Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 145(5): 1584-1597, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35262656

RESUMEN

There has been substantial progress in the development of regenerative medicine strategies for CNS disorders over the last decade, with progression to early clinical studies for some conditions. However, there are multiple challenges along the translational pipeline, many of which are common across diseases and pertinent to multiple donor cell types. These include defining the point at which the preclinical data are sufficiently compelling to permit progression to the first clinical studies; scaling-up, characterization, quality control and validation of the cell product; design, validation and approval of the surgical device; and operative procedures for safe and effective delivery of cell product to the brain. Furthermore, clinical trials that incorporate principles of efficient design and disease-specific outcomes are urgently needed (particularly for those undertaken in rare diseases, where relatively small cohorts are an additional limiting factor), and all processes must be adaptable in a dynamic regulatory environment. Here we set out the challenges associated with the clinical translation of cell therapy, using Huntington's disease as a specific example, and suggest potential strategies to address these challenges. Huntington's disease presents a clear unmet need, but, importantly, it is an autosomal dominant condition with a readily available gene test, full genetic penetrance and a wide range of associated animal models, which together mean that it is a powerful condition in which to develop principles and test experimental therapeutics. We propose that solving these challenges in Huntington's disease would provide a road map for many other neurological conditions. This white paper represents a consensus opinion emerging from a series of meetings of the international translational platforms Stem Cells for Huntington's Disease and the European Huntington's Disease Network Advanced Therapies Working Group, established to identify the challenges of cell therapy, share experience, develop guidance and highlight future directions, with the aim to expedite progress towards therapies for clinical benefit in Huntington's disease.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Animales , Encéfalo/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia
2.
Int J Mol Sci ; 23(3)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35163606

RESUMEN

Generation of relevant and robust models for neurological disorders is of main importance for both target identification and drug discovery. The non-cell autonomous effects of glial cells on neurons have been described in a broad range of neurodegenerative and neurodevelopmental disorders, pointing to neuroglial interactions as novel alternative targets for therapeutics development. Interestingly, the recent breakthrough discovery of human induced pluripotent stem cells (hiPSCs) has opened a new road for studying neurological and neurodevelopmental disorders "in a dish". Here, we provide an overview of the generation and modeling of both neuronal and glial cells from human iPSCs and a brief synthesis of recent work investigating neuroglial interactions using hiPSCs in a pathophysiological context.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/trasplante , Enfermedades Neurodegenerativas , Trastornos del Neurodesarrollo , Neuroglía/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/terapia
3.
Neurobiol Dis ; 130: 104484, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31132407

RESUMEN

As research progresses in the understanding of the molecular and cellular mechanisms underlying neurodegenerative diseases like Huntington's disease (HD) and expands towards preclinical work for the development of new therapies, highly relevant animal models are increasingly needed to test new hypotheses and to validate new therapeutic approaches. In this light, we characterized an excitotoxic lesion model of striatal dysfunction in non-human primates (NHPs) using cognitive and motor behaviour assessment as well as functional imaging and post-mortem anatomical analyses. NHPs received intra-striatal stereotaxic injections of quinolinic acid bilaterally in the caudate nucleus and unilaterally in the left sensorimotor putamen. Post-operative MRI scans showed atrophy of the caudate nucleus and a large ventricular enlargement in all 6 NHPs that correlated with post-mortem measurements. Behavioral analysis showed deficits in 2 analogues of the Wisconsin card sorting test (perseverative behavior) and in an executive task, while no deficits were observed in a visual recognition or an episodic memory task at 6 months following surgery. Spontaneous locomotor activity was decreased after lesion and the incidence of apomorphine-induced dyskinesias was significantly increased at 3 and 6 months following lesion. Positron emission tomography scans obtained at end-point showed a major deficit in glucose metabolism and D2 receptor density limited to the lesioned striatum of all NHPs compared to controls. Post-mortem analyses revealed a significant loss of medium-sized spiny neurons in the striatum, a loss of neurons and fibers in the globus pallidus, a unilateral decrease in dopaminergic neurons of the substantia nigra and a loss of neurons in the motor and dorsolateral prefrontal cortex. Overall, we show that this robust NHP model presents specific behavioral (learning, execution and retention of cognitive tests) and metabolic functional deficits that, to the best of our knowledge, are currently not mimicked in any available large animal model of striatal dysfunction. Moreover, we used non-invasive, translational techniques like behavior and imaging to quantify such deficits and found that they correlate to a significant cell loss in the striatum and its main input and output structures. This model can thus significantly contribute to the pre-clinical longitudinal evaluation of the ability of new therapeutic cell, gene or pharmacotherapy approaches in restoring the functionality of the striatal circuitry.


Asunto(s)
Disfunción Cognitiva , Modelos Animales de Enfermedad , Enfermedad de Huntington , Trastornos Motores , Animales , Disfunción Cognitiva/inducido químicamente , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Enfermedad de Huntington/inducido químicamente , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Estudios Longitudinales , Macaca fascicularis , Masculino , Trastornos Motores/inducido químicamente , Ácido Quinolínico/toxicidad
4.
Brain ; 141(5): 1434-1454, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29534157

RESUMEN

The neurobiological functions of a number of kinases expressed in the brain are unknown. Here, we report new findings on DCLK3 (doublecortin like kinase 3), which is preferentially expressed in neurons in the striatum and dentate gyrus. Its function has never been investigated. DCLK3 expression is markedly reduced in Huntington's disease. Recent data obtained in studies related to cancer suggest DCLK3 could have an anti-apoptotic effect. Thus, we hypothesized that early loss of DCLK3 in Huntington's disease may render striatal neurons more susceptible to mutant huntingtin (mHtt). We discovered that DCLK3 silencing in the striatum of mice exacerbated the toxicity of an N-terminal fragment of mHtt. Conversely, overexpression of DCLK3 reduced neurodegeneration produced by mHtt. DCLK3 also produced beneficial effects on motor symptoms in a knock-in mouse model of Huntington's disease. Using different mutants of DCLK3, we found that the kinase activity of the protein plays a key role in neuroprotection. To investigate the potential mechanisms underlying DCLK3 effects, we studied the transcriptional changes produced by the kinase domain in human striatal neurons in culture. Results show that DCLK3 regulates in a kinase-dependent manner the expression of many genes involved in transcription regulation and nucleosome/chromatin remodelling. Consistent with this, histological evaluation showed DCLK3 is present in the nucleus of striatal neurons and, protein-protein interaction experiments suggested that the kinase domain interacts with zinc finger proteins, including the transcriptional activator adaptor TADA3, a core component of the Spt-ada-Gcn5 acetyltransferase (SAGA) complex which links histone acetylation to the transcription machinery. Our novel findings suggest that the presence of DCLK3 in striatal neurons may play a key role in transcription regulation and chromatin remodelling in these brain cells, and show that reduced expression of the kinase in Huntington's disease could render the striatum highly vulnerable to neurodegeneration.


Asunto(s)
Cuerpo Estriado/enzimología , Proteína Huntingtina/genética , Enfermedad de Huntington/terapia , Mutación/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Quinasas Similares a Doblecortina , Regulación hacia Abajo/genética , Complejo IV de Transporte de Electrones/metabolismo , Fuerza de la Mano/fisiología , Enfermedad de Huntington/genética , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora , Neuronas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Hum Mol Genet ; 21(17): 3883-95, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22678061

RESUMEN

Huntington's disease (HD) is characterized by a late clinical onset despite ubiquitous expression of the mutant gene at all developmental stages. How mutant huntingtin impacts on signalling pathways in the pre-symptomatic period has remained essentially unexplored in humans due to a lack of appropriate models. Using multiple human embryonic stem cell lines derived from blastocysts diagnosed as carrying the mutant huntingtin gene by pre-implantation genetic diagnosis, we explored early developmental changes in gene expression using differential transcriptomics, combined with gain and loss of function strategies. We demonstrated a down-regulation of the HTT gene itself in HD neural cells and identified three genes, the expression of which differs significantly in HD cells when compared with wild-type controls, namely CHCHD2, TRIM4 and PKIB. Similar dysregulation had been observed previously for CHCDH2 and TRIM4 in blood cells from patients. CHCHD2 is involved in mitochondrial function and PKIB in protein kinase A-dependent pathway regulation, which suggests that these functions may be precociously impacted in HD.


Asunto(s)
Células Madre Embrionarias/metabolismo , Enfermedad de Huntington/genética , Mutación/genética , Neuronas/metabolismo , Transcripción Genética , Transcriptoma/genética , Línea Celular , Células Madre Embrionarias/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Proteína Huntingtina , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/patología , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Stem Cells ; 31(9): 1763-74, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23818270

RESUMEN

Wnt-ligands are among key morphogens that mediate patterning of the anterior territories of the developing brain in mammals. We qualified the role of Wnt-signals in regional specification and subregional organization of the human telencephalon using human pluripotent stem cells (hPSCs). One step neural conversion of hPSCs using SMAD inhibitors leads to progenitors with a default rostral identity. It provides an ideal biological substrate for investigating the role of Wnt signaling in both anteroposterior and dorso-ventral processes. Challenging hPSC-neural derivatives with Wnt-antagonists, alone or combined with sonic hedgehog (Shh), we found that Wnt-inhibition promote both telencephalic specification and ventral patterning of telencephalic neural precursors in a dose-dependent manner. Using optimal Wnt-antagonist and Shh-agonist signals we produced human ventral-telencephalic precursors, committed to differentiation into striatal projection neurons both in vitro and in vivo after homotypic transplantation in quinolinate-lesioned rats. This study indicates that sequentially organized Wnt-signals play a key role in the development of human ventral telencephalic territories from which the striatum arise. In addition, the optimized production of hPSC-derived striatal cells described here offers a relevant biological resource for exploring and curing Huntington disease.


Asunto(s)
Tipificación del Cuerpo , Diferenciación Celular , Células Madre Embrionarias/citología , Neuronas/citología , Especificidad de Órganos , Telencéfalo/citología , Vía de Señalización Wnt , Animales , Tipificación del Cuerpo/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Proteínas Hedgehog/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Enfermedad de Huntington/patología , Enfermedad de Huntington/terapia , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Especificidad de Órganos/efectos de los fármacos , Ratas , Vía de Señalización Wnt/efectos de los fármacos
8.
Stem Cells ; 31(9): 1816-28, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23712629

RESUMEN

Decreased expression of neuronal genes such as brain-derived neurotrophic factor (BDNF) is associated with several neurological disorders. One molecular mechanism associated with Huntington disease (HD) is a discrete increase in the nuclear activity of the transcriptional repressor REST/NRSF binding to repressor element-1 (RE1) sequences. High-throughput screening of a library of 6,984 compounds with luciferase-assay measuring REST activity in neural derivatives of human embryonic stem cells led to identify two benzoimidazole-5-carboxamide derivatives that inhibited REST silencing in a RE1-dependent manner. The most potent compound, X5050, targeted REST degradation, but neither REST expression, RNA splicing nor binding to RE1 sequence. Differential transcriptomic analysis revealed the upregulation of neuronal genes targeted by REST in wild-type neural cells treated with X5050. This activity was confirmed in neural cells produced from human induced pluripotent stem cells derived from a HD patient. Acute intraventricular delivery of X5050 increased the expressions of BDNF and several other REST-regulated genes in the prefrontal cortex of mice with quinolinate-induced striatal lesions. This study demonstrates that the use of pluripotent stem cell derivatives can represent a crucial step toward the identification of pharmacological compounds with therapeutic potential in neurological affections involving decreased expression of neuronal genes associated to increased REST activity, such as Huntington disease.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Genes Reporteros , Humanos , Enfermedad de Huntington/patología , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteínas Represoras/metabolismo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
9.
J Huntingtons Dis ; 13(1): 41-53, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38427495

RESUMEN

Background: Mutations in the Huntingtin (HTT) gene cause Huntington's disease (HD), a neurodegenerative disorder. As a scaffold protein, HTT is involved in numerous cellular functions, but its normal and pathogenic functions during human forebrain development are poorly understood. Objective: To investigate the developmental component of HD, with a specific emphasis on understanding the functions of wild-type and mutant HTT alleles during forebrain neuron development in individuals carrying HD mutations. Methods: We used CRISPR/Cas9 gene-editing technology to disrupt the ATG region of the HTT gene via non-homologous end joining to produce mono- or biallelic HTT knock-out human induced pluripotent stem cell (iPSC) clones. Results: We showed that the loss of wild-type, mutant, or both HTT isoforms does not affect the pluripotency of iPSCs or their transition into neural cells. However, we observed that HTT loss causes division impairments in forebrain neuro-epithelial cells and alters maturation of striatal projection neurons (SPNs) particularly in the acquisition of DARPP32 expression, a key functional marker of SPNs. Finally, young post-mitotic neurons derived from HTT-/- human iPSCs display cellular dysfunctions observed in adult HD neurons. Conclusions: We described a novel collection of isogenic clones with mono- and biallelic HTT inactivation that complement existing HD-hiPSC isogenic series to explore HTT functions and test therapeutic strategies in particular HTT-lowering drugs. Characterizing neural and neuronal derivatives from human iPSCs of this collection, we show evidence that HTT loss or mutation has impacts on neuro-epithelial and striatal neurons maturation, and on basal DNA damage and BDNF axonal transport in post-mitotic neurons.


Asunto(s)
Enfermedad de Huntington , Células Madre Pluripotentes Inducidas , Adulto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Huntington/metabolismo , Neuronas/metabolismo , Cuerpo Estriado/metabolismo , Alelos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
10.
Methods Mol Biol ; 2551: 357-378, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36310215

RESUMEN

Aggregated alpha-synuclein (α-Syn) in neurons is a hallmark of Parkinson's disease (PD) and other synucleinopathies. Recent advances (1) in the production and purification of synthetic assemblies of α-Syn, (2) in the design and production of microfluidic devices allowing the construction of oriented and compartmentalized neuronal network on a chip, and (3) in the differentiation of human pluripotent stem cells (hPSCs) into specific neuronal subtypes now allow the study of cellular and molecular determinants of the prion-like properties of α-Syn in vitro. Here, we described the methods we used to reconstruct a cortico-cortical human neuronal network in microfluidic devices and how to take advantage of this cellular model to characterize (1) the prion-like properties of different α-Syn strains and (2) the neuronal dysfunctions and the alterations associated with the exposure to α-Syn strains or the nucleation of endogenous α-Syn protein in vitro.


Asunto(s)
Enfermedad de Parkinson , Priones , Sinucleinopatías , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Priones/metabolismo
11.
Hum Gene Ther ; 34(17-18): 958-974, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37658843

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disorder caused by a toxic gain-of-function CAG expansion in the first exon of the huntingtin (HTT) gene. The monogenic nature of HD makes mutant HTT (mHTT) inactivation a promising therapeutic strategy. Single nucleotide polymorphisms frequently associated with CAG expansion have been explored to selectively inactivate mHTT allele using the CRISPR/Cas9 system. One of such allele-selective approaches consists of excising a region flanking the first exon of mHTT by inducing simultaneous double-strand breaks at upstream and downstream positions of the mHTT exon 1. The removal of the first exon of mHTT deletes the CAG expansion and important transcription regulatory sites, leading to mHTT inactivation. However, the frequency of deletion events is yet to be quantified either in vitro or in vivo. Here, we developed accurate quantitative digital polymerase chain reaction-based assays to assess HTT exon 1 deletion in vitro and in fully humanized HU97/18 mice. Our results demonstrate that dual-single guide RNA (sgRNA) strategies are efficient and that 67% of HTT editing events are leading to exon 1 deletion in HEK293T cells. In contrast, these sgRNA actively cleaved HTT in HU97/18 mice, but most editing events do not lead to exon 1 deletion (10% exon 1 deletion). We also showed that the in vivo editing pattern is not affected by CAG expansion but may potentially be due to the presence of multiple copies of wildtype (wt)/mHTT genes HU97/18 mice as well as the slow kinetics of AAV-mediated CRISPR/Cas9 delivery.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Enfermedad de Huntington , Humanos , Animales , Ratones , ARN Guía de Sistemas CRISPR-Cas , Células HEK293 , Exones/genética , Alelos , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Sistema Nervioso Central
12.
Am J Hum Genet ; 83(2): 208-18, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18656177

RESUMEN

The size and organization of the brain neocortex has dramatically changed during primate evolution. This is probably due to the emergence of novel genes after duplication events, evolutionary changes in gene expression, and/or acceleration in protein evolution. Here, we describe a human Ret finger protein-like (hRFPL)1,2,3 gene cluster on chromosome 22, which is transactivated by the corticogenic transcription factor Pax6. High hRFPL1,2,3 transcript levels were detected at the onset of neurogenesis in differentiating human embryonic stem cells and in the developing human neocortex, whereas the unique murine RFPL gene is expressed in liver but not in neural tissue. Study of the evolutionary history of the RFPL gene family revealed that the RFPL1,2,3 gene ancestor emerged after the Euarchonta-Glires split. Subsequent duplication events led to the presence of multiple RFPL1,2,3 genes in Catarrhini ( approximately 34 mya) resulting in an increase in gene copy number in the hominoid lineage. In Catarrhini, RFPL1,2,3 expression profile diverged toward the neocortex and cerebellum over the liver. Importantly, humans showed a striking increase in cortical RFPL1,2,3 expression in comparison to their cerebellum, and to chimpanzee and macaque neocortex. Acceleration in RFPL-protein evolution was also observed with signs of positive selection in the RFPL1,2,3 cluster and two neofunctionalization events (acquisition of a specific RFPL-Defining Motif in all RFPLs and of a N-terminal 29 amino-acid sequence in catarrhinian RFPL1,2,3). Thus, we propose that the recent emergence and multiplication of the RFPL1,2,3 genes contribute to changes in primate neocortex size and/or organization.


Asunto(s)
Proteínas Portadoras/biosíntesis , Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Neocórtex/embriología , Secuencias de Aminoácidos , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Células HeLa , Humanos , Hígado/metabolismo , Macaca , Neocórtex/metabolismo , Pan troglodytes
13.
Proc Natl Acad Sci U S A ; 105(43): 16707-12, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18922775

RESUMEN

Substitutive cell therapy using fetal striatal grafts has demonstrated preliminary clinical success in patients with Huntington's disease, but the logistics required for accessing fetal cells preclude its extension to the relevant population of patients. Human embryonic stem (hES) cells theoretically meet this challenge, because they can be expanded indefinitely and differentiated into any cell type. We have designed an in vitro protocol combining substrates, media, and cytokines to push hES cells along the neural lineage, up to postmitotic neurons expressing striatal markers. The therapeutic potential of such hES-derived cells was further substantiated by their in vivo differentiation into striatal neurons following xenotransplantation into adult rats. Our results open the way toward hES cell therapy for Huntington's disease. Long-term proliferation of human neural progenitors leads, however, to xenograft overgrowth in the rat brain, suggesting that the path to the clinic requires a way to switch them off after grafting.


Asunto(s)
Cuerpo Estriado/citología , Fosfoproteína 32 Regulada por Dopamina y AMPc , Células Madre Embrionarias/citología , Neuronas/citología , Trasplante de Células Madre , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Medios de Cultivo , Citocinas/farmacología , Células Madre Embrionarias/trasplante , Humanos , Enfermedad de Huntington/terapia , Ácido Quinolínico , Ratas , Trasplante Heterólogo
14.
Sci Adv ; 7(14)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33789888

RESUMEN

Huntington disease (HD) damages the corticostriatal circuitry in large part by impairing transport of brain-derived neurotrophic factor (BDNF). We hypothesized that improving vesicular transport of BDNF could slow or prevent disease progression. We therefore performed selective proteomic analysis of vesicles transported within corticostriatal projecting neurons followed by in silico screening and identified palmitoylation as a pathway that could restore defective huntingtin-dependent trafficking. Using a synchronized trafficking assay and an HD network-on-a-chip, we found that increasing brain palmitoylation via ML348, which inhibits the palmitate-removing enzyme acyl-protein thioesterase 1 (APT1), restores axonal transport, synapse homeostasis, and survival signaling to wild-type levels without toxicity. In human HD induced pluripotent stem cell-derived cortical neurons, ML348 increased BDNF trafficking. In HD knock-in mice, it efficiently crossed the blood-brain barrier to restore palmitoylation levels and reverse neuropathology, locomotor deficits, and anxio-depressive behaviors. APT1 and its inhibitor ML348 thus hold therapeutic interest for HD.


Asunto(s)
Enfermedad de Huntington , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Lipoilación , Ratones , Proteómica
15.
Stroke ; 41(1): 153-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19940279

RESUMEN

BACKGROUND AND PURPOSE: Risk of tumorigenesis is a major obstacle to human embryonic and induced pluripotent stem cell therapy. Likely linked to the stage of differentiation of the cells at the time of implantation, formation of teratoma/tumors can also be influenced by factors released by the host tissue. We have analyzed the relative effects of the stage of differentiation and the postischemic environment on the formation of adverse structures by transplanted human embryonic stem cell-derived neural progenitors. METHODS: Four differentiation stages were identified on the basis of quantitative polymerase chain reaction expression of pluripotency, proliferation, and differentiation markers. Neural progenitors were transplanted at these 4 stages into rats with no, small, or large middle cerebral artery occlusion lesions. The fate of each transplant was compared with their pretransplantation status 1 to 4 months posttransplantation. RESULTS: The influence of the postischemic environment was limited to graft survival and occurrence of nonneuroectodermal structures after transplantation of very immature neural progenitors. Both effects were lost with differentiation. We identified a particular stage of differentiation characterized in vitro by a rebound of proliferative activity that produced highly proliferative grafts susceptible to threaten surrounding host tissues. CONCLUSIONS: The effects of the ischemic environment on the formation of teratoma by transplanted human embryonic stem cell-derived neural progenitors are limited to early differentiation stages that will likely not be used for stem cell therapy. In contrast, hyperproliferation observed at later stages of differentiation corresponds to an intrinsic activity that should be monitored to avoid tumorigenesis.


Asunto(s)
Isquemia Encefálica/cirugía , Células Madre Embrionarias/trasplante , Ambiente , Neuronas/trasplante , Trasplante de Células Madre , Teratoma/patología , Factores de Edad , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Diferenciación Celular , Células Madre Embrionarias/citología , Humanos , Masculino , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Trasplante de Células Madre/efectos adversos , Teratoma/etiología
16.
Stem Cell Reports ; 12(2): 230-244, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30639210

RESUMEN

Reappraisal of neuropathological studies suggests that pathological hallmarks of Alzheimer's disease and Parkinson's disease (PD) spread progressively along predictable neuronal pathways in the human brain through unknown mechanisms. Although there is much evidence supporting the prion-like propagation and amplification of α-synuclein (α-Syn) in vitro and in rodent models, whether this scenario occurs in the human brain remains to be substantiated. Here we reconstructed in microfluidic devices corticocortical neuronal networks using human induced pluripotent stem cells derived from a healthy donor. We provide unique experimental evidence that different strains of human α-Syn disseminate in "wild-type" human neuronal networks in a prion-like manner. We show that two distinct α-Syn strains we named fibrils and ribbons are transported, traffic between neurons, and trigger to different extents, in a dose- and structure-dependent manner, the progressive accumulation of PD-like pathological hallmarks. We further demonstrate that seeded aggregation of endogenous soluble α-Syn affects synaptic integrity and mitochondria morphology.


Asunto(s)
Neuronas/metabolismo , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Parkinson/metabolismo
17.
Stem Cell Reports ; 13(3): 448-457, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31447328

RESUMEN

The mechanisms underlying the selective degeneration of medium spiny neurons (MSNs) in Huntington disease (HD) remain largely unknown. CTIP2, a transcription factor expressed by all MSNs, is implicated in HD pathogenesis because of its interactions with mutant huntingtin. Here, we report a key role for CTIP2 in protein phosphorylation via governing protein kinase A (PKA) signaling in human striatal neurons. Transcriptomic analysis of CTIP2-deficient MSNs implicates CTIP2 target genes at the heart of cAMP-Ca2+ signal integration in the PKA pathway. These findings are further supported by experimental evidence of a substantial reduction in phosphorylation of DARPP32 and GLUR1, two PKA targets in CTIP2-deficient MSNs. Moreover, we show that CTIP2-dependent dysregulation of protein phosphorylation is shared by HD hPSC-derived MSNs and striatal tissues of two HD mouse models. This study therefore establishes an essential role for CTIP2 in human MSN homeostasis and provides mechanistic and potential therapeutic insight into striatal neurodegeneration.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Cuerpo Estriado/metabolismo , Edición Génica , Células Madre Embrionarias Humanas/citología , Humanos , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Neuronas/citología , Estrés Oxidativo , Fosforilación , Receptores AMPA/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal , Transcriptoma , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
18.
Nat Commun ; 10(1): 4357, 2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31554807

RESUMEN

Cell therapy products (CTP) derived from pluripotent stem cells (iPSCs) may constitute a renewable, specifically differentiated source of cells to potentially cure patients with neurodegenerative disorders. However, the immunogenicity of CTP remains a major issue for therapeutic approaches based on transplantation of non-autologous stem cell-derived neural grafts. Despite its considerable side-effects, long-term immunosuppression, appears indispensable to mitigate neuro-inflammation and prevent rejection of allogeneic CTP. Matching iPSC donors' and patients' HLA haplotypes has been proposed as a way to access CTP with enhanced immunological compatibility, ultimately reducing the need for immunosuppression. In the present work, we challenge this paradigm by grafting autologous, MHC-matched and mis-matched neuronal grafts in a primate model of Huntington's disease. Unlike previous reports in unlesioned hosts, we show that in the absence of immunosuppression MHC matching alone is insufficient to grant long-term survival of neuronal grafts in the lesioned brain.


Asunto(s)
Rechazo de Injerto/inmunología , Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/trasplante , Complejo Mayor de Histocompatibilidad/inmunología , Neuronas/trasplante , Animales , Diferenciación Celular/inmunología , Citotoxicidad Inmunológica/inmunología , Modelos Animales de Enfermedad , Prueba de Histocompatibilidad , Humanos , Enfermedad de Huntington/inmunología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/inmunología , Neuronas/citología , Neuronas/inmunología , Primates , Ratas Desnudas , Trasplante Autólogo
19.
Neuron ; 33(2): 275-85, 2002 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-11804574

RESUMEN

As a tetramer, acetylcholinesterase (AChE) is anchored to the basal lamina of the neuromuscular junction and to the membrane of neuronal synapses. We have previously shown that collagen Q (ColQ) anchors AChE at the neuromuscular junction. We have now cloned the gene PRiMA (proline-rich membrane anchor) encoding the AChE anchor in mammalian brain. We show that PRiMA is able to organize AChE into tetramers and to anchor them at the surface of transfected cells. Furthermore, we demonstrate that AChE is actually anchored in neural cell membranes through its interaction with PRiMA. Finally, we propose that only PRiMA anchors AChE in mammalian brain and muscle cell membranes.


Asunto(s)
Acetilcolinesterasa/metabolismo , Encéfalo/enzimología , Proteínas de la Membrana/fisiología , Proteínas Musculares , Proteínas del Tejido Nervioso/fisiología , Acetilcolinesterasa/química , Animales , Butirilcolinesterasa/química , Membrana Celular/enzimología , Membrana Celular/metabolismo , Colágeno/química , ADN Complementario/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Músculo Esquelético/enzimología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Oocitos , Prolina , Estructura Terciaria de Proteína , Células Tumorales Cultivadas , Xenopus
20.
eNeuro ; 5(4)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30310861

RESUMEN

The neurodegenerative Huntington's disease (HD) is caused by a polyglutamine (polyQ) amplification in the huntingtin protein (HTT). Currently there is no effective therapy available for HD; however, several efforts are directed to develop and optimize HTT-lowering methods to improve HD phenotypes. To validate these approaches, there is an immediate need for reliable, sensitive, and easily accessible methods to quantify HTT expression. Using the AlphaLISA platform, we developed two novel sensitive and robust assays for quantification of HTT in biological samples using commercially available antibodies. The first, a polyQ-independent assay, measures the total pool of HTT, while the second, a polyQ-dependent assay, preferentially detects the mutant form of HTT. Using purified HTT protein standards and brain homogenates from an HD mouse model, we determine a lower limit of quantification of 1 and 3 pm and optimal reproducibility with CV values lower than 7% for intra- and 20% for interassay. In addition, we used the assays to quantify HTT in neural stem cells generated from patient-derived induced pluripotent stem cells in vitro and in human brain tissue lysates. Finally, we could detect changes in HTT levels in a mouse model where mutant HTT was conditionally deleted in neural tissue, verifying the potential to monitor the outcome of HTT-lowering strategies. This analytical platform is ideal for high-throughput screens and thus has an added value for the HD community as a tool to optimize novel therapeutic approaches aimed at modulating HTT protein levels.


Asunto(s)
Proteína Huntingtina/análisis , Enfermedad de Huntington/diagnóstico , Inmunoensayo/normas , Animales , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inmunoensayo/métodos , Ratones , Mutación , Células-Madre Neurales , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA