Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 256
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(20): 4404-4421.e20, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774679

RESUMEN

Persistent DNA double-strand breaks (DSBs) in neurons are an early pathological hallmark of neurodegenerative diseases including Alzheimer's disease (AD), with the potential to disrupt genome integrity. We used single-nucleus RNA-seq in human postmortem prefrontal cortex samples and found that excitatory neurons in AD were enriched for somatic mosaic gene fusions. Gene fusions were particularly enriched in excitatory neurons with DNA damage repair and senescence gene signatures. In addition, somatic genome structural variations and gene fusions were enriched in neurons burdened with DSBs in the CK-p25 mouse model of neurodegeneration. Neurons enriched for DSBs also had elevated levels of cohesin along with progressive multiscale disruption of the 3D genome organization aligned with transcriptional changes in synaptic, neuronal development, and histone genes. Overall, this study demonstrates the disruption of genome stability and the 3D genome organization by DSBs in neurons as pathological steps in the progression of neurodegenerative diseases.


Asunto(s)
Roturas del ADN de Doble Cadena , Enfermedades Neurodegenerativas , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , ADN , Reparación del ADN/genética , Enfermedades Neurodegenerativas/genética , Neuronas/fisiología , Análisis de la Célula Individual , Análisis de Secuencia de ARN , Inestabilidad Genómica
2.
Cell ; 186(20): 4386-4403.e29, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774678

RESUMEN

Altered microglial states affect neuroinflammation, neurodegeneration, and disease but remain poorly understood. Here, we report 194,000 single-nucleus microglial transcriptomes and epigenomes across 443 human subjects and diverse Alzheimer's disease (AD) pathological phenotypes. We annotate 12 microglial transcriptional states, including AD-dysregulated homeostatic, inflammatory, and lipid-processing states. We identify 1,542 AD-differentially-expressed genes, including both microglia-state-specific and disease-stage-specific alterations. By integrating epigenomic, transcriptomic, and motif information, we infer upstream regulators of microglial cell states, gene-regulatory networks, enhancer-gene links, and transcription-factor-driven microglial state transitions. We demonstrate that ectopic expression of our predicted homeostatic-state activators induces homeostatic features in human iPSC-derived microglia-like cells, while inhibiting activators of inflammation can block inflammatory progression. Lastly, we pinpoint the expression of AD-risk genes in microglial states and differential expression of AD-risk genes and their regulators during AD progression. Overall, we provide insights underlying microglial states, including state-specific and AD-stage-specific microglial alterations at unprecedented resolution.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Regulación de la Expresión Génica , Inflamación/patología , Microglía/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Epigenoma
3.
Cell ; 186(20): 4422-4437.e21, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774680

RESUMEN

Recent work has identified dozens of non-coding loci for Alzheimer's disease (AD) risk, but their mechanisms and AD transcriptional regulatory circuitry are poorly understood. Here, we profile epigenomic and transcriptomic landscapes of 850,000 nuclei from prefrontal cortexes of 92 individuals with and without AD to build a map of the brain regulome, including epigenomic profiles, transcriptional regulators, co-accessibility modules, and peak-to-gene links in a cell-type-specific manner. We develop methods for multimodal integration and detecting regulatory modules using peak-to-gene linking. We show AD risk loci are enriched in microglial enhancers and for specific TFs including SPI1, ELF2, and RUNX1. We detect 9,628 cell-type-specific ATAC-QTL loci, which we integrate alongside peak-to-gene links to prioritize AD variant regulatory circuits. We report differential accessibility of regulatory modules in late AD in glia and in early AD in neurons. Strikingly, late-stage AD brains show global epigenome dysregulation indicative of epigenome erosion and cell identity loss.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Regulación de la Expresión Génica , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Encéfalo/patología , Epigenoma , Epigenómica , Estudio de Asociación del Genoma Completo
4.
Cell ; 186(20): 4365-4385.e27, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37774677

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia worldwide, but the molecular and cellular mechanisms underlying cognitive impairment remain poorly understood. To address this, we generated a single-cell transcriptomic atlas of the aged human prefrontal cortex covering 2.3 million cells from postmortem human brain samples of 427 individuals with varying degrees of AD pathology and cognitive impairment. Our analyses identified AD-pathology-associated alterations shared between excitatory neuron subtypes, revealed a coordinated increase of the cohesin complex and DNA damage response factors in excitatory neurons and in oligodendrocytes, and uncovered genes and pathways associated with high cognitive function, dementia, and resilience to AD pathology. Furthermore, we identified selectively vulnerable somatostatin inhibitory neuron subtypes depleted in AD, discovered two distinct groups of inhibitory neurons that were more abundant in individuals with preserved high cognitive function late in life, and uncovered a link between inhibitory neurons and resilience to AD pathology.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Anciano , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Cognición , Disfunción Cognitiva/metabolismo , Neuronas/metabolismo
5.
Cell ; 177(2): 256-271.e22, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30879788

RESUMEN

We previously reported that inducing gamma oscillations with a non-invasive light flicker (gamma entrainment using sensory stimulus or GENUS) impacted pathology in the visual cortex of Alzheimer's disease mouse models. Here, we designed auditory tone stimulation that drove gamma frequency neural activity in auditory cortex (AC) and hippocampal CA1. Seven days of auditory GENUS improved spatial and recognition memory and reduced amyloid in AC and hippocampus of 5XFAD mice. Changes in activation responses were evident in microglia, astrocytes, and vasculature. Auditory GENUS also reduced phosphorylated tau in the P301S tauopathy model. Furthermore, combined auditory and visual GENUS, but not either alone, produced microglial-clustering responses, and decreased amyloid in medial prefrontal cortex. Whole brain analysis using SHIELD revealed widespread reduction of amyloid plaques throughout neocortex after multi-sensory GENUS. Thus, GENUS can be achieved through multiple sensory modalities with wide-ranging effects across multiple brain areas to improve cognitive function.


Asunto(s)
Estimulación Acústica/métodos , Enfermedad de Alzheimer/terapia , Cognición/fisiología , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Percepción Auditiva/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ritmo Gamma/fisiología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Placa Amiloide/metabolismo
6.
Cell ; 169(6): 1029-1041.e16, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575667

RESUMEN

We report a noninvasive strategy for electrically stimulating neurons at depth. By delivering to the brain multiple electric fields at frequencies too high to recruit neural firing, but which differ by a frequency within the dynamic range of neural firing, we can electrically stimulate neurons throughout a region where interference between the multiple fields results in a prominent electric field envelope modulated at the difference frequency. We validated this temporal interference (TI) concept via modeling and physics experiments, and verified that neurons in the living mouse brain could follow the electric field envelope. We demonstrate the utility of TI stimulation by stimulating neurons in the hippocampus of living mice without recruiting neurons of the overlying cortex. Finally, we show that by altering the currents delivered to a set of immobile electrodes, we can steerably evoke different motor patterns in living mice.


Asunto(s)
Estimulación Encefálica Profunda/métodos , Estimulación Transcraneal de Corriente Directa/métodos , Animales , Estimulación Encefálica Profunda/efectos adversos , Estimulación Encefálica Profunda/instrumentación , Electrodos , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Estimulación Transcraneal de Corriente Directa/efectos adversos , Estimulación Transcraneal de Corriente Directa/instrumentación
7.
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
8.
Nature ; 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39048816

RESUMEN

Alzheimer's disease is the leading cause of dementia worldwide, but the cellular pathways that underlie its pathological progression across brain regions remain poorly understood1-3. Here we report a single-cell transcriptomic atlas of six different brain regions in the aged human brain, covering 1.3 million cells from 283 post-mortem human brain samples across 48 individuals with and without Alzheimer's disease. We identify 76 cell types, including region-specific subtypes of astrocytes and excitatory neurons and an inhibitory interneuron population unique to the thalamus and distinct from canonical inhibitory subclasses. We identify vulnerable populations of excitatory and inhibitory neurons that are depleted in specific brain regions in Alzheimer's disease, and provide evidence that the Reelin signalling pathway is involved in modulating the vulnerability of these neurons. We develop a scalable method for discovering gene modules, which we use to identify cell-type-specific and region-specific modules that are altered in Alzheimer's disease and to annotate transcriptomic differences associated with diverse pathological variables. We identify an astrocyte program that is associated with cognitive resilience to Alzheimer's disease pathology, tying choline metabolism and polyamine biosynthesis in astrocytes to preserved cognitive function late in life. Together, our study develops a regional atlas of the ageing human brain and provides insights into cellular vulnerability, response and resilience to Alzheimer's disease pathology.

9.
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
10.
Mol Cell ; 82(20): 3794-3809.e8, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36206766

RESUMEN

Neuronal activity induces topoisomerase IIß (Top2B) to generate DNA double-strand breaks (DSBs) within the promoters of neuronal early response genes (ERGs) and facilitate their transcription, and yet, the mechanisms that control Top2B-mediated DSB formation are unknown. Here, we report that stimulus-dependent calcium influx through NMDA receptors activates the phosphatase calcineurin to dephosphorylate Top2B at residues S1509 and S1511, which stimulates its DNA cleavage activity and induces it to form DSBs. Exposing mice to a fear conditioning paradigm also triggers Top2B dephosphorylation at S1509 and S1511 in the hippocampus, indicating that calcineurin also regulates Top2B-mediated DSB formation following physiological neuronal activity. Furthermore, calcineurin-Top2B interactions following neuronal activity and sites that incur activity-induced DSBs are preferentially localized at the nuclear periphery in neurons. Together, these results reveal how radial gene positioning and the compartmentalization of activity-dependent signaling govern the position and timing of activity-induced DSBs and regulate gene expression patterns in neurons.


Asunto(s)
Calcineurina , Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo II , Neuronas , Animales , Ratones , Calcineurina/genética , Calcineurina/metabolismo , Calcio/metabolismo , ADN , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/genética
11.
Cell ; 156(1-2): 261-76, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439381

RESUMEN

Traumatic events generate some of the most enduring forms of memories. Despite the elevated lifetime prevalence of anxiety disorders, effective strategies to attenuate long-term traumatic memories are scarce. The most efficacious treatments to diminish recent (i.e., day-old) traumata capitalize on memory updating mechanisms during reconsolidation that are initiated upon memory recall. Here, we show that, in mice, successful reconsolidation-updating paradigms for recent memories fail to attenuate remote (i.e., month-old) ones. We find that, whereas recent memory recall induces a limited period of hippocampal neuroplasticity mediated, in part, by S-nitrosylation of HDAC2 and histone acetylation, such plasticity is absent for remote memories. However, by using an HDAC2-targeting inhibitor (HDACi) during reconsolidation, even remote memories can be persistently attenuated. This intervention epigenetically primes the expression of neuroplasticity-related genes, which is accompanied by higher metabolic, synaptic, and structural plasticity. Thus, applying HDACis during memory reconsolidation might constitute a treatment option for remote traumata.


Asunto(s)
Miedo , Memoria a Largo Plazo , Plasticidad Neuronal , Animales , Epigénesis Genética , Hipocampo/metabolismo , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Transcriptoma
12.
Cell ; 157(2): 486-498, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725413

RESUMEN

Cyclin-dependent kinase 5 regulates numerous neuronal functions with its activator, p35. Under neurotoxic conditions, p35 undergoes proteolytic cleavage to liberate p25, which has been implicated in various neurodegenerative diseases. Here, we show that p25 is generated following neuronal activity under physiological conditions in a GluN2B- and CaMKIIα-dependent manner. Moreover, we developed a knockin mouse model in which endogenous p35 is replaced with a calpain-resistant mutant p35 (Δp35KI) to prevent p25 generation. The Δp35KI mice exhibit impaired long-term depression and defective memory extinction, likely mediated through persistent GluA1 phosphorylation at Ser845. Finally, crossing the Δp35KI mice with the 5XFAD mouse model of Alzheimer's disease (AD) resulted in an amelioration of ß-amyloid (Aß)-induced synaptic depression and cognitive impairment. Together, these results reveal a physiological role of p25 production in synaptic plasticity and memory and provide new insights into the function of p25 in Aß-associated neurotoxicity and AD-like pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Calpaína/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cognición , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Endocitosis , Técnicas de Sustitución del Gen , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo , Depresión Sináptica a Largo Plazo , Ratones , Proteínas del Tejido Nervioso/genética , Fosfotransferasas , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis
13.
Genes Dev ; 35(3-4): 180-198, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33526585

RESUMEN

Oligodendrocyte precursor cells (OPCs) are not merely a transitory progenitor cell type, but rather a distinct and heterogeneous population of glia with various functions in the developing and adult central nervous system. In this review, we discuss the fate and function of OPCs in the brain beyond their contribution to myelination. OPCs are electrically sensitive, form synapses with neurons, support blood-brain barrier integrity, and mediate neuroinflammation. We explore how sex and age may influence OPC activity, and we review how OPC dysfunction may play a primary role in numerous neurological and neuropsychiatric diseases. Finally, we highlight areas of future research.


Asunto(s)
Encéfalo/citología , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/inmunología , Factores de Edad , Animales , Encéfalo/embriología , Encéfalo/inmunología , Encéfalo/metabolismo , Sinapsis Eléctricas/fisiología , Humanos , Trastornos Mentales/patología , Enfermedades del Sistema Nervioso/patología , Células Precursoras de Oligodendrocitos/patología , Células Precursoras de Oligodendrocitos/fisiología , Factores Sexuales
14.
Nature ; 603(7903): 893-899, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35158371

RESUMEN

Despite the importance of the cerebrovasculature in maintaining normal brain physiology and in understanding neurodegeneration and drug delivery to the central nervous system1, human cerebrovascular cells remain poorly characterized owing to their sparsity and dispersion. Here we perform single-cell characterization of the human cerebrovasculature using both ex vivo fresh tissue experimental enrichment and post mortem in silico sorting of human cortical tissue samples. We capture 16,681 cerebrovascular nuclei across 11 subtypes, including endothelial cells, mural cells and three distinct subtypes of perivascular fibroblast along the vasculature. We uncover human-specific expression patterns along the arteriovenous axis and determine previously uncharacterized cell-type-specific markers. We use these human-specific signatures to study changes in 3,945 cerebrovascular cells from patients with Huntington's disease, which reveal activation of innate immune signalling in vascular and glial cell types and a concomitant reduction in the levels of proteins critical for maintenance of blood-brain barrier integrity. Finally, our study provides a comprehensive molecular atlas of the human cerebrovasculature to guide future biological and therapeutic studies.


Asunto(s)
Células Endoteliales , Enfermedad de Huntington , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Sistema Inmunológico , Neuroglía , Proteínas/metabolismo
15.
Nature ; 611(7937): 769-779, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36385529

RESUMEN

APOE4 is the strongest genetic risk factor for Alzheimer's disease1-3. However, the effects of APOE4 on the human brain are not fully understood, limiting opportunities to develop targeted therapeutics for individuals carrying APOE4 and other risk factors for Alzheimer's disease4-8. Here, to gain more comprehensive insights into the impact of APOE4 on the human brain, we performed single-cell transcriptomics profiling of post-mortem human brains from APOE4 carriers compared with non-carriers. This revealed that APOE4 is associated with widespread gene expression changes across all cell types of the human brain. Consistent with the biological function of APOE2-6, APOE4 significantly altered signalling pathways associated with cholesterol homeostasis and transport. Confirming these findings with histological and lipidomic analysis of the post-mortem human brain, induced pluripotent stem-cell-derived cells and targeted-replacement mice, we show that cholesterol is aberrantly deposited in oligodendrocytes-myelinating cells that are responsible for insulating and promoting the electrical activity of neurons. We show that altered cholesterol localization in the APOE4 brain coincides with reduced myelination. Pharmacologically facilitating cholesterol transport increases axonal myelination and improves learning and memory in APOE4 mice. We provide a single-cell atlas describing the transcriptional effects of APOE4 on the aging human brain and establish a functional link between APOE4, cholesterol, myelination and memory, offering therapeutic opportunities for Alzheimer's disease.


Asunto(s)
Apolipoproteína E4 , Encéfalo , Colesterol , Fibras Nerviosas Mielínicas , Oligodendroglía , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Colesterol/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/patología , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/patología , Autopsia , Células Madre Pluripotentes Inducidas , Neuronas/metabolismo , Neuronas/patología , Heterocigoto , Transporte Biológico , Homeostasis , Análisis de la Célula Individual , Memoria , Envejecimiento/genética , Perfilación de la Expresión Génica , Vaina de Mielina/metabolismo , Vaina de Mielina/patología
16.
Immunity ; 48(3): 476-478, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29562197

RESUMEN

The epigenetic mechanisms controlling microglia functions are largely unknown. In this issue of Immunity, Datta et al. (2018) uncover surprising and distinct effects of deleting the epigenetic regulators HDAC1 and HDAC2 during microglial development versus during the course of neurodegeneration.


Asunto(s)
Histona Desacetilasa 2/genética , Microglía , Epigénesis Genética
17.
Proc Natl Acad Sci U S A ; 120(47): e2300308120, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37976261

RESUMEN

Spinal muscular atrophy (SMA), the top genetic cause of infant mortality, is characterized by motor neuron degeneration. Mechanisms underlying SMA pathogenesis remain largely unknown. Here, we report that the activity of cyclin-dependent kinase 5 (Cdk5) and the conversion of its activating subunit p35 to the more potent activator p25 are significantly up-regulated in mouse models and human induced pluripotent stem cell (iPSC) models of SMA. The increase of Cdk5 activity occurs before the onset of SMA phenotypes, suggesting that it may be an initiator of the disease. Importantly, aberrant Cdk5 activation causes mitochondrial defects and motor neuron degeneration, as the genetic knockout of p35 in an SMA mouse model rescues mitochondrial transport and fragmentation defects, and alleviates SMA phenotypes including motor neuron hyperexcitability, loss of excitatory synapses, neuromuscular junction denervation, and motor neuron degeneration. Inhibition of the Cdk5 signaling pathway reduces the degeneration of motor neurons derived from SMA mice and human SMA iPSCs. Altogether, our studies reveal a critical role for the aberrant activation of Cdk5 in SMA pathogenesis and suggest a potential target for therapeutic intervention.


Asunto(s)
Células Madre Pluripotentes Inducidas , Atrofia Muscular Espinal , Animales , Humanos , Ratones , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/metabolismo , Degeneración Nerviosa/patología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo
18.
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
19.
Ann Neurol ; 95(4): 625-634, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38180638

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder and one of the leading causes of disability worldwide. The apolipoprotein E4 gene (APOE4) is the strongest genetic risk factor for AD. In 2023, the APOE4 National Institute on Aging/Alzheimer's Disease Sequencing Project working group came together to gather data and discuss the question of whether to reduce or increase APOE4 as a therapeutic intervention for AD. It was the unanimous consensus that cumulative data from multiple studies in humans and animal models support that lowering APOE4 should be a target for therapeutic approaches for APOE4 carriers. ANN NEUROL 2024;95:625-634.


Asunto(s)
Enfermedad de Alzheimer , Animales , Estados Unidos , Humanos , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/tratamiento farmacológico , Apolipoproteína E4/genética , Objetivos , National Institute on Aging (U.S.)
20.
Cell ; 140(1): 30-2, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-20085703

RESUMEN

The microtubule network is crucial for the developing nervous system, and mutations in tubulin-encoding genes disrupt neuronal migration. Tischfield et al. (2010) now report that mutations in the tubulin-encoding gene TUBB3 have a striking impact on microtubule dynamics in neurons, resulting in a diverse set of disease symptoms.


Asunto(s)
Encéfalo/embriología , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Encéfalo/metabolismo , Humanos , Mutación , Neurogénesis , Tubulina (Proteína)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA