Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 146(12): 4949-4963, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37403195

RESUMEN

Learning and memory mainly rely on correct synaptic function in the hippocampus and other brain regions. In Parkinson's disease, subtle cognitive deficits may even precede motor signs early in the disease. Hence, we set out to unravel the earliest hippocampal synaptic alterations associated with human α-synuclein overexpression prior to and soon after the appearance of cognitive deficits in a parkinsonism model. We bilaterally injected adeno-associated viral vectors encoding A53T-mutated human α-synuclein into the substantia nigra of rats, and evaluated them 1, 2, 4 and 16 weeks post-inoculation by immunohistochemistry and immunofluorescence to study degeneration and distribution of α-synuclein in the midbrain and hippocampus. The object location test was used to evaluate hippocampal-dependent memory. Sequential window acquisition of all theoretical mass spectrometry-based proteomics and fluorescence analysis of single-synapse long-term potentiation were used to study alterations to protein composition and plasticity in isolated hippocampal synapses. The effect of L-DOPA and pramipexole on long-term potentiation was also tested. Human α-synuclein was found within dopaminergic and glutamatergic neurons of the ventral tegmental area, and in dopaminergic, glutamatergic and GABAergic axon terminals in the hippocampus from 1 week post-inoculation, concomitant with mild dopaminergic degeneration in the ventral tegmental area. In the hippocampus, differential expression of proteins involved in synaptic vesicle cycling, neurotransmitter release and receptor trafficking, together with impaired long-term potentiation were the first events observed (1 week post-inoculation), preceding cognitive deficits (4 weeks post-inoculation). Later on, at 16 weeks post-inoculation, there was a deregulation of proteins involved in synaptic function, particularly those involved in the regulation of membrane potential, ion balance and receptor signalling. Hippocampal long-term potentiation was impaired before and soon after the onset of cognitive deficits, at 1 and 4 weeks post-inoculation, respectively. L-DOPA recovered hippocampal long-term potentiation more efficiently at 4 weeks post-inoculation than pramipexole, which partially rescued it at both time points. Overall, we found impaired synaptic plasticity and proteome dysregulation at hippocampal terminals to be the first events that contribute to the development of cognitive deficits in experimental parkinsonism. Our results not only point to dopaminergic but also to glutamatergic and GABAergic dysfunction, highlighting the relevance of the three neurotransmitter systems in the ventral tegmental area-hippocampus interaction from the earliest stages of parkinsonism. The proteins identified in the current work may constitute potential biomarkers of early synaptic damage in the hippocampus and hence, therapies targeting these could potentially restore early synaptic malfunction and consequently, cognitive deficits in Parkinson's disease.


Asunto(s)
Enfermedad de Parkinson , Trastornos Parkinsonianos , Humanos , Ratas , Animales , alfa-Sinucleína/metabolismo , Levodopa/farmacología , Pramipexol/farmacología , Hipocampo/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neurotransmisores/metabolismo , Cognición
2.
Alzheimers Dement ; 17(11): 1808-1817, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34297895

RESUMEN

INTRODUCTION: Effective strategies to recruit older adults with mild cognitive impairment (MCI) into nonpharmacological intervention trials are lacking. METHODS: Recruitment for EXERT, a multisite randomized controlled 18-month trial examining the effects of aerobic exercise on cognitive trajectory in adults with amnestic MCI, involved a diverse portfolio of strategies to enroll 296 participants. RESULTS: Recruitment occurred September 2016 through March 2020 and was initially slow. After mass mailings of 490,323 age- and geo-targeted infographic postcards and brochures, recruitment rates increased substantially, peaking at 16 randomizations/month in early 2020. Mass mailings accounted for 52% of randomized participants, whereas 25% were recruited from memory clinic rosters, electronic health records, and national and local registries. Other sources included news broadcasts, public service announcements (PSA), local advertising, and community presentations. DISCUSSION: Age- and geo-targeted mass mailing of infographic materials was the most effective approach in recruiting older adults with amnestic MCI into an 18-month exercise trial.


Asunto(s)
Amnesia/terapia , Disfunción Cognitiva/terapia , Ejercicio Físico , Folletos , Selección de Paciente , Anciano , Cognición , Femenino , Humanos , Masculino , Servicios Postales
3.
Learn Mem ; 26(12): 485-492, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31732709

RESUMEN

The beneficial effects of exercise on cognition are well established; however specific exercise parameters regarding the frequency and duration of physical activity that provide optimal cognitive health have not been well defined. Here, we explore the effects of the duration of exercise and sedentary periods on long-term object location memory (OLM) in mice. We use a weak object location training paradigm that is subthreshold for long-term memory formation in sedentary controls, and demonstrate that exercise enables long-term memories to form. We show that 14- and 21-d of running wheel access enables mice to discriminate between familiar and novel object locations after a 24 h delay, while 2- or 7-d running wheel access provides insufficient exercise for such memory enhancement using the subthreshold learning paradigm. After 14- and 21-d of wheel running, exercise-induced cognitive enhancement then decays back to baseline performance following 3-d of sedentary activity. However, exercise-induced cognitive enhancement can be reactivated by an additional period of just 2 d exercise, previously shown to be insufficient to induce cognitive enhancement on its own. The reactivating period of exercise is capable of enhancing memory after three- or seven-sedentary days, but not 14-d. These data suggest a type of "molecular memory" for the exercise stimulus, in that once exercise duration reaches a certain threshold, it establishes a temporal window during which subsequent low-level exercise can capitalize on the neurobiological adaptations induced by the initial period of exercise, enabling it to maintain the benefits on cognitive function. These findings provide new information that may help to guide future clinical studies in exercise.


Asunto(s)
Adaptación Fisiológica/fisiología , Cognición/fisiología , Memoria a Largo Plazo/fisiología , Condicionamiento Físico Animal/fisiología , Memoria Espacial/fisiología , Animales , Conducta Animal/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
4.
Neurochem Res ; 44(1): 49-60, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29619614

RESUMEN

CNS inflammatory responses are linked to cognitive impairment in humans. Research in animal models supports this connection by showing that inflammatory cytokines suppress long-term potentiation (LTP), the best-known cellular correlate of memory. Cytokine-induced modulation of LTP has been previously studied in vivo or in brain slices, two experimental approaches containing multiple cell populations responsive to cytokines. In their target cells, cytokines commonly increase the expression of multiple cytokines, thus increasing the complexity of brain cytokine networks even after single-cytokine challenges. Whether cytokines suppress LTP by direct effects on neurons or by indirect mechanisms is still an open question. Here, we evaluated the effect of a major set of inflammatory cytokines including tumor necrosis factor-α (TNFα), interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) on chemically-induced LTP (cLTP) in isolated hippocampal synaptosomes of mice, using fluorescence analysis of single-synapse long-term potentiation (FASS-LTP). We found that TNFα and IL-1ß suppress synaptosomal cLTP. In contrast, cLTP was not affected by IL-18, at a concentration previously shown to block LTP in hippocampal slices. We also found that IL-18 does not impair cLTP or brain-derived neurotrophic factor (BDNF) signaling in primary hippocampal neuronal cultures. Thus, using both synaptosomes and neuron cultures, our data suggest that IL-18 impairs LTP by indirect mechanisms, which may depend on non-neuronal cells, such as glia. Notably, our results demonstrate that TNFα and IL-1ß directly suppress hippocampal plasticity via neuron-specific mechanisms. A better understanding of the brain's cytokine networks and their final molecular effectors is crucial to identify specific targets for intervention.


Asunto(s)
Hipocampo/fisiología , Interleucina-18/farmacología , Interleucina-1beta/farmacología , Potenciación a Largo Plazo/fisiología , Sinapsis/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Células Cultivadas , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Ratones , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos
5.
Alzheimer Dis Assoc Disord ; 33(4): 327-330, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31513029

RESUMEN

OBJECTIVE: A rare variant in TREM2 (p.R47H, rs75932628) has been consistently reported to increase the risk for Alzheimer disease (AD), while mixed evidence has been reported for association of the variant with other neurodegenerative diseases. Here, we investigated the frequency of the R47H variant in a diverse and well-characterized multicenter neurodegenerative disease cohort. METHODS: We examined the frequency of the R47H variant in a diverse neurodegenerative disease cohort, including a total of 3058 patients clinically diagnosed with AD, frontotemporal dementia spectrum syndromes, mild cognitive impairment, progressive supranuclear palsy syndrome, corticobasal syndrome, or amyotrophic lateral sclerosis and 5089 control subjects. RESULTS: We observed a significant association between the R47H variant and AD, while no association was observed with any other neurodegenerative disease included in this study. CONCLUSIONS: Our results support the consensus that the R47H variant is significantly associated with AD. However, we did not find evidence for association of the R47H variant with other neurodegenerative diseases.


Asunto(s)
Predisposición Genética a la Enfermedad , Variación Genética , Genotipo , Glicoproteínas de Membrana/genética , Enfermedades Neurodegenerativas/genética , Receptores Inmunológicos/genética , Anciano , Enfermedad de Alzheimer/genética , Esclerosis Amiotrófica Lateral/genética , Disfunción Cognitiva/genética , Estudios de Cohortes , Femenino , Demencia Frontotemporal/genética , Humanos , Internacionalidad , Masculino
6.
J Neurosci ; 37(5): 1197-1212, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27986924

RESUMEN

Long-term potentiation (LTP) is an activity-dependent and persistent increase in synaptic transmission. Currently available techniques to measure LTP are time-intensive and require highly specialized expertise and equipment, and thus are not well suited for screening of multiple candidate treatments, even in animal models. To expand and facilitate the analysis of LTP, here we use a flow cytometry-based method to track chemically induced LTP by detecting surface AMPA receptors in isolated synaptosomes: fluorescence analysis of single-synapse long-term potentiation (FASS-LTP). First, we demonstrate that FASS-LTP is simple, sensitive, and models electrically induced LTP recorded in intact circuitries. Second, we conducted FASS-LTP analysis in two well-characterized Alzheimer's disease (AD) mouse models (3xTg and Tg2576) and, importantly, in cryopreserved human AD brain samples. By profiling hundreds of synaptosomes, our data provide the first direct evidence to support the idea that synapses from AD brain are intrinsically defective in LTP. Third, we used FASS-LTP for drug evaluation in human synaptosomes. Testing a panel of modulators of cAMP and cGMP signaling pathways, FASS-LTP identified vardenafil and Bay-73-6691 (phosphodiesterase-5 and -9 inhibitors, respectively) as potent enhancers of LTP in synaptosomes from AD cases. These results indicate that our approach could provide the basis for protocols to study LTP in both healthy and diseased human brains, a previously unattainable goal. SIGNIFICANCE STATEMENT: Learning and memory depend on the ability of synapses to strengthen in response to activity. Long-term potentiation (LTP) is a rapid and persistent increase in synaptic transmission that is thought to be affected in Alzheimer's disease (AD). However, direct evidence of LTP deficits in human AD brain has been elusive, primarily due to methodological limitations. Here, we analyze LTP in isolated synapses from AD brain using a novel approach that allows testing LTP in cryopreserved brain. Our analysis of hundreds of synapses supports the idea that AD-diseased synapses are intrinsically defective in LTP. Further, we identified pharmacological agents that rescue LTP in AD, thus opening up a new avenue for drug screening and evaluation of strategies for alleviating memory impairments.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Potenciación a Largo Plazo/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , AMP Cíclico/fisiología , GMP Cíclico/fisiología , Estimulación Eléctrica , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de Fosfodiesterasa/farmacología , Ratas Sprague-Dawley , Receptores AMPA/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sinaptosomas/efectos de los fármacos
7.
J Neuroinflammation ; 15(1): 127, 2018 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712570

RESUMEN

BACKGROUND: Brain inflammation including increases in inflammatory cytokines such as IL-1ß is widely believed to contribute to the pathophysiology of Alzheimer's disease. Although IL-1ß-induced impairments in long-term potentiation (LTP) in acute hippocampal slices and memory functions in vivo have been well documented, the neuron-specific molecular mechanisms of IL-1ß-mediated impairments of LTP and memory remain unclear. METHODS: This study uses an in vitro approach in primary hippocampal neurons to evaluate the effect of IL-1ß on chemical LTP (cLTP)-induced structural plasticity and signaling. RESULTS: We found that IL-1ß reduces both the surface expression of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 and the spine growth following cLTP. These effects of IL-1ß were mediated by impairing actin polymerization during cLTP, as IL-1ß decreased the cLTP-induced formation of F-actin, and the effect of IL-1ß on cLTP-induced surface expression of GluA1 can be mimicked by latrunculin, a toxin that disrupts dynamics of actin filaments, and can be prevented by jasplakinolide, a cell-permeable peptide that stabilizes F-actin. Moreover, live-cell imaging demonstrated that IL-1ß decreased the stability of the actin cytoskeleton in spines, which is required for LTP consolidation. We further examined the role of sphingolipid signaling in the IL-1ß-mediated impairment of spine plasticity and found that both the neutral sphingomyelinase inhibitor GW4869 and the inhibitor of Src kinase PP2 attenuated the IL-1ß-mediated suppression of cLTP-induced surface expression of GluA1 and actin polymerization. CONCLUSIONS: These findings support a mechanism by which IL-1ß, via the sphingomyelinase/ceramide/Src pathway, impairs structural spine remodeling essential for LTP consolidation and memory.


Asunto(s)
Actinas/metabolismo , Ceramidas/farmacología , Genes src/fisiología , Interleucina-1beta/farmacología , Potenciación a Largo Plazo/fisiología , Receptores AMPA/biosíntesis , Animales , Células Cultivadas , Expresión Génica , Genes src/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Polimerizacion/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores AMPA/antagonistas & inhibidores
9.
Proc Natl Acad Sci U S A ; 112(36): E5078-87, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26305968

RESUMEN

In the aged brain, synaptic plasticity and memory show increased vulnerability to impairment by the inflammatory cytokine interleukin 1ß (IL-1ß). In this study, we evaluated the possibility that synapses may directly undergo maladaptive changes with age that augment sensitivity to IL-1ß impairment. In hippocampal neuronal cultures, IL-1ß increased the expression of the IL-1 receptor type 1 and the accessory coreceptor AcP (proinflammatory), but not of the AcPb (prosurvival) subunit, a reconfiguration that potentiates the responsiveness of neurons to IL-1ß. To evaluate whether synapses develop a similar heightened sensitivity to IL-1ß with age, we used an assay to track long-term potentiation (LTP) in synaptosomes. We found that IL-1ß impairs LTP directly at the synapse and that sensitivity to IL-1ß is augmented in aged hippocampal synapses. The increased synaptic sensitivity to IL-1ß was due to IL-1 receptor subunit reconfiguration, characterized by a shift in the AcP/AcPb ratio, paralleling our culture data. We suggest that the age-related increase in brain IL-1ß levels drives a shift in IL-1 receptor configuration, thus heightening the sensitivity to IL-1ß. Accordingly, selective blocking of AcP-dependent signaling with Toll-IL-1 receptor domain peptidomimetics prevented IL-1ß-mediated LTP suppression and blocked the memory impairment induced in aged mice by peripheral immune challenge (bacterial lipopolysaccharide). Overall, this study demonstrates that increased AcP signaling, specifically at the synapse, underlies the augmented vulnerability to cognitive impairment by IL-1ß that occurs with age.


Asunto(s)
Interleucina-1beta/farmacología , Neuronas/efectos de los fármacos , Receptores Tipo I de Interleucina-1/metabolismo , Sinapsis/metabolismo , Factores de Edad , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hipocampo/citología , Hipocampo/metabolismo , Proteína Accesoria del Receptor de Interleucina-1/genética , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Neuronas/metabolismo , Interferencia de ARN , Ratas Sprague-Dawley , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal/efectos de los fármacos , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo
10.
Learn Mem ; 24(3): 123-131, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28202716

RESUMEN

The pattern separation task has recently emerged as a behavioral model of hippocampus function and has been used in several pharmaceutical trials. The canine is a useful model to evaluate a multitude of hippocampal-dependent cognitive tasks that parallel those in humans. Thus, this study was designed to evaluate the suitability of pattern separation task(s) for detecting age-related changes in canines. We also assessed the dogs' ability to show pattern separation and discrimination reversal, which provides a novel extension of the pattern separation learning literature. Our data show that aged dogs are impaired on a complex pattern separation task (six-well task) relative to easier tasks (four-well or six-well pattern discrimination task), and that the age-related deficits are due to loss of perceptual and inhibitory control in addition to the loss of spatial discrimination and pattern separation ability. Our data also suggest that aged animals show pattern separation deficits when the objects are brought progressively closer together while changing the location of both correct and incorrect objects. However, if the location of any one object is fixed the animals tend to use alternate strategies. Overall, these data provide important insight into age-related pattern separation deficits in a higher animal model and offers additional means for evaluating the impact of lifestyle and pharmaceutical interventions on episodic memory in preclinical trials.


Asunto(s)
Envejecimiento/psicología , Cognición/fisiología , Aprendizaje Discriminativo/fisiología , Objetivos , Percepción Espacial/fisiología , Animales , Condicionamiento Operante/fisiología , Perros , Femenino , Masculino , Modelos Animales , Reconocimiento Visual de Modelos/fisiología , Tiempo de Reacción/fisiología
11.
J Neurosci ; 36(12): 3611-22, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-27013689

RESUMEN

An increasing number of studies show that an altered epigenetic landscape may cause impairments in regulation of learning and memory-related genes within the aged hippocampus, eventually resulting in cognitive deficits in the aged brain. One such epigenetic repressive mark is trimethylation of H3K9 (H3K9me3), which is typically implicated in gene silencing. Here, we identify, for the first time, an essential role for H3K9me3 and its histone methyl transferase (SUV39H1) in mediating hippocampal memory functions. Pharmacological inhibition of SUV39H1 using a novel and selective inhibitor decreased levels of H3K9me3 in the hippocampus of aged mice, and improved performance in the objection location memory and fear conditioning tasks and in a complex spatial environment learning task. The inhibition of SUV39H1 induced an increase in spine density of thin and stubby but not mushroom spines in the hippocampus of aged animals and increased surface GluR1 levels in hippocampal synaptosomes, a key index of spine plasticity. Furthermore, there were changes at BDNF exon I gene promoter, in concert with overall BDNF levels in the hippocampus of drug-treated animals compared with control animals. Together, these data demonstrate that SUV39H1 inhibition and the concomitant H3K9me3 downregulation mediate gene transcription in the hippocampus and reverse age-dependent deficits in hippocampal memory. SIGNIFICANCE STATEMENT: Cognitive decline is a debilitating condition associated with not only neurodegenerative diseases but also aging in general. However, effective treatments have been slow to emerge so far. In this study, we demonstrate that epigenetic regulation of key synaptic proteins may be an underlying, yet reversible, cause of this decline. Our findings suggest that histone 3 trimethylation is a probable target for pharmacological intervention that can counteract cognitive decline in the aging brain. Finally, we provide support to the hypothesis that, by manipulating the enzyme that regulates H3K9me3 (using a newly developed specific inhibitor of SUV39H1), it is possible to alter the chromatin state of subjects and restore memory and synaptic function in the aging brain.


Asunto(s)
Envejecimiento/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Espinas Dendríticas/fisiología , Hipocampo/fisiología , Histonas/metabolismo , Memoria/fisiología , Animales , Histonas/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba/fisiología
12.
J Neuroinflammation ; 14(1): 29, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28153028

RESUMEN

BACKGROUND: Pro-inflammatory cytokines accumulate in the brain with age and Alzheimer's disease and can impair neuron health and cognitive function. Brain-derived neurotrophic factor (BDNF) is a key neurotrophin that supports neuron health, function, and synaptic plasticity. The pro-inflammatory cytokine interleukin-1ß (IL-1ß) impairs BDNF signaling but whether it affects BDNF signaling endosome trafficking has not been studied. METHODS: This study uses an in vitro approach in primary hippocampal neurons to evaluate the effect of IL-1ß on BDNF signaling endosome trafficking. Neurons were cultured in microfluidic chambers that separate the environments of the cell body and its axon terminal, enabling us to specifically treat in axon compartments and trace vesicle trafficking in real-time. RESULTS: We found that IL-1ß attenuates BDNF signaling endosomes throughout networks in cultures. In IL-1ß-treated cells, overall BDNF endosomal density was decreased, and the colocalization of BDNF endosomes with presynaptic terminals was found to be more than two times higher than in control cultures. Selective IL-1ß treatment to the presynaptic compartment in microfluidic chamber attenuated BDNF endosome flux, as measured by reduced BDNF-GFP endosome counts in the somal compartment. Further, IL-1ß decreased the BDNF-induced phosphorylation of Erk5, a known BDNF retrograde trafficking target. Mechanistically, the deficiency in trafficking was not due to impaired endocytosis of the BDNF-TrkB complex, or impaired transport rate, since BDNF endosomes traveled at the same rate in both control and IL-1ß treatment groups. Among the regulators of presynaptic endosome sorting is the post-translational modification, ubiquitination. In support of this possibility, the IL-1ß-mediated suppression of BDNF-induced Erk5 phosphorylation can be rescued by exogenous ubiquitin C-terminal hydrolase L1 (UCH-L1), a deubiquitinating enzyme that regulates ubiquitin and endosomal trafficking. CONCLUSIONS: We observed a state of neurotrophic resistance whereby, in the prolonged presence of IL-1ß, BDNF is not effective in delivering long-distance signaling via the retrograde transport of signaling endosomes. Since IL-1ß accumulation is an invariant feature across many neurodegenerative diseases, our study suggest that compromised BDNF retrograde transport-dependent signaling may have important implications in neurodegenerative diseases.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Endosomas/metabolismo , Interleucina-1beta/farmacología , Transducción de Señal/fisiología , Animales , Células Cultivadas , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Endosomas/efectos de los fármacos , Humanos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Transducción de Señal/efectos de los fármacos
13.
J Neuroinflammation ; 13(1): 289, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27832807

RESUMEN

BACKGROUND: While exercise effects on the immune system have received increasing attention in recent years, it remains unclear to what extent gender and fluctuations in sex hormones during menstrual cycle influence immunological responses to exercise. METHODS: We investigated mRNA changes induced through exhaustive exercise (half-marathon; pre-exercise and post-exercise [30 min, 3 h, 24 h] on whole blood cultures ± lipopolysaccharide [LPS] [1 h]) with a specific focus on sex differences (men vs women in luteal phase) as an extension of our previous study. RESULTS: Inflammation related signaling pathways, TLRs, cytosolic DNA sensing and RIG-I like receptors were differentially activated between sexes in LPS-stimulated cultures. Genes differentially regulated between sexes included TNIP-1, TNIP-3, IL-6, HIVEP1, CXCL3, CCR3, IL-8, and CD69, revealing a bias towards less anti-inflammatory gene regulation in women compared to men. In addition, several genes relevant to brain function (KMO, DDIT4, VEGFA, IGF1R, IGF2R, and FGD4) showed differential activation between sexes. Some of these genes (e.g., KMO in women, DDIT4 in both sexes) potentially constitute neuroprotective mechanisms. CONCLUSIONS: These data reveal that the exercise-induced change in gene expression might be gender and menstrual cycle phase dependent.


Asunto(s)
Citocinas/metabolismo , Endotoxinas/farmacología , Ejercicio Físico , Expresión Génica/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Transducción de Señal/efectos de los fármacos , Adulto , Antropometría , Atletas , Células Cultivadas , Citocinas/genética , Femenino , Perfilación de la Expresión Génica , Hormonas , Humanos , Lipopolisacáridos/farmacología , Masculino , Ciclo Menstrual/fisiología , Factores Sexuales , Factores de Tiempo
14.
J Biol Chem ; 289(30): 20615-29, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24917666

RESUMEN

The mammalian target of rapamycin (mTOR) pathway has multiple important physiological functions, including regulation of protein synthesis, cell growth, autophagy, and synaptic plasticity. Activation of mTOR is necessary for the many beneficial effects of brain-derived neurotrophic factor (BDNF), including dendritic translation and memory formation in the hippocampus. At present, however, the role of mTOR in BDNF's support of survival is not clear. We report that mTOR activation is necessary for BDNF-dependent survival of primary rat hippocampal neurons, as either mTOR inhibition by rapamycin or genetic manipulation of the downstream molecule p70S6K specifically blocked BDNF rescue. Surprisingly, however, BDNF did not promote neuron survival by up-regulating mTOR-dependent protein synthesis or through mTOR-dependent suppression of caspase-3 activation. Instead, activated mTOR was responsible for BDNF's suppression of autophagic flux. shRNA against the autophagic machinery Atg7 or Atg5 prolonged the survival of neurons co-treated with BDNF and rapamycin, suggesting that suppression of mTOR in BDNF-treated cells resulted in excessive autophagy. Finally, acting as a physiological analog of rapamycin, IL-1ß impaired BDNF signaling by way of inhibiting mTOR activation as follows: the cytokine induced caspase-independent neuronal death and accelerated autophagic flux in BDNF-treated cells. These findings reveal a novel mechanism of BDNF neuroprotection; BDNF not only prevents apoptosis through inhibiting caspase activation but also promotes neuron survival through modulation of autophagy. This protection mechanism is vulnerable under chronic inflammation, which deregulates autophagy through impairing mTOR signaling. These results may be relevant to age-related changes observed in neurodegenerative diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Inmunosupresores/farmacología , Interleucina-1beta/metabolismo , Neuronas/metabolismo , Sirolimus/farmacología , Animales , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Masculino , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo
15.
J Biol Chem ; 288(23): 16937-16948, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23599427

RESUMEN

We previously found that BDNF-dependent retrograde trafficking is impaired in AD transgenic mouse neurons. Utilizing a novel microfluidic culture chamber, we demonstrate that Aß oligomers compromise BDNF-mediated retrograde transport by impairing endosomal vesicle velocities, resulting in impaired downstream signaling driven by BDNF/TrkB, including ERK5 activation, and CREB-dependent gene regulation. Our data suggest that a key mechanism mediating the deficit involves ubiquitin C-terminal hydrolase L1 (UCH-L1), a deubiquitinating enzyme that functions to regulate cellular ubiquitin. Aß-induced deficits in BDNF trafficking and signaling are mimicked by LDN (an inhibitor of UCH-L1) and can be reversed by increasing cellular UCH-L1 levels, demonstrated here using a transducible TAT-UCH-L1 strategy. Finally, our data reveal that UCH-L1 mRNA levels are decreased in the hippocampi of AD brains. Taken together, our data implicate that UCH-L1 is important for regulating neurotrophin receptor sorting to signaling endosomes and supporting retrograde transport. Further, our results support the idea that in AD, Aß may down-regulate UCH-L1 in the AD brain, which in turn impairs BDNF/TrkB-mediated retrograde signaling, compromising synaptic plasticity and neuronal survival.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Supervivencia Celular/genética , Hipocampo/patología , Humanos , Ratones , Ratones Transgénicos , Plasticidad Neuronal/genética , Neuronas/metabolismo , Neuronas/patología , Transporte de Proteínas/genética , Ratas , Receptor trkB/genética , Receptor trkB/metabolismo , Transducción de Señal/genética , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética
16.
Hum Mol Genet ; 21(15): 3500-12, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22556362

RESUMEN

Rare mutations in the gene encoding for tau (MAPT, microtubule-associated protein tau) cause frontotemporal dementia-spectrum (FTD-s) disorders, including FTD, progressive supranuclear palsy (PSP) and corticobasal syndrome, and a common extended haplotype spanning across the MAPT locus is associated with increased risk of PSP and Parkinson's disease. We identified a rare tau variant (p.A152T) in a patient with a clinical diagnosis of PSP and assessed its frequency in multiple independent series of patients with neurodegenerative conditions and controls, in a total of 15 369 subjects. Tau p.A152T significantly increases the risk for both FTD-s (n = 2139, OR = 3.0, CI: 1.6-5.6, P = 0.0005) and Alzheimer's disease (AD) (n = 3345, OR = 2.3, CI: 1.3-4.2, P = 0.004) compared with 9047 controls. Functionally, p.A152T (i) decreases the binding of tau to microtubules and therefore promotes microtubule assembly less efficiently; and (ii) reduces the tendency to form abnormal fibers. However, there is a pronounced increase in the formation of tau oligomers. Importantly, these findings suggest that other regions of the tau protein may be crucial in regulating normal function, as the p.A152 residue is distal to the domains considered responsible for microtubule interactions or aggregation. These data provide both the first genetic evidence and functional studies supporting the role of MAPT p.A152T as a rare risk factor for both FTD-s and AD and the concept that rare variants can increase the risk for relatively common, complex neurodegenerative diseases, but since no clear significance threshold for rare genetic variation has been established, some caution is warranted until the findings are further replicated.


Asunto(s)
Enfermedad de Alzheimer/genética , Demencia Frontotemporal/genética , Variación Genética , Proteínas tau/genética , Anciano , Enfermedad de Alzheimer/epidemiología , Demencia Frontotemporal/epidemiología , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Humanos , Persona de Mediana Edad , Riesgo
17.
Am J Pathol ; 183(2): 369-81, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747948

RESUMEN

Amyloid-ß (Aß) peptides, starting with pyroglutamate at the third residue (pyroGlu-3 Aß), are a major species deposited in the brain of Alzheimer disease (AD) patients. Recent studies suggest that this isoform shows higher toxicity and amyloidogenecity when compared to full-length Aß peptides. Here, we report the first comprehensive and comparative IHC evaluation of pyroGlu-3 Aß deposition in humans and animal models. PyroGlu-3 Aß immunoreactivity (IR) is abundant in plaques and cerebral amyloid angiopathy of AD and Down syndrome patients, colocalizing with general Aß IR. PyroGlu-3 Aß is further present in two nontransgenic mammalian models of cerebral amyloidosis, Caribbean vervets, and beagle canines. In addition, pyroGlu-3 Aß deposition was analyzed in 12 different AD-like transgenic mouse models. In contrast to humans, all transgenic models showed general Aß deposition preceding pyroGlu-3 Aß deposition. The findings varied greatly among the mouse models concerning age of onset and cortical brain region. In summary, pyroGlu-3 Aß is a major species of ß-amyloid deposited early in diffuse and focal plaques and cerebral amyloid angiopathy in humans and nonhuman primates, whereas it is deposited later in a subset of focal and vascular amyloid in AD-like transgenic mouse models. Given the proposed decisive role of pyroGlu-3 Aß peptides for the development of human AD pathology, this study provides insights into the usage of animal models in AD studies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Ácido Pirrolidona Carboxílico/metabolismo , Edad de Inicio , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide , Animales , Encéfalo/patología , Angiopatía Amiloide Cerebral/metabolismo , Chlorocebus aethiops , Modelos Animales de Enfermedad , Perros , Síndrome de Down/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Placa Amiloide/metabolismo
18.
Nat Commun ; 15(1): 3836, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714691

RESUMEN

Exercise has beneficial effects on cognition throughout the lifespan. Here, we demonstrate that specific exercise patterns transform insufficient, subthreshold training into long-term memory in mice. Our findings reveal a potential molecular memory window such that subthreshold training within this window enables long-term memory formation. We performed RNA-seq on dorsal hippocampus and identify genes whose expression correlate with conditions in which exercise enables long-term memory formation. Among these genes we found Acvr1c, a member of the TGF ß family. We find that exercise, in any amount, alleviates epigenetic repression at the Acvr1c promoter during consolidation. Additionally, we find that ACVR1C can bidirectionally regulate synaptic plasticity and long-term memory in mice. Furthermore, Acvr1c expression is impaired in the aging human and mouse brain, as well as in the 5xFAD mouse model, and over-expression of Acvr1c enables learning and facilitates plasticity in mice. These data suggest that promoting ACVR1C may protect against cognitive impairment.


Asunto(s)
Receptores de Activinas Tipo I , Epigénesis Genética , Hipocampo , Memoria a Largo Plazo , Condicionamiento Físico Animal , Animales , Femenino , Humanos , Masculino , Ratones , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Envejecimiento/genética , Envejecimiento/fisiología , Hipocampo/metabolismo , Memoria a Largo Plazo/fisiología , Ratones Endogámicos C57BL , Plasticidad Neuronal/genética , Condicionamiento Físico Animal/fisiología , Regiones Promotoras Genéticas
19.
J Neurosci ; 32(49): 17714-24, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23223292

RESUMEN

Evolving evidence suggests that brain inflammation and the buildup of proinflammatory cytokine increases the risk for cognitive decline and cognitive dysfunction. Interleukin-1ß (IL-1ß), acting via poorly understood mechanisms, appears to be a key cytokine in causing these deleterious effects along with a presumably related loss of long-term potentiation (LTP)-type synaptic plasticity. We hypothesized that IL-1ß disrupts brain-derived neurotrophic factor (BDNF) signaling cascades and thereby impairs the formation of filamentous actin (F-actin) in dendritic spines, an event that is essential for the stabilization of LTP. Actin polymerization in spines requires phosphorylation of the filament severing protein cofilin and is modulated by expression of the immediate early gene product Arc. Using rat organotypic hippocampal cultures, we found that IL-1ß suppressed BDNF-dependent regulation of Arc and phosphorylation of cofilin and cAMP response element-binding protein (CREB), a transcription factor regulating Arc expression. IL-1ß appears to act on BDNF signal transduction by impairing the phosphorylation of insulin receptor substrate 1, a protein that couples activation of the BDNF receptor TrkB to downstream signaling pathways regulating CREB, Arc, and cofilin. IL-1ß upregulated p38 mitogen-activated protein kinase (MAPK), and inhibiting p38 MAPK prevented IL-1ß from disrupting BDNF signaling. IL-1ß also prevented the formation of F-actin in spines and impaired the consolidation, but not the induction, of BDNF-dependent LTP in acute hippocampal slices. The suppressive effect of IL-1ß on F-actin and LTP was prevented by inhibiting p38 MAPK. These findings define a new mechanism for the action of IL-1ß on LTP and point to a potential therapeutic target to restore synaptic plasticity.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Interleucina-1beta/fisiología , Plasticidad Neuronal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Actinas/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Factor Neurotrófico Derivado del Encéfalo/fisiología , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Cofilina 1/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Regulación de la Expresión Génica/fisiología , Hipocampo/fisiología , Interleucina-1beta/farmacología , Potenciación a Largo Plazo/fisiología , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Neurobiol Dis ; 57: 47-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22750524

RESUMEN

Alzheimer's disease (AD) afflicts more than 5.4 million Americans and ranks as the most common type of dementia (Thies and Bleiler, 2011), yet effective pharmacological treatments have not been identified. Substantial evidence indicates that physical activity enhances learning and memory for people of all ages, including individuals that suffer from cognitive impairment. The mechanisms that underlie these benefits have been explored using animal models, including transgenic models of AD. Accumulating research shows that physical activity reinstates hippocampal function by enhancing the expression of brain-derived neurotrophic factor (BDNF) and other growth factors that promote neurogenesis, angiogenesis, and synaptic plasticity. In addition, several studies have found that physical activity counteracts age- and AD-associated declines in mitochondrial and immune system function. A growing body of evidence also suggests that exercise interventions hold the potential to reduce the pathological features associated with AD. Taken together, animal and human studies indicate that exercise provides a powerful stimulus that can countervail the molecular changes that underlie the progressive loss of hippocampal function in advanced age and AD.


Asunto(s)
Envejecimiento/fisiología , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Hipocampo/fisiopatología , Memoria/fisiología , Enfermedad de Alzheimer/inmunología , Animales , Modelos Animales de Enfermedad , Terapia por Ejercicio , Hipocampo/fisiología , Humanos , Sistema Inmunológico/inmunología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA