Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 133(4): 666-80, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18485874

RESUMEN

The role of cell size and shape in controlling local intracellular signaling reactions, and how this spatial information originates and is propagated, is not well understood. We have used partial differential equations to model the flow of spatial information from the beta-adrenergic receptor to MAPK1,2 through the cAMP/PKA/B-Raf/MAPK1,2 network in neurons using real geometries. The numerical simulations indicated that cell shape controls the dynamics of local biochemical activity of signal-modulated negative regulators, such as phosphodiesterases and protein phosphatases within regulatory loops to determine the size of microdomains of activated signaling components. The model prediction that negative regulators control the flow of spatial information to downstream components was verified experimentally in rat hippocampal slices. These results suggest a mechanism by which cellular geometry, the presence of regulatory loops with negative regulators, and key reaction rates all together control spatial information transfer and microdomain characteristics within cells.


Asunto(s)
Forma de la Célula , Sistema de Señalización de MAP Quinasas , Neuronas/metabolismo , Animales , Aplysia , AMP Cíclico/metabolismo , Retroalimentación Fisiológica , Feto , Hipocampo/citología , Isoproterenol/metabolismo , Redes y Vías Metabólicas , Modelos Biológicos , Neuronas/citología , Neuronas/enzimología , Ratas , Receptores Adrenérgicos beta 2/metabolismo
2.
PLoS Genet ; 14(1): e1007169, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29364887

RESUMEN

Dystonia is characterized by involuntary muscle contractions. Its many forms are genetically, phenotypically and etiologically diverse and it is unknown whether their pathogenesis converges on shared pathways. Mutations in THAP1 [THAP (Thanatos-associated protein) domain containing, apoptosis associated protein 1], a ubiquitously expressed transcription factor with DNA binding and protein-interaction domains, cause dystonia, DYT6. There is a unique, neuronal 50-kDa Thap1-like immunoreactive species, and Thap1 levels are auto-regulated on the mRNA level. However, THAP1 downstream targets in neurons, and the mechanism via which it causes dystonia are largely unknown. We used RNA-Seq to assay the in vivo effect of a heterozygote Thap1 C54Y or ΔExon2 allele on the gene transcription signatures in neonatal mouse striatum and cerebellum. Enriched pathways and gene ontology terms include eIF2α Signaling, Mitochondrial Dysfunction, Neuron Projection Development, Axonal Guidance Signaling, and Synaptic LongTerm Depression, which are dysregulated in a genotype and tissue-dependent manner. Electrophysiological and neurite outgrowth assays were consistent with those enrichments, and the plasticity defects were partially corrected by salubrinal. Notably, several of these pathways were recently implicated in other forms of inherited dystonia, including DYT1. We conclude that dysfunction of these pathways may represent a point of convergence in the pathophysiology of several forms of inherited dystonia.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al ADN/genética , Distonía/genética , Mutación , Red Nerviosa/fisiología , Neuronas/fisiología , Proteínas Nucleares/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Humanos , Células K562 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/metabolismo , Plasticidad Neuronal/genética
3.
Acta Neuropathol ; 140(3): 295-315, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32666270

RESUMEN

MicroRNAs are recognized as important regulators of many facets of physiological brain function while also being implicated in the pathogenesis of several neurological disorders. Dysregulation of miR155 is widely reported across a variety of neurodegenerative conditions, including Alzheimer's disease (AD), Parkinson's disease, amyotrophic lateral sclerosis, and traumatic brain injury. In previous work, we observed that experimentally validated miR155 gene targets were consistently enriched among genes identified as differentially expressed across multiple brain tissue and disease contexts. In particular, we found that human herpesvirus-6A (HHV-6A) suppressed miR155, recapitulating reports of miR155 inhibition by HHV-6A in infected T-cells, thyrocytes, and natural killer cells. In earlier studies, we also reported the effects of constitutive deletion of miR155 on accelerating the accumulation of Aß deposits in 4-month-old APP/PSEN1 mice. Herein, we complete the cumulative characterization of transcriptomic, electrophysiological, neuropathological, and learning behavior profiles from 4-, 8- and 10-month-old WT and APP/PSEN1 mice in the absence or presence of miR155. We also integrated human post-mortem brain RNA-sequences from four independent AD consortium studies, together comprising 928 samples collected from six brain regions. We report that gene expression perturbations associated with miR155 deletion in mouse cortex are in aggregate observed to be concordant with AD-associated changes across these independent human late-onset AD (LOAD) data sets, supporting the relevance of our findings to human disease. LOAD has recently been formulated as the clinicopathological manifestation of a multiplex of genetic underpinnings and pathophysiological mechanisms. Our accumulated data are consistent with such a formulation, indicating that miR155 may be uniquely positioned at the intersection of at least four components of this LOAD "multiplex": (1) innate immune response pathways; (2) viral response gene networks; (3) synaptic pathology; and (4) proamyloidogenic pathways involving the amyloid ß peptide (Aß).


Asunto(s)
Enfermedad de Alzheimer/genética , Encéfalo/patología , MicroARNs/genética , Transcriptoma/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Redes Reguladoras de Genes/genética , Humanos , Ratones Transgénicos , Enfermedades del Sistema Nervioso/patología , Placa Amiloide/patología
4.
Mol Psychiatry ; 24(9): 1383-1397, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30283031

RESUMEN

TYROBP/DAP12 forms complexes with ectodomains of immune receptors (TREM2, SIRPß1, CR3) associated with Alzheimer's disease (AD) and is a network hub and driver in the complement subnetwork identified by multi-scale gene network studies of postmortem human AD brain. Using transgenic or viral approaches, we characterized in mice the effects of TYROBP deficiency on the phenotypic and pathological evolution of tauopathy. Biomarkers usually associated with worsening clinical phenotype (i.e., hyperphosphorylation and increased tauopathy spreading) were unexpectedly increased in MAPTP301S;Tyrobp-/- mice despite the improved learning behavior and synaptic function relative to controls with normal levels of TYROBP. Notably, levels of complement cascade initiator C1q were reduced in MAPTP301S;Tyrobp-/- mice, consistent with the prediction that C1q reduction exerts a neuroprotective effect. These observations suggest a model wherein TYROBP-KO-(knock-out)-associated reduction in C1q is associated with normalized learning behavior and electrophysiological properties in tauopathy model mice despite a paradoxical evolution of biomarker signatures usually associated with neurological decline.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/metabolismo , Complemento C1q/metabolismo , Complemento C1q/fisiología , Modelos Animales de Enfermedad , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Fenotipo , Fosforilación , Placa Amiloide/metabolismo , Tauopatías/genética , Proteínas tau/metabolismo
5.
Mol Psychiatry ; 24(3): 431-446, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30283032

RESUMEN

Integrative gene network approaches enable new avenues of exploration that implicate causal genes in sporadic late-onset Alzheimer's disease (LOAD) pathogenesis, thereby offering novel insights for drug-discovery programs. We previously constructed a probabilistic causal network model of sporadic LOAD and identified TYROBP/DAP12, encoding a microglial transmembrane signaling polypeptide and direct adapter of TREM2, as the most robust key driver gene in the network. Here, we show that absence of TYROBP/DAP12 in a mouse model of AD-type cerebral Aß amyloidosis (APPKM670/671NL/PSEN1Δexon9) recapitulates the expected network characteristics by normalizing the transcriptome of APP/PSEN1 mice and repressing the induction of genes involved in the switch from homeostatic microglia to disease-associated microglia (DAM), including Trem2, complement (C1qa, C1qb, C1qc, and Itgax), Clec7a and Cst7. Importantly, we show that constitutive absence of TYROBP/DAP12 in the amyloidosis mouse model prevented appearance of the electrophysiological and learning behavior alterations associated with the phenotype of APPKM670/671NL/PSEN1Δexon9 mice. Our results suggest that TYROBP/DAP12 could represent a novel therapeutic target to slow, arrest, or prevent the development of sporadic LOAD. These data establish that the network pathology observed in postmortem human LOAD brain can be faithfully recapitulated in the brain of a genetically manipulated mouse. These data also validate our multiscale gene networks by demonstrating how the networks intersect with the standard neuropathological features of LOAD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Amiloidosis/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/genética , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Redes Reguladoras de Genes , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Patología Molecular/métodos , Fenotipo , Placa Amiloide/patología , Transcriptoma
6.
Mol Psychiatry ; 24(3): 472, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30464330

RESUMEN

This article was originally published under standard licence, but has now been made available under a CC BY 4.0 license. The PDF and HTML versions of the paper have been modified accordingly.

7.
Acta Neuropathol ; 134(5): 769-788, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28612290

RESUMEN

Conventional genetic approaches and computational strategies have converged on immune-inflammatory pathways as key events in the pathogenesis of late onset sporadic Alzheimer's disease (LOAD). Mutations and/or differential expression of microglial specific receptors such as TREM2, CD33, and CR3 have been associated with strong increased risk for developing Alzheimer's disease (AD). DAP12 (DNAX-activating protein 12)/TYROBP, a molecule localized to microglia, is a direct partner/adapter for TREM2, CD33, and CR3. We and others have previously shown that TYROBP expression is increased in AD patients and in mouse models. Moreover, missense mutations in the coding region of TYROBP have recently been identified in some AD patients. These lines of evidence, along with computational analysis of LOAD brain gene expression, point to DAP12/TYROBP as a potential hub or driver protein in the pathogenesis of AD. Using a comprehensive panel of biochemical, physiological, behavioral, and transcriptomic assays, we evaluated in a mouse model the role of TYROBP in early stage AD. We crossed an Alzheimer's model mutant APP KM670/671NL /PSEN1 Δexon9 (APP/PSEN1) mouse model with Tyrobp -/- mice to generate AD model mice deficient or null for TYROBP (APP/PSEN1; Tyrobp +/- or APP/PSEN1; Tyrobp -/-). While we observed relatively minor effects of TYROBP deficiency on steady-state levels of amyloid-ß peptides, there was an effect of Tyrobp deficiency on the morphology of amyloid deposits resembling that reported by others for Trem2 -/- mice. We identified modulatory effects of TYROBP deficiency on the level of phosphorylation of TAU that was accompanied by a reduction in the severity of neuritic dystrophy. TYROBP deficiency also altered the expression of several AD related genes, including Cd33. Electrophysiological abnormalities and learning behavior deficits associated with APP/PSEN1 transgenes were greatly attenuated on a Tyrobp-null background. Some modulatory effects of TYROBP on Alzheimer's-related genes were only apparent on a background of mice with cerebral amyloidosis due to overexpression of mutant APP/PSEN1. These results suggest that reduction of TYROBP gene expression and/or protein levels could represent an immune-inflammatory therapeutic opportunity for modulating early stage LOAD, potentially leading to slowing or arresting the progression to full-blown clinical and pathological LOAD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Enfermedad de Alzheimer/genética , Encéfalo/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Mutación , Fosforilación , Proteínas tau/metabolismo
8.
Nature ; 469(7331): 491-7, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21270887

RESUMEN

We report that, in the rat, administering insulin-like growth factor II (IGF-II, also known as IGF2) significantly enhances memory retention and prevents forgetting. Inhibitory avoidance learning leads to an increase in hippocampal expression of IGF-II, which requires the transcription factor CCAAT enhancer binding protein ß and is essential for memory consolidation. Furthermore, injections of recombinant IGF-II into the hippocampus after either training or memory retrieval significantly enhance memory retention and prevent forgetting. To be effective, IGF-II needs to be administered within a sensitive period of memory consolidation. IGF-II-dependent memory enhancement requires IGF-II receptors, new protein synthesis, the function of activity-regulated cytoskeletal-associated protein and glycogen-synthase kinase 3 (GSK3). Moreover, it correlates with a significant activation of synaptic GSK3ß and increased expression of GluR1 (also known as GRIA1) α-amino-3-hydroxy-5-methyl-4-isoxasolepropionic acid receptor subunits. In hippocampal slices, IGF-II promotes IGF-II receptor-dependent, persistent long-term potentiation after weak synaptic stimulation. Thus, IGF-II may represent a novel target for cognitive enhancement therapies.


Asunto(s)
Hipocampo/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Memoria/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Hipocampo/efectos de los fármacos , Factor II del Crecimiento Similar a la Insulina/farmacología , Potenciación a Largo Plazo/fisiología , Masculino , Memoria/efectos de los fármacos , Ratas , Ratas Long-Evans , Factores de Tiempo
9.
J Neurosci ; 35(32): 11190-5, 2015 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-26269629

RESUMEN

Parkinson's disease (PD) is a major movement disorder characterized by the loss of dopamine neurons and formation of Lewy bodies. Clinical and pathological evidence indicates that multiple brain regions are affected in PD in a spatiotemporal manner and are associated with a variety of motor and nonmotor symptoms, including disturbances in mood, executive function, and memory. The common PD-associated gene for leucine-rich repeat kinase, leucine-rich repeat kinase 2 (LRRK2), is highly expressed in brain regions that are involved with nonmotor functions, including the neocortex and hippocampus, but whether mutant LRRK2 contributes to neuronal dysfunction in these regions is unknown. Here, we use bacterial artificial chromosome transgenic mouse models of LRRK2 to explore potential nonmotor mechanisms of PD. Through electrophysiological analysis of the Schaffer collateral-CA1 synapse in dorsal hippocampus, we find that overexpression of LRRK2-G2019S increases basal synaptic efficiency through a postsynaptic mechanism, and disrupts long-term depression. Furthermore, these effects of the G2019S mutation are age dependent and can be normalized by acute inhibition of LRRK2 kinase activity. In contrast, overexpression of wild-type LRRK2 has no effect under the same conditions, suggesting a specific phenotype for the G2019S mutation. These results identify a pathogenic function of LRRK2 in the hippocampus that may contribute to nonmotor symptoms of PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is among the most common neurological diseases and is best known for its adverse effects on brain regions that control motor function, resulting in tremor, rigidity, and gait abnormalities. Less well appreciated are the psychiatric symptoms experienced by many PD patients, including depression and memory loss, which do not respond well to currently available treatments for PD. Here, we describe functional effects of a common PD-linked mutation of leucine-rich repeat kinase 2 in the mouse hippocampus, an area of the brain that is responsible for encoding and retaining memories. By providing a potential mechanism for some of the cognitive symptoms produced by this mutation, our findings may lead to novel approaches for the treatment of nonmotor symptoms of PD.


Asunto(s)
Hipocampo/fisiopatología , Mutación , Plasticidad Neuronal/genética , Enfermedad de Parkinson/fisiopatología , Proteínas Serina-Treonina Quinasas/genética , Factores de Edad , Animales , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/genética , Fenotipo , Transmisión Sináptica/genética
10.
Pain ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38985454

RESUMEN

ABSTRACT: Preclinical and clinical work has demonstrated altered plasticity and activity in the nucleus accumbens (NAc) under chronic pain states, highlighting critical therapeutic avenues for the management of chronic pain conditions. In this study, we demonstrate that myocyte enhancer factor 2C (MEF2C), a master regulator of neuronal activity and plasticity, is repressed in NAc neurons after prolonged spared nerve injury (SNI). Viral-mediated overexpression of Mef2c in NAc neurons partially ameliorated sensory hypersensitivity and emotional behaviors in mice with SNI, while also altering transcriptional pathways associated with synaptic signaling. Mef2c overexpression also reversed SNI-induced potentiation of phasic dopamine release and neuronal hyperexcitability in the NAc. Transcriptional changes induced by Mef2c overexpression were different than those observed after desipramine treatment, suggesting a mechanism of action different from antidepressants. Overall, we show that interventions in MEF2C-regulated mechanisms in the NAc are sufficient to disrupt the maintenance of chronic pain states, providing potential new treatment avenues for neuropathic pain.

11.
Front Behav Neurosci ; 17: 1130840, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37830039

RESUMEN

The hippocampus is critical for the precise formation of contextual memories. Overlapping inputs coming from the entorhinal cortex are processed by the trisynaptic pathway to form distinct memories. Disruption in any step of the circuit flow can lead to a lack of memory precision, and to memory interference. We have identified the transcriptional repressor Wilm's Tumor 1 (WT1) as an important regulator of synaptic plasticity involved in memory discrimination in the hippocampus. In male mice, using viral and transgenic approaches, we showed that WT1 deletion in granule cells of the dentate gyrus (DG) disrupts memory discrimination. With electrophysiological methods, we then identified changes in granule cells' excitability and DG synaptic transmission indicating that WT1 knockdown in DG granule cells disrupts the inhibitory feedforward input from mossy fibers to CA3 by decreasing mIPSCs and shifting the normal excitatory/inhibitory (E/I) balance in the DG → CA3 circuit in favor of excitation. Finally, using a chemogenetic approach, we established a causal link between granule cell hyperexcitability and memory discrimination impairments. Our results suggest that WT1 enables a circuit-level computation that drives pattern discrimination behavior.

12.
bioRxiv ; 2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37546856

RESUMEN

The hippocampus 1-7, as well as dopamine circuits 8-11, coordinate decision-making in anxiety-eliciting situations. Yet, little is known about how dopamine modulates hippocampal representations of emotionally-salient stimuli to inform appropriate resolution of approach versus avoidance conflicts. We here study dopaminoceptive neurons in mouse ventral hippocampus (vHipp), molecularly distinguished by their expression of dopamine D1 or D2 receptors. We show that these neurons are transcriptionally distinct and topographically organized across vHipp subfields and cell types. In the ventral subiculum where they are enriched, both D1 and D2 neurons are recruited during anxiogenic exploration, yet with distinct profiles related to investigation and behavioral selection. In turn, they mediate opposite approach/avoidance responses, and are differentially modulated by dopaminergic transmission in that region. Together, these results suggest that vHipp dopamine dynamics gate exploratory behaviors under contextual uncertainty, implicating dopaminoception in the complex computation engaged in vHipp to govern emotional states.

13.
Front Pharmacol ; 14: 1225759, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37799971

RESUMEN

There are no known drugs or drug combinations that promote substantial central nervous system axonal regeneration after injury. We used systems pharmacology approaches to model pathways underlying axonal growth and identify a four-drug combination that regulates multiple subcellular processes in the cell body and axons using the optic nerve crush model in rats. We intravitreally injected agonists HU-210 (cannabinoid receptor-1) and IL-6 (interleukin 6 receptor) to stimulate retinal ganglion cells for axonal growth. We applied, in gel foam at the site of nerve injury, Taxol to stabilize growing microtubules, and activated protein C to clear the debris field since computational models predicted that this drug combination regulating two subcellular processes at the growth cone produces synergistic growth. Physiologically, drug treatment restored or preserved pattern electroretinograms and some of the animals had detectable visual evoked potentials in the brain and behavioral optokinetic responses. Morphology experiments show that the four-drug combination protects axons or promotes axonal regrowth to the optic chiasm and beyond. We conclude that spatially targeted drug treatment is therapeutically relevant and can restore limited functional recovery.

14.
bioRxiv ; 2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37205394

RESUMEN

Hyperexcitability in the orbitofrontal cortex (OFC) is a key clinical feature of anhedonic domains of Major Depressive Disorder (MDD). However, the cellular and molecular substrates underlying this dysfunction remain unknown. Here, cell-population-specific chromatin accessibility profiling in human OFC unexpectedly mapped genetic risk for MDD exclusively to non-neuronal cells, and transcriptomic analyses revealed significant glial dysregulation in this region. Characterization of MDD-specific cis-regulatory elements identified ZBTB7A - a transcriptional regulator of astrocyte reactivity - as an important mediator of MDD-specific chromatin accessibility and gene expression. Genetic manipulations in mouse OFC demonstrated that astrocytic Zbtb7a is both necessary and sufficient to promote behavioral deficits, cell-type-specific transcriptional and chromatin profiles, and OFC neuronal hyperexcitability induced by chronic stress - a major risk factor for MDD. These data thus highlight a critical role for OFC astrocytes in stress vulnerability and pinpoint ZBTB7A as a key dysregulated factor in MDD that mediates maladaptive astrocytic functions driving OFC hyperexcitability.

15.
J Neurosci ; 31(48): 17537-46, 2011 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-22131415

RESUMEN

The persistent or "late" phase of long-term potentiation (L-LTP), which requires protein synthesis, can be induced by relatively intense synaptic activity. The ability of such strong synaptic protocols to engage the translational machinery and produce plasticity-related proteins, while weaker protocols activate only posttranslational processes and transient potentiation (early LTP; E-LTP), is not understood. Among the major translation control pathways in neurons, the stimulation of mammalian target of rapamycin (mTOR) is a key event in the induction of L-LTP. We report that mTOR is tonically suppressed in rat hippocampus under resting conditions, a consequence of the basal activity of glycogen synthetase kinase 3 (GSK3). This suppression could be overcome by weak synaptic stimulation in the presence of the ß-adrenergic agonist isoproterenol, a combination that induced L-LTP, and activation of mTOR coincided with the Akt-mediated phosphorylation of GSK3. Surprisingly, while isoproterenol alone elevated Akt activity, it failed to increase GSK3 phosphorylation or mTOR signaling, showing that Akt was uncoupled from these effectors in the absence of synaptic stimulation. With the addition of weak stimulation, Akt signaled to GSK3 and mTOR, a gating effect that was mediated by voltage-dependent Ca(2+) channels and the Wnt pathway. mTOR could be stimulated by pharmacological inhibition, enabling weak HFS to induce L-LTP. These results establish GSK3 as an integrator of Akt and Wnt signals and suggest that overcoming GSK3-mediated suppression of mTOR is a key event in the induction of L-LTP by synaptic activity.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Sinapsis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Masculino , Neuronas/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Nat Commun ; 13(1): 4039, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35864121

RESUMEN

The dorsal raphe nucleus (DRN) is known to control aggressive behavior in mice. Here, we found that glutamatergic projections from the lateral habenula (LHb) to the DRN were activated in male mice that experienced pre-exposure to a rival male mouse ("social instigation") resulting in heightened intermale aggression. Both chemogenetic and optogenetic suppression of the LHb-DRN projection blocked heightened aggression after social instigation in male mice. In contrast, inhibition of this pathway did not affect basal levels of aggressive behavior, suggesting that the activity of the LHb-DRN projection is not necessary for the expression of species-typical aggressive behavior, but required for the increase of aggressive behavior resulting from social instigation. Anatomical analysis showed that LHb neurons synapse on non-serotonergic DRN neurons that project to the ventral tegmental area (VTA), and optogenetic activation of the DRN-VTA projection increased aggressive behaviors. Our results demonstrate that the LHb glutamatergic inputs to the DRN promote aggressive arousal induced by social instigation, which contributes to aggressive behavior by activating VTA-projecting non-serotonergic DRN neurons as one of its potential targets.


Asunto(s)
Núcleo Dorsal del Rafe , Habénula , Agresión/fisiología , Animales , Nivel de Alerta , Núcleo Dorsal del Rafe/fisiología , Habénula/fisiología , Masculino , Ratones , Vías Nerviosas/fisiología , Neuronas/metabolismo
17.
Biol Psychiatry ; 91(4): 346-358, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34130857

RESUMEN

BACKGROUND: Obstructive sleep apnea, characterized by sleep fragmentation and chronic intermittent hypoxia (CIH), is a risk factor for Alzheimer's disease (AD) progression. Recent epidemiological studies point to CIH as the best predictor of developing cognitive decline and AD in older adults with obstructive sleep apnea. However, the precise underlying mechanisms remain unknown. This study was undertaken to evaluate the effect of CIH on pathological human tau seeding, propagation, and accumulation; cognition; synaptic plasticity; neuronal network excitability; and gene expression profiles in a P301S human mutant tau mouse model of AD and related tauopathies. METHODS: We exposed 4- to 4.5-month-old male P301S and wild-type mice to an 8-week CIH protocol (6-min cycle: 21% O2 to 8% O2 to 21% O2, 80 cycles per 8 hours during daytime) and assessed its effect on tau pathology and various AD-related phenotypic and molecular signatures. Age- and sex-matched P301S and wild-type mice were reared in normoxia (21% O2) as experimental controls. RESULTS: CIH significantly enhanced pathological human tau seeding and spread across connected brain circuitry in P301S mice; it also increased phosphorylated tau load. CIH also exacerbated memory and synaptic plasticity deficits in P301S mice. However, CIH had no effect on seizure susceptibility and network hyperexcitability in these mice. Finally, CIH exacerbated AD-related pathogenic molecular signaling in P301S mice. CONCLUSIONS: CIH-induced increase in pathologic human tau seeding and spread and exacerbation of other AD-related impairments provide new insights into the role of CIH and obstructive sleep apnea in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Animales , Modelos Animales de Enfermedad , Hipoxia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal
18.
Nat Commun ; 13(1): 6384, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36289231

RESUMEN

With an incidence of ~1 in 800 births, Down syndrome (DS) is the most common chromosomal condition linked to intellectual disability worldwide. While the genetic basis of DS has been identified as a triplication of chromosome 21 (HSA21), the genes encoded from HSA21 that directly contribute to cognitive deficits remain incompletely understood. Here, we found that the HSA21-encoded chromatin effector, BRWD1, was upregulated in neurons derived from iPS cells from an individual with Down syndrome and brain of trisomic mice. We showed that selective copy number restoration of Brwd1 in trisomic animals rescued deficits in hippocampal LTP, cognition and gene expression. We demonstrated that Brwd1 tightly binds the BAF chromatin remodeling complex, and that increased Brwd1 expression promotes BAF genomic mistargeting. Importantly, Brwd1 renormalization rescued aberrant BAF localization, along with associated changes in chromatin accessibility and gene expression. These findings establish BRWD1 as a key epigenomic mediator of normal neurodevelopment and an important contributor to DS-related phenotypes.


Asunto(s)
Trastornos del Conocimiento , Síndrome de Down , Ratones , Animales , Síndrome de Down/genética , Síndrome de Down/metabolismo , Variaciones en el Número de Copia de ADN/genética , Modelos Animales de Enfermedad , Trastornos del Conocimiento/genética , Cromatina/genética , Ratones Transgénicos
19.
Biochem Biophys Res Commun ; 400(4): 679-83, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20816664

RESUMEN

Testosterone increases the size and strength of skeletal muscle. This study further characterized the molecular mechanisms of the anabolic actions of testosterone on a rat myoblast cell line (L6 cells). Testosterone did not induce hypertrophy in L6 cells lacking the androgen receptor (AR). Hypertrophy was prevented by the AR antagonist bicalutamide and the mTOR inhibitor rapamycin. Testosterone induced Erk phosphorylation by 2h, and mTOR autophosphorylation was elevated within 20min; phosphorylation of p70S6 kinase was increased by 2h. Inhibitors of Erk or PI3K blocked tesotosterone-induced hypertrophy. Erk phosphorylation returned to baseline when media containing testosterone was replaced at 16h with fresh media lacking testosterone; when bicalutamide was added to testosterone-enriched media at 16h, Erk phosphorylation remained elevated. Autophosphorylation of the IGF-1 receptor was minimally altered by testosterone at 20min and unaffected at later time points; PI3K/PDK1-dependent phosphorylation of Akt was not altered by testosterone. These findings indicate that testosterone stimulates hypertrophy of L6 myoblasts through a mechanism that requires its binding to the AR and involves a signaling cascade dependent upon Erk and mTOR which is likely activated by substances released into the extracellular space which are not IGF-1 or other ligands for receptor tyrosine kinases.


Asunto(s)
Aumento de la Célula , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Músculo Esquelético/fisiología , Mioblastos/fisiología , Receptores Androgénicos/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Testosterona/fisiología , Animales , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/genética , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Mioblastos/efectos de los fármacos , Mioblastos/enzimología , Fosforilación , Ratas , Receptores Androgénicos/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Testosterona/farmacología
20.
Mol Neurodegener ; 14(1): 43, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775806

RESUMEN

BACKGROUND: Dysfunctional autophagy is implicated in Alzheimer's Disease (AD) pathogenesis. The alterations in the expression of many autophagy related genes (ATGs) have been reported in AD brains; however, the disparity of the changes confounds the role of autophagy in AD. METHODS: To further understand the autophagy alteration in AD brains, we analyzed transcriptomic (RNAseq) datasets of several brain regions (BA10, BA22, BA36 and BA44 in 223 patients compared to 59 healthy controls) and measured the expression of 130 ATGs. We used autophagy-deficient mouse models to assess the impact of the identified ATGs depletion on memory, autophagic activity and amyloid-ß (Aß) production. RESULTS: We observed significant downregulation of multiple components of two autophagy kinase complexes BECN1-PIK3C3 and ULK1/2-FIP200 specifically in the parahippocampal gyrus (BA36). Most importantly, we demonstrated that deletion of NRBF2, a component of the BECN1-PIK3C3 complex, which also associates with ULK1/2-FIP200 complex, impairs memory in mice, alters long-term potentiation (LTP), reduces autophagy in mouse hippocampus, and promotes Aß accumulation. Furthermore, AAV-mediated NRBF2 overexpression in the hippocampus not only rescues the impaired autophagy and memory deficits in NRBF2-depleted mice, but also reduces ß-amyloid levels and improves memory in an AD mouse model. CONCLUSIONS: Our data not only implicates NRBF2 deficiency as a risk factor for cognitive impairment associated with AD, but also support the idea of NRBF2 as a potential therapeutic target for AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Autofagia/fisiología , Memoria/fisiología , Transactivadores/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Trastornos de la Memoria/metabolismo , Ratones , Ratones Transgénicos , Neuronas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA