Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-38559190

RESUMEN

Age is the strongest risk factor for developing Alzheimer's disease, the most common neurodegenerative disorder. However, the mechanisms connecting advancing age to neurodegeneration in Alzheimer's disease are incompletely understood. We conducted an unbiased, genome-scale, forward genetic screen for age-associated neurodegeneration in Drosophila to identify the underlying biological processes required for maintenance of aging neurons. To connect genetic screen hits to Alzheimer's disease pathways, we measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease. We further identified Alzheimer's disease human genetic variants that modify expression in disease-vulnerable neurons. Through multi-omic, multi-species network integration of these data, we identified relationships between screen hits and tau-mediated neurotoxicity. Furthermore, we computationally and experimentally identified relationships between screen hits and DNA damage in Drosophila and human iPSC-derived neural progenitor cells. Our work identifies candidate pathways that could be targeted to attenuate the effects of age on neurodegeneration and Alzheimer's disease.

2.
Nature ; 627(8002): 149-156, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38418876

RESUMEN

The glymphatic movement of fluid through the brain removes metabolic waste1-4. Noninvasive 40 Hz stimulation promotes 40 Hz neural activity in multiple brain regions and attenuates pathology in mouse models of Alzheimer's disease5-8. Here we show that multisensory gamma stimulation promotes the influx of cerebrospinal fluid and the efflux of interstitial fluid in the cortex of the 5XFAD mouse model of Alzheimer's disease. Influx of cerebrospinal fluid was associated with increased aquaporin-4 polarization along astrocytic endfeet and dilated meningeal lymphatic vessels. Inhibiting glymphatic clearance abolished the removal of amyloid by multisensory 40 Hz stimulation. Using chemogenetic manipulation and a genetically encoded sensor for neuropeptide signalling, we found that vasoactive intestinal peptide interneurons facilitate glymphatic clearance by regulating arterial pulsatility. Our findings establish novel mechanisms that recruit the glymphatic system to remove brain amyloid.


Asunto(s)
Enfermedad de Alzheimer , Amiloide , Encéfalo , Líquido Cefalorraquídeo , Líquido Extracelular , Ritmo Gamma , Sistema Glinfático , Animales , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Amiloide/metabolismo , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Encéfalo/patología , Líquido Cefalorraquídeo/metabolismo , Modelos Animales de Enfermedad , Líquido Extracelular/metabolismo , Sistema Glinfático/fisiología , Interneuronas/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Estimulación Eléctrica
3.
Glia ; 72(2): 452-469, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37969043

RESUMEN

Genetic findings have highlighted key roles for microglia in the pathology of neurodegenerative conditions such as Alzheimer's disease (AD). A number of mutations in the microglial protein triggering receptor expressed on myeloid cells 2 (TREM2) have been associated with increased risk for developing AD, most notably the R47H/+ substitution. We employed gene editing and stem cell models to gain insight into the effects of the TREM2 R47H/+ mutation on human-induced pluripotent stem cell-derived microglia. We found transcriptional changes affecting numerous cellular processes, with R47H/+ cells exhibiting a proinflammatory gene expression signature. TREM2 R47H/+ also caused impairments in microglial movement and the uptake of multiple substrates, as well as rendering microglia hyperresponsive to inflammatory stimuli. We developed an in vitro laser-induced injury model in neuron-microglia cocultures, finding an impaired injury response by TREM2 R47H/+ microglia. Furthermore, mouse brains transplanted with TREM2 R47H/+ microglia exhibited reduced synaptic density, with upregulation of multiple complement cascade components in TREM2 R47H/+ microglia suggesting inappropriate synaptic pruning as one potential mechanism. These findings identify a number of potentially detrimental effects of the TREM2 R47H/+ mutation on microglial gene expression and function likely to underlie its association with AD.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , Ratones , Animales , Humanos , Microglía/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mutación/genética , Enfermedad de Alzheimer/patología , Sinapsis/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
4.
Proc Natl Acad Sci U S A ; 120(16): e2217864120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37043533

RESUMEN

Aberrant activity of cyclin-dependent kinase (Cdk5) has been implicated in various neurodegenerative diseases. This deleterious effect is mediated by pathological cleavage of the Cdk5 activator p35 into the truncated product p25, leading to prolonged Cdk5 activation and altered substrate specificity. Elevated p25 levels have been reported in humans and rodents with neurodegeneration, and the benefit of genetically blocking p25 production has been demonstrated previously in rodent and human neurodegenerative models. Here, we report a 12-amino-acid-long peptide fragment derived from Cdk5 (Cdk5i) that is considerably smaller than existing peptide inhibitors of Cdk5 (P5 and CIP) but shows high binding affinity toward the Cdk5/p25 complex, disrupts the interaction of Cdk5 with p25, and lowers Cdk5/p25 kinase activity. When tagged with a fluorophore (FITC) and the cell-penetrating transactivator of transcription (TAT) sequence, the Cdk5i-FT peptide exhibits cell- and brain-penetrant properties and confers protection against neurodegenerative phenotypes associated with Cdk5 hyperactivity in cell and mouse models of neurodegeneration, highlighting Cdk5i's therapeutic potential.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina , Péptidos , Ratones , Animales , Humanos , Quinasa 5 Dependiente de la Ciclina/metabolismo , Fosforilación , Péptidos/metabolismo , Fragmentos de Péptidos/metabolismo , Fenotipo
5.
Ibrain ; 9(4): 359-368, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38680504

RESUMEN

Since the brain structure of neonatal rats was not fully formed during the first 4 days, it cannot be detected using ultrasound. The objective of this study was to investigate the use of ultrasound to guide puncture in the normal coronal brain structure and determine the puncture depth of the location of the cortex, hippocampus, lateral ventricle, and striatum of newborn rats of 5-15 days. The animal was placed in a prone position. The specific positions of the cortex, hippocampus, lateral ventricle, and striatum were measured under ultrasound. Then, the rats were punctured with a stereotaxic instrument, and dye was injected. Finally, the brains of rats were taken to make frozen sections to observe the puncture results. By ultrasound, the image of the cortex, hippocampus, lateral ventricle, and striatum of the rat can be obtained and the puncture depth of the cortex (8 days: 1.02 ± 0.12, 10 days: 1.02 ± 0.08, 13 days: 1.43 ± 0.05), hippocampus (8 days: 2.63 ± 0.07, 10 days: 2.77 ± 0.14, 13 days: 2.82 ± 0.09), lateral ventricle (8 days: 2.08 ± 0.04, 10 days: 2.26 ± 0.03, 13 days: 2.40 ± 0.06), and corpus striatum (8 days: 4.57 ± 0.09, 10 days: 4.94 ± 0.31, 13 days: 5.13 ± 0.10) can be accurately measured. The rat brain structure and puncture depth changed with the age of the rats. Ultrasound technology can not only clarify the brain structure characteristics of 5-15-day-old rats but also guide the puncture and injection of the rat brain structure. The results of this study laid the foundation for the future use of ultrasound in experimental animal models of neurological diseases.

6.
ACS Chem Neurosci ; 13(7): 848-858, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35263084

RESUMEN

Histone deacetylases (HDACs) have been implicated in learning and memory, and their dysregulation has been linked to cognitive impairment in brain aging and neurodegenerative diseases. In this review, we focus on HDAC1 and HDAC2, highlighting recent progress on their roles in regulating brain function through distinct mechanisms, including gene repression and DNA repair pathways. Moreover, we discuss evidence demonstrating how HDAC1 and HDAC2 could be modulated and their potential as targets to combat memory deficits.


Asunto(s)
Histona Desacetilasa 1 , Histona Desacetilasa 2 , Enfermedades Neurodegenerativas , Encéfalo/metabolismo , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Aprendizaje
7.
J Biomed Sci ; 28(1): 79, 2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34814918

RESUMEN

Cdk5 is a proline-directed serine/threonine protein kinase that governs a variety of cellular processes in neurons, the dysregulation of which compromises normal brain function. The mechanisms underlying the modulation of Cdk5, its modes of action, and its effects on the nervous system have been a great focus in the field for nearly three decades. In this review, we provide an overview of the discovery and regulation of Cdk5, highlighting recent findings revealing its role in neuronal/synaptic functions, circadian clocks, DNA damage, cell cycle reentry, mitochondrial dysfunction, as well as its non-neuronal functions under physiological and pathological conditions. Moreover, we discuss evidence underscoring aberrant Cdk5 activity as a common theme observed in many neurodegenerative diseases.


Asunto(s)
Ciclo Celular/genética , Relojes Circadianos/fisiología , Quinasa 5 Dependiente de la Ciclina/genética , Daño del ADN/genética , Enfermedades Neurodegenerativas/genética , Neuronas/fisiología , Sinapsis/fisiología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Humanos
8.
ACS Chem Neurosci ; 12(2): 271-284, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33417763

RESUMEN

Genomic instability caused by a deficiency in the DNA damage response and repair has been linked to age-related cognitive decline and neurodegenerative diseases. Preventing genomic instability that ultimately leads to neuronal death may provide a broadly effective strategy to protect against multiple potential genotoxic stressors. Recently, the zinc-dependent class I histone deacetylase (HDAC1) has been identified as a critical factor for protecting neurons from deleterious effects of DNA damage in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and frontotemporal dementia (FTD). Translating these observations to a novel neuroprotective therapy for AD, ALS, and FTD may be advanced by the identification of small molecules capable of increasing the deacetylase activity of HDAC1 selectively over other structurally similar HDACs. Here, we demonstrate that exifone, a drug previously shown to be effective in treating cognitive deficits associated with AD and Parkinson's disease, the molecular mechanism of which has remained poorly understood, potently activates the deacetylase activity of HDAC1. We show that exifone acts as a mixed, nonessential activator of HDAC1 that is capable of binding to both free and substrate-bound enzyme, resulting in an increased relative maximal rate of HDAC1-catalyzed deacetylation. Exifone can directly bind to HDAC1 based upon biolayer interferometry assays with kinetic and selectivity profiling, suggesting that HDAC1 is preferentially targeted compared to other class I HDACs and the kinase CDK5, which have also been implicated in neurodegeneration. Consistent with a mechanism of deacetylase activation intracellularly, the treatment of human induced pluripotent stem cell (iPSC)-derived neuronal cells resulted in globally decreased histone acetylation. Moreover, exifone treatment was neuroprotective in a tauopathy patient iPSC-derived neuronal model subject to oxidative stress. Taken together, these findings reveal exifone as a potent activator of HDAC1-mediated deacetylation, thereby offering a lead for novel therapeutic development aiming to protect genomic integrity in the context of neurodegeneration and aging.


Asunto(s)
Histona Desacetilasas , Células Madre Pluripotentes Inducidas , Benzofenonas , Histona Desacetilasa 1 , Humanos , Neuronas
9.
Mol Syst Biol ; 16(12): e9819, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33289969

RESUMEN

Alzheimer's disease (AD) is characterized by the appearance of amyloid-ß plaques, neurofibrillary tangles, and inflammation in brain regions involved in memory. Using mass spectrometry, we have quantified the phosphoproteome of the CK-p25, 5XFAD, and Tau P301S mouse models of neurodegeneration. We identified a shared response involving Siglec-F which was upregulated on a subset of reactive microglia. The human paralog Siglec-8 was also upregulated on microglia in AD. Siglec-F and Siglec-8 were upregulated following microglial activation with interferon gamma (IFNγ) in BV-2 cell line and human stem cell-derived microglia models. Siglec-F overexpression activates an endocytic and pyroptotic inflammatory response in BV-2 cells, dependent on its sialic acid substrates and immunoreceptor tyrosine-based inhibition motif (ITIM) phosphorylation sites. Related human Siglecs induced a similar response in BV-2 cells. Collectively, our results point to an important role for mouse Siglec-F and human Siglec-8 in regulating microglial activation during neurodegeneration.


Asunto(s)
Inflamación/patología , Microglía/metabolismo , Degeneración Nerviosa/patología , Fosfoproteínas/metabolismo , Proteómica , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Animales , Anticuerpos/metabolismo , Muerte Celular , Línea Celular , Humanos , Inflamación/metabolismo , Interferón gamma/metabolismo , Ratones Transgénicos , Microglía/patología , Degeneración Nerviosa/metabolismo , Péptidos/metabolismo , Fagocitosis , Fosfotirosina/metabolismo , Proteoma/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/química , Transducción de Señal , Regulación hacia Arriba
10.
Cell Rep ; 32(2): 107908, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32668255

RESUMEN

We present a consensus atlas of the human brain transcriptome in Alzheimer's disease (AD), based on meta-analysis of differential gene expression in 2,114 postmortem samples. We discover 30 brain coexpression modules from seven regions as the major source of AD transcriptional perturbations. We next examine overlap with 251 brain differentially expressed gene sets from mouse models of AD and other neurodegenerative disorders. Human-mouse overlaps highlight responses to amyloid versus tau pathology and reveal age- and sex-dependent expression signatures for disease progression. Human coexpression modules enriched for neuronal and/or microglial genes broadly overlap with mouse models of AD, Huntington's disease, amyotrophic lateral sclerosis, and aging. Other human coexpression modules, including those implicated in proteostasis, are not activated in AD models but rather following other, unexpected genetic manipulations. Our results comprise a cross-species resource, highlighting transcriptional networks altered by human brain pathophysiology and identifying correspondences with mouse models for AD preclinical studies.


Asunto(s)
Enfermedad de Alzheimer/genética , Encéfalo/metabolismo , Encéfalo/patología , Transcriptoma/genética , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Masculino , Ratones , Caracteres Sexuales , Especificidad de la Especie , Transcripción Genética
11.
Nat Commun ; 11(1): 2484, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424276

RESUMEN

DNA damage contributes to brain aging and neurodegenerative diseases. However, the factors stimulating DNA repair to stave off functional decline remain obscure. We show that HDAC1 modulates OGG1-initated 8-oxoguanine (8-oxoG) repair in the brain. HDAC1-deficient mice display age-associated DNA damage accumulation and cognitive impairment. HDAC1 stimulates OGG1, a DNA glycosylase known to remove 8-oxoG lesions that are associated with transcriptional repression. HDAC1 deficiency causes impaired OGG1 activity, 8-oxoG accumulation at the promoters of genes critical for brain function, and transcriptional repression. Moreover, we observe elevated 8-oxoG along with reduced HDAC1 activity and downregulation of a similar gene set in the 5XFAD mouse model of Alzheimer's disease. Notably, pharmacological activation of HDAC1 alleviates the deleterious effects of 8-oxoG in aged wild-type and 5XFAD mice. Our work uncovers important roles for HDAC1 in 8-oxoG repair and highlights the therapeutic potential of HDAC1 activation to counter functional decline in brain aging and neurodegeneration.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/patología , Encéfalo/patología , Daño del ADN , ADN Glicosilasas/metabolismo , Histona Desacetilasa 1/metabolismo , Estrés Oxidativo , Acetilación , Envejecimiento/genética , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Secuencia de Bases , Benzofenonas/farmacología , Cognición/efectos de los fármacos , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/patología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Ontología de Genes , Guanina/análogos & derivados , Guanina/metabolismo , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Regiones Promotoras Genéticas/genética
12.
Aquat Toxicol ; 222: 105469, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32179334

RESUMEN

Phthalates are commonly used in plastic products in daily life. The endocrine-disrupting effects of phthalates have been widely reported. Accumulating evidence from human cohorts and lab animals indicate exposure to phthalates might impair neurodevelopment. However, the direct causal relationship and mechanism between phthalates with neurodevelopment and neurotoxicity have not been firmly established. We found that phthalates (i.e. DBP, DINP, BBP) disrupted the expression of estrogen receptors (esr1, esr2a, esr2b), and impaired neurogenesis in the brain of zebrafish during embryonic development. Moreover, the abnormal expression of estrogen receptors, especially esr2a, was partly rescued in zebrafish which exposed to phthalates, with the estrogen receptor antagonist tamoxifen. Hence, impaired neurogenesis of zebrafish exposed to phthalates was partly reversed by tamoxifen treatment. Moreover, our results show that induced pluripotent stem cells (iPSC)-derived human neurons exposed to phthalates triggered double-strand DNA breaks in vitro. Overall, this study demonstrates that exposure to phthalates affects neurodevelopment in zebrafish embryos and induces neurotoxicity in human neurons partly through disrupting the expression of estrogen receptors.


Asunto(s)
Roturas del ADN de Doble Cadena , Desarrollo Embrionario/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Neuronas/efectos de los fármacos , Ácidos Ftálicos/toxicidad , Receptores de Estrógenos/genética , Contaminantes Químicos del Agua/toxicidad , Animales , Células Cultivadas , Embrión no Mamífero/efectos de los fármacos , Desarrollo Embrionario/genética , Antagonistas del Receptor de Estrógeno/farmacología , Humanos , Neuronas/metabolismo , Neuronas/patología , Pez Cebra
13.
Methods Mol Biol ; 1983: 225-234, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31087301

RESUMEN

Histone deacetylases (HDACs) modulate chromatin structure by removing acetyl groups from histones. Upon DNA double-strand breaks (DSBs), deacetylation of H3K56 and H4K16 by HDACs occurs immediately at break sites, and is crucial for DSB repair. Here we describe two assays that examine defective DSB repair caused by HDAC inhibition in primary cortical neurons: single-cell gel electrophoresis to assay DNA integrity (the comet assay) and western blot analysis for γH2AX, a phosphorylated histone variant associated with DSBs.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Histona Desacetilasas/metabolismo , Neuronas/metabolismo , Acetilación , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Ensayo Cometa , Inestabilidad Genómica , Histonas/metabolismo , Humanos , Ratones , Imagen Molecular
14.
Neuron ; 102(5): 929-943.e8, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31076275

RESUMEN

Neuronal and synaptic loss is characteristic in many neurodegenerative diseases, such as frontotemporal dementia and Alzheimer's disease. Recently, we showed that inducing gamma oscillations with visual stimulation (gamma entrainment using sensory stimuli, or GENUS) reduced amyloid plaques and phosphorylated tau in multiple mouse models. Whether GENUS can affect neurodegeneration or cognitive performance remains unknown. Here, we demonstrate that GENUS can entrain gamma oscillations in the visual cortex, hippocampus, and prefrontal cortex in Tau P301S and CK-p25 mouse models of neurodegeneration. Tau P301S and CK-p25 mice subjected to chronic, daily GENUS from the early stages of neurodegeneration showed a preservation of neuronal and synaptic density across multiple brain areas and modified cognitive performance. Our transcriptomic and phosphoproteomic data suggest that chronic GENUS shifts neurons to a less degenerative state, improving synaptic function, enhancing neuroprotective factors, and reducing DNA damage in neurons while also reducing inflammatory response in microglia.


Asunto(s)
Ritmo Gamma/fisiología , Hipocampo/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/patología , Neuroprotección/fisiología , Estimulación Luminosa/métodos , Corteza Prefrontal/fisiopatología , Corteza Visual/fisiopatología , Animales , Daño del ADN , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Inflamación , Ratones , Microglía/inmunología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Proteómica , Aprendizaje Espacial/fisiología , Memoria Espacial/fisiología , Sinapsis/metabolismo , Sinapsis/patología , Corteza Visual/metabolismo , Corteza Visual/patología
15.
J Biomed Sci ; 26(1): 30, 2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31027502

RESUMEN

BACKGROUND: Promyelocytic leukemia zinc finger (Plzf), a transcriptional regulator involved in a lot of important biological processes during development, has been implied to maintain neural stem cells and inhibit their differentiation into neurons. However, the effects of Plzf on brain structures and functions are still not clarified. RESULTS: We showed that Plzf expression was detected as early as embryonic day (E) 9.5 in Pax6+ cells in the mouse brain, and was completely disappeared in telencephalon before the initiation of cortical neurogenesis. Loss of Plzf resulted in a smaller cerebral cortex with a decrease in the number of Tbr1+ deep layer neurons due to a decrease of mitotic cell number in the ventricular zone of forebrain at early developmental stage. Microarray, qRT-PCR, and flow cytometry analysis identified dysregulation of Mash1 proneural gene expression. We also observed an impairment of recognition memory in Plzf-deficient mice. CONCLUSIONS: Plzf is expressed at early stages of brain development and involved in the formation of deep layer cortical neurons. Loss of Plzf results in dysregulation of Mash1, microcephaly with reduced numbers of early-born neurons, and impairment of recognition memory.


Asunto(s)
Expresión Génica/fisiología , Neurogénesis/genética , Neuronas/fisiología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Animales , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiología , Ratones , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo
16.
J Neurosci ; 37(36): 8655-8666, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878098

RESUMEN

Diverse molecular mechanisms regulate synaptic composition and function in the mammalian nervous system. The multifunctional protein arginine methyltransferase 8 (PRMT8) possesses both methyltransferase and phospholipase activities. Here we examine the role of this neuron-specific protein in hippocampal plasticity and cognitive function. PRMT8 protein localizes to synaptic sites, and conditional whole-brain Prmt8 deletion results in altered levels of multiple synaptic proteins in the hippocampus, using both male and female mice. Interestingly, these altered protein levels are due to post-transcriptional mechanisms as the corresponding mRNA levels are unaffected. Strikingly, electrophysiological recordings from hippocampal slices of mice lacking PRMT8 reveal multiple defects in excitatory synaptic function and plasticity. Furthermore, behavioral analyses show that PRMT8 conditional knock-out mice exhibit impaired hippocampal-dependent fear learning. Together, these findings establish PRMT8 as an important component of the molecular machinery required for hippocampal neuronal function.SIGNIFICANCE STATEMENT Numerous molecular processes are critically required for normal brain function. Here we use mice lacking protein arginine methyltransferase 8 (PRMT8) in the brain to examine how loss of this protein affects the structure and function of neurons in the hippocampus. We find that PRMT8 localizes to the sites of communication between neurons. Hippocampal neurons from mice lacking PRMT8 have no detectable structural differences compared with controls; however, multiple aspects of their function are altered. Consistently, we find that mice lacking PRMT8 also exhibit reduced hippocampus-dependent memory. Together, our findings establish important roles for PRMT8 in regulating neuron function and cognition in the mammalian brain.


Asunto(s)
Hipocampo/fisiopatología , Trastornos de la Memoria/fisiopatología , Trastornos Mentales/fisiopatología , Proteína-Arginina N-Metiltransferasas/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica , Animales , Femenino , Hipocampo/patología , Masculino , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/patología , Trastornos Mentales/complicaciones , Trastornos Mentales/patología , Ratones , Ratones Noqueados , Plasticidad Neuronal , Proteína-Arginina N-Metiltransferasas/genética , Sinapsis/patología
17.
Proc Natl Acad Sci U S A ; 114(17): 4388-4393, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396416

RESUMEN

The mitochondrial calcium uniporter is a Ca2+-activated Ca2+ channel complex mediating mitochondrial Ca2+ uptake, a process crucial for Ca2+ signaling, bioenergetics, and cell death. The uniporter is composed of the pore-forming MCU protein, the gatekeeping MICU1 and MICU2 subunits, and EMRE, a single-pass membrane protein that links MCU and MICU1 together. As a bridging subunit required for channel function, EMRE could paradoxically inhibit uniporter complex formation if expressed in excess. Here, we show that mitochondrial mAAA proteases AFG3L2 and SPG7 rapidly degrade unassembled EMRE using the energy of ATP hydrolysis. Once EMRE is incorporated into the complex, its turnover is inhibited >15-fold. Protease-resistant EMRE mutants produce uniporter subcomplexes that induce constitutive Ca2+ leakage into mitochondria, a condition linked to debilitating neuromuscular disorders in humans. The results highlight the dynamic nature of uniporter subunit assembly, which must be tightly regulated to ensure proper mitochondrial responses to intracellular Ca2+ signals.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Regulación de la Expresión Génica/fisiología , Canales de Calcio/genética , Eliminación de Gen , Células HEK293 , Células HeLa , Humanos , Péptido Hidrolasas/metabolismo , Subunidades de Proteína
18.
Mol Neurobiol ; 54(3): 2286-2300, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26951452

RESUMEN

Rnf112 is a member of the RING finger protein family. The expression of Rnf112 is abundant in the brain and is regulated during brain development. Our previous study has revealed that Rnf112 can promote neuronal differentiation by inhibiting the progression of the cell cycle in cell models. In this study, we further revealed the important functions of Rnf112 in embryo development and in adult brain. Our data showed that most of the Rnf112 -/- embryos exhibited blood vascular defects and died in utero. Upon further investigation, we found that the survival rate of homozygous Rnf112 knockout mice in 129/sv and C57BL/6 mixed genetic background was increased. The survived newborns of Rnf112 -/- mice manifested growth retardation as indicated by smaller size and a reduced weight. Although the overall organization of the brain did not appear to be severely affected in Rnf112 -/- mice, using in vivo 3D MRI imaging, we found that when compared to wild-type littermates, brains of Rnf112 -/- mice were smaller. In addition, Rnf112 -/- mice displayed impairment of brain functions including motor balance, and spatial learning and memory. Our results provide important aspects for the study of Rnf112 gene functions.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/irrigación sanguínea , Encéfalo/embriología , Proteínas de Unión al ADN/metabolismo , Animales , Proteínas de Unión al ADN/genética , Imagenología Tridimensional/métodos , Aprendizaje/fisiología , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
19.
Cell ; 161(7): 1592-605, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26052046

RESUMEN

Neuronal activity causes the rapid expression of immediate early genes that are crucial for experience-driven changes to synapses, learning, and memory. Here, using both molecular and genome-wide next-generation sequencing methods, we report that neuronal activity stimulation triggers the formation of DNA double strand breaks (DSBs) in the promoters of a subset of early-response genes, including Fos, Npas4, and Egr1. Generation of targeted DNA DSBs within Fos and Npas4 promoters is sufficient to induce their expression even in the absence of an external stimulus. Activity-dependent DSB formation is likely mediated by the type II topoisomerase, Topoisomerase IIß (Topo IIß), and knockdown of Topo IIß attenuates both DSB formation and early-response gene expression following neuronal stimulation. Our results suggest that DSB formation is a physiological event that rapidly resolves topological constraints to early-response gene expression in neurons.


Asunto(s)
Roturas del ADN de Doble Cadena , Neuronas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factor de Unión a CCCTC , ADN-Topoisomerasas de Tipo II/análisis , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Etopósido/farmacología , Regulación de la Expresión Génica , Genes fos , Estudio de Asociación del Genoma Completo , Ratones , Proteínas Represoras/metabolismo , Transcriptoma/efectos de los fármacos
20.
Mol Neurobiol ; 51(1): 370-82, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24788683

RESUMEN

Reactive astrogliosis is a cellular manifestation of neuroinflammation and occurs in response to all forms and severities of the central nervous system (CNS)'s injury and disease. Both astroglial proliferation and antiapoptotic processes are aspects of astrogliosis. However, the underlying mechanism of this response remains poorly understood. In addition, little is known about why activated astrocytes are more resistant to stress and inflammation. CCAAT/enhancer binding protein delta (CEBPD) is a transcription factor found in activated astrocytes that surround ß-amyloid plaques. In this study, we found that astrocytes activation was attenuated in the cortex and hippocampus of APPswe/PS1 E9 (AppTg)/Cebpd (-/-)mice. Furthermore, an increase in apoptotic astrocytes was observed in AppTg/Cebpd (-/-)mice, suggesting that CEBPD plays a functional role in enhancing the antiapoptotic ability of astrocytes. We found that Zinc Finger Protein 179 (ZNF179) was a CEBPD-regulated gene that played an antiapoptotic, but not proliferative, role in astrocytes. The transcriptions of the proapoptotic genes, insulin-like growth factor binding protein 3 (IGFBP3) and BCL2-interacting killer (BIK), were suppressed by ZNF179 via its interaction with the promyelocytic leukemia zinc finger (PLZF) protein in astrocytes. This study provides the first evidence that ZNF179, PLZF, IGFBP3, and BIK contributed to the novel CEBPD-induced antiapoptotic feature of astrocytes.


Asunto(s)
Enfermedad de Alzheimer/patología , Apoptosis , Astrocitos/metabolismo , Astrocitos/patología , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Astrocitos/efectos de los fármacos , Secuencia de Bases , Proteína delta de Unión al Potenciador CCAAT/deficiencia , Línea Celular Tumoral , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Interleucina-1beta/farmacología , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Metilmetanosulfonato/farmacología , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Placa Amiloide/complicaciones , Placa Amiloide/metabolismo , Placa Amiloide/patología , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...