Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(6): e2315804121, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38294937

RESUMEN

Spontaneously occurring miniature excitatory postsynaptic currents (mEPSCs) are fundamental electrophysiological events produced by quantal vesicular transmitter release at synapses. Their analysis can provide important information regarding pre- and postsynaptic function. However, the small signal relative to recording noise requires expertise and considerable time for their identification. Furthermore, many mEPSCs smaller than ~8 pA are not well resolved (e.g., those produced at distant synapses or synapses with few receptor channels). Here, we describe an automated approach to detect mEPSCs using a machine learning-based tool. This method, which can be easily generalized to other one-dimensional signals, eliminates inter-observer bias, provides an estimate of its sensitivity and specificity and permits reliable detection of small (e.g., 5 pA) spontaneous unitary synaptic events.


Asunto(s)
Sinapsis , Transmisión Sináptica , Sinapsis/fisiología , Transmisión Sináptica/fisiología
2.
Front Genet ; 12: 647436, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34194466

RESUMEN

There is hope that genomic information will assist prediction, treatment, and understanding of Alzheimer's disease (AD). Here, using exome data from ∼10,000 individuals, we explore machine learning neural network (NN) methods to estimate the impact of SNPs (i.e., genetic variants) on AD risk. We develop an NN-based method (netSNP) that identifies hundreds of novel potentially protective or at-risk AD-associated SNPs (along with an effect measure); the majority with frequency under 0.01. For case individuals, the number of "protective" (or "at-risk") netSNP-identified SNPs in their genome correlates positively (or inversely) with their age of AD diagnosis and inversely (or positively) with autopsy neuropathology. The effect measure increases correlations. Simulations suggest our results are not due to genetic linkage, overfitting, or bias introduced by netSNP. These findings suggest that netSNP can identify SNPs associated with AD pathophysiology that may assist with the diagnosis and mechanistic understanding of the disease.

3.
Cell Rep ; 35(9): 109194, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34077732

RESUMEN

Beta-amyloid (Aß) depresses excitatory synapses by a poorly understood mechanism requiring NMDA receptor (NMDAR) function. Here, we show that increased PSD-95, a major synaptic scaffolding molecule, blocks the effects of Aß on synapses. The protective effect persists in tissue lacking the AMPA receptor subunit GluA1, which prevents the confounding synaptic potentiation by increased PSD-95. Aß modifies the conformation of the NMDAR C-terminal domain (CTD) and its interaction with protein phosphatase 1 (PP1), producing synaptic weakening. Higher endogenous levels or overexpression of PSD-95 block Aß-induced effects on the NMDAR CTD conformation, its interaction with PP1, and synaptic weakening. Our results indicate that increased PSD-95 protects synapses from Aß toxicity, suggesting that low levels of synaptic PSD-95 may be a molecular sign indicating synapse vulnerability to Aß. Importantly, pharmacological inhibition of its depalmitoylation increases PSD-95 at synapses and rescues deficits caused by Aß, possibly opening a therapeutic avenue against Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Homólogo 4 de la Proteína Discs Large/metabolismo , Neuroprotección , Sinapsis/metabolismo , Animales , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Homólogo 4 de la Proteína Discs Large/antagonistas & inhibidores , Transferencia Resonante de Energía de Fluorescencia , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroprotección/efectos de los fármacos , Ácido Palmítico/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Dominios Proteicos , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/efectos de los fármacos
4.
Neuroscience ; 456: 43-49, 2021 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32114099

RESUMEN

We recently demonstrated that NMDA receptors (NMDARs) are capable of ion-flux independent signaling through conformational change in the NMDAR intracellular domain resulting in long-term depression of synaptic transmission (LTD). Here we show that PSD-95 overexpression blocks agonist induced conformational movement in the NMDAR intracellular domain as well as LTD that is NMDAR-dependent and ion-flux independent. Interestingly, previous studies indicate that overexpressed PSD-95 does not block NMDAR-dependent LTD. These data support a model where ion-flux independent LTD is predominant in young animals, which have synapses with low amounts of PSD-95, whereas only ion flux dependent LTD occurs at more mature synapses, which have more PSD-95 that would block ion-flux independent LTD. These results may reconcile different findings regarding ion-flux independent LTD.


Asunto(s)
Hipocampo , Plasticidad Neuronal , Animales , Homólogo 4 de la Proteína Discs Large , Hipocampo/metabolismo , Depresión Sináptica a Largo Plazo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica
6.
Proc Natl Acad Sci U S A ; 117(6): 3214-3219, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-31974314

RESUMEN

Which neural circuits undergo synaptic changes when an animal learns? Although it is widely accepted that changes in synaptic strength underlie many forms of learning and memory, it remains challenging to connect changes in synaptic strength at specific neural pathways to specific behaviors and memories. Here we introduce SYNPLA (synaptic proximity ligation assay), a synapse-specific, high-throughput, and potentially brain-wide method capable of detecting circuit-specific learning-induced synaptic plasticity.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Aprendizaje/fisiología , Plasticidad Neuronal/fisiología , Mapeo de Interacción de Proteínas/métodos , Sinapsis , Animales , Corteza Auditiva/química , Corteza Auditiva/citología , Corteza Auditiva/metabolismo , Células Cultivadas , Condicionamiento Psicológico/fisiología , Cuerpos Geniculados/química , Cuerpos Geniculados/citología , Cuerpos Geniculados/metabolismo , Hipocampo/química , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Ratas , Sinapsis/química , Sinapsis/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(5): 2656-2662, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31941713

RESUMEN

Slow response to the standard treatment for depression increases suffering and risk of suicide. Ketamine, an N-methyl-d-aspartate (NMDA) receptor antagonist, can rapidly alleviate depressive symptoms and reduce suicidality, possibly by decreasing hyperactivity in the lateral habenula (LHb) brain nucleus. Here we find that in a rat model of human depression, opioid antagonists abolish the ability of ketamine to reduce the depression-like behavioral and LHb hyperactive cellular phenotypes. However, activation of opiate receptors alone is not sufficient to produce ketamine-like effects, nor does ketamine mimic the hedonic effects of an opiate, indicating that the opioid system does not mediate the actions of ketamine but rather is permissive. Thus, ketamine does not act as an opiate but its effects require both NMDA and opiate receptor signaling, suggesting that interactions between these two neurotransmitter systems are necessary to achieve an antidepressant effect.


Asunto(s)
Antidepresivos/administración & dosificación , Depresión/tratamiento farmacológico , Ketamina/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Animales , Depresión/genética , Depresión/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides/genética , Receptores Opioides/metabolismo
8.
Cell Rep ; 29(7): 1789-1799.e6, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31722197

RESUMEN

The transport and translation of dendritic mRNAs by RNA-binding proteins (RBPs) allows for spatially restricted gene expression in neuronal processes. Although local translation in neuronal dendrites is now well documented, there is little evidence for corresponding effects on local synaptic function. Here, we report that the RBP Sam68 promotes the localization and translation of Arc mRNA preferentially in distal dendrites of rodent hippocampal CA1 pyramidal neurons. Consistent with Arc function in translation-dependent synaptic plasticity, we find that Sam68 knockout (KO) mice display impaired metabotropic glutamate-receptor-dependent long-term depression (mGluR-LTD) and impaired structural plasticity exclusively at distal Schaffer-collateral synapses. Moreover, by using quantitative proteomics, we find that the Sam68 interactome contains numerous regulators of mRNA translation and synaptic function. This work identifies an important player in Arc expression, provides a general framework for Sam68 regulation of protein synthesis, and uncovers a mechanism that enables the precise spatiotemporal expression of long-term plasticity throughout neurons.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Región CA1 Hipocampal/metabolismo , Dendritas/metabolismo , Depresión Sináptica a Largo Plazo , Biosíntesis de Proteínas , Células Piramidales/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Región CA1 Hipocampal/citología , Femenino , Ratones , Ratones Noqueados , Células Piramidales/citología , Proteínas de Unión al ARN/genética
9.
Proc Natl Acad Sci U S A ; 116(25): 12488-12493, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31152135

RESUMEN

Neuronal activity in the lateral habenula (LHb), a brain region implicated in depression [C. D. Proulx, O. Hikosaka, R. Malinow, Nat. Neurosci. 17, 1146-1152 (2014)], decreases during reward and increases during punishment or reward omission [M. Matsumoto, O. Hikosaka, Nature 447, 1111-1115 (2007)]. While stress is a major risk factor for depression and strongly impacts the LHb, its effect on LHb reward signals is unknown. Here we image LHb neuronal activity in behaving mice and find that acute stress transforms LHb reward responses into punishment-like neural signals; punishment-like responses to reward omission also increase. These neural changes matched the onset of anhedonic behavior and were specific to LHb neurons that distinguished reward and its omission. Thus, stress distorts LHb responsivity to positive and negative feedback, which could bias individuals toward negative expectations, a key aspect of the proposed pathogenesis of depression [A. T. Beck, Depression: Clinical, Experimental, and Theoretical Aspects, sixth Ed (1967)].


Asunto(s)
Habénula/fisiología , Castigo , Recompensa , Estrés Psicológico , Animales , Ratones , Neuronas/fisiología
10.
Biochem Pharmacol ; 159: 140-153, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30503374

RESUMEN

N-methyl-d-aspartate receptors (NMDARs) have multiple prominent roles in CNS function but their excessive or insufficient activity contributes to neuropathological/psychiatric disorders. Consequently, a variety of positive and negative allosteric modulators (PAMs and NAMs, respectively) have recently been developed. Although these modulators bind to extracellular domains, in the present report we find that the NMDAR's intracellular C-terminal domains (CTDs) significantly influence PAM/NAM activity. GluN2 CTD deletion robustly affected NAM and PAM activity with both enhancing and inhibiting effects that were compound-specific and NMDAR subunit-specific. In three cases, individual PAMs became NAMs at specific GluN2-truncated receptors. In contrast to GluN2, GluN1 CTD removal only reduced PAM activity of UBP684 and CIQ, and did not affect NAM activity. Consistent with these findings, agents altering phosphorylation state or intracellular calcium levels displayed receptor-specific and compound-specific effects on PAM activity. It is possible that the GluN2's M4 domain transmits intracellular modulatory signals from the CTD to the M1/M4 channel gating machinery and that this site is a point of convergence in the direct or indirect actions of several PAMs/NAMs thus rendering them sensitive to CTD status. Thus, allosteric modulators are likely to have a marked and varied sensitivity to post-translational modifications, protein-protein associations, and intracellular ions. The interaction between PAM activity and NMDAR CTDs appears reciprocal. GluN1 CTD-deletion eliminated UBP684, but not pregnenolone sulfate (PS), PAM activity. And, in the absence of agonists, UBP684, but not PS, was able to promote movement of fluorescently-tagged GluN1-CTDs. Thus, it may be possible to pharmacologically target NMDAR metabotropic activity in the absence of channel activation.


Asunto(s)
Ácidos Carboxílicos/farmacología , Naftalenos/farmacología , Pregnenolona/farmacología , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Calcio/metabolismo , Femenino , Transferencia Resonante de Energía de Fluorescencia , Neuronas/citología , Neuronas/fisiología , Oocitos/efectos de los fármacos , Dominios Proteicos , Subunidades de Proteína , Ratas , Receptores de N-Metil-D-Aspartato/genética , Xenopus laevis
11.
Acta Neuropathol Commun ; 6(1): 110, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30352630

RESUMEN

Amyloid beta (Aß)-mediated synapse dysfunction and spine loss are considered to be early events in Alzheimer's disease (AD) pathogenesis. N-methyl-D-aspartate receptors (NMDARs) have previously been suggested to play a role for Amyloid beta (Aß) toxicity. Pharmacological block of NMDAR subunits in cultured neurons and mice suggested that NMDARs containing the GluN2B subunit are necessary for Aß-mediated changes in synapse number and function in hippocampal neurons. Interestingly, NMDARs undergo a developmental switch from GluN2B- to GluN2A-containing receptors. This indicates different functional roles of NMDARs in young mice compared to older animals. In addition, the lack of pharmacological tools to efficiently dissect the role of NMDARs containing the different subunits complicates the interpretation of their specific role. In order to address this problem and to investigate the specific role for Aß toxicity of the distinct NMDAR subunits in dentate gyrus granule cells of adult mice, we used conditional knockout mouse lines for the subunits GluN1, GluN2A and GluN2B. Aß-mediated changes in synaptic function and neuronal anatomy were investigated in several-months old mice with virus-mediated overproduction of Aß and in 1-year old 5xFAD mice. We found that all three NMDAR subunits contribute to the Aß-mediated decrease in the number of functional synapses. However, NMDARs are not required for the spine number reduction in dentate gyrus granule cells after chronic Aß-overproduction in 5xFAD mice. Furthermore, the amplitude of synaptic and extrasynaptic NMDAR-mediated currents was reduced in dentate gyrus granule of 5xFAD mice without changes in current kinetics, suggesting that a redistribution or change in subunit composition of NMDARs does not play a role in mediating Amyloid beta (Aß) toxicity. Our study indicates that NMDARs are involved in AD pathogenesis by compromising synapse function but not by affecting neuron morphology.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Espinas Dendríticas/patología , Giro Dentado/citología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Modelos Animales de Enfermedad , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/ultraestructura , Presenilina-1/genética , Receptores de N-Metil-D-Aspartato/genética , Sinapsis/efectos de los fármacos
12.
Proc Natl Acad Sci U S A ; 115(22): 5792-5797, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29752382

RESUMEN

The neural mechanisms conferring reduced motivation, as observed in depressed individuals, is poorly understood. Here, we examine in rodents if reduced motivation to exert effort is controlled by transmission from the lateral habenula (LHb), a nucleus overactive in depressed-like states, to the rostromedial tegmental nucleus (RMTg), a nucleus that inhibits dopaminergic neurons. In an aversive test wherein immobility indicates loss of effort, LHb→RMTg transmission increased during transitions into immobility, driving LHb→RMTg increased immobility, and inhibiting LHb→RMTg produced the opposite effects. In an appetitive test, driving LHb→RMTg reduced the effort exerted to receive a reward, without affecting the reward's hedonic property. Notably, LHb→RMTg stimulation only affected specific aspects of these motor tasks, did not affect all motor tasks, and promoted avoidance, indicating that LHb→RMTg activity does not generally reduce movement but appears to carry a negative valence that reduces effort. These results indicate that LHb→RMTg activity controls the motivation to exert effort and may contribute to the reduced motivation in depression.


Asunto(s)
Habénula/fisiología , Motivación/fisiología , Vías Nerviosas/fisiología , Tegmento Mesencefálico/fisiología , Animales , Depresión , Humanos , Movimiento/fisiología , Optogenética , Fotometría , Ratas , Análisis y Desempeño de Tareas
13.
J Neurosci ; 37(25): 6021-6030, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28539417

RESUMEN

The lateral habenula (LHb) is a small part of the epithalamus that projects to monoamine centers in the brain. Previously, neurotransmission onto the LHb was shown to be abnormally potentiated in animal models of depression. However, synaptic plasticity in this brain area and the effect of stressor exposure on synaptic plasticity of the LHb have not been investigated. Thus, we explored whether the LHb undergoes dynamic changes in synaptic efficacy or not. First, we observed that a moderate LTP occurs in a fraction of LHb neurons obtained from naive Sprague Dawley rats. Interestingly, a single exposure to acute stressors, such as inescapable foot shock or restraint plus tail shock (RTS), significantly enhances the magnitude of LTP in the LHb. We also observed an increased number of LHb neurons expressing phosphorylated cAMP response element-binding protein (pCREB) after exposure to stressors, which may contribute to determine the threshold for LTP induction. LTP induction in the LHb resulted in an additional increase in the number of pCREB-expressing neurons in stress-exposed animals but not in naive control animals. Together, we showed that LHb neurons have heterogeneous propensity for synaptic potentiation at rest; however, a single exposure to stressors greatly facilitates LTP induction in the LHb, suggesting that fundamental alterations in synaptic plasticity in the LHb may occur in animal models of depression or post-traumatic stress disorder.SIGNIFICANCE STATEMENT Stress exposure is known to cause depression in human patients and animal models, although explanations at the cellular level remain to be elaborated. Here, we show that the lateral habenula (LHb) exhibits LTP after a pattern of brief strong stimulation. In addition, we show that stress exposure facilitates LTP in the LHb by lowering the threshold for LTP induction. We observed a selective increase in the number of neurons expressing pCREB in the LHb of animal models of depression. LTP induction results in a further increase in the density of pCREB-expressing neurons only after stress exposure. Our study provides the first evidence that animal models of depression exhibit altered synaptic plasticity of the LHb.


Asunto(s)
Habénula/fisiopatología , Potenciación a Largo Plazo , Estrés Psicológico/fisiopatología , Sinapsis , Animales , Ansiedad/fisiopatología , Ansiedad/psicología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Electrochoque , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Restricción Física , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/fisiopatología , Estrés Psicológico/psicología
14.
J Biol Methods ; 4(3)2017.
Artículo en Inglés | MEDLINE | ID: mdl-29457040

RESUMEN

Robust assays for detecting the effects of elevated concentrations of amyloid-ß (Aß) may facilitate Alzheimer's disease research. An appropriate assay would be high-throughput and enable identification of drugs and genetic mutations that block the effects of Aß, potentially leading to treatments for Alzheimer's disease. We discovered that the commonly used cytomegalovirus (CMV) enhancer/promoter is sensitive to the effects of Aß. By combining the CMV enhancer/promoter with a fluorescent protein, we created a reporter system that produces changes in intracellular fluorescence in response to Aß. Using hippocampal neurons, we quantified the ability of a CMV-fluorescent protein recombinant reporter to detect both exogenously applied and overexpressed Aß. This is the first report of a high-throughput enhancer/promoter-based Aß detection method. The reporter is able to detect the effects of elevated concentrations of Aß in a high-throughput fashion, providing a new tool for Alzheimer's disease research and important knowledge about the commonly used CMV enhancer/promoter.

15.
Proc Natl Acad Sci U S A ; 113(42): E6526-E6534, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27708157

RESUMEN

Amyloid-ß (Aß) is a prime suspect for causing cognitive deficits during the early phases of Alzheimer's disease (AD). Experiments in AD mouse models have shown that soluble oligomeric clusters of Aß degrade synapses and impair memory formation. We show that all Aß-driven effects measured in these mice depend on AMPA receptor (AMPAR) subunit GluA3. Hippocampal neurons that lack GluA3 were resistant against Aß-mediated synaptic depression and spine loss. In addition, Aß oligomers blocked long-term synaptic potentiation only in neurons that expressed GluA3. Furthermore, although Aß-overproducing mice showed significant memory impairment, memories in GluA3-deficient congenics remained unaffected. These experiments indicate that the presence of GluA3-containing AMPARs is critical for Aß-mediated synaptic and cognitive deficits.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Memoria , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/mortalidad , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Análisis de Varianza , Animales , Conducta Animal , Células CHO , Condicionamiento Psicológico , Cricetulus , Espinas Dendríticas , Miedo/psicología , Femenino , Hipocampo/citología , Hipocampo/fisiología , Potenciación a Largo Plazo , Masculino , Potenciales de la Membrana , Ratones , Ratones Noqueados , Ratones Transgénicos , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Células Piramidales/citología , Células Piramidales/metabolismo , Receptores AMPA/genética
16.
Artículo en Inglés | MEDLINE | ID: mdl-27516738

RESUMEN

The NMDA receptor (R) participates in many important physiological and pathological processes. For example, its activation is required for both long-term potentiation (LTP) and long-term depression (LTD) of synaptic transmission, cellular models of learning and memory. Furthermore, it may play a role in the actions of amyloid-beta on synapses as well as in the signaling leading to cell death following stroke. Until recently, these processes were thought to be mediated by ion-flux through the receptor. Using a combination of imaging and electrophysiological approaches, ion-flux independent functions of the NMDAR were recently examined. In this review, we will discuss the role of metabotropic NMDAR function in LTD and synaptic dysfunction.

17.
Sci Signal ; 9(427): ra47, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27165780

RESUMEN

Alzheimer's disease (AD) is a progressive dementia disorder characterized by synaptic degeneration and amyloid-ß (Aß) accumulation in the brain. Through whole-genome sequencing of 1345 individuals from 410 families with late-onset AD (LOAD), we identified three highly penetrant variants in PRKCA, the gene that encodes protein kinase Cα (PKCα), in five of the families. All three variants linked with LOAD displayed increased catalytic activity relative to wild-type PKCα as assessed in live-cell imaging experiments using a genetically encoded PKC activity reporter. Deleting PRKCA in mice or adding PKC antagonists to mouse hippocampal slices infected with a virus expressing the Aß precursor CT100 revealed that PKCα was required for the reduced synaptic activity caused by Aß. In PRKCA(-/-) neurons expressing CT100, introduction of PKCα, but not PKCα lacking a PDZ interaction moiety, rescued synaptic depression, suggesting that a scaffolding interaction bringing PKCα to the synapse is required for its mediation of the effects of Aß. Thus, enhanced PKCα activity may contribute to AD, possibly by mediating the actions of Aß on synapses. In contrast, reduced PKCα activity is implicated in cancer. Hence, these findings reinforce the importance of maintaining a careful balance in the activity of this enzyme.


Asunto(s)
Enfermedad de Alzheimer/genética , Mutación , Proteína Quinasa C-alfa/genética , Sinapsis/patología , Animales , Células COS , Chlorocebus aethiops , Salud de la Familia , Genoma , Genoma Humano , Hipocampo/metabolismo , Humanos , Ratones , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Dominios Proteicos
19.
Biol Psychiatry ; 80(11): 827-835, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27113500

RESUMEN

BACKGROUND: Major depressive disorder is associated with disturbed circadian rhythms. To investigate the causal relationship between mood disorders and circadian clock disruption, previous studies in animal models have employed light/dark manipulations, global mutations of clock genes, or brain area lesions. However, light can impact mood by noncircadian mechanisms; clock genes have pleiotropic, clock-independent functions; and brain lesions not only disrupt cellular circadian rhythms but also destroy cells and eliminate important neuronal connections, including light reception pathways. Thus, a definitive causal role for functioning circadian clocks in mood regulation has not been established. METHODS: We stereotactically injected viral vectors encoding short hairpin RNA to knock down expression of the essential clock gene Bmal1 into the brain's master circadian pacemaker, the suprachiasmatic nucleus (SCN). RESULTS: In these SCN-specific Bmal1-knockdown (SCN-Bmal1-KD) mice, circadian rhythms were greatly attenuated in the SCN, while the mice were maintained in a standard light/dark cycle, SCN neurons remained intact, and neuronal connections were undisturbed, including photic inputs. In the learned helplessness paradigm, the SCN-Bmal1-KD mice were slower to escape, even before exposure to inescapable stress. They also spent more time immobile in the tail suspension test and less time in the lighted section of a light/dark box. The SCN-Bmal1-KD mice also showed greater weight gain, an abnormal circadian pattern of corticosterone, and an attenuated increase of corticosterone in response to stress. CONCLUSIONS: Disrupting SCN circadian rhythms is sufficient to cause helplessness, behavioral despair, and anxiety-like behavior in mice, establishing SCN-Bmal1-KD mice as a new animal model of depression.


Asunto(s)
Ansiedad/etiología , Conducta Animal/fisiología , Trastornos Cronobiológicos/complicaciones , Ritmo Circadiano/fisiología , Depresión/etiología , Modelos Animales de Enfermedad , Núcleo Supraquiasmático/fisiopatología , Factores de Transcripción ARNTL , Animales , Trastornos Cronobiológicos/genética , Ritmo Circadiano/genética , Desamparo Adquirido , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
20.
Proc Natl Acad Sci U S A ; 112(47): 14711-6, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26553983

RESUMEN

The NMDA receptor (NMDAR) is known to transmit important information by conducting calcium ions. However, some recent studies suggest that activation of NMDARs can trigger synaptic plasticity in the absence of ion flow. Does ligand binding transmit information to signaling molecules that mediate synaptic plasticity? Using Förster resonance energy transfer (FRET) imaging of fluorescently tagged proteins expressed in neurons, conformational signaling is identified within the NMDAR complex that is essential for downstream actions. Ligand binding transiently reduces FRET between the NMDAR cytoplasmic domain (cd) and the associated protein phosphatase 1 (PP1), requiring NMDARcd movement, and persistently reduces FRET between the NMDARcd and calcium/calmodulin-dependent protein kinase II (CaMKII), a process requiring PP1 activity. These studies directly monitor agonist-driven conformational signaling at the NMDAR complex required for synaptic plasticity.


Asunto(s)
Plasticidad Neuronal , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Animales , Anticuerpos/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Transporte Iónico/efectos de los fármacos , Modelos Biológicos , N-Metilaspartato/farmacología , Plasticidad Neuronal/efectos de los fármacos , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/química , Transducción de Señal/efectos de los fármacos , Sinapsis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...