in early life, the present study restored leptin signaling either at the fourth or tenth week of age in mice formerly null for the leptin receptor (LepR) gene. We found that some defects previously considered irreversible due to neonatal deficiency of leptin signaling, including the poor development of arcuate nucleus neural projections, were recovered by LepR reactivation in adulthood. However, LepR deficiency in early life led to irreversible obesity via suppression of energy expenditure. LepR reactivation in adulthood also led to persistent reduction in hypothalamic Pomc, Cartpt and Prlh mRNA expression and to defects in the reproductive system and brain growth. Our findings revealed that early defects in leptin signaling cause permanent metabolic, neuroendocrine and developmental problems.


Asunto(s)
Envejecimiento/genética , Regulación del Desarrollo de la Expresión Génica , Leptina/genética , Obesidad/genética , Receptores de Leptina/genética , Envejecimiento/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/patología , Metabolismo Energético/genética , Femenino , Gónadas/crecimiento & desarrollo , Gónadas/metabolismo , Gónadas/patología , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Hipotálamo/patología , Leptina/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Obesidad/metabolismo , Obesidad/patología , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Hormona Liberadora de Prolactina/genética , Hormona Liberadora de Prolactina/metabolismo , Receptores de Leptina/deficiencia , Transducción de Señal
9.
Brain Struct Funct ; 223(5): 2229-2241, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29460051

RESUMEN

The signal transducer and activator of transcription 5 (STAT5) is a transcription factor recruited by numerous cytokines. STAT5 is important for several physiological functions, including body and tissue growth, mammary gland development, immune system and lipid metabolism. However, the role of STAT5 signaling for brain functions is still poorly investigated, especially regarding cognitive aspects. Therefore, the objective of the present study was to investigate whether brain STAT5 signaling modulates learning and memory formation. For this purpose, brain-specific STAT5 knockout (STAT5 KO) mice were studied in well-established memory tests. Initially, we confirmed a robust reduction in STAT5a and STAT5b mRNA levels in different brain structures of STAT5 KO mice. STAT5 KO mice showed no significant alterations in metabolism, growth, somatotropic axis and spontaneous locomotor activity. In contrast, brain-specific STAT5 ablation impaired learning and memory formation in the novel object recognition, Barnes maze and contextual fear conditioning tests. To unravel possible mechanisms that might underlie the memory deficits of STAT5 KO mice, we assessed neurogenesis in the hippocampus, but no significant differences were observed between groups. On the other hand, reduced insulin-like growth factor-1 (IGF-1) mRNA expression was found in the hippocampus and hypothalamus of STAT5 KO mice. These findings collectively indicate that brain STAT5 signaling is required to attain normal learning and memory. Therefore, STAT5 is an important downstream cellular mechanism shared by several cytokines to regulate cognitive functions.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica/genética , Aprendizaje por Laberinto/fisiología , Reconocimiento en Psicología/fisiología , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/fisiología , Animales , Condicionamiento Psicológico , Citocinas/metabolismo , Conducta Exploratoria/fisiología , Miedo/psicología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Discapacidades para el Aprendizaje/genética , Ratones , Ratones Transgénicos , Nestina/genética , Nestina/metabolismo , Neurogénesis/genética , ARN Mensajero/metabolismo , Tiempo de Reacción/genética , Factor de Transcripción STAT5/genética
10.
J Neurosci ; 37(28): 6797-6809, 2017 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-28607171

RESUMEN

Alzheimer's disease (AD) is characterized by progressive cognitive decline, increasingly attributed to neuronal dysfunction induced by amyloid-ß oligomers (AßOs). Although the impact of AßOs on neurons has been extensively studied, only recently have the possible effects of AßOs on astrocytes begun to be investigated. Given the key roles of astrocytes in synapse formation, plasticity, and function, we sought to investigate the impact of AßOs on astrocytes, and to determine whether this impact is related to the deleterious actions of AßOs on synapses. We found that AßOs interact with astrocytes, cause astrocyte activation and trigger abnormal generation of reactive oxygen species, which is accompanied by impairment of astrocyte neuroprotective potential in vitro We further show that both murine and human astrocyte conditioned media (CM) increase synapse density, reduce AßOs binding, and prevent AßO-induced synapse loss in cultured hippocampal neurons. Both a neutralizing anti-transforming growth factor-ß1 (TGF-ß1) antibody and siRNA-mediated knockdown of TGF-ß1, previously identified as an important synaptogenic factor secreted by astrocytes, abrogated the protective action of astrocyte CM against AßO-induced synapse loss. Notably, TGF-ß1 prevented hippocampal dendritic spine loss and memory impairment in mice that received an intracerebroventricular infusion of AßOs. Results suggest that astrocyte-derived TGF-ß1 is part of an endogenous mechanism that protects synapses against AßOs. By demonstrating that AßOs decrease astrocyte ability to protect synapses, our results unravel a new mechanism underlying the synaptotoxic action of AßOs in AD.SIGNIFICANCE STATEMENT Alzheimer's disease is characterized by progressive cognitive decline, mainly attributed to synaptotoxicity of the amyloid-ß oligomers (AßOs). Here, we investigated the impact of AßOs in astrocytes, a less known subject. We show that astrocytes prevent synapse loss induced by AßOs, via production of transforming growth factor-ß1 (TGF-ß1). We found that AßOs trigger morphological and functional alterations in astrocytes, and impair their neuroprotective potential. Notably, TGF-ß1 reduced hippocampal dendritic spine loss and memory impairment in mice that received intracerebroventricular infusions of AßOs. Our results describe a new mechanism underlying the toxicity of AßOs and indicate novel therapeutic targets for Alzheimer's disease, mainly focused on TGF-ß1 and astrocytes.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Factor de Crecimiento Transformador beta1/metabolismo , Péptidos beta-Amiloides , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Especies Reactivas de Oxígeno/metabolismo
11.
J Biol Chem ; 292(18): 7327-7337, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28283575

RESUMEN

Brain accumulation of the amyloid-ß protein (Aß) and synapse loss are neuropathological hallmarks of Alzheimer disease (AD). Aß oligomers (AßOs) are synaptotoxins that build up in the brains of patients and are thought to contribute to memory impairment in AD. Thus, identification of novel synaptic components that are targeted by AßOs may contribute to the elucidation of disease-relevant mechanisms. Trans-synaptic interactions between neurexins (Nrxs) and neuroligins (NLs) are essential for synapse structure, stability, and function, and reduced NL levels have been associated recently with AD. Here we investigated whether the interaction of AßOs with Nrxs or NLs mediates synapse damage and cognitive impairment in AD models. We found that AßOs interact with different isoforms of Nrx and NL, including Nrx2α and NL1. Anti-Nrx2α and anti-NL1 antibodies reduced AßO binding to hippocampal neurons and prevented AßO-induced neuronal oxidative stress and synapse loss. Anti-Nrx2α and anti-NL1 antibodies further blocked memory impairment induced by AßOs in mice. The results indicate that Nrx2α and NL1 are targets of AßOs and that prevention of this interaction reduces the deleterious impact of AßOs on synapses and cognition. Identification of Nrx2α and NL1 as synaptic components that interact with AßOs may pave the way for development of novel approaches aimed at halting synapse failure and cognitive loss in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Encéfalo/patología , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Fragmentos de Péptidos/genética , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Ratas , Ratas Wistar , Sinapsis/genética
12.
J Biol Chem ; 292(18): 7395-7406, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28302722

RESUMEN

AMP-activated kinase (AMPK) is a key player in energy sensing and metabolic reprogramming under cellular energy restriction. Several studies have linked impaired AMPK function to peripheral metabolic diseases such as diabetes. However, the impact of neurological disorders, such as Alzheimer disease (AD), on AMPK function and downstream effects of altered AMPK activity on neuronal metabolism have been investigated only recently. Here, we report the impact of Aß oligomers (AßOs), synaptotoxins that accumulate in AD brains, on neuronal AMPK activity. Short-term exposure of cultured rat hippocampal neurons or ex vivo human cortical slices to AßOs transiently decreased intracellular ATP levels and AMPK activity, as evaluated by its phosphorylation at threonine residue 172 (AMPK-Thr(P)172). The AßO-dependent reduction in AMPK-Thr(P)172 levels was mediated by glutamate receptors of the N-methyl-d-aspartate (NMDA) subtype and resulted in removal of glucose transporters (GLUTs) from the surfaces of dendritic processes in hippocampal neurons. Importantly, insulin prevented the AßO-induced inhibition of AMPK. Our results establish a novel toxic impact of AßOs on neuronal metabolism and suggest that AßO-induced, NMDA receptor-mediated AMPK inhibition may play a key role in early brain metabolic defects in AD.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Adenosina Trifosfato/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Hipocampo/patología , Humanos , Insulina/farmacología , Neuronas/patología , Fragmentos de Péptidos/genética , Ratas , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
13.
J Neurosci ; 36(48): 12106-12116, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27903721

RESUMEN

Considerable clinical and epidemiological evidence links Alzheimer's disease (AD) and depression. However, the molecular mechanisms underlying this connection are largely unknown. We reported recently that soluble Aß oligomers (AßOs), toxins that accumulate in AD brains and are thought to instigate synapse damage and memory loss, induce depressive-like behavior in mice. Here, we report that the mechanism underlying this action involves AßO-induced microglial activation, aberrant TNF-α signaling, and decreased brain serotonin levels. Inactivation or ablation of microglia blocked the increase in brain TNF-α and abolished depressive-like behavior induced by AßOs. Significantly, we identified serotonin as a negative regulator of microglial activation. Finally, AßOs failed to induce depressive-like behavior in Toll-like receptor 4-deficient mice and in mice harboring a nonfunctional TLR4 variant in myeloid cells. Results establish that AßOs trigger depressive-like behavior via a double impact on brain serotonin levels and microglial activation, unveiling a cross talk between brain innate immunity and serotonergic signaling as a key player in mood alterations in AD. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the main cause of dementia in the world. Brain accumulation of amyloid-ß oligomers (AßOs) is a major feature in the pathogenesis of AD. Although clinical and epidemiological data suggest a strong connection between AD and depression, the underlying mechanisms linking these two disorders remain largely unknown. Here, we report that aberrant activation of the brain innate immunity and decreased serotonergic tonus in the brain are key players in AßO-induced depressive-like behavior in mice. Our findings may open up new possibilities for the development of effective therapeutics for AD and depression aimed at modulating microglial function.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Encéfalo/inmunología , Depresión/inmunología , Inmunidad Innata/inmunología , Receptor Cross-Talk/inmunología , Serotonina/inmunología , Animales , Conducta Animal , Depresión/etiología , Masculino , Ratones , Ratones Endogámicos C3H , Microglía/inmunología , Factor de Necrosis Tumoral alfa/inmunología
14.
Glia ; 62(12): 1917-31, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25042347

RESUMEN

The balance between excitatory and inhibitory synaptic inputs is critical for the control of brain function. Astrocytes play important role in the development and maintenance of neuronal circuitry. Whereas astrocytes-derived molecules involved in excitatory synapses are recognized, molecules and molecular mechanisms underlying astrocyte-induced inhibitory synapses remain unknown. Here, we identified transforming growth factor beta 1 (TGF-ß1), derived from human and murine astrocytes, as regulator of inhibitory synapse in vitro and in vivo. Conditioned media derived from human and murine astrocytes induce inhibitory synapse formation in cerebral cortex neurons, an event inhibited by pharmacologic and genetic manipulation of the TGF-ß pathway. TGF-ß1-induction of inhibitory synapse depends on glutamatergic activity and activation of CaM kinase II, which thus induces localization and cluster formation of the synaptic adhesion protein, Neuroligin 2, in inhibitory postsynaptic terminals. Additionally, intraventricular injection of TGF-ß1 enhanced inhibitory synapse number in the cerebral cortex. Our results identify TGF-ß1/CaMKII pathway as a novel molecular mechanism underlying astrocyte control of inhibitory synapse formation. We propose here that the balance between excitatory and inhibitory inputs might be provided by astrocyte signals, at least partly achieved via TGF-ß1 downstream pathways. Our work contributes to the understanding of the GABAergic synapse formation and may be of relevance to further the current knowledge on the mechanisms underlying the development of various neurological disorders, which commonly involve impairment of inhibitory synapse transmission.


Asunto(s)
Astrocitos/química , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Medios de Cultivo Condicionados/farmacología , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Humanos , Inyecciones Intraventriculares , Masculino , Ratones , N-Metilaspartato/farmacología , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Neuronas/ultraestructura , Receptores de GABA-A/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
15.
J Neurosci ; 33(23): 9626-34, 2013 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23739959

RESUMEN

Brain accumulation of soluble amyloid-ß oligomers (AßOs) has been implicated in synapse failure and cognitive impairment in Alzheimer's disease (AD). However, whether and how oligomers of different sizes induce synapse dysfunction is a matter of controversy. Here, we report that low-molecular-weight (LMW) and high-molecular-weight (HMW) Aß oligomers differentially impact synapses and memory. A single intracerebroventricular injection of LMW AßOs (10 pmol) induced rapid and persistent cognitive impairment in mice. On the other hand, memory deficit induced by HMW AßOs (10 pmol) was found to be reversible. While memory impairment in LMW oligomer-injected mice was associated with decreased hippocampal synaptophysin and GluN2B immunoreactivities, synaptic pathology was not detected in the hippocampi of HMW oligomer-injected mice. On the other hand, HMW oligomers, but not LMW oligomers, induced oxidative stress in hippocampal neurons. Memantine rescued both neuronal oxidative stress and the transient memory impairment caused by HMW oligomers, but did not prevent the persistent cognitive deficit induced by LMW oligomers. Results establish that different Aß oligomer assemblies act in an orchestrated manner, inducing different pathologies and leading to synapse dysfunction. Furthermore, results suggest a mechanistic explanation for the limited efficacy of memantine in preventing memory loss in AD.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/tratamiento farmacológico , Memantina/farmacología , Fragmentos de Péptidos/farmacología , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Células Cultivadas , Trastornos del Conocimiento/metabolismo , Masculino , Ratones , Peso Molecular , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Ratas
16.
J Clin Invest ; 122(4): 1339-53, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22476196

RESUMEN

Defective brain insulin signaling has been suggested to contribute to the cognitive deficits in patients with Alzheimer's disease (AD). Although a connection between AD and diabetes has been suggested, a major unknown is the mechanism(s) by which insulin resistance in the brain arises in individuals with AD. Here, we show that serine phosphorylation of IRS-1 (IRS-1pSer) is common to both diseases. Brain tissue from humans with AD had elevated levels of IRS-1pSer and activated JNK, analogous to what occurs in peripheral tissue in patients with diabetes. We found that amyloid-ß peptide (Aß) oligomers, synaptotoxins that accumulate in the brains of AD patients, activated the JNK/TNF-α pathway, induced IRS-1 phosphorylation at multiple serine residues, and inhibited physiological IRS-1pTyr in mature cultured hippocampal neurons. Impaired IRS-1 signaling was also present in the hippocampi of Tg mice with a brain condition that models AD. Importantly, intracerebroventricular injection of Aß oligomers triggered hippocampal IRS-1pSer and JNK activation in cynomolgus monkeys. The oligomer-induced neuronal pathologies observed in vitro, including impaired axonal transport, were prevented by exposure to exendin-4 (exenatide), an anti-diabetes agent. In Tg mice, exendin-4 decreased levels of hippocampal IRS-1pSer and activated JNK and improved behavioral measures of cognition. By establishing molecular links between the dysregulated insulin signaling in AD and diabetes, our results open avenues for the investigation of new therapeutics in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/toxicidad , Hipocampo/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Insulina/fisiología , Péptidos/uso terapéutico , Ponzoñas/uso terapéutico , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/psicología , Animales , Anticuerpos Monoclonales/farmacología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Exenatida , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Hipoglucemiantes/farmacología , Infliximab , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macaca fascicularis , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Péptidos/farmacología , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Ponzoñas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA