Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 152(4): 831-43, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23415230

RESUMEN

p11, through unknown mechanisms, is required for behavioral and cellular responses to selective serotonin reuptake inhibitors (SSRIs). We show that SMARCA3, a chromatin-remodeling factor, is a target for the p11/annexin A2 heterotetrameric complex. Determination of the crystal structure indicates that SMARCA3 peptide binds to a hydrophobic pocket in the heterotetramer. Formation of this complex increases the DNA-binding affinity of SMARCA3 and its localization to the nuclear matrix fraction. In the dentate gyrus, both p11 and SMARCA3 are highly enriched in hilar mossy cells and basket cells. The SSRI fluoxetine induces expression of p11 in both cell types and increases the amount of the ternary complex of p11/annexin A2/SMARCA3. SSRI-induced neurogenesis and behavioral responses are abolished by constitutive knockout of SMARCA3. Our studies indicate a central role for a chromatin-remodeling factor in the SSRI/p11 signaling pathway and suggest an approach to the development of improved antidepressant therapies. PAPERCLIP:


Asunto(s)
Anexina A2/metabolismo , Proteínas de Unión al ADN/metabolismo , Giro Dentado/metabolismo , Proteínas S100/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/química , Femenino , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Fibras Musgosas del Hipocampo/metabolismo , Alineación de Secuencia , Transducción de Señal , Factores de Transcripción/química , Difracción de Rayos X
2.
Cell ; 149(5): 1152-63, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22632977

RESUMEN

Our understanding of current treatments for depression, and the development of more specific therapies, is limited by the complexity of the circuits controlling mood and the distributed actions of antidepressants. Although the therapeutic efficacy of serotonin-specific reuptake inhibitors (SSRIs) is correlated with increases in cortical activity, the cell types crucial for their action remain unknown. Here we employ bacTRAP translational profiling to show that layer 5 corticostriatal pyramidal cells expressing p11 (S100a10) are strongly and specifically responsive to chronic antidepressant treatment. This response requires p11 and includes the specific induction of Htr4 expression. Cortex-specific deletion of p11 abolishes behavioral responses to SSRIs, but does not lead to increased depression-like behaviors. Our data identify corticostriatal projection neurons as critical for the response to antidepressants, and suggest that the regulation of serotonergic tone in this single cell type plays a pivotal role in antidepressant therapy.


Asunto(s)
Antidepresivos/metabolismo , Depresión/tratamiento farmacológico , Neuronas/citología , Corteza Prefrontal/citología , Inhibidores Selectivos de la Recaptación de Serotonina/metabolismo , Animales , Antidepresivos/farmacología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
3.
Nature ; 586(7831): 735-740, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32879487

RESUMEN

Innate immunity is associated with Alzheimer's disease1, but the influence of immune activation on the production of amyloid-ß is unknown2,3. Here we identify interferon-induced transmembrane protein 3 (IFITM3) as a γ-secretase modulatory protein, and establish a mechanism by which inflammation affects the generation of amyloid-ß. Inflammatory cytokines induce the expression of IFITM3 in neurons and astrocytes, which binds to γ-secretase and upregulates its activity, thereby increasing the production of amyloid-ß. The expression of IFITM3 is increased with ageing and in mouse models that express familial Alzheimer's disease genes. Furthermore, knockout of IFITM3 reduces γ-secretase activity and the formation of amyloid plaques in a transgenic mouse model (5xFAD) of early amyloid deposition. IFITM3 protein is upregulated in tissue samples from a subset of patients with late-onset Alzheimer's disease that exhibit higher γ-secretase activity. The amount of IFITM3 in the γ-secretase complex has a strong and positive correlation with γ-secretase activity in samples from patients with late-onset Alzheimer's disease. These findings reveal a mechanism in which γ-secretase is modulated by neuroinflammation via IFITM3 and the risk of Alzheimer's disease is thereby increased.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Edad de Inicio , Anciano de 80 o más Años , Envejecimiento/genética , Envejecimiento/inmunología , Envejecimiento/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/química , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/metabolismo , Dominio Catalítico , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Inflamación , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/metabolismo , Proteínas de Unión al ARN/genética , Riesgo , Regulación hacia Arriba
4.
Proc Natl Acad Sci U S A ; 119(12): e2122292119, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35298330

RESUMEN

Aberrant cleavage of amyloid precursor protein (APP) by γ-secretase is closely associated with Alzheimer's disease (AD). γ-secretase activating protein (GSAP) specifically promotes γ-secretase­mediated cleavage of APP. However, the underlying mechanism remains enigmatic. Here, we demonstrate that the 16-kDa C-terminal fragment of GSAP (GSAP-16K) undergoes phase separation in vitro and forms puncta-like condensates in cells. GSAP-16K exerts dual modulation on γ-secretase cleavage; GSAP-16K in dilute phase increases APP­C-terminal 99-residue fragment (C99) cleavage toward preferred production of ß-amyloid peptide 42 (Aß42), but GSAP-16K condensates reduce APP-C99 cleavage through substrate sequestration. Notably, the Aß42/Aß40 ratio is markedly elevated with increasing concentrations of GSAP-16K. GSAP-16K stably associates with APP-C99 through specific sequence elements. These findings mechanistically explain GSAP-mediated modulation of γ-secretase activity that may have ramifications on the development of potential therapeutics.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Humanos , Fragmentos de Péptidos/metabolismo
5.
Mol Psychiatry ; 27(4): 2068-2079, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35177825

RESUMEN

Forebrain dopamine-sensitive (dopaminoceptive) neurons play a key role in movement, action selection, motivation, and working memory. Their activity is altered in Parkinson's disease, addiction, schizophrenia, and other conditions, and drugs that stimulate or antagonize dopamine receptors have major therapeutic applications. Yet, similarities and differences between the various neuronal populations sensitive to dopamine have not been systematically explored. To characterize them, we compared translating mRNAs in the dorsal striatum and nucleus accumbens neurons expressing D1 or D2 dopamine receptor and prefrontal cortex neurons expressing D1 receptor. We identified genome-wide cortico-striatal, striatal D1/D2 and dorso/ventral differences in the translating mRNA and isoform landscapes, which characterize dopaminoceptive neuronal populations. Expression patterns and network analyses identified novel transcription factors with presumptive roles in these differences. Prostaglandin E2 (PGE2) was a candidate upstream regulator in the dorsal striatum. We pharmacologically explored this hypothesis and showed that misoprostol, a PGE2 receptor agonist, decreased the excitability of D2 striatal projection neurons in slices, and diminished their activity in vivo during novel environment exploration. We found that misoprostol also modulates mouse behavior including by facilitating reversal learning. Our study provides powerful resources for characterizing dopamine target neurons, new information about striatal gene expression patterns and regulation. It also reveals the unforeseen role of PGE2 in the striatum as a potential neuromodulator and an attractive therapeutic target.


Asunto(s)
Dinoprostona , Misoprostol , Animales , Cuerpo Estriado/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacología , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Exones , Expresión Génica , Ratones , Misoprostol/metabolismo , Misoprostol/farmacología , ARN Mensajero/metabolismo , Receptores de Dopamina D1/metabolismo
6.
Cell ; 135(4): 738-48, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-19013281

RESUMEN

The cellular heterogeneity of the brain confounds efforts to elucidate the biological properties of distinct neuronal populations. Using bacterial artificial chromosome (BAC) transgenic mice that express EGFP-tagged ribosomal protein L10a in defined cell populations, we have developed a methodology for affinity purification of polysomal mRNAs from genetically defined cell populations in the brain. The utility of this approach is illustrated by the comparative analysis of four types of neurons, revealing hundreds of genes that distinguish these four cell populations. We find that even two morphologically indistinguishable, intermixed subclasses of medium spiny neurons display vastly different translational profiles and present examples of the physiological significance of such differences. This genetically targeted translating ribosome affinity purification (TRAP) methodology is a generalizable method useful for the identification of molecular changes in any genetically defined cell type in response to genetic alterations, disease, or pharmacological perturbations.


Asunto(s)
Encéfalo/metabolismo , Técnicas Genéticas , Biosíntesis de Proteínas , Animales , Sistema Nervioso Central/metabolismo , Cromosomas Artificiales Bacterianos/metabolismo , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica/métodos , Ratones , Ratones Transgénicos , Modelos Biológicos , Neuronas/metabolismo , Ribosomas/metabolismo
7.
Cell ; 135(4): 749-62, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-19013282

RESUMEN

Comparative analysis can provide important insights into complex biological systems. As demonstrated in the accompanying paper, translating ribosome affinity purification (TRAP) permits comprehensive studies of translated mRNAs in genetically defined cell populations after physiological perturbations. To establish the generality of this approach, we present translational profiles for 24 CNS cell populations and identify known cell-specific and enriched transcripts for each population. We report thousands of cell-specific mRNAs that were not detected in whole-tissue microarray studies and provide examples that demonstrate the benefits deriving from comparative analysis. To provide a foundation for further biological and in silico studies, we provide a resource of 16 transgenic mouse lines, their corresponding anatomic characterization, and translational profiles for cell types from a variety of central nervous system structures. This resource will enable a wide spectrum of molecular and mechanistic studies of both well-known and previously uncharacterized neural cell populations.


Asunto(s)
Encéfalo/metabolismo , Técnicas Genéticas , Biosíntesis de Proteínas , Animales , Sistema Nervioso Central/metabolismo , Cromosomas Artificiales Bacterianos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica/métodos , Ratones , Ratones Transgénicos , Modelos Biológicos , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribosomas/metabolismo
8.
Mol Psychiatry ; 26(12): 7308-7315, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34234280

RESUMEN

Major depressive disorder (MDD) is a severe, common mood disorder. While reduced cerebrospinal fluid (CSF) flow adversely affects brain metabolism and fluid balance in the aging population and during development, only indirect evidence links aberrant CSF circulation with many diseases including neurological, neurodegenerative, and psychiatric disorders, such as anxiety and depression. Here we show a very high concentration of p11 as a key molecular determinant for depression in ependymal cells, which is significantly decreased in patients with MDD, and in two mouse models of depression induced by chronic stress, such as restraint and social isolation. The loss of p11 in ependymal cells causes disoriented ependymal planar cell polarity (PCP), reduced CSF flow, and depression-like and anxiety-like behaviors. p11 intrinsically controls PCP core genes, which mediates CSF flow. Viral expression of p11 in ependymal cells specifically rescues the pathophysiological and behavioral deficits caused by loss of p11. Taken together, our results identify a new role and a key molecular determinant for ependymal cell-driven CSF flow in mood disorders and suggest a novel strategy for development of treatments for stress-associated neurological, neurodegenerative, and psychiatric disorders.


Asunto(s)
Trastorno Depresivo Mayor , Anciano , Animales , Trastornos de Ansiedad , Depresión/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Neuroglía
9.
Mol Psychiatry ; 26(7): 3350-3362, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33723417

RESUMEN

The delayed behavioral response to chronic antidepressants depends on dynamic changes in the hippocampus. It was suggested that the antidepressant protein p11 and the chromatin remodeling factor SMARCA3 mediate this delayed response by inducing transcriptional changes in hippocampal neurons. However, what target genes are regulated by the p11/SMARCA3 complex to mediate the behavioral response to antidepressants, and what cell type mediates these molecular changes remain unknown. Here we report that the p11/SMARCA3 complex represses Neurensin-2 transcription in hippocampal parvalbumin-expressing interneurons after chronic treatment with Selective Serotonin Reuptake Inhibitors (SSRI). The behavioral response to antidepressants requires upregulation of p11, accumulation of SMARCA3 in the cell nucleus, and a consequent repression of Neurensin-2 transcription in these interneurons. We elucidate a functional role for p11/SMARCA3/Neurensin-2 pathway in regulating AMPA-receptor signaling in parvalbumin-expressing interneurons, a function that is enhanced by chronic treatment with SSRIs. These results link SSRIs to dynamic glutamatergic changes and implicate p11/SMARCA3/Neurensin-2 pathway in the development of more specific and efficient therapeutic strategies for neuropsychiatric disorders.


Asunto(s)
Antidepresivos , Proteínas de Unión al ADN/metabolismo , Interneuronas/efectos de los fármacos , Parvalbúminas , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Factores de Transcripción/metabolismo , Animales , Antidepresivos/farmacología , Hipocampo/metabolismo , Interneuronas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Parvalbúminas/metabolismo
10.
Mol Psychiatry ; 26(7): 2872-2885, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33742167

RESUMEN

Among the hallmarks of major depressive disorders (MDD) are molecular, functional, and morphological impairments in the hippocampus. Recent studies suggested a key role for hippocampal GABAergic interneurons both in depression and in the response to its treatments. These interneurons highly express the chromatin-remodeler SMARCA3 which mediates the response to chronic antidepressants in an unknown mechanism. Using cell-type-specific molecular and physiological approaches, we report that SMARCA3 mediates the glutamatergic signaling in interneurons by repressing the expression of the neuronal protein, Neurensin-2. This vesicular protein associates with endosomes and postsynaptic proteins and is highly and selectively expressed in subpopulations of GABAergic interneurons. Upregulation of Neurensin-2 in the hippocampus either by stress, viral overexpression, or by SMARCA3 deletion, results in depressive-like behaviors. In contrast, the deletion of Neurensin-2 confers resilience to stress and induces AMPA receptor localization to synapses. This pathway which bidirectionally affects emotional behavior could be involved in neuropsychiatric disorders, and suggests novel therapeutic approaches.


Asunto(s)
Trastorno Depresivo Mayor , Hipocampo , Humanos , Interneuronas , Neuronas , Sinapsis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA