Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Neurosci ; 26(12): 2104-2121, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37957317

RESUMEN

Apolipoprotein E4 (APOE4) is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD), leading to earlier age of clinical onset and exacerbating pathologies. There is a critical need to identify protective targets. Recently, a rare APOE variant, APOE3-R136S (Christchurch), was found to protect against early-onset AD in a PSEN1-E280A carrier. In this study, we sought to determine if the R136S mutation also protects against APOE4-driven effects in LOAD. We generated tauopathy mouse and human iPSC-derived neuron models carrying human APOE4 with the homozygous or heterozygous R136S mutation. We found that the homozygous R136S mutation rescued APOE4-driven Tau pathology, neurodegeneration and neuroinflammation. The heterozygous R136S mutation partially protected against APOE4-driven neurodegeneration and neuroinflammation but not Tau pathology. Single-nucleus RNA sequencing revealed that the APOE4-R136S mutation increased disease-protective and diminished disease-associated cell populations in a gene dose-dependent manner. Thus, the APOE-R136S mutation protects against APOE4-driven AD pathologies, providing a target for therapeutic development against AD.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Mutación/genética , Enfermedades Neuroinflamatorias , Tauopatías/genética
2.
bioRxiv ; 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-38014339

RESUMEN

Despite strong evidence supporting the involvement of both apolipoprotein E4 (APOE4) and microglia in Alzheimer's Disease (AD) pathogenesis, the effects of microglia on neuronal APOE4-driven AD pathogenesis remain elusive. Here, we examined such effects utilizing microglial depletion in a chimeric model with human neurons in mouse hippocampus. Specifically, we transplanted homozygous APOE4, isogenic APOE3, and APOE-knockout (APOE-KO) induced pluripotent stem cell (iPSC)-derived human neurons into the hippocampus of human APOE3 or APOE4 knock-in mice, and depleted microglia in half the chimeric mice. We found that both neuronal APOE and microglial presence were important for the formation of Aß and tau pathologies in an APOE isoform-dependent manner (APOE4 > APOE3). Single-cell RNA-sequencing analysis identified two pro-inflammatory microglial subtypes with high MHC-II gene expression that are enriched in chimeric mice with human APOE4 neuron transplants. These findings highlight the concerted roles of neuronal APOE, especially APOE4, and microglia in AD pathogenesis.

3.
bioRxiv ; 2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37873080

RESUMEN

ApoE4 is the primary risk factor for Alzheimer's Disease. While apoE is primarily expressed by astrocytes, AD pathology including endosomal abnormalities and mitochondrial dysfunction first occurs in neurons. Lysosomes are poised at the convergence point between these features. We find that apoE4-expressing cells exhibit lysosomal alkalinization, reduced lysosomal proteolysis, and impaired mitophagy. To identify driving factors for this lysosomal dysfunction, we performed quantitative lysosomal proteome profiling. This revealed that apoE4 expression results in lysosomal depletion of Lgals3bp and accumulation of Tmed5 in both Neuro-2a cells and postmitotic human neurons. Modulating the expression of both proteins affected lysosomal function, with Tmed5 knockdown rescuing lysosomal alkalinization in apoE4 cells, and Lgals3bp knockdown causing lysosomal alkalinization and reduced lysosomal density in apoE3 cells. Taken together, our work reveals that apoE4 exerts gain-of-toxicity by alkalinizing the lysosomal lumen, pinpointing lysosomal Tmed5 accumulation and Lgals3bp depletion as apoE4-associated drivers for this phenotype.

4.
Cell Rep ; 42(10): 113252, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37863057

RESUMEN

Apolipoprotein E4 (APOE4) is an important driver of Tau pathology, gliosis, and degeneration in Alzheimer's disease (AD). Still, the mechanisms underlying these APOE4-driven pathological effects remain elusive. Here, we report in a tauopathy mouse model that APOE4 promoted the nucleocytoplasmic translocation and release of high-mobility group box 1 (HMGB1) from hippocampal neurons, which correlated with the severity of hippocampal microgliosis and degeneration. Injection of HMGB1 into the hippocampus of young APOE4-tauopathy mice induced considerable and persistent gliosis. Selective removal of neuronal APOE4 reduced HMGB1 translocation and release. Treatment of APOE4-tauopathy mice with HMGB1 inhibitors effectively blocked the intraneuronal translocation and release of HMGB1 and ameliorated the development of APOE4-driven gliosis, Tau pathology, neurodegeneration, and myelin deficits. Single-nucleus RNA sequencing revealed that treatment with HMGB1 inhibitors diminished disease-associated and enriched disease-protective subpopulations of neurons, microglia, and astrocytes in APOE4-tauopathy mice. Thus, HMGB1 inhibitors represent a promising approach for treating APOE4-related AD.


Asunto(s)
Enfermedad de Alzheimer , Proteína HMGB1 , Tauopatías , Animales , Ratones , Enfermedad de Alzheimer/patología , Apolipoproteína E4/genética , Gliosis , Ratones Transgénicos , Tauopatías/tratamiento farmacológico
5.
Nat Aging ; 3(3): 275-296, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37118426

RESUMEN

Apolipoprotein E4 (APOE4) is the strongest known genetic risk factor for late-onset Alzheimer's disease (AD). Conditions of stress or injury induce APOE expression within neurons, but the role of neuronal APOE4 in AD pathogenesis is still unclear. Here we report the characterization of neuronal APOE4 effects on AD-related pathologies in an APOE4-expressing tauopathy mouse model. The selective genetic removal of APOE4 from neurons led to a significant reduction in tau pathology, gliosis, neurodegeneration, neuronal hyperexcitability and myelin deficits. Single-nucleus RNA-sequencing revealed that the removal of neuronal APOE4 greatly diminished neurodegenerative disease-associated subpopulations of neurons, oligodendrocytes, astrocytes and microglia whose accumulation correlated to the severity of tau pathology, neurodegeneration and myelin deficits. Thus, neuronal APOE4 plays a central role in promoting the development of major AD pathologies and its removal can mitigate the progressive cellular and tissue alterations occurring in this model of APOE4-driven tauopathy.


Asunto(s)
Enfermedades Neurodegenerativas , Tauopatías , Ratones , Animales , Apolipoproteína E4/genética , Enfermedades Neurodegenerativas/genética , Vaina de Mielina/metabolismo , Gliosis/genética , Tauopatías/genética , Neuronas/metabolismo
6.
Annu Rev Pathol ; 17: 73-99, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-34460318

RESUMEN

Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder that involves dysregulation of many cellular and molecular processes. It is notoriously difficult to develop therapeutics for AD due to its complex nature. Nevertheless, recent advancements in imaging technology and the development of innovative experimental techniques have allowed researchers to perform in-depth analyses to uncover the pathogenic mechanisms of AD. An important consideration when studying late-onset AD is its major genetic risk factor, apolipoprotein E4 (apoE4). Although the exact mechanisms underlying apoE4 effects on AD initiation and progression are not fully understood, recent studies have revealed critical insights into the apoE4-induced deficits that occur in AD. In this review, we highlight notable studies that detail apoE4 effects on prominent AD pathologies, including amyloid-ß, tau pathology, neuroinflammation, and neural network dysfunction. We also discuss evidence that defines the physiological functions of apoE and outlines how these functions are disrupted in apoE4-related AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Humanos
7.
Cell Rep ; 37(13): 110159, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34965435

RESUMEN

Specific classes of GABAergic neurons play specific roles in regulating information processing in the brain. In the hippocampus, two major classes, parvalbumin-expressing (PV+) and somatostatin-expressing (SST+), differentially regulate endogenous firing patterns and target subcellular compartments of principal cells. How these classes regulate the flow of information throughout the hippocampus is poorly understood. We hypothesize that PV+ and SST+ interneurons in the dentate gyrus (DG) and CA3 differentially modulate CA3 patterns of output, thereby altering the influence of CA3 on CA1. We find that while suppressing either interneuron class increases DG and CA3 output, the effects on CA1 were very different. Suppressing PV+ interneurons increases local field potential signatures of coupling from CA3 to CA1 and decreases signatures of coupling from entorhinal cortex to CA1; suppressing SST+ interneurons has the opposite effect. Thus, DG and CA3 PV+ and SST+ interneurons bidirectionally modulate the flow of information through the hippocampal circuit.


Asunto(s)
Región CA1 Hipocampal/fisiología , Región CA3 Hipocampal/fisiología , Giro Dentado/fisiología , Corteza Entorrinal/fisiología , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Somatostatina/metabolismo , Potenciales de Acción , Animales , Región CA1 Hipocampal/citología , Región CA3 Hipocampal/citología , Giro Dentado/citología , Corteza Entorrinal/citología , Femenino , Neuronas GABAérgicas/citología , Interneuronas/citología , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Nat Neurosci ; 24(6): 786-798, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33958804

RESUMEN

Selective neurodegeneration is a critical causal factor in Alzheimer's disease (AD); however, the mechanisms that lead some neurons to perish, whereas others remain resilient, are unknown. We sought potential drivers of this selective vulnerability using single-nucleus RNA sequencing and discovered that ApoE expression level is a substantial driver of neuronal variability. Strikingly, neuronal expression of ApoE-which has a robust genetic linkage to AD-correlated strongly, on a cell-by-cell basis, with immune response pathways in neurons in the brains of wild-type mice, human ApoE knock-in mice and humans with or without AD. Elimination or over-expression of neuronal ApoE revealed a causal relationship among ApoE expression, neuronal MHC-I expression, tau pathology and neurodegeneration. Functional reduction of MHC-I ameliorated tau pathology in ApoE4-expressing primary neurons and in mouse hippocampi expressing pathological tau. These findings suggest a mechanism linking neuronal ApoE expression to MHC-I expression and, subsequently, to tau pathology and selective neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Apolipoproteínas E/biosíntesis , Antígenos de Histocompatibilidad Clase I/biosíntesis , Neuronas/metabolismo , Regulación hacia Arriba/fisiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Apolipoproteínas E/genética , Células Cultivadas , Bases de Datos Genéticas/tendencias , Femenino , Expresión Génica , Técnicas de Sustitución del Gen/métodos , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/patología
9.
Cell Rep ; 32(4): 107962, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32726626

RESUMEN

Despite its clear impact on Alzheimer's disease (AD) risk, apolipoprotein (apo) E4's contributions to AD etiology remain poorly understood. Progress in answering this and other questions in AD research has been limited by an inability to model human-specific phenotypes in an in vivo environment. Here we transplant human induced pluripotent stem cell (hiPSC)-derived neurons carrying normal apoE3 or pathogenic apoE4 into human apoE3 or apoE4 knockin mouse hippocampi, enabling us to disentangle the effects of apoE4 produced in human neurons and in the brain environment. Using single-nucleus RNA sequencing (snRNA-seq), we identify key transcriptional changes specific to human neuron subtypes in response to endogenous or exogenous apoE4. We also find that Aß from transplanted human neurons forms plaque-like aggregates, with differences in localization and interaction with microglia depending on the transplant and host apoE genotype. These findings highlight the power of in vivo chimeric disease modeling for studying AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Apolipoproteína E4/metabolismo , Neuronas/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacología , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Quimera/genética , Quimera/metabolismo , Técnicas de Sustitución del Gen , Hipocampo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Modelos Biológicos , Proteínas tau/metabolismo
10.
Cell Stem Cell ; 26(3): 297-299, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32142656

RESUMEN

Understanding why adult hippocampal neurogenesis (AHN) is impaired in Alzheimer's disease (AD) is essential for unravelling its role in pathogenesis. In this issue of Cell Stem Cell, Zheng et al. (2020) report that human tau accumulation in dentate gyrus GABAergic interneurons disrupts AHN and strengthening GABAergic signaling restores AHN and improves cognition in an AD mouse model.


Asunto(s)
Enfermedad de Alzheimer , Adulto , Animales , Cognición , Modelos Animales de Enfermedad , Hipocampo , Humanos , Interneuronas , Ratones , Neurogénesis
11.
Stem Cell Reports ; 10(5): 1481-1491, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29628395

RESUMEN

Huntington disease (HD) is an inherited, progressive neurological disorder characterized by degenerating striatal medium spiny neurons (MSNs). One promising approach for treating HD is cell replacement therapy, where lost cells are replaced by MSN progenitors derived from human pluripotent stem cells (hPSCs). While there has been remarkable progress in generating hPSC-derived MSNs, current production methods rely on two-dimensional culture systems that can include poorly defined components, limit scalability, and yield differing preclinical results. To facilitate clinical translation, here, we generated striatal progenitors from hPSCs within a fully defined and scalable PNIPAAm-PEG three-dimensional (3D) hydrogel. Transplantation of 3D-derived striatal progenitors into a transgenic mouse model of HD slowed disease progression, improved motor coordination, and increased survival. In addition, the transplanted cells developed an MSN-like phenotype and formed synaptic connections with host cells. Our results illustrate the potential of scalable 3D biomaterials for generating striatal progenitors for HD cell therapy.


Asunto(s)
Cuerpo Estriado/patología , Enfermedad de Huntington/patología , Enfermedad de Huntington/terapia , Hidrogeles/farmacología , Células Madre Pluripotentes/trasplante , Potenciales de Acción/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
12.
Stem Cell Reports ; 8(6): 1770-1783, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28552605

RESUMEN

Oligodendrocyte precursor cells (OPCs) offer considerable potential for the treatment of demyelinating diseases and injuries of the CNS. However, generating large quantities of high-quality OPCs remains a substantial challenge that impedes their therapeutic application. Here, we show that OPCs can be generated from human pluripotent stem cells (hPSCs) in a three-dimensional (3D), scalable, and fully defined thermoresponsive biomaterial system. We used CRISPR/Cas9 to create a NKX2.2-EGFP human embryonic stem cell reporter line that enabled fine-tuning of early OPC specification and identification of conditions that markedly increased the number of OLIG2+ and NKX2.2+ cells generated from hPSCs. Transplantation of 50-day-old OPCs into the brains of NOD/SCID mice revealed that progenitors generated in 3D without cell selection or purification subsequently engrafted, migrated, and matured into myelinating oligodendrocytes in vivo. These results demonstrate the potential of harnessing lineage reporter lines to develop 3D platforms for rapid and large-scale production of OPCs.


Asunto(s)
Diferenciación Celular , Células Precursoras de Oligodendrocitos/citología , Células Madre Pluripotentes/citología , Animales , Materiales Biocompatibles/química , Encéfalo/metabolismo , Sistemas CRISPR-Cas/genética , Técnicas de Cultivo de Célula , Línea Celular , Reprogramación Celular , Genes Reporteros , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Nucleares , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/trasplante , Factor de Transcripción 2 de los Oligodendrocitos/genética , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Células Madre Pluripotentes/metabolismo , Andamios del Tejido/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Trasplante Heterólogo , Proteínas de Pez Cebra
13.
Biomaterials ; 136: 1-11, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28505596

RESUMEN

Cell replacement therapies have broad biomedical potential; however, low cell survival and poor functional integration post-transplantation are major hurdles that hamper clinical benefit. For example, following striatal transplantation of midbrain dopaminergic (mDA) neurons for the treatment of Parkinson's disease (PD), only 1-5% of the neurons typically survive in preclinical models and in clinical trials. In general, resource-intensive generation and implantation of larger numbers of cells are used to compensate for the low post-transplantation cell-survival. Poor graft survival is often attributed to adverse biochemical, mechanical, and/or immunological stress that cells experience during and after implantation. To address these challenges, we developed a functionalized hyaluronic acid (HA)-based hydrogel for in vitro maturation and central nervous system (CNS) transplantation of human pluripotent stem cell (hPSC)-derived neural progenitors. Specifically, we functionalized the HA hydrogel with RGD and heparin (hep) via click-chemistry and tailored its stiffness to encourage neuronal maturation, survival, and long-term maintenance of the desired mDA phenotype. Importantly, ∼5 times more hydrogel-encapsulated mDA neurons survived after transplantation in the rat striatum, compared to unencapsulated neurons harvested from commonly used 2D surfaces. This engineered biomaterial may therefore increase the therapeutic potential and reduce the manufacturing burden for successful neuronal implantation.


Asunto(s)
Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/trasplante , Células Madre Embrionarias/citología , Ácido Hialurónico/química , Hidrogeles/química , Andamios del Tejido/química , Animales , Línea Celular , Supervivencia Celular , Células Cultivadas , Femenino , Heparina/química , Humanos , Mesencéfalo/citología , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Neurogénesis , Oligopéptidos/química , Ratas Endogámicas F344
14.
Sci Rep ; 7: 40573, 2017 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-28091566

RESUMEN

Pluripotent stem cells (PSCs) have major potential as an unlimited source of functional cells for many biomedical applications; however, the development of cell manufacturing systems to enable this promise faces many challenges. For example, there have been major recent advances in the generation of midbrain dopaminergic (mDA) neurons from stem cells for Parkinson's Disease (PD) therapy; however, production of these cells typically involves undefined components and difficult to scale 2D culture formats. Here, we used a fully defined, 3D, thermoresponsive biomaterial platform to rapidly generate large numbers of action-potential firing mDA neurons after 25 days of differentiation (~40% tyrosine hydroxylase (TH) positive, maturing into 25% cells exhibiting mDA neuron-like spiking behavior). Importantly, mDA neurons generated in 3D exhibited a 30-fold increase in viability upon implantation into rat striatum compared to neurons generated on 2D, consistent with the elevated expression of survival markers FOXA2 and EN1 in 3D. A defined, scalable, and resource-efficient cell culture platform can thus rapidly generate high quality differentiated cells, both neurons and potentially other cell types, with strong potential to accelerate both basic and translational research.


Asunto(s)
Materiales Biocompatibles/farmacología , Técnicas de Cultivo de Célula/métodos , Neuronas Dopaminérgicas/citología , Células Madre Pluripotentes Inducidas/citología , Mesencéfalo/citología , Resinas Acrílicas/química , Animales , Biomarcadores/metabolismo , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Fenómenos Electrofisiológicos , Femenino , Células Madre Embrionarias Humanas/citología , Humanos , Implantes Experimentales , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Polietilenglicoles/química , Ratas Endogámicas F344 , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
15.
J Neurosci ; 35(9): 3946-58, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25740523

RESUMEN

Layer (L)2 is a major output of primary sensory cortex that exhibits very sparse spiking, but the structure of sensory representation in L2 is not well understood. We combined two-photon calcium imaging with deflection of many whiskers to map whisker receptive fields, characterize sparse coding, and quantitatively define the point representation in L2 of mouse somatosensory cortex. Neurons within a column-sized imaging field showed surprisingly heterogeneous, salt-and-pepper tuning to many different whiskers. Single whisker deflection elicited low-probability spikes in highly distributed, shifting neural ensembles spanning multiple cortical columns. Whisker-evoked response probability correlated strongly with spontaneous firing rate, but weakly with tuning properties, indicating a spectrum of inherent responsiveness across pyramidal cells. L2 neurons projecting to motor and secondary somatosensory cortex differed in whisker tuning and responsiveness, and carried different amounts of information about columnar whisker deflection. From these data, we derive a quantitative, fine-scale picture of the distributed point representation in L2.


Asunto(s)
Vías Nerviosas/anatomía & histología , Vías Nerviosas/fisiología , Corteza Somatosensorial/anatomía & histología , Corteza Somatosensorial/fisiología , Vibrisas/inervación , Animales , Mapeo Encefálico , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estimulación Física
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA