Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther Methods Clin Dev ; 29: 381-394, 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37251982

RESUMEN

Cell therapy for Parkinson's disease has experienced substantial growth in the past decades with several ongoing clinical trials. Despite increasing refinement of differentiation protocols and standardization of the transplanted neural precursors, the transcriptomic analysis of cells in the transplant after its full maturation in vivo has not been thoroughly investigated. Here, we present spatial transcriptomics analysis of fully differentiated grafts in their host tissue. Unlike earlier transcriptomics analyses using single-cell technologies, we observe that cells derived from human embryonic stem cells (hESCs) in the grafts adopt mature dopaminergic signatures. We show that the presence of phenotypic dopaminergic genes, which were found to be differentially expressed in the transplants, is concentrated toward the edges of the grafts, in agreement with the immunohistochemical analyses. Deconvolution shows dopamine neurons being the dominating cell type in many features beneath the graft area. These findings further support the preferred environmental niche of TH-positive cells and confirm their dopaminergic phenotype through the presence of multiple dopaminergic markers.

2.
Stem Cell Reports ; 17(1): 159-172, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34971563

RESUMEN

Transplantation in Parkinson's disease using human embryonic stem cell (hESC)-derived dopaminergic (DA) neurons is a promising future treatment option. However, many of the mechanisms that govern their differentiation, maturation, and integration into the host circuitry remain elusive. Here, we engrafted hESCs differentiated toward a ventral midbrain DA phenotype into the midbrain of a preclinical rodent model of Parkinson's disease. We then injected a novel DA-neurotropic retrograde MNM008 adeno-associated virus vector capsid, into specific DA target regions to generate starter cells based on their axonal projections. Using monosynaptic rabies-based tracing, we demonstrated for the first time that grafted hESC-derived DA neurons receive distinctly different afferent inputs depending on their projections. The similarities to the host DA system suggest a previously unknown directed circuit integration. By evaluating the differential host-to-graft connectivity based on projection patterns, this novel approach offers a tool to answer outstanding questions regarding the integration of grafted hESC-derived DA neurons.


Asunto(s)
Diferenciación Celular , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/metabolismo , Biomarcadores , Rastreo Celular , Expresión Génica , Genes Reporteros , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Mesencéfalo/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Trasplante de Células Madre
4.
Nat Biotechnol ; 38(11): 1265-1273, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32451506

RESUMEN

The study of brain development in humans is limited by the lack of tissue samples and suitable in vitro models. Here, we model early human neural tube development using human embryonic stem cells cultured in a microfluidic device. The approach, named microfluidic-controlled stem cell regionalization (MiSTR), exposes pluripotent stem cells to signaling gradients that mimic developmental patterning. Using a WNT-activating gradient, we generated a neural tissue exhibiting progressive caudalization from forebrain to midbrain to hindbrain, including formation of isthmic organizer characteristics. Single-cell transcriptomics revealed that rostro-caudal organization was already established at 24 h of differentiation, and that the first markers of a neural-specific transcription program emerged in the rostral cells at 48 h. The transcriptomic hallmarks of rostro-caudal organization recapitulated gene expression patterns of the early rostro-caudal neural plate in mouse embryos. Thus, MiSTR will facilitate research on the factors and processes underlying rostro-caudal neural tube patterning.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias Humanas/citología , Microfluídica/métodos , Tubo Neural/embriología , Proteínas Wnt/metabolismo , Tipificación del Cuerpo , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Humanos , Análisis de la Célula Individual , Transcriptoma/genética , Vía de Señalización Wnt
5.
Proc Natl Acad Sci U S A ; 116(52): 27053-27062, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31818949

RESUMEN

Adeno-associated virus (AAV) capsid modification enables the generation of recombinant vectors with tailored properties and tropism. Most approaches to date depend on random screening, enrichment, and serendipity. The approach explored here, called BRAVE (barcoded rational AAV vector evolution), enables efficient selection of engineered capsid structures on a large scale using only a single screening round in vivo. The approach stands in contrast to previous methods that require multiple generations of enrichment. With the BRAVE approach, each virus particle displays a peptide, derived from a protein, of known function on the AAV capsid surface, and a unique molecular barcode in the packaged genome. The sequencing of RNA-expressed barcodes from a single-generation in vivo screen allows the mapping of putative binding sequences from hundreds of proteins simultaneously. Using the BRAVE approach and hidden Markov model-based clustering, we present 25 synthetic capsid variants with refined properties, such as retrograde axonal transport in specific subtypes of neurons, as shown for both rodent and human dopaminergic neurons.

6.
Methods Mol Biol ; 1937: 59-87, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30706390

RESUMEN

Chemogenetics is the process of genetically expressing a macromolecule receptor capable of modulating the activity of the cell in response to selective chemical ligand. This chapter will cover the chemogenetic technologies that are available to date, focusing on the commonly available engineered or otherwise modified ligand-gated ion channels and G-protein-coupled receptors in the context of neuromodulation. First, we will give a brief overview of each chemogenetic approach as well as in vitro/in vivo applications, then we will list their strengths and weaknesses. Finally, we will provide tips for ligand application in each case.Each technology has specific limitations that make them more or less suitable for different applications in neuroscience although we will focus mainly on the most commonly used and versatile family named designer receptors exclusively activated by designer drugs or DREADDs. We here describe the most common cases where these can be implemented and provide tips on how and where these technologies can be applied in the field of neuroscience.


Asunto(s)
Encéfalo/fisiología , Drogas de Diseño/administración & dosificación , Ingeniería de Proteínas/métodos , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Drogas de Diseño/farmacología , Humanos , Canales Iónicos Activados por Ligandos/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/fisiología , Receptores Acoplados a Proteínas G/metabolismo
7.
Brain ; 141(7): 2014-2031, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29788236

RESUMEN

Recombinant adeno-associated viruses (AAVs) are popular in vivo gene transfer vehicles. However, vector doses needed to achieve therapeutic effect are high and some target tissues in the central nervous system remain difficult to transduce. Gene therapy trials using AAV for the treatment of neurological disorders have seldom led to demonstrated clinical efficacy. Important contributing factors are low transduction rates and inefficient distribution of the vector. To overcome these hurdles, a variety of capsid engineering methods have been utilized to generate capsids with improved transduction properties. Here we describe an alternative approach to capsid engineering, which draws on the natural evolution of the virus and aims to yield capsids that are better suited to infect human tissues. We generated an AAV capsid to include amino acids that are conserved among natural AAV2 isolates and tested its biodistribution properties in mice and rats. Intriguingly, this novel variant, AAV-TT, demonstrates strong neurotropism in rodents and displays significantly improved distribution throughout the central nervous system as compared to AAV2. Additionally, sub-retinal injections in mice revealed markedly enhanced transduction of photoreceptor cells when compared to AAV2. Importantly, AAV-TT exceeds the distribution abilities of benchmark neurotropic serotypes AAV9 and AAVrh10 in the central nervous system of mice, and is the only virus, when administered at low dose, that is able to correct the neurological phenotype in a mouse model of mucopolysaccharidosis IIIC, a transmembrane enzyme lysosomal storage disease, which requires delivery to every cell for biochemical correction. These data represent unprecedented correction of a lysosomal transmembrane enzyme deficiency in mice and suggest that AAV-TT-based gene therapies may be suitable for treatment of human neurological diseases such as mucopolysaccharidosis IIIC, which is characterized by global neuropathology.


Asunto(s)
Cápside/fisiología , Terapia Genética/métodos , Ingeniería de Proteínas/métodos , Animales , Dependovirus/genética , Femenino , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/terapia , Células Fotorreceptoras/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Retina/fisiología , Distribución Tisular , Transducción Genética
8.
RNA ; 24(5): 673-687, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29386333

RESUMEN

Genome editing has proven to be highly potent in the generation of functional gene knockouts in dividing cells. In the CNS however, efficient technologies to repair sequences are yet to materialize. Reprogramming on the mRNA level is an attractive alternative as it provides means to perform in situ editing of coding sequences without nuclease dependency. Furthermore, de novo sequences can be inserted without the requirement of homologous recombination. Such reprogramming would enable efficient editing in quiescent cells (e.g., neurons) with an attractive safety profile for translational therapies. In this study, we applied a novel molecular-barcoded screening assay to investigate RNA trans-splicing in mammalian neurons. Through three alternative screening systems in cell culture and in vivo, we demonstrate that factors determining trans-splicing are reproducible regardless of the screening system. With this screening, we have located the most permissive trans-splicing sequences targeting an intron in the Synapsin I gene. Using viral vectors, we were able to splice full-length fluorophores into the mRNA while retaining very low off-target expression. Furthermore, this approach also showed evidence of functionality in the mouse striatum. However, in its current form, the trans-splicing events are stochastic and the overall activity lower than would be required for therapies targeting loss-of-function mutations. Nevertheless, the herein described barcode-based screening assay provides a unique possibility to screen and map large libraries in single animals or cell assays with very high precision.


Asunto(s)
Dependovirus/genética , Vectores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Lentivirus/genética , Análisis de Secuencia de ARN/métodos , Trans-Empalme , Animales , Encéfalo/metabolismo , Femenino , Biblioteca de Genes , Células HEK293 , Células HeLa , Humanos , Intrones , Ratones Endogámicos C57BL , Sinapsinas/genética
9.
Neurobiol Dis ; 109(Pt A): 148-162, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29037828

RESUMEN

The intricate balance between dopaminergic and cholinergic neurotransmission in the striatum has been thoroughly difficult to characterize. It was initially described as a seesaw with a competing function of dopamine versus acetylcholine. Recent technical advances however, have brought this view into question suggesting that the two systems work rather in concert with the cholinergic interneurons (ChIs) driving dopamine release. In this study, we have utilized two transgenic Cre-driver rat lines, a choline acetyl transferase ChAT-Cre transgenic rat and a novel double-transgenic tyrosine hydroxylase TH-Cre/ChAT-Cre rat to further elucidate the role of striatal ChIs in normal motor function and in Parkinson's disease. Here we show that selective and reversible activation of ChIs using chemogenetic (DREADD) receptors increases locomotor function in intact rats and potentiate the therapeutic effect of L-DOPA in the rats with lesions of the nigral dopamine system. However, the potentiation of the L-DOPA effect is accompanied by an aggravation of L-DOPA induced dyskinesias (LIDs). These LIDs appear to be driven primarily through the indirect striato-pallidal pathway since the same effect can be induced by the D2 agonist Quinpirole. Taken together, the results highlight the intricate regulation of balance between the two output pathways from the striatum orchestrated by the ChIs.


Asunto(s)
Neuronas Colinérgicas/fisiología , Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/fisiología , Interneuronas/fisiología , Enfermedad de Parkinson/fisiopatología , Animales , Colina O-Acetiltransferasa/genética , Neuronas Colinérgicas/citología , Neuronas Colinérgicas/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Discinesia Inducida por Medicamentos/metabolismo , Discinesia Inducida por Medicamentos/fisiopatología , Femenino , Interneuronas/citología , Interneuronas/metabolismo , Levodopa/administración & dosificación , Locomoción , Masculino , Enfermedad de Parkinson/metabolismo , Ratas Long-Evans , Ratas Sprague-Dawley , Ratas Transgénicas , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D1/fisiología , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/fisiología , Tirosina 3-Monooxigenasa/genética
10.
Neuron ; 90(5): 955-68, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27161524

RESUMEN

Transplantation of DA neurons is actively pursued as a restorative therapy in Parkinson's disease (PD). Pioneering clinical trials using transplants of fetal DA neuroblasts have given promising results, although a number of patients have developed graft-induced dyskinesias (GIDs), and the mechanism underlying this troublesome side effect is still unknown. Here we have used a new model where the activity of the transplanted DA neurons can be selectively modulated using a bimodal chemogenetic (DREADD) approach, allowing either enhancement or reduction of the therapeutic effect. We show that exclusive activation of a cAMP-linked (Gs-coupled) DREADD or serotonin 5-HT6 receptor, located on the grafted DA neurons, is sufficient to induce GIDs. These findings establish a mechanistic link between the 5-HT6 receptor, intracellular cAMP, and GIDs in transplanted PD patients. This effect is thought to be mediated through counteraction of the D2 autoreceptor feedback inhibition, resulting in a dysplastic DA release from the transplant.


Asunto(s)
Neuronas Dopaminérgicas/trasplante , Discinesia Inducida por Medicamentos/fisiopatología , Trasplante de Tejido Fetal/efectos adversos , Trastornos Parkinsonianos/metabolismo , Receptores de Serotonina/fisiología , Animales , Clozapina/análogos & derivados , Clozapina/farmacología , AMP Cíclico/metabolismo , Diterpenos/farmacología , Diterpenos de Tipo Clerodano , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Etilaminas/farmacología , Femenino , Técnicas de Sustitución del Gen , Humanos , Indoles/farmacología , Oxidopamina , Trastornos Parkinsonianos/cirugía , Complicaciones Posoperatorias , Ratas , Receptores de Serotonina/biosíntesis , Receptores de Serotonina/efectos de los fármacos
11.
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
12.
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
...