Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Clin Exp Pharmacol Physiol ; 50(3): 228-237, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36398458

RESUMEN

Metabolic syndrome (MetS) is a rapidly increasing health concern during midlife and is an emerging risk factor for the development of neurodegenerative diseases, such as Alzheimer's disease (AD). While angiotensin receptor blockers (ARB) are widely used for MetS-associated hypertension and kidney disease, its therapeutic potential in the brain during MetS are not well-described. Here, we tested whether treatment with ARB could alleviate the brain pathology and inflammation associated with MetS using the Otsuka Long-Evans Tokushima Fatty (OLETF) rat. Here, we report that chronic ARB treatment with olmesartan (10 mg/kg/day by oral gavage for 6 weeks) partially but significantly ameliorated accumulation of oxidized and ubiquitinated proteins, astrogliosis and transformation to neurotoxic astrocytes in the brain of old OLETF rats, which otherwise exhibit the progression of these pathological hallmarks associated with MetS. Additionally, olmesartan treatment restored claudin-5 and ZO-1, markers of the structural integrity of the blood-brain barrier as well as synaptic protein PSD-95, which were otherwise decreased in old OLETF rats, particularly in the hippocampus, a critical region in cognition, memory and AD. These data demonstrate that the progression of MetS in OLETF rats is associated with deterioration of various aspects of neuronal integrity that may manifest neurodegenerative conditions and that overactivation of angiotensin receptor directly or indirectly contributes to these detriments. Thus, olmesartan treatment may slow or delay the onset of degenerative process in the brain and subsequent neurological disorders associated with MetS.


Asunto(s)
Diabetes Mellitus Tipo 2 , Síndrome Metabólico , Ratas , Animales , Ratas Endogámicas OLETF , Antagonistas de Receptores de Angiotensina , Receptores de Angiotensina , Inhibidores de la Enzima Convertidora de Angiotensina , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ratas Long-Evans , Síndrome Metabólico/metabolismo , Encéfalo/metabolismo , Glucemia/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(42): 21198-21206, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570577

RESUMEN

Defects in interleukin-1ß (IL-1ß)-mediated cellular responses contribute to Alzheimer's disease (AD). To decipher the mechanism associated with its pathogenesis, we investigated the molecular events associated with the termination of IL-1ß inflammatory responses by focusing on the role played by the target of Myb1 (TOM1), a negative regulator of the interleukin-1ß receptor-1 (IL-1R1). We first show that TOM1 steady-state levels are reduced in human AD hippocampi and in the brain of an AD mouse model versus respective controls. Experimentally reducing TOM1 affected microglia activity, substantially increased amyloid-beta levels, and impaired cognition, whereas enhancing its levels was therapeutic. These data show that reparation of the TOM1-signaling pathway represents a therapeutic target for brain inflammatory disorders such as AD. A better understanding of the age-related changes in the immune system will allow us to craft therapies to limit detrimental aspects of inflammation, with the broader purpose of sharply reducing the number of people afflicted by AD.

3.
Glia ; 66(12): 2700-2718, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30277607

RESUMEN

Emerging evidence have posited that dysregulated microglia impair clearance and containment of amyloid-ß (Aß) species in the brain, resulting in aberrant buildup of Aß and onset of Alzheimer's disease (AD). Hematopoietic cell kinase (Hck) is one of the key regulators of phagocytosis among the Src family tyrosine kinases (SFKs) in myeloid cells, and its expression is found to be significantly altered in AD brains. However, the role of Hck signaling in AD pathogenesis is unknown. We employed pharmacological inhibition and genetic ablation of Hck in BV2 microglial cells and J20 mouse model of AD, respectively, to evaluate the impact of Hck deficiency on Aß-stimulated microglial phagocytosis, Aß clearance, and resultant AD-like neuropathology. Our in vitro data reveal that pharmacological inhibition of SFKs/Hck in BV2 cells and genetic ablation of their downstream kinase, spleen tyrosine kinase (Syk), in primary microglia significantly attenuate Aß oligomers-stimulated microglial phagocytosis. Whereas in Hck-deficient J20 mice, we observed exacerbated Aß plaque burden, reduced microglial coverage, containment, and phagocytosis of Aß plaques, and induced iNOS expression in plaque-associated microglial clusters. These multifactorial changes in microglial activities led to attenuated PSD95 levels in hippocampal DG and CA3 regions, but did not alter the postsynaptic dendritic spine morphology at the CA1 region nor cognitive function of the mice. Hck inhibition thus accelerates early stage AD-like neuropathology by dysregulating microglial function and inducing neuroinflammation. Our data implicate that Hck pathway plays a prominent role in regulating microglial neuroprotective function during the early stage of AD development.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Regulación de la Expresión Génica/genética , Microglía/enzimología , Proteínas Proto-Oncogénicas c-hck/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetulus , Modelos Animales de Enfermedad , Antagonistas de Estrógenos/farmacología , Conducta Exploratoria/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/ultraestructura , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Proteínas Proto-Oncogénicas c-hck/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Quinasa Syk/genética , Quinasa Syk/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Transfección
4.
Am J Pathol ; 186(6): 1623-34, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27106764

RESUMEN

Valosin-containing protein (VCP) mutations cause inclusion body myopathy with Paget disease and frontotemporal dementia. However, the mechanisms by which mutant VCP triggers degeneration remain unknown. Here, we investigated the role of VCP in cellular stress and found that the oxidative stressor arsenite and heat shock-activated stress responses evident by T-intracellular antigen-1-positive granules in C2C12 myoblasts. Granules also contained phosphorylated transactive response DNA-binding protein 43, ubiquitin, microtubule-associated protein 1A/1B light chains 3, and lysosome-associated membrane protein 2. Mutant VCP produced more T-intracellular antigen-1-positive granules than wild-type in the postarsenite exposure period. Similar results were observed for other granule components, indicating that mutant VCP delayed clearance of stress granules. Furthermore, stress granule resolution was impaired on differentiated C2C12 cells expressing mutant VCP. To address whether mutant VCP triggers dysregulation of the stress granule pathway in vivo, we analyzed skeletal muscle of aged VCPR155H-knockin mice. We found significant increments in oxidated proteins but observed the stress granule markers RasGAP SH3-binding protein and phosphorylated eukaryotic translation initiation factor 2α unchanged. The mixed results indicate that mutant VCP together with aging lead to higher oxidative stress in skeletal muscle but were insufficient to disrupt the stress granule pathway. Our findings support that deficiencies in recovery from stressors may result in attenuated tolerance to stress that could trigger muscle degeneration.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Ciclo Celular/genética , Demencia Frontotemporal/patología , Distrofia Muscular de Cinturas/patología , Mioblastos/patología , Miositis por Cuerpos de Inclusión/patología , Osteítis Deformante/patología , Estrés Oxidativo/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Demencia Frontotemporal/genética , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Distrofia Muscular de Cinturas/genética , Mioblastos/metabolismo , Miositis por Cuerpos de Inclusión/genética , Osteítis Deformante/genética , Transfección , Proteína que Contiene Valosina
5.
Neurobiol Learn Mem ; 142(Pt A): 4-12, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28011191

RESUMEN

Long-term memories can undergo destabilization/restabilization processes, collectively called reconsolidation. However, the parameters that trigger memory reconsolidation are poorly understood and are a matter of intense investigation. Particularly, memory retrieval is widely held as requisite to initiate reconsolidation. This assumption makes sense since only relevant cues will induce reconsolidation of a specific memory. However, recent studies show that pharmacological inhibition of retrieval does not avoid memory from undergoing reconsolidation, indicating that memory reconsolidation occurs through a process that can be dissociated from retrieval. We propose that retrieval is not a unitary process but has two dissociable components; one leading to the expression of memory and the other to reconsolidation, referred herein as executer and integrator respectively. The executer would lead to the behavioral expression of the memory. This component would be the one disrupted on the studies that show reconsolidation independence from retrieval. The integrator would deal with reconsolidation. This component of retrieval would lead to long-term memory destabilization when specific conditions are met. We think that an important number of reports are consistent with the hypothesis that reconsolidation is only initiated when updating information is acquired. We suggest that the integrator would initiate reconsolidation to integrate updating information into long-term memory.


Asunto(s)
Aprendizaje por Asociación/fisiología , Consolidación de la Memoria/fisiología , Memoria/fisiología , Animales , Señales (Psicología) , Memoria a Largo Plazo/fisiología
6.
J Neurochem ; 134(5): 915-26, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26077803

RESUMEN

Alzheimer's disease (AD) is a progressive neurological disorder that impairs memory and other cognitive functions in the elderly. The social and financial impacts of AD are overwhelming and are escalating exponentially as a result of population aging. Therefore, identifying AD-related risk factors and the development of more efficacious therapeutic approaches are critical to cure this neurological disorder. Current epidemiological evidence indicates that life experiences, including chronic stress, are a risk for AD. However, it is unknown if short-term stress, lasting for hours, influences the onset or progression of AD. Here, we determined the effect of short-term, multi-modal 'modern life-like' stress on AD pathogenesis and synaptic plasticity in mice bearing three AD mutations (the 3xTg-AD mouse model). We found that combined emotional and physical stress lasting 5 h severely impaired memory in wild-type mice and tended to impact it in already low-performing 3xTg-AD mice. This stress reduced the number of synapse-bearing dendritic spines in 3xTg-AD mice and increased Aß levels by augmenting AßPP processing. Thus, short-term stress simulating modern-life conditions may exacerbate cognitive deficits in preclinical AD by accelerating amyloid pathology and reducing synapse numbers. Epidemiological evidence indicates that life experiences, including chronic stress, are a risk for Alzheimer disease (AD). However, it is unknown if short stress in the range of hours influences the onset or progression of AD. Here, we determined the effect of short, multi-modal 'modern-lifelike'stress on AD pathogenesis and synaptic plasticity in mice bearing three AD mutations (the 3xTg-AD mouse model). We found that combined emotional and physical stress lasting 5 h severely impaired memory in wild-type mice and tended to impact it in already low-performing 3xTg-AD mice. This stress reduced the number of synapse-bearing dendritic spines in 3xTg-AD mice and increased Aß levels by augmenting AßPP processing. Thus, short stress simulating modern-life conditions may exacerbate cognitive deficits in preclinical AD by accelerating amyloid pathology and reducing synapse numbers.


Asunto(s)
Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Ruido/efectos adversos , Estrés Psicológico/complicaciones , Vibración/efectos adversos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Células Cultivadas , Corticosterona/sangre , Hormona Liberadora de Corticotropina/fisiología , Dendritas/metabolismo , Dendritas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Emociones , Conducta Exploratoria , Glucocorticoides/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal , Reconocimiento en Psicología , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Sinapsis/patología , Proteínas tau/genética
7.
Am J Pathol ; 184(3): 819-26, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24412516

RESUMEN

Patients affected by diabetes show an increased risk of developing Alzheimer disease (AD). Similarly, patients with AD show impaired insulin function and glucose metabolism. However, the underlying molecular mechanisms connecting these two disorders are still not well understood. Herein, we investigated the microtubule-associated protein tau as a new link between AD and diabetes. To determine whether diabetes causes cognitive decline by a tau-dependent mechanism, we treated non-transgenic (Ntg) and tau-knockout mice with streptozotocin, causing type 1 diabetes-like disease (T1D). Interestingly, although induction of T1D in Ntg mice led to cellular and behavioral deficits, it did not do so in tau-knockout mice. Thus, data suggest that tau is a fundamental mediator of the induction of cognitive impairments in T1D. Tau dysregulation, which causes a reduction in synaptic protein levels, may be responsible for the cognitive decline observed in Ntg streptozotocin-treated mice. Concomitantly, we demonstrate the novel finding that depletion of endogenous tau mitigates behavioral impairment and synaptic deficits induced in T1D-like mice. Overall, our data reveal that tau is a key molecular factor responsible for the induction of cognitive deficits observed in T1D and represents a potential therapeutic target for diabetes and patients with AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Trastornos del Conocimiento/etiología , Diabetes Mellitus Tipo 1/complicaciones , Insulina/metabolismo , Proteínas tau/metabolismo , Animales , Cognición , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Estreptozocina/metabolismo , Proteínas tau/genética
8.
Learn Mem ; 21(9): 452-6, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25128536

RESUMEN

Memory retrieval has been considered as requisite to initiate memory reconsolidation; however, some studies indicate that blocking retrieval does not prevent memory from undergoing reconsolidation. Since N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors in the perirhinal cortex have been involved in object recognition memory formation, the present study evaluated whether retrieval and reconsolidation are independent processes by manipulating these glutamate receptors. The results showed that AMPA receptor antagonist infusions in the perirhinal cortex blocked retrieval, but did not affect memory reconsolidation, although NMDA receptor antagonist infusions disrupted reconsolidation even if retrieval was blocked. Importantly, neither of these antagonists disrupted short-term memory. These data suggest that memory underwent reconsolidation even in the absence of retrieval.


Asunto(s)
Recuerdo Mental/fisiología , Reconocimiento en Psicología/fisiología , Lóbulo Temporal/fisiología , 2-Amino-5-fosfonovalerato/farmacología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Animales , Infusiones Intraventriculares , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Memoria a Largo Plazo/fisiología , Recuerdo Mental/efectos de los fármacos , Ratas Wistar , Receptores AMPA/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Reconocimiento en Psicología/efectos de los fármacos , Lóbulo Temporal/efectos de los fármacos
9.
Neurobiol Dis ; 62: 407-15, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24176788

RESUMEN

Recent studies on tauopathy animal models suggest that the concomitant expression of the endogenous murine tau delays the pathological accumulation of human tau, and interferes with the disease progression. To elucidate the role of endogenous murine tau in a model with both plaques and tangles, we developed a novel transgenic mouse model by crossing 3xTg-AD with mtauKO mice (referred to as 3xTg-AD/mtauKO mice). Therefore, this new model allows us to determine the pathological consequences of the murine tau. Here, we show that 3xTg-AD/mtauKO mice have lower tau loads in both soluble and insoluble fractions, and lower tau hyperphosphorylation level in the soluble fraction relative to 3xTg-AD mice. In the 3xTg-AD model endogenous mouse tau is hyperphosphorylated and significantly co-aggregates with human tau. Despite the deletion of the endogenous tau gene in 3xTg-AD/mtauKO mice, cognitive dysfunction was equivalent to 3xTg-AD mice, as there was no additional impairment on a spatial memory task, and thus despite increased tau phosphorylation, accumulation and NFTs in 3xTg-AD mice no further effects on cognition are seen. These findings provide better understanding about the role of endogenous tau to Alzheimer's disease (AD) pathology and for developing new AD models.


Asunto(s)
Cognición/fisiología , Ovillos Neurofibrilares/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Femenino , Hipocampo/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas tau/genética
10.
Am J Pathol ; 183(2): 504-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747512

RESUMEN

Mutations in valosin-containing protein (VCP) cause a rare, autosomal dominant disease called inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia (IBMPFD). One-third of patients with IBMPFD develop frontotemporal dementia, characterized by an extensive neurodegeneration in the frontal and temporal lobes. Neuropathologic hallmarks include nuclear and cytosolic inclusions positive to ubiquitin and transactive response DNA-binding protein 43 (TDP-43) in neurons and glial activation in affected regions. However, the pathogenic mechanisms by which mutant VCP triggers neurodegeneration remain unknown. Herein, we generated a mouse model selectively overexpressing a human mutant VCP in neurons to study pathogenic mechanisms of mutant VCP-mediated neurodegeneration and cognitive impairment. The overexpression of VCPA232E mutation in forebrain regions produced significant progressive impairments of cognitive function, including deficits in spatial memory, object recognition, and fear conditioning. Although overexpressed or endogenous VCP did not seem to focally aggregate inside neurons, TDP-43 and ubiquitin accumulated with age in transgenic mouse brains. TDP-43 was also found to co-localize with stress granules in the cytosolic compartment. Together with the appearance of high-molecular-weight TDP-43 in cytosolic fractions, these findings demonstrate the mislocalization and accumulation of abnormal TDP-43 in the cytosol of transgenic mice, which likely lead to an increase in cellular stress and cognitive impairment. Taken together, these results highlight an important pathologic link between VCP and cognition.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Trastornos del Conocimiento/metabolismo , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/genética , Distrofia Muscular de Cinturas/genética , Mutación/genética , Miositis por Cuerpos de Inclusión/genética , Osteítis Deformante/genética , Ubiquitina/metabolismo , Adenosina Trifosfatasas/genética , Animales , Proteínas de Ciclo Celular/genética , Corteza Cerebral/metabolismo , Trastornos del Conocimiento/genética , Reacción de Fuga , Miedo , Demencia Frontotemporal/psicología , Habituación Psicofisiológica , Humanos , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Distrofia Muscular de Cinturas/psicología , Miositis por Cuerpos de Inclusión/psicología , Neuronas/metabolismo , Osteítis Deformante/psicología , Prosencéfalo/metabolismo , Reconocimiento en Psicología , Proteína que Contiene Valosina
11.
Toxicol Sci ; 193(2): 175-191, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37074955

RESUMEN

Exposure to traffic-related air pollution consisting of particulate matter (PM) is associated with cognitive decline leading to Alzheimer's disease (AD). In this study, we sought to examine the neurotoxic effects of exposure to ultrafine PM and how it exacerbates neuronal loss and AD-like neuropathology in wildtype (WT) mice and a knock-in mouse model of AD (AppNL-G-F/+-KI) when the exposure occurs at a prepathologic stage or at a later age with the presence of neuropathology. AppNL-G-F/+-KI and WT mice were exposed to concentrated ultrafine PM from local ambient air in Irvine, California, for 12 weeks, starting at 3 or 9 months of age. Particulate matter-exposed animals received concentrated ultrafine PM up to 8 times above the ambient levels, whereas control animals were exposed to purified air. Particulate matter exposure resulted in a marked impairment of memory tasks in prepathologic AppNL-G-F/+-KI mice without measurable changes in amyloid-ß pathology, synaptic degeneration, and neuroinflammation. At aged, both WT and AppNL-G-F/+-KI mice exposed to PM showed a significant memory impairment along with neuronal loss. In AppNL-G-F/+-KI mice, we also detected an increased amyloid-ß buildup and potentially harmful glial activation including ferritin-positive microglia and C3-positive astrocytes. Such glial activation could promote the cascade of degenerative consequences in the brain. Our results suggest that exposure to PM impairs cognitive function at both ages while exacerbation of AD-related pathology and neuronal loss may depend on the stage of pathology, aging, and/or state of glial activation. Further studies will be required to unveil the neurotoxic role of glial activation activated by PM exposure.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/patología , Material Particulado/toxicidad , Péptidos beta-Amiloides/metabolismo , Modelos Animales de Enfermedad , Encéfalo/metabolismo , Trastornos de la Memoria/inducido químicamente , Ratones Transgénicos
12.
Neurobiol Learn Mem ; 98(3): 215-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22910716

RESUMEN

Reconsolidation refers to the destabilization/re-stabilization memory process upon its activation. However, the conditions needed to undergo reconsolidation, as well as its functional significance is quite unclear and a matter of intense investigation. Even so, memory retrieval is held as requisite to initiate reconsolidation. Therefore, in the present work we examined whether transient pharmacological disruption of memory retrieval impedes reconsolidation of stored memory in the widely used associative conditioning task, taste aversion. We found that AMPA receptors inhibition in the amygdala impaired retrieval of taste aversion memory. Furthermore, AMPA receptors blockade impeded retrieval regardless of memory strength. However, inhibition of retrieval did not affect anisomycin-mediated disruption of reconsolidation. These results indicate that retrieval is a dispensable condition to undergo reconsolidation and provide evidence of molecular dissociation between retrieval and activation of memory in the non-declarative memory model taste aversion.


Asunto(s)
Reacción de Prevención/fisiología , Condicionamiento Clásico/fisiología , Memoria/fisiología , Gusto/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Reacción de Prevención/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Masculino , Memoria/efectos de los fármacos , Microinyecciones , Quinoxalinas/farmacología , Ratas , Ratas Wistar , Receptores AMPA/antagonistas & inhibidores , Gusto/efectos de los fármacos
13.
Neurobiol Learn Mem ; 97(4): 418-24, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22452926

RESUMEN

In this work we probed the effects of post-trial infusions of the muscarinic receptor antagonist scopolamine on object recognition memory formation. Scopolamine was infused bilaterally immediately after the sample phase in the perirhinal cortex or dorsal hippocampus and animals were tested for short-term (90 min) or long-term (24 h) memory. Results showed that scopolamine impaired short-term memory when injected in either the perirhinal cortex or hippocampus. Nevertheless, scopolamine disrupted long-term memory when administrated in the perirhinal cortex but not when applied in the hippocampus. Long-term memory was unaffected when scopolamine was infused 160 min after the sample phase or 90 min before test phase. Our data indicate that short-term recognition memory requires muscarinic receptors signaling in both the perirhinal cortex and hippocampus, whereas long-term recognition memory depends on muscarinic receptors in the perirhinal cortex but not hippocampus. These results support a differential involvement of muscarinic activity in these two medial temporal lobe structures in the formation of recognition memory.


Asunto(s)
Hipocampo/fisiología , Memoria a Largo Plazo/fisiología , Memoria a Corto Plazo/fisiología , Receptores Muscarínicos/fisiología , Reconocimiento en Psicología/fisiología , Lóbulo Temporal/fisiología , Animales , Hipocampo/efectos de los fármacos , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Memoria a Corto Plazo/efectos de los fármacos , Antagonistas Muscarínicos/farmacología , Ratas , Ratas Wistar , Receptores Muscarínicos/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos , Escopolamina/farmacología , Lóbulo Temporal/efectos de los fármacos
14.
Neurobiol Learn Mem ; 95(3): 311-5, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21193052

RESUMEN

Some reports have shown that the ubiquitin-proteasome system (UPS) is necessary to degrade repressor factors to produce new proteins essential to memory consolidation. Furthermore, recent evidence suggests that memory updating also relies on protein degradation through the UPS. To evaluate whether degradation of proteins is part of the cellular events needed for long-term storage of taste aversion, we injected lactacystin--an UPS inhibitor--into the amygdala and/or insular cortex 30 min before the first or second training trials. The results revealed that degradation of proteins in either the amygdala or insular cortex suffices for long-term stabilization of first-time encounter taste aversion. On the other hand, lactacystin applied in the insula, but not in the amygdala, before the second training prevented long-term storage of updated information. Our results support that degradation of proteins by means of the UPS is required every time taste aversion is to be stored in long-term memory.


Asunto(s)
Acetilcisteína/análogos & derivados , Amígdala del Cerebelo/metabolismo , Reacción de Prevención/fisiología , Corteza Cerebral/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Memoria a Largo Plazo/fisiología , Acetilcisteína/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Aprendizaje por Asociación/efectos de los fármacos , Aprendizaje por Asociación/fisiología , Reacción de Prevención/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Ratas , Ratas Wistar , Gusto , Ubiquitina/antagonistas & inhibidores , Ubiquitina/metabolismo
15.
Neuroscience ; 453: 69-80, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33246059

RESUMEN

Effective clearance of neurotoxic amyloid-beta (Aß) from the brain is a critical process to prevent Alzheimer's disease (AD). One major clearance mechanism is Aß transcytosis mediated by low-density lipoprotein receptor-related protein 1 (LRP1) in capillary endothelial cells. A marked loss of endothelial LRP1 is found in AD brains and is believed to significantly impair Aß clearance. Recently, we demonstrated that pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α, significantly down-regulated LRP1 in human primary microvascular endothelial cells (MVECs). In this study, we sought to determine the underlying molecular mechanism by which IL-1ß led to LRP1 loss in MVECs. Reduced LRP1 protein and transcript were detected up to 24 h post-exposure and returned to the baseline levels after 48 h post-exposure with 1 ng/ml IL-1ß. This reduction was in part mediated by microRNA-205-5p, -200b-3p, and -200c-3p, as these microRNAs were concomitantly upregulated in MVECs exposed to IL-1ß. Synthetic microRNA-205-5p, -200b-3p, and -200c-3p mimics recapitulated LRP1 loss in MVECs without IL-1ß, and their synthetic antagomirs effectively reversed IL-1ß-mediated LRP1 loss. Importantly, we found that the expression of these three microRNAs was controlled by NF-κB as pharmacological NF-κB inhibitor, BMS-345541, inhibited the IL-1ß-mediated upregulation of these microRNAs and rescued LRP1 expression. siRNA-mediated silencing of IκB in MVECs elevated microRNA-200b-3p and decreased LRP1 transcript, partially confirming our overall findings. In conclusion, our study provides a mechanism by which pro-inflammatory IL-1ß instigates the suppression of LRP1 expression in MVECs. Our findings could implicate spatiotemporal loss of LRP1 and impairment of the LRP1-mediated clearance mechanism by endothelial cells.


Asunto(s)
Células Endoteliales , Silenciador del Gen , Interleucina-1beta/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , MicroARNs , Péptidos beta-Amiloides/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Humanos , MicroARNs/genética
16.
Int J Biol Macromol ; 192: 1178-1184, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34673103

RESUMEN

Ascorbic acid (AA) uptake in neurons occurs via a Na+-dependent carrier-mediated process mediated by the sodium-dependent vitamin C transporter-2 (SVCT2). Relatively little information is available concerning the network of interacting proteins that support human (h)SVCT2 trafficking and cell surface expression in neuronal cells. Here we identified the synaptogenic adhesion protein, calsyntenin-3 (CLSTN3) as an hSVCT2 interacting protein from yeast two-hybrid (Y2H) screening of a human adult brain cDNA library. This interaction was confirmed by co-immunoprecipitation, mammalian two-hybrid (M2H), and co-localization in human cell lines. Co-expression of hCLSTN3 with hSVCT2 in SH-SY5Y cells led to a marked increase in AA uptake. Reciprocally, siRNA targeting hCLSTN3 inhibited AA uptake. In the J20 mouse model of Alzheimer's disease (AD), mouse (m)SVCT2 and mCLSTN3 expression levels in hippocampus were decreased. Similarly, expression levels of hSVCT2 and hCLSTN3 were markedly decreased in hippocampal samples from AD patients. These findings establish CLSTN3 as a novel hSVCT2 interactor in neuronal cells with potential pathophysiological significance.


Asunto(s)
Ácido Ascórbico/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Transportadores de Sodio Acoplados a la Vitamina C/metabolismo , Animales , Línea Celular , Expresión Génica , Hipocampo/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Unión Proteica , Técnicas del Sistema de Dos Híbridos
17.
Nat Commun ; 12(1): 2421, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893290

RESUMEN

The majority of Alzheimer's disease (AD) cases are late-onset and occur sporadically, however most mouse models of the disease harbor pathogenic mutations, rendering them better representations of familial autosomal-dominant forms of the disease. Here, we generated knock-in mice that express wildtype human Aß under control of the mouse App locus. Remarkably, changing 3 amino acids in the mouse Aß sequence to its wild-type human counterpart leads to age-dependent impairments in cognition and synaptic plasticity, brain volumetric changes, inflammatory alterations, the appearance of Periodic Acid-Schiff (PAS) granules and changes in gene expression. In addition, when exon 14 encoding the Aß sequence was flanked by loxP sites we show that Cre-mediated excision of exon 14 ablates hAß expression, rescues cognition and reduces the formation of PAS granules.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Mutación , Plasticidad Neuronal/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/genética
18.
Eur J Neurosci ; 32(6): 1018-23, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20722718

RESUMEN

The extinction process has been described as the decline in the frequency or intensity of the conditioned response following the withdrawal of reinforcement. Hence, experimental extinction does not reflect loss of the original memory, but rather reflects new learning, which in turn requires consolidation in order to be maintained in the long term. During extinction of conditioned taste aversion (CTA), a taste previously associated with aversive consequences acquires a safe status through continuous presentations of the flavor with no aversive consequence. In addition, reconsolidation has been defined as the labile state of a consolidated memory after its reactivation by the presentation of relevant information. In this study, we analyzed structures from the temporal lobe that could be involved in consolidation and reconsolidation of extinction of CTA by means of new protein synthesis. Our results showed that protein synthesis in the hippocampus (HC), the perirhinal cortex (PR) and the insular cortex (IC) of rats participate in extinction consolidation, whereas the basolateral amygdala plays no part in this phenomenon. Furthermore, we found that inhibition of protein synthesis in the IC in a third extinction trial had an effect on reconsolidation of extinction. The participation of the HC in taste memory has been described as a downmodulator for CTA consolidation, and has been related to a context-taste association. Altogether, these data suggest that extinction of aversive taste memories are subserved by the IC, HC and PR, and that extinction can undergo reconsolidation, a process depending only on the IC.


Asunto(s)
Reacción de Prevención/fisiología , Condicionamiento Clásico/fisiología , Extinción Psicológica/fisiología , Gusto/fisiología , Lóbulo Temporal/fisiología , Animales , Masculino , Ratas , Ratas Wistar
19.
Learn Mem ; 16(9): 514-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19706834

RESUMEN

Reconsolidation has been described as a process where a consolidated memory returns to a labile state when retrieved. Growing evidence suggests that reconsolidation is, in fact, a destabilization/stabilization process that incorporates updated information to a previously consolidated memory. We used the conditioned taste aversion (CTA) task in order to test this theory. On the first trial, the conditioned stimulus (CS) (saccharin) was associated to the unconditioned stimulus (US) (LiCl injection), and as a result, aversion to saccharin was obtained. The following day, animals were injected with anisomycin in either the insular cortex (IC), central amygdala (CeA), basolateral amygdala (BLA), or simultaneously in IC and CeA or IC and BLA, and a second CTA trial was carried out in which updated information was acquired. Animals were tested 24 h later. When protein synthesis was inhibited in either the IC or CeA, consolidation was affected and previously consolidated memory was unimpaired. However, when both the IC and CeA were simultaneously anisomycin injected, the previously consolidated memory was affected. After repeated association trials, protein synthesis inhibition in the IC and CeA did not have an effect on taste memory. These results suggest that the IC and the CeA are necessary for taste-aversion consolidation, and that both share the previously consolidated memory trace. In addition, our data demonstrated that protein synthesis in either the IC or the CeA suffices to stabilize previously consolidated taste memory when destabilized by incorporation of updated information.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Anisomicina/farmacología , Corteza Cerebral/efectos de los fármacos , Memoria/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Gusto/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Corteza Cerebral/fisiología , Condicionamiento Clásico/efectos de los fármacos , Cloruro de Litio/efectos adversos , Masculino , Ratas , Ratas Wistar , Sacarina/administración & dosificación , Edulcorantes/administración & dosificación , Gusto/fisiología
20.
Aging Cell ; 19(3): e13118, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32087004

RESUMEN

MicroRNAs play a pivotal role in rapid, dynamic, and spatiotemporal modulation of synaptic functions. Among them, recent emerging evidence highlights that microRNA-181a (miR-181a) is particularly abundant in hippocampal neurons and controls the expression of key plasticity-related proteins at synapses. We have previously demonstrated that miR-181a was upregulated in the hippocampus of a mouse model of Alzheimer's disease (AD) and correlated with reduced levels of plasticity-related proteins. Here, we further investigated the underlying mechanisms by which miR-181a negatively modulated synaptic plasticity and memory. In primary hippocampal cultures, we found that an activity-dependent upregulation of the microRNA-regulating protein, translin, correlated with reduction of miR-181a upon chemical long-term potentiation (cLTP), which induced upregulation of GluA2, a predicted target for miR-181a, and other plasticity-related proteins. Additionally, Aß treatment inhibited cLTP-dependent induction of translin and subsequent reduction of miR-181a, and cotreatment with miR-181a antagomir effectively reversed the effects elicited by Aß but did not rescue translin levels, suggesting that the activity-dependent upregulation of translin was upstream of miR-181a. In mice, a learning episode markedly decreased miR-181a in the hippocampus and raised the protein levels of GluA2. Lastly, we observed that inhibition of miR-181a alleviated memory deficits and increased GluA2 and GluA1 levels, without restoring translin, in the 3xTg-AD model. Taken together, our results indicate that miR-181a is a major negative regulator of the cellular events that underlie synaptic plasticity and memory through AMPA receptors, and importantly, Aß disrupts this process by suppressing translin and leads to synaptic dysfunction and memory impairments in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo/genética , Trastornos de la Memoria/metabolismo , MicroARNs/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Aprendizaje/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Neuronas/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores AMPA/metabolismo , Transducción de Señal/efectos de los fármacos , Sinapsis/metabolismo , Transfección , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA