Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
2.
Nature ; 573(7772): 75-82, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31316211

RESUMEN

Multiple sclerosis (MS) is a neuroinflammatory disease with a relapsing-remitting disease course at early stages, distinct lesion characteristics in cortical grey versus subcortical white matter and neurodegeneration at chronic stages. Here we used single-nucleus RNA sequencing to assess changes in expression in multiple cell lineages in MS lesions and validated the results using multiplex in situ hybridization. We found selective vulnerability and loss of excitatory CUX2-expressing projection neurons in upper-cortical layers underlying meningeal inflammation; such MS neuron populations exhibited upregulation of stress pathway genes and long non-coding RNAs. Signatures of stressed oligodendrocytes, reactive astrocytes and activated microglia mapped most strongly to the rim of MS plaques. Notably, single-nucleus RNA sequencing identified phagocytosing microglia and/or macrophages by their ingestion and perinuclear import of myelin transcripts, confirmed by functional mouse and human culture assays. Our findings indicate lineage- and region-specific transcriptomic changes associated with selective cortical neuron damage and glial activation contributing to progression of MS lesions.


Asunto(s)
Linaje de la Célula , Esclerosis Múltiple/patología , Neuronas/patología , Adulto , Animales , Astrocitos/metabolismo , Astrocitos/patología , Autopsia , Criopreservación , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Esclerosis Múltiple/genética , Vaina de Mielina/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/patología , Fagocitosis , ARN Nuclear Pequeño/análisis , ARN Nuclear Pequeño/genética , RNA-Seq , Transcriptoma/genética
3.
Nature ; 573(7772): 130-134, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31413369

RESUMEN

Ageing causes a decline in tissue regeneration owing to a loss of function of adult stem cell and progenitor cell populations1. One example is the deterioration of the regenerative capacity of the widespread and abundant population of central nervous system (CNS) multipotent stem cells known as oligodendrocyte progenitor cells (OPCs)2. A relatively overlooked potential source of this loss of function is the stem cell 'niche'-a set of cell-extrinsic cues that include chemical and mechanical signals3,4. Here we show that the OPC microenvironment stiffens with age, and that this mechanical change is sufficient to cause age-related loss of function of OPCs. Using biological and synthetic scaffolds to mimic the stiffness of young brains, we find that isolated aged OPCs cultured on these scaffolds are molecularly and functionally rejuvenated. When we disrupt mechanical signalling, the proliferation and differentiation rates of OPCs are increased. We identify the mechanoresponsive ion channel PIEZO1 as a key mediator of OPC mechanical signalling. Inhibiting PIEZO1 overrides mechanical signals in vivo and allows OPCs to maintain activity in the ageing CNS. We also show that PIEZO1 is important in regulating cell number during CNS development. Thus we show that tissue stiffness is a crucial regulator of ageing in OPCs, and provide insights into how the function of adult stem and progenitor cells changes with age. Our findings could be important not only for the development of regenerative therapies, but also for understanding the ageing process itself.


Asunto(s)
Células Madre Adultas/patología , Envejecimiento/patología , Sistema Nervioso Central/patología , Células Madre Multipotentes/patología , Nicho de Células Madre , Animales , Animales Recién Nacidos , Recuento de Células , Matriz Extracelular/patología , Femenino , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Oligodendroglía/patología , Ratas , Nicho de Células Madre/fisiología
4.
Nature ; 563(7731): 347-353, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30429548

RESUMEN

During early human pregnancy the uterine mucosa transforms into the decidua, into which the fetal placenta implants and where placental trophoblast cells intermingle and communicate with maternal cells. Trophoblast-decidual interactions underlie common diseases of pregnancy, including pre-eclampsia and stillbirth. Here we profile the transcriptomes of about 70,000 single cells from first-trimester placentas with matched maternal blood and decidual cells. The cellular composition of human decidua reveals subsets of perivascular and stromal cells that are located in distinct decidual layers. There are three major subsets of decidual natural killer cells that have distinctive immunomodulatory and chemokine profiles. We develop a repository of ligand-receptor complexes and a statistical tool to predict the cell-type specificity of cell-cell communication via these molecular interactions. Our data identify many regulatory interactions that prevent harmful innate or adaptive immune responses in this environment. Our single-cell atlas of the maternal-fetal interface reveals the cellular organization of the decidua and placenta, and the interactions that are critical for placentation and reproductive success.


Asunto(s)
Comunicación Celular , Feto/citología , Histocompatibilidad Materno-Fetal/inmunología , Placenta/citología , Placenta/metabolismo , Embarazo/inmunología , Análisis de la Célula Individual , Comunicación Celular/inmunología , Diferenciación Celular/genética , Decidua/citología , Decidua/inmunología , Decidua/metabolismo , Femenino , Feto/inmunología , Feto/metabolismo , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Ligandos , Placenta/inmunología , ARN Citoplasmático Pequeño/genética , Análisis de Secuencia de ARN , Células del Estroma/citología , Células del Estroma/metabolismo , Transcriptoma , Trofoblastos/citología , Trofoblastos/inmunología , Trofoblastos/metabolismo
5.
J Neurosci ; 37(49): 11835-11853, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29089438

RESUMEN

Many lines of evidence suggest that the Parkinson's disease (PD)-related protein α-synuclein (α-SYN) can propagate from cell to cell in a prion-like manner. However, the cellular mechanisms behind the spreading remain elusive. Here, we show that human astrocytes derived from embryonic stem cells actively transfer aggregated α-SYN to nearby astrocytes via direct contact and tunneling nanotubes (TNTs). Failure in the astrocytes' lysosomal digestion of excess α-SYN oligomers results in α-SYN deposits in the trans-Golgi network followed by endoplasmic reticulum swelling and mitochondrial disturbances. The stressed astrocytes respond by conspicuously sending out TNTs, enabling intercellular transfer of α-SYN to healthy astrocytes, which in return deliver mitochondria, indicating a TNT-mediated rescue mechanism. Using a pharmacological approach to inhibit TNT formation, we abolished the transfer of both α-SYN and mitochondria. Together, our results highlight the role of astrocytes in α-SYN cell-to-cell transfer, identifying possible pathophysiological events in the PD brain that could be of therapeutic relevance.SIGNIFICANCE STATEMENT Astrocytes are the major cell type in the brain, yet their role in Parkinson's disease progression remains elusive. Here, we show that human astrocytes actively transfer aggregated α-synuclein (α-SYN) to healthy astrocytes via direct contact and tunneling nanotubes (TNTs), rather than degrade it. The astrocytes engulf large amounts of oligomeric α-SYN that are subsequently stored in the trans-Golgi network region. The accumulation of α-SYN in the astrocytes affects their lysosomal machinery and induces mitochondrial damage. The stressed astrocytes respond by sending out TNTs, enabling intercellular transfer of α-SYN to healthy astrocytes. Our findings highlight an unexpected role of astrocytes in the propagation of α-SYN pathology via TNTs, revealing astrocytes as a potential target for therapeutic intervention.


Asunto(s)
Astrocitos/química , Astrocitos/metabolismo , Nanotubos , alfa-Sinucleína/análisis , alfa-Sinucleína/metabolismo , Astrocitos/ultraestructura , Comunicación Celular/fisiología , Células Cultivadas , Células Madre Embrionarias/química , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/ultraestructura , Humanos , alfa-Sinucleína/ultraestructura
6.
Acta Neuropathol ; 128(6): 805-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25296989

RESUMEN

The cellular hallmarks of Parkinson's disease (PD) are the loss of nigral dopaminergic neurons and the formation of α-synuclein-enriched Lewy bodies and Lewy neurites in the remaining neurons. Based on the topographic distribution of Lewy bodies established after autopsy of brains from PD patients, Braak and coworkers hypothesized that Lewy pathology primes in the enteric nervous system and spreads to the brain, suggesting an active retrograde transport of α-synuclein (the key protein component in Lewy bodies), via the vagal nerve. This hypothesis, however, has not been tested experimentally thus far. Here, we use a human PD brain lysate containing different forms of α-synuclein (monomeric, oligomeric and fibrillar), and recombinant α-synuclein in an in vivo animal model to test this hypothesis. We demonstrate that α-synuclein present in the human PD brain lysate and distinct recombinant α-synuclein forms are transported via the vagal nerve and reach the dorsal motor nucleus of the vagus in the brainstem in a time-dependent manner after injection into the intestinal wall. Using live cell imaging in a differentiated neuroblastoma cell line, we determine that both slow and fast components of axonal transport are involved in the transport of aggregated α-synuclein. In conclusion, we here provide the first experimental evidence that different α-synuclein forms can propagate from the gut to the brain, and that microtubule-associated transport is involved in the translocation of aggregated α-synuclein in neurons.


Asunto(s)
Encéfalo/fisiopatología , Tracto Gastrointestinal/fisiopatología , Enfermedad de Parkinson/fisiopatología , alfa-Sinucleína/metabolismo , Animales , Transporte Axonal , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Tracto Gastrointestinal/patología , Humanos , Enfermedad de Parkinson/patología , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Nervio Vago/patología , Nervio Vago/fisiopatología
7.
Cell Rep ; 36(2): 109362, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260928

RESUMEN

The mediobasal hypothalamus (MBH; arcuate nucleus of the hypothalamus [ARH] and median eminence [ME]) is a key nutrient sensing site for the production of the complex homeostatic feedback responses required for the maintenance of energy balance. Here, we show that refeeding after an overnight fast rapidly triggers proliferation and differentiation of oligodendrocyte progenitors, leading to the production of new oligodendrocytes in the ME specifically. During this nutritional paradigm, ME perineuronal nets (PNNs), emerging regulators of ARH metabolic functions, are rapidly remodeled, and this process requires myelin regulatory factor (Myrf) in oligodendrocyte progenitors. In genetically obese ob/ob mice, nutritional regulations of ME oligodendrocyte differentiation and PNN remodeling are blunted, and enzymatic digestion of local PNN increases food intake and weight gain. We conclude that MBH PNNs are required for the maintenance of energy balance in lean mice and are remodeled in the adult ME by the nutritional control of oligodendrocyte differentiation.


Asunto(s)
Diferenciación Celular , Eminencia Media/citología , Red Nerviosa/fisiología , Fenómenos Fisiológicos de la Nutrición , Oligodendroglía/citología , Adulto , Animales , Linaje de la Célula , Proliferación Celular , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Oligodendroglía/ultraestructura , Análisis de la Célula Individual , Transcriptoma/genética
8.
Neuron ; 105(5): 855-866.e5, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31924446

RESUMEN

Recent interest in astrocyte activation states has raised the fundamental question of how these cells, normally essential for synapse and neuronal maintenance, become pathogenic. Here, we show that activation of the unfolded protein response (UPR), specifically phosphorylated protein kinase R-like endoplasmic reticulum (ER) kinase (PERK-P) signaling-a pathway that is widely dysregulated in neurodegenerative diseases-generates a distinct reactivity state in astrocytes that alters the astrocytic secretome, leading to loss of synaptogenic function in vitro. Further, we establish that the same PERK-P-dependent astrocyte reactivity state is harmful to neurons in vivo in mice with prion neurodegeneration. Critically, targeting this signaling exclusively in astrocytes during prion disease is alone sufficient to prevent neuronal loss and significantly prolongs survival. Thus, the astrocyte reactivity state resulting from UPR over-activation is a distinct pathogenic mechanism that can by itself be effectively targeted for neuroprotection.


Asunto(s)
Astrocitos/metabolismo , Factor 2B Eucariótico de Iniciación/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades por Prión/metabolismo , Sinapsis/metabolismo , Respuesta de Proteína Desplegada/fisiología , eIF-2 Quinasa/metabolismo , Animales , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnicas In Vitro , Memoria , Ratones , Fosforilación , Biosíntesis de Proteínas , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Transducción de Señal , Tapsigargina/farmacología , Transcriptoma , Tunicamicina/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
9.
Nat Neurosci ; 23(4): 500-509, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32203496

RESUMEN

Although the cerebral cortex is organized into six excitatory neuronal layers, it is unclear whether glial cells show distinct layering. In the present study, we developed a high-content pipeline, the large-area spatial transcriptomic (LaST) map, which can quantify single-cell gene expression in situ. Screening 46 candidate genes for astrocyte diversity across the mouse cortex, we identified superficial, mid and deep astrocyte identities in gradient layer patterns that were distinct from those of neurons. Astrocyte layer features, established in the early postnatal cortex, mostly persisted in adult mouse and human cortex. Single-cell RNA sequencing and spatial reconstruction analysis further confirmed the presence of astrocyte layers in the adult cortex. Satb2 and Reeler mutations that shifted neuronal post-mitotic development were sufficient to alter glial layering, indicating an instructive role for neuronal cues. Finally, astrocyte layer patterns diverged between mouse cortical regions. These findings indicate that excitatory neurons and astrocytes are organized into distinct lineage-associated laminae.


Asunto(s)
Astrocitos/citología , Corteza Cerebral/citología , Neuronas/citología , Transcriptoma , Animales , Astrocitos/metabolismo , Mapeo Encefálico , Corteza Cerebral/metabolismo , Humanos , Ratones , Neuronas/metabolismo
10.
Integr Biol (Camb) ; 8(3): 332-40, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26915333

RESUMEN

Concentration of viable cell populations in suspension is of interest for several clinical and pre-clinical applications. Here, we report that microfluidic acoustophoresis is an effective method to efficiently concentrate live and viable cells with high target purity without any need for protein fluorescent labeling using antibodies or over-expression. We explored the effect of the acoustic field acoustic energy density and systematically used different protocols to induce apoptosis or cell death and then determined the efficiency of live and dead cell separation. We used the breast cancer cell line MCF-7, the mouse neuroblastoma N2a as well as human embryonic stem cells (hESCs) to demonstrate that this method is gentle and can be applied to different cell populations. First, we induced cell death by means of high osmotic shock using a high concentration of PBS (10×), the protein kinase inhibitor staurosporine, high concentrations of dimethyl sulfoxide (DMSO, 10%), and finally, cell starvation. In all the methods employed, we successfully induced cell death and were able to purify and concentrate the remaining live cells using acoustophoresis. Importantly, the concentration of viable cells was not dependent on a specific cell type. Further, we demonstrate that different death inducing stimuli have different effects on the intrinsic cell properties and therefore affect the efficiency of the acoustophoretic separation.


Asunto(s)
Acústica/instrumentación , Separación Celular/instrumentación , Supervivencia Celular , Dispositivos Laboratorio en un Chip , Animales , Apoptosis , Línea Celular , Línea Celular Tumoral , Separación Celular/métodos , Femenino , Células Madre Embrionarias Humanas/citología , Humanos , Células MCF-7 , Ratones , Neuronas/citología , Presión Osmótica
11.
NPJ Parkinsons Dis ; 2: 16009, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28725696

RESUMEN

Induced pluripotent stem cells (iPSCs) are becoming an important source of pre-clinical models for research focusing on neurodegeneration. They offer the possibility for better understanding of common and divergent pathogenic mechanisms of brain diseases. Moreover, iPSCs provide a unique opportunity to develop personalized therapeutic strategies, as well as explore early pathogenic mechanisms, since they rely on the use of patients' own cells that are otherwise accessible only post-mortem, when neuronal death-related cellular pathways and processes are advanced and adaptive. Neurodegenerative diseases are in majority of unknown cause, but mutations in specific genes can lead to familial forms of these diseases. For example, mutations in the superoxide dismutase 1 gene lead to the motor neuron disease amyotrophic lateral sclerosis (ALS), while mutations in the SNCA gene encoding for alpha-synuclein protein lead to familial Parkinson's disease (PD). The generations of libraries of familial human ALS iPSC lines have been described, and the iPSCs rapidly became useful models for studying cell autonomous and non-cell autonomous mechanisms of the disease. Here we report the generation of a comprehensive library of iPSC lines of familial PD and an associated synucleinopathy, multiple system atrophy (MSA). In addition, we provide examples of relevant neural cell types these iPSC can be differentiated into, and which could be used to further explore early disease mechanisms. These human cellular models will be a valuable resource for identifying common and divergent mechanisms leading to neurodegeneration in PD and MSA.

12.
Sci Rep ; 6: 33289, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27628239

RESUMEN

α-Synuclein is a presynaptic protein associated to Parkinson's disease, which is unstructured when free in the cytoplasm and adopts α helical conformation when bound to vesicles. After decades of intense studies, α-Synuclein physiology is still difficult to clear up due to its interaction with multiple partners and its involvement in a pletora of neuronal functions. Here, we looked at the remarkably neglected interplay between α-Synuclein and microtubules, which potentially impacts on synaptic functionality. In order to identify the mechanisms underlying these actions, we investigated the interaction between purified α-Synuclein and tubulin. We demonstrated that α-Synuclein binds to microtubules and tubulin α2ß2 tetramer; the latter interaction inducing the formation of helical segment(s) in the α-Synuclein polypeptide. This structural change seems to enable α-Synuclein to promote microtubule nucleation and to enhance microtubule growth rate and catastrophe frequency, both in vitro and in cell. We also showed that Parkinson's disease-linked α-Synuclein variants do not undergo tubulin-induced folding and cause tubulin aggregation rather than polymerization. Our data enable us to propose α-Synuclein as a novel, foldable, microtubule-dynamase, which influences microtubule organisation through its binding to tubulin and its regulating effects on microtubule nucleation and dynamics.


Asunto(s)
Enfermedad de Parkinson/genética , Agregación Patológica de Proteínas/genética , Tubulina (Proteína)/metabolismo , alfa-Sinucleína/metabolismo , Humanos , Microtúbulos/química , Microtúbulos/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Unión Proteica , Pliegue de Proteína , Multimerización de Proteína/genética , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , alfa-Sinucleína/química , alfa-Sinucleína/genética
13.
Stem Cell Reports ; 5(2): 174-84, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26235891

RESUMEN

In this study, we sought evidence for alpha-synuclein (ASYN) expression in oligodendrocytes, as a possible endogenous source of ASYN to explain its presence in glial inclusions found in multiple system atrophy (MSA) and Parkinson's disease (PD). We identified ASYN in oligodendrocyte lineage progenitors isolated from the rodent brain, in oligodendrocytes generated from embryonic stem cells, and in induced pluripotent stem cells produced from fibroblasts of a healthy individual and patients diagnosed with MSA or PD, in cultures in vitro. Notably, we observed a significant decrease in ΑSYN during oligodendrocyte maturation. Additionally, we show the presence of transcripts in PDGFRΑ/CD140a(+) cells and SOX10(+) oligodendrocyte lineage nuclei isolated by FACS from rodent and human healthy and diseased brains, respectively. Our work identifies ASYN in oligodendrocyte lineage cells, and it offers additional in vitro cellular models that should provide significant insights of the functional implication of ASYN during oligodendrocyte development and disease.


Asunto(s)
Linaje de la Célula , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , alfa-Sinucleína/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Células-Madre Neurales/citología , Oligodendroglía/citología , Especificidad de la Especie , alfa-Sinucleína/genética
14.
Stem Cell Res ; 15(1): 203-20, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26100233

RESUMEN

Astrocytes play a critical role during the development and the maintenance of the CNS in health and disease. Yet, their lack of accessibility from fetuses and from the brain of diseased patients has hindered our understanding of their full implication in developmental and pathogenic processes. Human pluripotent stem cells (PSCs) are an alternative source to obtain large quantities of astrocytes in vitro, for mechanistic studies of development and disease. However, these studies often require highly pure populations of astrocytes, which are not always achieved, depending on the PSC lines and protocols used. Here, we describe the generation and characterization of human PSC reporter lines expressing TagRFP driven by the ABC1D region of the human GFAP promoter, as new cellular model for generating homogenous population of astrocytes generated from CNS regionally defined PSC-derived neural progenitors. GFA(ABC1D)::TagRFP-expressing astrocytes can be purified by fluorescent-activated cell sorting and maintain a bright expression for several additional weeks. These express canonical astrocyte markers NF1A, S100ß, CX43, GLAST, GS and CD44. These new cellular models, from which highly pure populations of fluorescence-expressing astrocytes can be obtained, provide a new platform for studies where pure or fluorescently labeled astrocyte populations are necessary, for example to assess pro-inflammatory cytokine and chemokine release in response to specific treatment, and uptake and degradation of fluorescently labeled pathogenic proteins, as reported in this study.


Asunto(s)
Astrocitos/citología , Separación Celular/métodos , Genes Reporteros , Mesencéfalo/citología , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Médula Espinal/citología , Astrocitos/metabolismo , Línea Celular , Células Clonales , Citometría de Flujo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inflamación/patología , Factores de Tiempo , Transgenes
15.
PLoS One ; 9(7): e100869, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24999658

RESUMEN

Synucleinopathies, characterized by intracellular aggregation of α-synuclein protein, share a number of features in pathology and disease progression. However, the vulnerable cell population differs significantly between the disorders, despite being caused by the same protein. While the vulnerability of dopamine cells in the substantia nigra to α-synuclein over-expression, and its link to Parkinson's disease, is well studied, animal models recapitulating the cortical degeneration in dementia with Lewy-bodies (DLB) are much less mature. The aim of this study was to develop a first rat model of widespread progressive synucleinopathy throughout the forebrain using adeno-associated viral (AAV) vector mediated gene delivery. Through bilateral injection of an AAV6 vector expressing human wild-type α-synuclein into the forebrain of neonatal rats, we were able to achieve widespread, robust α-synuclein expression with preferential expression in the frontal cortex. These animals displayed a progressive emergence of hyper-locomotion and dysregulated response to the dopaminergic agonist apomorphine. The animals receiving the α-synuclein vector displayed significant α-synuclein pathology including intra-cellular inclusion bodies, axonal pathology and elevated levels of phosphorylated α-synuclein, accompanied by significant loss of cortical neurons and a progressive reduction in both cortical and striatal ChAT positive interneurons. Furthermore, we found evidence of α-synuclein sequestered by IBA-1 positive microglia, which was coupled with a distinct change in morphology. In areas of most prominent pathology, the total α-synuclein levels were increased to, on average, two-fold, which is similar to the levels observed in patients with SNCA gene triplication, associated with cortical Lewy body pathology. This study provides a novel rat model of progressive cortical synucleinopathy, showing for the first time that cholinergic interneurons are vulnerable to α-synuclein over-expression. This animal model provides a powerful new tool for studies of neuronal degeneration in conditions of widespread cortical α-synuclein pathology, such as DLB, as well an attractive model for the exploration of novel biomarkers.


Asunto(s)
Neuronas Colinérgicas/patología , Dependovirus/genética , Progresión de la Enfermedad , Interneuronas/patología , Enfermedad por Cuerpos de Lewy/patología , Prosencéfalo/metabolismo , alfa-Sinucleína/genética , Animales , Animales Recién Nacidos , Axones/efectos de los fármacos , Axones/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Neuronas Colinérgicas/efectos de los fármacos , Modelos Animales de Enfermedad , Agonistas de Dopamina/farmacología , Conducta Exploratoria/efectos de los fármacos , Femenino , Vectores Genéticos/genética , Humanos , Interneuronas/efectos de los fármacos , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/fisiopatología , Microglía/efectos de los fármacos , Microglía/patología , Actividad Motora/efectos de los fármacos , Neostriado/efectos de los fármacos , Neostriado/patología , Fenotipo , Embarazo , Prosencéfalo/patología , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , alfa-Sinucleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA