Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
EBioMedicine ; 103: 105124, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38701619

RESUMEN

BACKGROUND: PolyQ diseases are autosomal dominant neurodegenerative disorders caused by the expansion of CAG repeats. While of slow progression, these diseases are ultimately fatal and lack effective therapies. METHODS: A high-throughput chemical screen was conducted to identify drugs that lower the toxicity of a protein containing the first exon of Huntington's disease (HD) protein huntingtin (HTT) harbouring 94 glutamines (Htt-Q94). Candidate drugs were tested in a wide range of in vitro and in vivo models of polyQ toxicity. FINDINGS: The chemical screen identified the anti-leprosy drug clofazimine as a hit, which was subsequently validated in several in vitro models. Computational analyses of transcriptional signatures revealed that the effect of clofazimine was due to the stimulation of mitochondrial biogenesis by peroxisome proliferator-activated receptor gamma (PPARγ). In agreement with this, clofazimine rescued mitochondrial dysfunction triggered by Htt-Q94 expression. Importantly, clofazimine also limited polyQ toxicity in developing zebrafish and neuron-specific worm models of polyQ disease. INTERPRETATION: Our results support the potential of repurposing the antimicrobial drug clofazimine for the treatment of polyQ diseases. FUNDING: A full list of funding sources can be found in the acknowledgments section.


Asunto(s)
Clofazimina , Modelos Animales de Enfermedad , Proteína Huntingtina , Leprostáticos , PPAR gamma , Péptidos , Pez Cebra , Clofazimina/farmacología , PPAR gamma/metabolismo , PPAR gamma/genética , Animales , Humanos , Péptidos/farmacología , Leprostáticos/farmacología , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/metabolismo
2.
Nat Commun ; 14(1): 8040, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086799

RESUMEN

Epigenetic dysregulation may influence disease progression. Here we explore whether epigenetic alterations in human pancreatic islets impact insulin secretion and type 2 diabetes (T2D). In islets, 5,584 DNA methylation sites exhibit alterations in T2D cases versus controls and are associated with HbA1c in individuals not diagnosed with T2D. T2D-associated methylation changes are found in enhancers and regions bound by ß-cell-specific transcription factors and associated with reduced expression of e.g. CABLES1, FOXP1, GABRA2, GLR1A, RHOT1, and TBC1D4. We find RHOT1 (MIRO1) to be a key regulator of insulin secretion in human islets. Rhot1-deficiency in ß-cells leads to reduced insulin secretion, ATP/ADP ratio, mitochondrial mass, Ca2+, and respiration. Regulators of mitochondrial dynamics and metabolites, including L-proline, glycine, GABA, and carnitines, are altered in Rhot1-deficient ß-cells. Islets from diabetic GK rats present Rhot1-deficiency. Finally, RHOT1methylation in blood is associated with future T2D. Together, individuals with T2D exhibit epigenetic alterations linked to mitochondrial dysfunction in pancreatic islets.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Ratas , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Secreción de Insulina , Insulina/metabolismo , Metilación de ADN , Islotes Pancreáticos/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción/metabolismo , Epigénesis Genética , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción Forkhead/metabolismo
3.
Front Aging Neurosci ; 15: 1125739, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37065462

RESUMEN

As our society ages inexorably, geroscience and research focusing on healthy aging is becoming increasingly urgent. Macroautophagy (referred to as autophagy), a highly conserved process of cellular clearance and rejuvenation has attracted much attention due to its universal role in organismal life and death. Growing evidence points to autophagy process as being one of the key players in the determination of lifespan and health. Autophagy inducing interventions show significant improvement in organismal lifespan demonstrated in several experimental models. In line with this, preclinical models of age-related neurodegenerative diseases demonstrate pathology modulating effect of autophagy induction, implicating its potential to treat such disorders. In humans this specific process seems to be more complex. Recent clinical trials of drugs targeting autophagy point out some beneficial effects for clinical use, although with limited effectiveness, while others fail to show any significant improvement. We propose that using more human-relevant preclinical models for testing drug efficacy would significantly improve clinical trial outcomes. Lastly, the review discusses the available cellular reprogramming techniques used to model neuronal autophagy and neurodegeneration while exploring the existing evidence of autophagy's role in aging and pathogenesis in human-derived in vitro models such as embryonic stem cells (ESCs), induced pluripotent stem cell derived neurons (iPSC-neurons) or induced neurons (iNs).

4.
Cell Reprogram ; 25(1): 32-44, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36719998

RESUMEN

Mesenchymal stem/stromal cells (MSCs) are found in almost all postnatal organs. Under appropriate environmental cues, multipotency enables MSCs to serve as progenitors for several lineage-specific, differentiated cell types. In vitro expansion and differentiation of MSCs give the opportunity to obtain hardly available somatic cells, such as neurons. The neurogenic potential of MSCs makes them a promising, autologous source to restore damaged tissue and as such, they have received much attention in the field of regenerative medicine. Several stem cell pool candidates have been studied thus far, but only a few of them showed neurogenic differentiation potential. Due to their embryonic ontology, stem cells residing in the stroma of the dental pulp chamber are an exciting source for in vitro neural cell differentiation. In this study, we review the key properties of dental pulp stem cells (DPSCs), with a particular focus on their neurogenic potential. Moreover, we summarize the various presently available methods used for neural differentiation of human DPSCs also emphasizing the difficulties in reproducibly high production of such cells. We postulate that because DPSCs are stem cells with very close ontology to neurogenic lineages, they may serve as excellent targets for neuronal differentiation in vitro and even for direct reprogramming.


Asunto(s)
Pulpa Dental , Células Madre Mesenquimatosas , Humanos , Células Madre , Diferenciación Celular/fisiología , Neurogénesis , Células Cultivadas
5.
Stem Cell Reports ; 17(10): 2203-2219, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36150382

RESUMEN

We have developed an efficient approach to generate functional induced dopaminergic (DA) neurons from adult human dermal fibroblasts. When performing DA neuronal conversion of patient fibroblasts with idiopathic Parkinson's disease (PD), we could specifically detect disease-relevant pathology in these cells. We show that the patient-derived neurons maintain age-related properties of the donor and exhibit lower basal chaperone-mediated autophagy compared with healthy donors. Furthermore, stress-induced autophagy resulted in an age-dependent accumulation of macroautophagic structures. Finally, we show that these impairments in patient-derived DA neurons leads to an accumulation of phosphorylated alpha-synuclein, the classical hallmark of PD pathology. This pathological phenotype is absent in neurons generated from induced pluripotent stem cells from the same patients. Taken together, our results show that direct neural reprogramming can be used for obtaining patient-derived DA neurons, which uniquely function as a cellular model to study age-related pathology relevant to idiopathic PD.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Parkinson , Adulto , Autofagia/fisiología , Neuronas Dopaminérgicas/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Enfermedad de Parkinson/genética , alfa-Sinucleína/genética
6.
Acta Neuropathol Commun ; 10(1): 106, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35869509

RESUMEN

Altered microRNA (miRNA) expression is a common feature of Huntington's disease (HD) and could participate in disease onset and progression. However, little is known about the underlying causes of miRNA disruption in HD. We and others have previously shown that mutant Huntingtin binds to Ago2, a central component of miRNA biogenesis, and disrupts mature miRNA levels. In this study, we sought to determine if miRNA maturation per se was compromised in HD. Towards this end, we characterized major miRNA biogenesis pathway components and miRNA maturation products (pri-miRNA, pre-miRNA, and mature) in human HD (N = 41, Vonsattel grades HD2-4) and healthy control (N = 25) subjects. Notably, the striatum (putamen) and cortex (BA39) from the same individuals were analyzed in parallel. We show that Ago2, Drosha, and Dicer were strongly downregulated in human HD at the early stages of the disease. Using a panel of HD-related miRNAs (miR-10b, miR-196b, miR-132, miR-212, miR-127, miR-128), we uncovered various types of maturation defects in the HD brain, the most prominent occurring at the pre-miRNA to mature miRNA maturation step. Consistent with earlier findings, we provide evidence that alterations in autophagy could participate in miRNA maturation defects. Notably, most changes occurred in the striatum, which is more prone to HTT aggregation and neurodegeneration. Likewise, we observed no significant alterations in miRNA biogenesis in human HD cortex and blood, strengthening tissue-specific effects. Overall, these data provide important clues into the underlying mechanisms behind miRNA alterations in HD-susceptible tissues. Further investigations are now required to understand the biological, diagnostic, and therapeutic implications of miRNA/RNAi biogenesis defects in HD and related neurodegenerative disorders.


Asunto(s)
Enfermedad de Huntington , MicroARNs , Encéfalo/metabolismo , Cuerpo Estriado/metabolismo , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , MicroARNs/metabolismo , Putamen/metabolismo
7.
Autophagy ; 18(12): 3031-3032, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35468035

RESUMEN

Huntington disease is an inherited, progressive, incurable neurodegenerative disorder that primarily affects cells in the brain. Although the genetic basis for this condition has been known for nearly 30 years, how this causes disease is still unresolved. Of late there has been increasing evidence suggesting that dysfunction in macroautophagic/autophagic pathways may contribute to cellular dysfunction and death. In our recent work we highlight more precisely how and where this problem might arise in this pathway using directly reprogrammed neurons.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Humanos , Autofagia/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Degeneración Nerviosa/patología , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo
8.
Cell Stem Cell ; 29(1): 52-69.e8, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34624206

RESUMEN

The human forebrain has expanded in size and complexity compared to chimpanzees despite limited changes in protein-coding genes, suggesting that gene expression regulation is an important driver of brain evolution. Here, we identify a KRAB-ZFP transcription factor, ZNF558, that is expressed in human but not chimpanzee forebrain neural progenitor cells. ZNF558 evolved as a suppressor of LINE-1 transposons but has been co-opted to regulate a single target, the mitophagy gene SPATA18. ZNF558 plays a role in mitochondrial homeostasis, and loss-of-function experiments in cerebral organoids suggests that ZNF558 influences developmental timing during early human brain development. Expression of ZNF558 is controlled by the size of a variable number tandem repeat that is longer in chimpanzees compared to humans, and variable in the human population. Thus, this work provides mechanistic insight into how a cis-acting structural variation establishes a regulatory network that affects human brain evolution.


Asunto(s)
Redes Reguladoras de Genes , Organoides , Encéfalo/metabolismo , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Humanos , Organoides/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Brain ; 145(9): 3035-3057, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34936701

RESUMEN

Huntington's disease is a neurodegenerative disorder caused by CAG expansions in the huntingtin (HTT) gene. Modelling Huntington's disease is challenging, as rodent and cellular models poorly recapitulate the disease as seen in ageing humans. To address this, we generated induced neurons through direct reprogramming of human skin fibroblasts, which retain age-dependent epigenetic characteristics. Huntington's disease induced neurons (HD-iNs) displayed profound deficits in autophagy, characterized by reduced transport of late autophagic structures from the neurites to the soma. These neurite-specific alterations in autophagy resulted in shorter, thinner and fewer neurites specifically in HD-iNs. CRISPRi-mediated silencing of HTT did not rescue this phenotype but rather resulted in additional autophagy alterations in control induced neurons, highlighting the importance of wild-type HTT in normal neuronal autophagy. In summary, our work identifies a distinct subcellular autophagy impairment in adult patient derived Huntington's disease neurons and provides a new rationale for future development of autophagy activation therapies.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Adulto , Autofagia/fisiología , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Neuronas
10.
Nat Commun ; 12(1): 2431, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893273

RESUMEN

Insulin resistance and lower muscle quality (strength divided by mass) are hallmarks of type 2 diabetes (T2D). Here, we explore whether alterations in muscle stem cells (myoblasts) from individuals with T2D contribute to these phenotypes. We identify VPS39 as an important regulator of myoblast differentiation and muscle glucose uptake, and VPS39 is downregulated in myoblasts and myotubes from individuals with T2D. We discover a pathway connecting VPS39-deficiency in human myoblasts to impaired autophagy, abnormal epigenetic reprogramming, dysregulation of myogenic regulators, and perturbed differentiation. VPS39 knockdown in human myoblasts has profound effects on autophagic flux, insulin signaling, epigenetic enzymes, DNA methylation and expression of myogenic regulators, and gene sets related to the cell cycle, muscle structure and apoptosis. These data mimic what is observed in myoblasts from individuals with T2D. Furthermore, the muscle of Vps39+/- mice display reduced glucose uptake and altered expression of genes regulating autophagy, epigenetic programming, and myogenesis. Overall, VPS39-deficiency contributes to impaired muscle differentiation and reduced glucose uptake. VPS39 thereby offers a therapeutic target for T2D.


Asunto(s)
Proteínas Relacionadas con la Autofagia/genética , Autofagia/genética , Diferenciación Celular/genética , Diabetes Mellitus Tipo 2/genética , Epigenómica/métodos , Mioblastos/metabolismo , Células Madre/metabolismo , Proteínas de Transporte Vesicular/genética , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Epigénesis Genética/genética , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Desarrollo de Músculos/genética , Proteínas de Transporte Vesicular/deficiencia
11.
EMBO J ; 40(9): e106423, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33644903

RESUMEN

Endogenous retroviruses (ERVs) make up a large fraction of mammalian genomes and are thought to contribute to human disease, including brain disorders. In the brain, aberrant activation of ERVs is a potential trigger for an inflammatory response, but mechanistic insight into this phenomenon remains lacking. Using CRISPR/Cas9-based gene disruption of the epigenetic co-repressor protein Trim28, we found a dynamic H3K9me3-dependent regulation of ERVs in proliferating neural progenitor cells (NPCs), but not in adult neurons. In vivo deletion of Trim28 in cortical NPCs during mouse brain development resulted in viable offspring expressing high levels of ERVs in excitatory neurons in the adult brain. Neuronal ERV expression was linked to activated microglia and the presence of ERV-derived proteins in aggregate-like structures. This study demonstrates that brain development is a critical period for the silencing of ERVs and provides causal in vivo evidence demonstrating that transcriptional activation of ERV in neurons results in an inflammatory response.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encefalitis/genética , Retrovirus Endógenos/genética , Eliminación de Gen , Proteína 28 que Contiene Motivos Tripartito/genética , Animales , Encéfalo/inmunología , Encéfalo/virología , Sistemas CRISPR-Cas , Células Cultivadas , Encefalitis/inmunología , Encefalitis/virología , Retrovirus Endógenos/inmunología , Epigénesis Genética , Regulación de la Expresión Génica , Histonas/metabolismo , Ratones , Activación Transcripcional
12.
Autophagy ; 17(6): 1316-1329, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32374203

RESUMEN

Activation of macroautophagy/autophagy, a key mechanism involved in the degradation and removal of aggregated proteins, can successfully reverse Huntington disease phenotypes in various model systems. How neuronal autophagy impairments need to be considered in Huntington disease progression to achieve a therapeutic effect is currently not known. In this study, we used a mouse model of HTT (huntingtin) protein aggregation to investigate how different methods and timing of autophagy activation influence the efficacy of autophagy-activating treatment in vivo. We found that overexpression of human TFEB, a master regulator of autophagy, did not decrease mutant HTT aggregation. On the other hand, Becn1 overexpression, an autophagic regulator that plays a key role in autophagosome formation, partially cleared mutant HTT aggregates and restored neuronal pathology, but only when administered early in the disease progression. When Becn1 was administered at a later stage, when prominent mutant HTT accumulation and autophagy impairments have occurred, Becn1 overexpression did not rescue the mutant HTT-associated phenotypes. Together, these results demonstrate that the targets used to activate autophagy, as well as the timing of autophagy activation, are crucial for achieving efficient therapeutic effects.Abbreviations: AAV: adeno-associated viral vectors; ACTB: actin beta; BECN1: beclin 1, autophagy related; DAPI: 4',6-diamidino-2-phenylindole; GO: gene ontology; HD: Huntington disease; HTT: huntingtin; ICQ: Li's intensity correlation quotient; IHC: immunohistochemistry; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; mHTT: mutant huntingtin; PCA: principal component analysis; PPP1R1B/DARPP-32: protein phosphatase 1 regulatory inhibitor subunit 1B; SQSTM1: sequestosome 1; TFEB: transcription factor EB; WB: western blot; WT: wild-type.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/fisiología , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Animales , Beclina-1/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Factores de Tiempo
13.
Nat Commun ; 10(1): 3182, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-31320637

RESUMEN

DNA methylation contributes to the maintenance of genomic integrity in somatic cells, in part through the silencing of transposable elements. In this study, we use CRISPR-Cas9 technology to delete DNMT1, the DNA methyltransferase key for DNA methylation maintenance, in human neural progenitor cells (hNPCs). We observe that inactivation of DNMT1 in hNPCs results in viable, proliferating cells despite a global loss of DNA CpG-methylation. DNA demethylation leads to specific transcriptional activation and chromatin remodeling of evolutionarily young, hominoid-specific LINE-1 elements (L1s), while older L1s and other classes of transposable elements remain silent. The activated L1s act as alternative promoters for many protein-coding genes involved in neuronal functions, revealing a hominoid-specific L1-based transcriptional network controlled by DNA methylation that influences neuronal protein-coding genes. Our results provide mechanistic insight into the role of DNA methylation in silencing transposable elements in somatic human cells, as well as further implicating L1s in human brain development and disease.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/genética , Desmetilación del ADN , Metilación de ADN/genética , Elementos de Nucleótido Esparcido Largo/genética , Células-Madre Neurales/citología , Encéfalo/embriología , Sistemas CRISPR-Cas/genética , Ensamble y Desensamble de Cromatina/genética , Islas de CpG/genética , Silenciador del Gen/fisiología , Humanos , Células-Madre Neurales/metabolismo , Activación Transcripcional/genética
14.
PLoS Genet ; 15(3): e1008036, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30865625

RESUMEN

Transposable elements (TEs) are dynamically expressed at high levels in multiple human tissues, but the function of TE-derived transcripts remains largely unknown. In this study, we identify numerous TE-derived microRNAs (miRNAs) by conducting Argonaute2 RNA immunoprecipitation followed by small RNA sequencing (AGO2 RIP-seq) on human brain tissue. Many of these miRNAs originated from LINE-2 (L2) elements, which entered the human genome around 100-300 million years ago. L2-miRNAs derived from the 3' end of the L2 consensus sequence and thus shared very similar sequences, indicating that L2-miRNAs could target transcripts with L2s in their 3'UTR. In line with this, many protein-coding genes carried fragments of L2-derived sequences in their 3'UTR: these sequences served as target sites for L2-miRNAs. L2-miRNAs and their targets were generally ubiquitously expressed at low levels in multiple human tissues, suggesting a role for this network in buffering transcriptional levels of housekeeping genes. In addition, we also found evidence that this network is perturbed in glioblastoma. In summary, our findings uncover a TE-based post-transcriptional network that shapes transcriptional regulation in human cells.


Asunto(s)
Elementos Transponibles de ADN , Elementos de Nucleótido Esparcido Largo , MicroARNs/genética , Regiones no Traducidas 3' , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Secuencia de Bases , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genoma Humano , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , MicroARNs/metabolismo
15.
Cell Syst ; 7(4): 438-452.e8, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30292704

RESUMEN

Non-coding RNAs regulate many biological processes including neurogenesis. The brain-enriched miR-124 has been assigned as a key player of neuronal differentiation via its complex but little understood regulation of thousands of annotated targets. To systematically chart its regulatory functions, we used CRISPR/Cas9 gene editing to disrupt all six miR-124 alleles in human induced pluripotent stem cells. Upon neuronal induction, miR-124-deleted cells underwent neurogenesis and became functional neurons, albeit with altered morphology and neurotransmitter specification. Using RNA-induced-silencing-complex precipitation, we identified 98 high-confidence miR-124 targets, of which some directly led to decreased viability. By performing advanced transcription-factor-network analysis, we identified indirect miR-124 effects on apoptosis, neuronal subtype differentiation, and the regulation of previously uncharacterized zinc finger transcription factors. Our data emphasize the need for combined experimental- and system-level analyses to comprehensively disentangle and reveal miRNA functions, including their involvement in the neurogenesis of diverse neuronal cell types found in the human brain.


Asunto(s)
Redes Reguladoras de Genes , MicroARNs/genética , Neurogénesis/genética , Células Cultivadas , Células HEK293 , Humanos , MicroARNs/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Brain Plast ; 3(2): 195-203, 2018 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-30151343

RESUMEN

Adult neurogenesis in the mammalian brain, including in humans, occurs throughout life in distinct brain regions. Alterations in adult neurogenesis is a common phenomenon in several different neurodegenerative disorders, which is likely to contribute to the pathophysiology of these disorders. This review summarizes novel concepts related to the interplay between autophagy and microRNAs in control of adult neurogenesis, with a specific focus on its relevance to neurodegenerative diseases.

17.
Cell Rep ; 24(6): 1397-1406, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30089251

RESUMEN

Many neurodegenerative diseases are characterized by the presence of intracellular protein aggregates, resulting in alterations in autophagy. However, the consequences of impaired autophagy for neuronal function remain poorly understood. In this study, we used cell culture and mouse models of huntingtin protein aggregation as well as post-mortem material from patients with Huntington's disease to demonstrate that Argonaute-2 (AGO2) accumulates in the presence of neuronal protein aggregates and that this is due to impaired autophagy. Accumulation of AGO2, a key factor of the RNA-induced silencing complex that executes microRNA functions, results in global alterations of microRNA levels and activity. Together, these results demonstrate that impaired autophagy found in neurodegenerative diseases not only influences protein aggregation but also directly contributes to global alterations of intracellular post-transcriptional networks.


Asunto(s)
Proteínas Argonautas/genética , Autofagia/fisiología , Enfermedad de Huntington/genética , MicroARNs/metabolismo , Humanos
18.
J Vis Exp ; (132)2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29443113

RESUMEN

Induced neurons (iNs), the product of somatic cells directly converted to neurons, are a way to obtain patient-derived neurons from tissue that is easily accessible. Through this route, mature neurons can be obtained in a matter of a few weeks. Here, we describe a straightforward and rapid one-step protocol to obtain iNs from dermal fibroblasts obtained through biopsy samples from adult human donors. We explain each step of the process, including the maintenance of the dermal fibroblasts, the freezing procedure to build a stock of the cell line, seeding of the cells for reprogramming, as well as the culture conditions during the conversion process. In addition, we describe the preparation of glass coverslips for electrophysiological recordings, long-term coating conditions, and fluorescence activated cell sorting (FACS). We also illustrate examples of the results to be expected. The protocol described here is easy to perform and can be applied to human fibroblasts derived from human skin biopsies from patients with various different diagnoses and ages. This protocol generates a sufficient amount of iNs which can be used for a wide array of biomedical applications, including disease modeling, drug screening, and target validation.


Asunto(s)
Reprogramación Celular/fisiología , Fibroblastos/metabolismo , Neuronas/metabolismo , Técnicas de Cultivo de Célula , Fibroblastos/citología , Humanos
19.
Front Neurosci ; 11: 530, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29033781

RESUMEN

Direct neuronal reprogramming, by which a neuron is formed via direct conversion from a somatic cell without going through a pluripotent intermediate stage, allows for the possibility of generating patient-derived neurons. A unique feature of these so-called induced neurons (iNs) is the potential to maintain aging and epigenetic signatures of the donor, which is critical given that many diseases of the CNS are age related. Here, we review the published literature on the work that has been undertaken using iNs to model human brain disorders. Furthermore, as disease-modeling studies using this direct neuronal reprogramming approach are becoming more widely adopted, it is important to assess the criteria that are used to characterize the iNs, especially in relation to the extent to which they are mature adult neurons. In particular: i) what constitutes an iN cell, ii) which stages of conversion offer the earliest/optimal time to assess features that are specific to neurons and/or a disorder and iii) whether generating subtype-specific iNs is critical to the disease-related features that iNs express. Finally, we discuss the range of potential biomedical applications that can be explored using patient-specific models of neurological disorders with iNs, and the challenges that will need to be overcome in order to realize these applications.

20.
EMBO Mol Med ; 9(8): 1117-1131, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28646119

RESUMEN

Direct conversion of human fibroblasts into mature and functional neurons, termed induced neurons (iNs), was achieved for the first time 6 years ago. This technology offers a promising shortcut for obtaining patient- and disease-specific neurons for disease modeling, drug screening, and other biomedical applications. However, fibroblasts from adult donors do not reprogram as easily as fetal donors, and no current reprogramming approach is sufficiently efficient to allow the use of this technology using patient-derived material for large-scale applications. Here, we investigate the difference in reprogramming requirements between fetal and adult human fibroblasts and identify REST as a major reprogramming barrier in adult fibroblasts. Via functional experiments where we overexpress and knockdown the REST-controlled neuron-specific microRNAs miR-9 and miR-124, we show that the effect of REST inhibition is only partially mediated via microRNA up-regulation. Transcriptional analysis confirmed that REST knockdown activates an overlapping subset of neuronal genes as microRNA overexpression and also a distinct set of neuronal genes that are not activated via microRNA overexpression. Based on this, we developed an optimized one-step method to efficiently reprogram dermal fibroblasts from elderly individuals using a single-vector system and demonstrate that it is possible to obtain iNs of high yield and purity from aged individuals with a range of familial and sporadic neurodegenerative disorders including Parkinson's, Huntington's, as well as Alzheimer's disease.


Asunto(s)
Transdiferenciación Celular , Fibroblastos/fisiología , Técnicas de Silenciamiento del Gen , Neuronas/fisiología , Proteínas Represoras/biosíntesis , Adulto , Técnicas Citológicas/métodos , Perfilación de la Expresión Génica , Humanos , MicroARNs/análisis , Proteínas Represoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA