Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 42.634
Filtrar
Más filtros

Publication year range
1.
Cell ; 186(19): 4117-4133.e22, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37591239

RESUMEN

Aging is the key risk factor for cognitive decline, yet the molecular changes underlying brain aging remain poorly understood. Here, we conducted spatiotemporal RNA sequencing of the mouse brain, profiling 1,076 samples from 15 regions across 7 ages and 2 rejuvenation interventions. Our analysis identified a brain-wide gene signature of aging in glial cells, which exhibited spatially defined changes in magnitude. By integrating spatial and single-nucleus transcriptomics, we found that glial aging was particularly accelerated in white matter compared with cortical regions, whereas specialized neuronal populations showed region-specific expression changes. Rejuvenation interventions, including young plasma injection and dietary restriction, exhibited distinct effects on gene expression in specific brain regions. Furthermore, we discovered differential gene expression patterns associated with three human neurodegenerative diseases, highlighting the importance of regional aging as a potential modulator of disease. Our findings identify molecular foci of brain aging, providing a foundation to target age-related cognitive decline.


Asunto(s)
Envejecimiento , Disfunción Cognitiva , Sustancia Blanca , Animales , Humanos , Ratones , Disfunción Cognitiva/genética , Perfilación de la Expresión Génica , Núcleo Solitario , Sustancia Blanca/patología , Análisis de Expresión Génica de una Sola Célula , Encéfalo/patología
2.
Cell ; 186(5): 1026-1038.e20, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36868208

RESUMEN

Down syndrome (DS) is a neurological disorder with multiple immune-related symptoms; however, crosstalk between the CNS and peripheral immune system remains unexplored. Using parabiosis and plasma infusion, we found that blood-borne factors drive synaptic deficits in DS. Proteomic analysis revealed elevation of ß2-microglobulin (B2M), a major histocompatibility complex class I (MHC-I) component, in human DS plasma. Systemic administration of B2M in wild-type mice led to synaptic and memory defects similar to those observed in DS mice. Moreover, genetic ablation of B2m or systemic administration of an anti-B2M antibody counteracts synaptic impairments in DS mice. Mechanistically, we demonstrate that B2M antagonizes NMDA receptor (NMDAR) function through interactions with the GluN1-S2 loop; blocking B2M-NMDAR interactions using competitive peptides restores NMDAR-dependent synaptic function. Our findings identify B2M as an endogenous NMDAR antagonist and reveal a pathophysiological role for circulating B2M in NMDAR dysfunction in DS and related cognitive disorders.


Asunto(s)
Síndrome de Down , Receptores de N-Metil-D-Aspartato , Microglobulina beta-2 , Animales , Humanos , Ratones , Microglobulina beta-2/metabolismo , Microglobulina beta-2/farmacología , Disfunción Cognitiva/metabolismo , Reacciones Cruzadas , Parabiosis , Proteómica , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Síndrome de Down/sangre , Síndrome de Down/metabolismo
3.
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
4.
Cell ; 185(26): 5028-5039.e13, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36516855

RESUMEN

Cerebrospinal fluid (CSF) contains a tightly regulated immune system. However, knowledge is lacking about how CSF immunity is altered with aging or neurodegenerative disease. Here, we performed single-cell RNA sequencing on CSF from 45 cognitively normal subjects ranging from 54 to 82 years old. We uncovered an upregulation of lipid transport genes in monocytes with age. We then compared this cohort with 14 cognitively impaired subjects. In cognitively impaired subjects, downregulation of lipid transport genes in monocytes occurred concomitantly with altered cytokine signaling to CD8 T cells. Clonal CD8 T effector memory cells upregulated C-X-C motif chemokine receptor 6 (CXCR6) in cognitively impaired subjects. The CXCR6 ligand, C-X-C motif chemokine ligand 16 (CXCL16), was elevated in the CSF of cognitively impaired subjects, suggesting CXCL16-CXCR6 signaling as a mechanism for antigen-specific T cell entry into the brain. Cumulatively, these results reveal cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedades Neurodegenerativas , Humanos , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Ligandos , Encéfalo , Envejecimiento , Lípidos , Biomarcadores
5.
Cell ; 185(14): 2391-2393, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35768007

RESUMEN

Acute mild respiratory SARS-CoV-2 infection can lead to a more chronic cognitive syndrome known as "COVID fog." New findings from Fernández-Castañeda et al. reveal how glial dysregulation and consequent neural circuit dysfunction may contribute to cognitive impairments in long COVID.


Asunto(s)
COVID-19 , Disfunción Cognitiva , COVID-19/complicaciones , Humanos , SARS-CoV-2 , Síndrome Post Agudo de COVID-19
6.
Cell ; 180(5): 833-846.e16, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32142677

RESUMEN

Cognitive dysfunction and reactive microglia are hallmarks of traumatic brain injury (TBI), yet whether these cells contribute to cognitive deficits and secondary inflammatory pathology remains poorly understood. Here, we show that removal of microglia from the mouse brain has little effect on the outcome of TBI, but inducing the turnover of these cells through either pharmacologic or genetic approaches can yield a neuroprotective microglial phenotype that profoundly aids recovery. The beneficial effects of these repopulating microglia are critically dependent on interleukin-6 (IL-6) trans-signaling via the soluble IL-6 receptor (IL-6R) and robustly support adult neurogenesis, specifically by augmenting the survival of newborn neurons that directly support cognitive function. We conclude that microglia in the mammalian brain can be manipulated to adopt a neuroprotective and pro-regenerative phenotype that can aid repair and alleviate the cognitive deficits arising from brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Interleucina-6/genética , Receptores de Interleucina-6/genética , Regeneración/genética , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Disfunción Cognitiva/terapia , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/patología , Ratones , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/genética
7.
Cell ; 176(1-2): 2-4, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30633904

RESUMEN

With the advent of more successful chemotherapies, the number of cancer survivors continues to increase. Unfortunately, many of these patients will exhibit long-term sequelae from their treatments, including serious cognitive deficits that impair daily function. In this issue of Cell, Gibson et al. (2019) demonstrate that chemotherapy-related cognitive impairment is orchestrated by microglia.


Asunto(s)
Trastornos del Conocimiento , Disfunción Cognitiva , Humanos , Metotrexato
8.
Cell ; 178(6): 1282-1284, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31474365

RESUMEN

Can we one day prevent mental disorders? Mukherjee et al. (2019) use a genetic mouse model of schizophrenia-risk with established abnormalities in adult hippocampal-prefrontal circuit function and cognitive behaviors to identify circuit-specific treatments during adolescence that prevent the onset of the adult deficits.


Asunto(s)
Disfunción Cognitiva , Esquizofrenia , Adolescente , Adulto , Animales , Modelos Animales de Enfermedad , Hipocampo , Humanos , Ratones , Corteza Prefrontal
9.
Cell ; 176(1-2): 43-55.e13, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30528430

RESUMEN

Chemotherapy results in a frequent yet poorly understood syndrome of long-term neurological deficits. Neural precursor cell dysfunction and white matter dysfunction are thought to contribute to this debilitating syndrome. Here, we demonstrate persistent depletion of oligodendrocyte lineage cells in humans who received chemotherapy. Developing a mouse model of methotrexate chemotherapy-induced neurological dysfunction, we find a similar depletion of white matter OPCs, increased but incomplete OPC differentiation, and a persistent deficit in myelination. OPCs from chemotherapy-naive mice similarly exhibit increased differentiation when transplanted into the microenvironment of previously methotrexate-exposed brains, indicating an underlying microenvironmental perturbation. Methotrexate results in persistent activation of microglia and subsequent astrocyte activation that is dependent on inflammatory microglia. Microglial depletion normalizes oligodendroglial lineage dynamics, myelin microstructure, and cognitive behavior after methotrexate chemotherapy. These findings indicate that methotrexate chemotherapy exposure is associated with persistent tri-glial dysregulation and identify inflammatory microglia as a therapeutic target to abrogate chemotherapy-related cognitive impairment. VIDEO ABSTRACT.


Asunto(s)
Disfunción Cognitiva/inducido químicamente , Metotrexato/efectos adversos , Oligodendroglía/efectos de los fármacos , Animales , Encéfalo/metabolismo , Diferenciación Celular , Linaje de la Célula , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Quimioterapia , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Metotrexato/farmacología , Ratones , Microglía/metabolismo , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas , Neurogénesis/fisiología , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Oligodendroglía/metabolismo , Sustancia Blanca/metabolismo
10.
Cell ; 178(6): 1387-1402.e14, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31474363

RESUMEN

Although sensitizing processes occur earlier, schizophrenia is diagnosed in young adulthood, which suggests that it might involve a pathological transition during late brain development in predisposed individuals. Parvalbumin (PV) interneuron alterations have been noticed, but their role in the disease is unclear. Here we demonstrate that adult LgDel+/- mice, a genetic model of schizophrenia, exhibit PV neuron hypo-recruitment and associated chronic PV neuron plasticity together with network and cognitive deficits. All these deficits can be permanently rescued by chemogenetic activation of PV neurons or D2R antagonist treatments, specifically in the ventral hippocampus (vH) or medial-prefrontal cortex during a late-adolescence-sensitive time window. PV neuron alterations were initially restricted to the hippocampal CA1/subiculum, where they became responsive to treatment in late adolescence. Therefore, progression to disease in schizophrenia-model mice can be prevented by treatments supporting vH-mPFC PV network function during a sensitive time window late in adolescence, suggesting therapeutic strategies to prevent the outbreak of schizophrenia.


Asunto(s)
Disfunción Cognitiva/terapia , Antagonistas de los Receptores de Dopamina D2/farmacología , Hipocampo/efectos de los fármacos , Interneuronas/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Esquizofrenia/terapia , Adolescente , Animales , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Parvalbúminas/metabolismo , Corteza Prefrontal/patología
13.
Nat Immunol ; 19(2): 151-161, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29292385

RESUMEN

Memory impairment following West Nile virus neuroinvasive disease (WNND) is associated with loss of hippocampal synapses with lack of recovery. Adult neurogenesis and synaptogenesis are fundamental features of hippocampal repair, which suggests that viruses affect these processes. Here, in an established model of WNND-induced cognitive dysfunction, transcriptional profiling revealed alterations in the expression of genes encoding molecules that limit adult neurogenesis, including interleukin 1 (IL-1). Mice that had recovered from WNND exhibited fewer neuroblasts and increased astrogenesis without recovery of hippocampal neurogenesis at 30 d. Analysis of cytokine production in microglia and astrocytes isolated ex vivo revealed that the latter were the predominant source of IL-1. Mice deficient in the IL-1 receptor IL-1R1 and that had recovered from WNND exhibited normal neurogenesis, recovery of presynaptic termini and resistance to spatial learning defects, the last of which likewise occurred after treatment with an IL-1R1 antagonist. Thus, 'preferential' generation of proinflammatory astrocytes impaired the homeostasis of neuronal progenitor cells via expression of IL-1; this might underlie the long-term cognitive consequences of WNND but also provides a therapeutic target.


Asunto(s)
Astrocitos/metabolismo , Interleucina-1/biosíntesis , Neurogénesis/fisiología , Fiebre del Nilo Occidental/complicaciones , Células Madre Adultas/metabolismo , Animales , Astrocitos/inmunología , Diferenciación Celular/fisiología , Disfunción Cognitiva/etiología , Trastornos de la Memoria/etiología , Ratones , Células-Madre Neurales/metabolismo
14.
Nature ; 628(8006): 145-153, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538785

RESUMEN

As hippocampal neurons respond to diverse types of information1, a subset assembles into microcircuits representing a memory2. Those neurons typically undergo energy-intensive molecular adaptations, occasionally resulting in transient DNA damage3-5. Here we found discrete clusters of excitatory hippocampal CA1 neurons with persistent double-stranded DNA (dsDNA) breaks, nuclear envelope ruptures and perinuclear release of histone and dsDNA fragments hours after learning. Following these early events, some neurons acquired an inflammatory phenotype involving activation of TLR9 signalling and accumulation of centrosomal DNA damage repair complexes6. Neuron-specific knockdown of Tlr9 impaired memory while blunting contextual fear conditioning-induced changes of gene expression in specific clusters of excitatory CA1 neurons. Notably, TLR9 had an essential role in centrosome function, including DNA damage repair, ciliogenesis and build-up of perineuronal nets. We demonstrate a novel cascade of learning-induced molecular events in discrete neuronal clusters undergoing dsDNA damage and TLR9-mediated repair, resulting in their recruitment to memory circuits. With compromised TLR9 function, this fundamental memory mechanism becomes a gateway to genomic instability and cognitive impairments implicated in accelerated senescence, psychiatric disorders and neurodegenerative disorders. Maintaining the integrity of TLR9 inflammatory signalling thus emerges as a promising preventive strategy for neurocognitive deficits.


Asunto(s)
Región CA1 Hipocampal , Roturas del ADN de Doble Cadena , Reparación del ADN , Inflamación , Memoria , Receptor Toll-Like 9 , Animales , Femenino , Masculino , Ratones , Envejecimiento/genética , Envejecimiento/patología , Región CA1 Hipocampal/fisiología , Centrosoma/metabolismo , Disfunción Cognitiva/genética , Condicionamiento Clásico , Matriz Extracelular/metabolismo , Miedo , Inestabilidad Genómica/genética , Histonas/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Memoria/fisiología , Trastornos Mentales/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neuroinflamatorias/genética , Neuronas/metabolismo , Neuronas/patología , Membrana Nuclear/patología , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo
15.
Nature ; 620(7973): 374-380, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37532932

RESUMEN

Low-grade inflammation is a hallmark of old age and a central driver of ageing-associated impairment and disease1. Multiple factors can contribute to ageing-associated inflammation2; however, the molecular pathways that transduce aberrant inflammatory signalling and their impact in natural ageing remain unclear. Here we show that the cGAS-STING signalling pathway, which mediates immune sensing of DNA3, is a critical driver of chronic inflammation and functional decline during ageing. Blockade of STING suppresses the inflammatory phenotypes of senescent human cells and tissues, attenuates ageing-related inflammation in multiple peripheral organs and the brain in mice, and leads to an improvement in tissue function. Focusing on the ageing brain, we reveal that activation of STING triggers reactive microglial transcriptional states, neurodegeneration and cognitive decline. Cytosolic DNA released from perturbed mitochondria elicits cGAS activity in old microglia, defining a mechanism by which cGAS-STING signalling is engaged in the ageing brain. Single-nucleus RNA-sequencing analysis of microglia and hippocampi of a cGAS gain-of-function mouse model demonstrates that engagement of cGAS in microglia is sufficient to direct ageing-associated transcriptional microglial states leading to bystander cell inflammation, neurotoxicity and impaired memory capacity. Our findings establish the cGAS-STING pathway as a driver of ageing-related inflammation in peripheral organs and the brain, and reveal blockade of cGAS-STING signalling as a potential strategy to halt neurodegenerative processes during old age.


Asunto(s)
Envejecimiento , Encéfalo , Disfunción Cognitiva , Inflamación , Proteínas de la Membrana , Enfermedades Neurodegenerativas , Nucleotidiltransferasas , Animales , Humanos , Ratones , Envejecimiento/metabolismo , Envejecimiento/patología , Encéfalo/metabolismo , Encéfalo/patología , Efecto Espectador , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , ADN/inmunología , Inflamación/enzimología , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/metabolismo , Microglía/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/metabolismo , Nucleotidiltransferasas/metabolismo , Especificidad de Órganos , Transducción de Señal , Hipocampo/metabolismo , Hipocampo/patología
16.
Nature ; 620(7976): 1071-1079, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37587343

RESUMEN

Identifying therapeutics to delay, and potentially reverse, age-related cognitive decline is critical in light of the increased incidence of dementia-related disorders forecasted in the growing older population1. Here we show that platelet factors transfer the benefits of young blood to the ageing brain. Systemic exposure of aged male mice to a fraction of blood plasma from young mice containing platelets decreased neuroinflammation in the hippocampus at the transcriptional and cellular level and ameliorated hippocampal-dependent cognitive impairments. Circulating levels of the platelet-derived chemokine platelet factor 4 (PF4) (also known as CXCL4) were elevated in blood plasma preparations of young mice and humans relative to older individuals. Systemic administration of exogenous PF4 attenuated age-related hippocampal neuroinflammation, elicited synaptic-plasticity-related molecular changes and improved cognition in aged mice. We implicate decreased levels of circulating pro-ageing immune factors and restoration of the ageing peripheral immune system in the beneficial effects of systemic PF4 on the aged brain. Mechanistically, we identified CXCR3 as a chemokine receptor that, in part, mediates the cellular, molecular and cognitive benefits of systemic PF4 on the aged brain. Together, our data identify platelet-derived factors as potential therapeutic targets to abate inflammation and rescue cognition in old age.


Asunto(s)
Envejecimiento , Cognición , Disfunción Cognitiva , Enfermedades Neuroinflamatorias , Nootrópicos , Factor Plaquetario 4 , Animales , Masculino , Ratones , Envejecimiento/sangre , Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Cognición/efectos de los fármacos , Cognición/fisiología , Enfermedades Neuroinflamatorias/sangre , Enfermedades Neuroinflamatorias/complicaciones , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/prevención & control , Factor Plaquetario 4/sangre , Factor Plaquetario 4/metabolismo , Factor Plaquetario 4/farmacología , Factor Plaquetario 4/uso terapéutico , Nootrópicos/sangre , Nootrópicos/metabolismo , Nootrópicos/farmacología , Nootrópicos/uso terapéutico , Plasma/química , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Disfunción Cognitiva/sangre , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/prevención & control , Transcripción Genética/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos
17.
Nature ; 624(7990): 164-172, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38057571

RESUMEN

Animal studies show aging varies between individuals as well as between organs within an individual1-4, but whether this is true in humans and its effect on age-related diseases is unknown. We utilized levels of human blood plasma proteins originating from specific organs to measure organ-specific aging differences in living individuals. Using machine learning models, we analysed aging in 11 major organs and estimated organ age reproducibly in five independent cohorts encompassing 5,676 adults across the human lifespan. We discovered nearly 20% of the population show strongly accelerated age in one organ and 1.7% are multi-organ agers. Accelerated organ aging confers 20-50% higher mortality risk, and organ-specific diseases relate to faster aging of those organs. We find individuals with accelerated heart aging have a 250% increased heart failure risk and accelerated brain and vascular aging predict Alzheimer's disease (AD) progression independently from and as strongly as plasma pTau-181 (ref. 5), the current best blood-based biomarker for AD. Our models link vascular calcification, extracellular matrix alterations and synaptic protein shedding to early cognitive decline. We introduce a simple and interpretable method to study organ aging using plasma proteomics data, predicting diseases and aging effects.


Asunto(s)
Envejecimiento , Biomarcadores , Enfermedad , Salud , Especificidad de Órganos , Proteoma , Proteómica , Adulto , Humanos , Envejecimiento/sangre , Enfermedad de Alzheimer/sangre , Biomarcadores/sangre , Encéfalo/metabolismo , Disfunción Cognitiva/sangre , Proteoma/análisis , Aprendizaje Automático , Estudios de Cohortes , Progresión de la Enfermedad , Insuficiencia Cardíaca/sangre , Matriz Extracelular/metabolismo , Sinapsis/metabolismo , Calcificación Vascular/sangre , Corazón
18.
Annu Rev Neurosci ; 43: 297-314, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32097575

RESUMEN

An enduring problem in neuroscience is determining whether cases of amnesia result from eradication of the memory trace (storage impairment) or if the trace is present but inaccessible (retrieval impairment). The most direct approach to resolving this question is to quantify changes in the brain mechanisms of long-term memory (BM-LTM). This approach argues that if the amnesia is due to a retrieval failure, BM-LTM should remain at levels comparable to trained, unimpaired animals. Conversely, if memories are erased, BM-LTM should be reduced to resemble untrained levels. Here we review the use of BM-LTM in a number of studies that induced amnesia by targeting memory maintenance or reconsolidation. The literature strongly suggests that such amnesia is due to storage rather than retrieval impairments. We also describe the shortcomings of the purely behavioral protocol that purports to show recovery from amnesia as a method of understanding the nature of amnesia.


Asunto(s)
Amnesia/fisiopatología , Encéfalo/fisiopatología , Disfunción Cognitiva/fisiopatología , Memoria a Largo Plazo/fisiología , Animales , Humanos , Mantenimiento , Memoria a Corto Plazo/fisiología
19.
Nature ; 612(7938): 123-131, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36385530

RESUMEN

Aggregates of medin amyloid (a fragment of the protein MFG-E8, also known as lactadherin) are found in the vasculature of almost all humans over 50 years of age1,2, making it the most common amyloid currently known. We recently reported that medin also aggregates in blood vessels of ageing wild-type mice, causing cerebrovascular dysfunction3. Here we demonstrate in amyloid-ß precursor protein (APP) transgenic mice and in patients with Alzheimer's disease that medin co-localizes with vascular amyloid-ß deposits, and that in mice, medin deficiency reduces vascular amyloid-ß deposition by half. Moreover, in both the mouse and human brain, MFG-E8 is highly enriched in the vasculature and both MFG-E8 and medin levels increase with the severity of vascular amyloid-ß burden. Additionally, analysing data from 566 individuals in the ROSMAP cohort, we find that patients with Alzheimer's disease have higher MFGE8 expression levels, which are attributable to vascular cells and are associated with increased measures of cognitive decline, independent of plaque and tau pathology. Mechanistically, we demonstrate that medin interacts directly with amyloid-ß to promote its aggregation, as medin forms heterologous fibrils with amyloid-ß, affects amyloid-ß fibril structure, and cross-seeds amyloid-ß aggregation both in vitro and in vivo. Thus, medin could be a therapeutic target for prevention of vascular damage and cognitive decline resulting from amyloid-ß deposition in the blood vessels of the brain.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Precursor de Proteína beta-Amiloide , Animales , Humanos , Ratones , Persona de Mediana Edad , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Disfunción Cognitiva , Ratones Transgénicos , Placa Amiloide/metabolismo , Proteínas tau/metabolismo
20.
Mol Cell ; 77(6): 1176-1192.e16, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-31999954

RESUMEN

Microexons represent the most highly conserved class of alternative splicing, yet their functions are poorly understood. Here, we focus on closely related neuronal microexons overlapping prion-like domains in the translation initiation factors, eIF4G1 and eIF4G3, the splicing of which is activity dependent and frequently disrupted in autism. CRISPR-Cas9 deletion of these microexons selectively upregulates synaptic proteins that control neuronal activity and plasticity and further triggers a gene expression program mirroring that of activated neurons. Mice lacking the Eif4g1 microexon display social behavior, learning, and memory deficits, accompanied by altered hippocampal synaptic plasticity. We provide evidence that the eIF4G microexons function as a translational brake by causing ribosome stalling, through their propensity to promote the coalescence of cytoplasmic granule components associated with translation repression, including the fragile X mental retardation protein FMRP. The results thus reveal an autism-disrupted mechanism by which alternative splicing specializes neuronal translation to control higher order cognitive functioning.


Asunto(s)
Trastorno Autístico/fisiopatología , Disfunción Cognitiva/patología , Factor 4G Eucariótico de Iniciación/fisiología , Exones/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Neuroblastoma/patología , Neuronas/patología , Animales , Conducta Animal , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neurogénesis , Neuronas/metabolismo , Biosíntesis de Proteínas , Empalme del ARN , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda