Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 14(1): 189, 2023 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-37507794

RESUMO

BACKGROUND: Huntington's disease (HD) is a motor and cognitive neurodegenerative disorder due to prominent loss of striatal medium spiny neurons (MSNs). Cell replacement using human embryonic stem cells (hESCs) derivatives may offer new therapeutic opportunities to replace degenerated neurons and repair damaged circuits. METHODS: With the aim to develop effective cell replacement for HD, we assessed the long-term therapeutic value of hESC-derived striatal progenitors by grafting the cells into the striatum of a preclinical model of HD [i.e., adult immunodeficient rats in which the striatum was lesioned by monolateral injection of quinolinic acid (QA)]. We examined the survival, maturation, self-organization and integration of the graft as well as its impact on lesion-dependent motor alterations up to 6 months post-graft. Moreover, we tested whether exposing a cohort of QA-lesioned animals to environmental enrichment (EE) could improve graft integration and function. RESULTS: Human striatal progenitors survived up to 6 months after transplantation and showed morphological and neurochemical features typical of human MSNs. Donor-derived interneurons were also detected. Grafts wired in both local and long-range striatal circuits, formed domains suggestive of distinct ganglionic eminence territories and displayed emerging striosome features. Moreover, over time grafts improved complex motor performances affected by QA. EE selectively increased cell differentiation into MSN phenotype and promoted host-to-graft connectivity. However, when combined to the graft, the EE paradigm used in this study was insufficient to produce an additive effect on task execution. CONCLUSIONS: The data support the long-term therapeutic potential of ESC-derived human striatal progenitor grafts for the replacement of degenerated striatal neurons in HD and suggest that EE can effectively accelerate the maturation and promote the integration of human striatal cells.


Assuntos
Transplante de Tecido Encefálico , Células-Tronco Embrionárias Humanas , Doença de Huntington , Ratos , Animais , Humanos , Doença de Huntington/terapia , Corpo Estriado/fisiologia , Neurônios , Modelos Animais de Doenças
2.
Methods Mol Biol ; 2655: 211-220, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37212999

RESUMO

Several studies highlighted the importance of the polycomb repressive complex 2 (PRC2) already at the beginning of development. Although the crucial function of PRC2 in regulating lineage commitment and cell-fate specification has been well-established, the in vitro study of the exact mechanisms for which H3K27me3 is indispensable for proper differentiation is still challenging. In this chapter, we report a well-established and reproducible differentiation protocol to generate striatal medium spiny neurons as a tool to explore PRC2 role in brain development.


Assuntos
Complexo Repressor Polycomb 2 , Complexo Repressor Polycomb 2/genética , Diferenciação Celular/fisiologia
3.
Int J Mol Sci ; 23(21)2022 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-36361956

RESUMO

In vitro models of corticogenesis from pluripotent stem cells (PSCs) have greatly improved our understanding of human brain development and disease. Among these, 3D cortical organoid systems are able to recapitulate some aspects of in vivo cytoarchitecture of the developing cortex. Here, we tested three cortical organoid protocols for brain regional identity, cell type specificity and neuronal maturation. Overall, all protocols gave rise to organoids that displayed a time-dependent expression of neuronal maturation genes such as those involved in the establishment of synapses and neuronal function. Comparatively, guided differentiation methods without WNT activation generated the highest degree of cortical regional identity, whereas default conditions produced the broadest range of cell types such as neurons, astrocytes and hematopoietic-lineage-derived microglia cells. These results suggest that cortical organoid models produce diverse outcomes of brain regional identity and cell type specificity and emphasize the importance of selecting the correct model for the right application.


Assuntos
Organoides , Células-Tronco Pluripotentes , Humanos , Células-Tronco Pluripotentes/metabolismo , Diferenciação Celular , Neurônios/metabolismo , Encéfalo
4.
Cell Rep Methods ; 2(12): 100367, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-36590694

RESUMO

Stem cell engineering of striatal medium spiny neurons (MSNs) is a promising strategy to understand diseases affecting the striatum and for cell-replacement therapies in different neurological diseases. Protocols to generate cells from human pluripotent stem cells (PSCs) are scarce and how well they recapitulate the endogenous fetal cells remains poorly understood. We have developed a protocol that modulates cell seeding density and exposure to specific morphogens that generates authentic and functional D1- and D2-MSNs with a high degree of reproducibility in 25 days of differentiation. Single-cell RNA sequencing (scRNA-seq) shows that our cells can mimic the cell-fate acquisition steps observed in vivo in terms of cell type composition, gene expression, and signaling pathways. Finally, by modulating the midkine pathway we show that we can increase the yield of MSNs. We expect that this protocol will help decode pathogenesis factors in striatal diseases and eventually facilitate cell-replacement therapies for Huntington's disease (HD).


Assuntos
Neurônios Espinhosos Médios , Células-Tronco Pluripotentes , Humanos , Reprodutibilidade dos Testes , Neurogênese , Corpo Estriado , Células-Tronco Pluripotentes/metabolismo
5.
Science ; 372(6542)2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33958447

RESUMO

Deciphering how the human striatum develops is necessary for understanding the diseases that affect this region. To decode the transcriptional modules that regulate this structure during development, we compiled a catalog of 1116 long intergenic noncoding RNAs (lincRNAs) identified de novo and then profiled 96,789 single cells from the early human fetal striatum. We found that D1 and D2 medium spiny neurons (D1- and D2-MSNs) arise from a common progenitor and that lineage commitment is established during the postmitotic transition, across a pre-MSN phase that exhibits a continuous spectrum of fate determinants. We then uncovered cell type-specific gene regulatory networks that we validated through in silico perturbation. Finally, we identified human-specific lincRNAs that contribute to the phylogenetic divergence of this structure in humans. This work delineates the cellular hierarchies governing MSN lineage commitment.


Assuntos
Atlas como Assunto , Corpo Estriado/citologia , Corpo Estriado/embriologia , Neurogênese/genética , RNA Longo não Codificante/genética , Análise de Célula Única , Fatores de Transcrição/genética , Feto , Neurônios GABAérgicos/metabolismo , Humanos , RNA-Seq , Transcrição Gênica
6.
Brain ; 144(10): 3175-3190, 2021 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-33974044

RESUMO

Brain cholesterol is produced mainly by astrocytes and is important for neuronal function. Its biosynthesis is severely reduced in mouse models of Huntington's disease. One possible mechanism is a diminished nuclear translocation of the transcription factor sterol regulatory element-binding protein 2 (SREBP2) and, consequently, reduced activation of SREBP2-controlled genes in the cholesterol biosynthesis pathway. Here we evaluated the efficacy of a gene therapy based on the unilateral intra-striatal injection of a recombinant adeno-associated virus 2/5 (AAV2/5) targeting astrocytes specifically and carrying the transcriptionally active N-terminal fragment of human SREBP2 (hSREBP2). Robust hSREBP2 expression in striatal glial cells in R6/2 Huntington's disease mice activated the transcription of cholesterol biosynthesis pathway genes, restored synaptic transmission, reversed dopamine receptor D2 (Drd2) transcript levels decline, cleared mutant huntingtin aggregates and attenuated behavioural deficits. We conclude that glial SREBP2 participates in Huntington's disease brain pathogenesis in vivo and that AAV-based delivery of SREBP2 to astrocytes counteracts key features of the disease.


Assuntos
Astrócitos/metabolismo , Corpo Estriado/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Doença de Huntington/terapia , Proteína de Ligação a Elemento Regulador de Esterol 2/administração & dosagem , Animais , Astrócitos/patologia , Corpo Estriado/patologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Fenótipo , Proteína de Ligação a Elemento Regulador de Esterol 2/biossíntese , Proteína de Ligação a Elemento Regulador de Esterol 2/genética
7.
Nat Rev Neurol ; 17(6): 381-392, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33658662

RESUMO

Human induced pluripotent stem cells (hiPSCs) were first generated in 2007, but the full translational potential of this valuable tool has yet to be realized. The potential applications of hiPSCs are especially relevant to neurology, as brain cells from patients are rarely available for research. hiPSCs from individuals with neuropsychiatric or neurodegenerative diseases have facilitated biological and multi-omics studies as well as large-scale screening of chemical libraries. However, researchers are struggling to improve the scalability, reproducibility and quality of this descriptive disease modelling. Addressing these limitations will be the first step towards a new era in hiPSC research - that of predictive disease modelling - involving the correlation and integration of in vitro experimental data with longitudinal clinical data. This approach is a key element of the emerging precision medicine paradigm, in which hiPSCs could become a powerful diagnostic and prognostic tool. Here, we consider the steps necessary to achieve predictive modelling of neurodegenerative disease with hiPSCs, using Huntington disease as an example.


Assuntos
Epigênese Genética/genética , Testes Genéticos/tendências , Células-Tronco Pluripotentes Induzidas/fisiologia , Doenças Neurodegenerativas/diagnóstico por imagem , Doenças Neurodegenerativas/genética , Ensaios Clínicos como Assunto/métodos , Testes Genéticos/métodos , Humanos , Doença de Huntington/diagnóstico por imagem , Doença de Huntington/genética , Doença de Huntington/terapia , Doenças Neurodegenerativas/terapia , Valor Preditivo dos Testes
8.
EMBO Mol Med ; 13(4): e12433, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33665914

RESUMO

MECP2 mutations cause Rett syndrome (RTT), a severe and progressive neurodevelopmental disorder mainly affecting females. Although RTT patients exhibit delayed onset of symptoms, several evidences demonstrate that MeCP2 deficiency alters early development of the brain. Indeed, during early maturation, Mecp2 null cortical neurons display widespread transcriptional changes, reduced activity, and defective morphology. It has been proposed that during brain development these elements are linked in a feed-forward cycle where neuronal activity drives transcriptional and morphological changes that further increase network maturity. We hypothesized that the enhancement of neuronal activity during early maturation might prevent the onset of RTT-typical molecular and cellular phenotypes. Accordingly, we show that the enhancement of excitability, obtained by adding to neuronal cultures Ampakine CX546, rescues transcription of several genes, neuronal morphology, and responsiveness to stimuli. Greater effects are achieved in response to earlier treatments. In vivo, short and early administration of CX546 to Mecp2 null mice prolongs lifespan, delays the disease progression, and rescues motor abilities and spatial memory, thus confirming the value for RTT of an early restoration of neuronal activity.


Assuntos
Proteína 2 de Ligação a Metil-CpG , Síndrome de Rett , Animais , Encéfalo/metabolismo , Feminino , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Neurônios/metabolismo , Fenótipo , Síndrome de Rett/genética
9.
Hum Mol Genet ; 30(13): 1175-1187, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33601422

RESUMO

Synaptic dysfunction and cognitive decline in Huntington's disease (HD) involve hyperactive A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10). To identify the molecular mechanisms through which ADAM10 is associated with synaptic dysfunction in HD, we performed an immunoaffinity purification-mass spectrometry (IP-MS) study of endogenous ADAM10 in the brains of wild-type and HD mice. We found that proteins implicated in synapse organization, synaptic plasticity, and vesicle and organelles trafficking interact with ADAM10, suggesting that it may act as hub protein at the excitatory synapse. Importantly, the ADAM10 interactome is enriched in presynaptic proteins and ADAM10 co-immunoprecipitates with piccolo (PCLO), a key player in the recycling and maintenance of synaptic vesicles. In contrast, reduced ADAM10/PCLO immunoprecipitation occurs in the HD brain, with decreased density of synaptic vesicles in the reserve and docked pools at the HD presynaptic terminal. Conditional heterozygous deletion of ADAM10 in the forebrain of HD mice reduces active ADAM10 to wild-type level and normalizes ADAM10/PCLO complex formation and synaptic vesicle density and distribution. The results indicate that presynaptic ADAM10 and PCLO are a relevant component of HD pathogenesis.


Assuntos
Proteína ADAM10/metabolismo , Proteínas do Citoesqueleto/metabolismo , Doença de Huntington/metabolismo , Neuropeptídeos/metabolismo , Vesículas Sinápticas/metabolismo , Proteína ADAM10/genética , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/ultraestrutura , Humanos , Doença de Huntington/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Terminações Pré-Sinápticas/metabolismo , Ligação Proteica , Mapas de Interação de Proteínas/genética , Proteômica/métodos , Vesículas Sinápticas/ultraestrutura , Sinaptossomos/metabolismo , Espectrometria de Massas em Tandem/métodos
10.
Stem Cell Res ; 49: 102016, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33039807

RESUMO

GSX2 is a homeobox transcription factor (TF) controlling the specification of the ventral lateral ganglionic eminence and its major derivative, the corpus striatum. Medium spiny neurons (MSNs) represent the largest cell component of the striatum and they are primarily affected in Huntington disease (HD). Here, we used CRISPR technology to generate a pluripotent GSX2-reporter human embryonic stem cell (hESC) line that can be leveraged to monitor striatal differentiation in real-time and to enrich for MSN-committed progenitors.


Assuntos
Células-Tronco Embrionárias Humanas , Diferenciação Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Corpo Estriado , Células-Tronco Embrionárias , Humanos , Neurônios
11.
Neurobiol Dis ; 146: 105140, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33065279

RESUMO

RUES2 cell lines represent the first collection of isogenic human embryonic stem cells (hESCs) carrying different pathological CAG lengths in the HTT gene. However, their neuronal differentiation potential has yet to be thoroughly evaluated. Here, we report that RUES2 during ventral telencephalic differentiation is biased towards medial ganglionic eminence (MGE). We also show that HD-RUES2 cells exhibit an altered MGE transcriptional signature in addition to recapitulating known HD phenotypes, with reduced expression of the neurodevelopmental regulators NEUROD1 and BDNF and increased cleavage of synaptically enriched N-cadherin. Finally, we identified the transcription factor SP1 as a common potential detrimental co-partner of muHTT by de novo motif discovery analysis on the LGE, MGE, and cortical genes differentially expressed in HD human pluripotent stem cells in our and additional datasets. Taken together, these observations suggest a broad deleterious effect of muHTT in the early phases of neuronal development that may unfold through its altered interaction with SP1.


Assuntos
Biomarcadores Tumorais/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Pluripotentes/citologia , Receptores Imunológicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/patologia , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo
12.
EMBO Mol Med ; 12(10): e12519, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32959531

RESUMO

A variety of pathophysiological mechanisms are implicated in Huntington's disease (HD). Among them, reduced cholesterol biosynthesis has been detected in the HD mouse brain from pre-symptomatic stages, leading to diminished cholesterol synthesis, particularly in the striatum. In addition, systemic injection of cholesterol-loaded brain-permeable nanoparticles ameliorates synaptic and cognitive function in a transgenic mouse model of HD. To identify an appropriate treatment regimen and gain mechanistic insights into the beneficial activity of exogenous cholesterol in the HD brain, we employed osmotic mini-pumps to infuse three escalating doses of cholesterol directly into the striatum of HD mice in a continuous and rate-controlled manner. All tested doses prevented cognitive decline, while amelioration of disease-related motor defects was dose-dependent. In parallel, we found morphological and functional recovery of synaptic transmission involving both excitatory and inhibitory synapses of striatal medium spiny neurons. The treatment also enhanced endogenous cholesterol biosynthesis and clearance of mutant Huntingtin aggregates. These results indicate that cholesterol infusion to the striatum can exert a dose-dependent, disease-modifying effect and may be therapeutically relevant in HD.


Assuntos
Doença de Huntington , Animais , Colesterol , Corpo Estriado , Modelos Animais de Doenças , Proteína Huntingtina/genética , Doença de Huntington/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Sinapses
13.
Stem Cell Reports ; 14(5): 876-891, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32302555

RESUMO

Huntington disease (HD) is an inherited late-onset neurological disorder characterized by progressive neuronal loss and disruption of cortical and basal ganglia circuits. Cell replacement using human embryonic stem cells may offer the opportunity to repair the damaged circuits and significantly ameliorate disease conditions. Here, we showed that in-vitro-differentiated human striatal progenitors undergo maturation and integrate into host circuits upon intra-striatal transplantation in a rat model of HD. By combining graft-specific immunohistochemistry, rabies virus-mediated synaptic tracing, and ex vivo electrophysiology, we showed that grafts can extend projections to the appropriate target structures, including the globus pallidus, the subthalamic nucleus, and the substantia nigra, and receive synaptic contact from both host and graft cells with 6.6 ± 1.6 inputs cell per transplanted neuron. We have also shown that transplants elicited a significant improvement in sensory-motor tasks up to 2 months post-transplant further supporting the therapeutic potential of this approach.


Assuntos
Corpo Estriado/citologia , Células-Tronco Embrionárias Humanas/transplante , Doença de Huntington/terapia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Animais , Células Cultivadas , Corpo Estriado/fisiologia , Células-Tronco Embrionárias Humanas/citologia , Humanos , Locomoção , Masculino , Células-Tronco Neurais/citologia , Neurogênese , Ratos , Regeneração , Sensação , Substância Negra/citologia , Substância Negra/fisiologia , Núcleo Subtalâmico/citologia , Núcleo Subtalâmico/fisiologia , Sinapses/metabolismo , Sinapses/fisiologia
14.
Mol Cell ; 78(2): 346-358.e9, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32268123

RESUMO

CAG-repeat expansions in at least eight different genes cause neurodegeneration. The length of the extended polyglutamine stretches in the corresponding proteins is proportionally related to their aggregation propensity. Although these proteins are ubiquitously expressed, they predominantly cause toxicity to neurons. To understand this neuronal hypersensitivity, we generated induced pluripotent stem cell (iPSC) lines of spinocerebellar ataxia type 3 and Huntington's disease patients. iPSC generation and neuronal differentiation are unaffected by polyglutamine proteins and show no spontaneous aggregate formation. However, upon glutamate treatment, aggregates form in neurons but not in patient-derived neural progenitors. During differentiation, the chaperone network is drastically rewired, including loss of expression of the anti-amyloidogenic chaperone DNAJB6. Upregulation of DNAJB6 in neurons antagonizes glutamate-induced aggregation, while knockdown of DNAJB6 in progenitors results in spontaneous polyglutamine aggregation. Loss of DNAJB6 expression upon differentiation is confirmed in vivo, explaining why stem cells are intrinsically protected against amyloidogenesis and protein aggregates are dominantly present in neurons.


Assuntos
Proteínas Amiloidogênicas/genética , Diferenciação Celular/genética , Proteínas de Choque Térmico HSP40/genética , Chaperonas Moleculares/genética , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/metabolismo , Regulação da Expressão Gênica/genética , Técnicas de Inativação de Genes , Ácido Glutâmico/metabolismo , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Doença de Machado-Joseph/genética , Doença de Machado-Joseph/metabolismo , Doença de Machado-Joseph/patologia , Células-Tronco Neurais/patologia , Neurônios/metabolismo , Neurônios/patologia , Agregados Proteicos/genética , Expansão das Repetições de Trinucleotídeos/genética
15.
Stem Cell Reports ; 13(5): 847-861, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31607568

RESUMO

The regulation of the proliferation and polarity of neural progenitors is crucial for the development of the brain cortex. Animal studies have implicated glycogen synthase kinase 3 (GSK3) as a pivotal regulator of both proliferation and polarity, yet the functional relevance of its signaling for the unique features of human corticogenesis remains to be elucidated. We harnessed human cortical brain organoids to probe the longitudinal impact of GSK3 inhibition through multiple developmental stages. Chronic GSK3 inhibition increased the proliferation of neural progenitors and caused massive derangement of cortical tissue architecture. Single-cell transcriptome profiling revealed a direct impact on early neurogenesis and uncovered a selective role of GSK3 in the regulation of glutamatergic lineages and outer radial glia output. Our dissection of the GSK3-dependent transcriptional network in human corticogenesis underscores the robustness of the programs determining neuronal identity independent of tissue architecture.


Assuntos
Córtex Cerebral/citologia , Quinase 3 da Glicogênio Sintase/metabolismo , Neurogênese , Neurônios/citologia , Organoides/citologia , Linhagem Celular , Proliferação de Células , Córtex Cerebral/metabolismo , Deleção de Genes , Quinase 3 da Glicogênio Sintase/genética , Humanos , Neurônios/metabolismo , Organoides/metabolismo , Transcriptoma
16.
J Clin Invest ; 129(6): 2390-2403, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-31063986

RESUMO

A disintegrine and metalloproteinase 10 (ADAM10) is implicated in synaptic function through its interaction with postsynaptic receptors and adhesion molecules. Here, we report that levels of active ADAM10 are increased in Huntington's disease (HD) mouse cortices and striata and in human postmortem caudate. We show that, in the presence of polyglutamine-expanded (polyQ-expanded) huntingtin (HTT), ADAM10 accumulates at the postsynaptic densities (PSDs) and causes excessive cleavage of the synaptic protein N-cadherin (N-CAD). This aberrant phenotype is also detected in neurons from HD patients where it can be reverted by selective silencing of mutant HTT. Consistently, ex vivo delivery of an ADAM10 synthetic inhibitor reduces N-CAD proteolysis and corrects electrophysiological alterations in striatal medium-sized spiny neurons (MSNs) of 2 HD mouse models. Moreover, we show that heterozygous conditional deletion of ADAM10 or delivery of a competitive TAT-Pro-ADAM10709-729 peptide in R6/2 mice prevents N-CAD proteolysis and ameliorates cognitive deficits in the mice. Reduction in synapse loss was also found in R6/2 mice conditionally deleted for ADAM10. Taken together, these results point to a detrimental role of hyperactive ADAM10 at the HD synapse and provide preclinical evidence of the therapeutic potential of ADAM10 inhibition in HD.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Disfunção Cognitiva/enzimologia , Doença de Huntington/enzimologia , Proteínas de Membrana/metabolismo , Densidade Pós-Sináptica/enzimologia , Proteína ADAM10/genética , Adulto , Idoso , Secretases da Proteína Precursora do Amiloide/genética , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Doença de Huntington/genética , Doença de Huntington/patologia , Masculino , Proteínas de Membrana/genética , Camundongos Transgênicos , Pessoa de Meia-Idade , Densidade Pós-Sináptica/genética , Densidade Pós-Sináptica/patologia
17.
Proc Natl Acad Sci U S A ; 114(50): E10809-E10818, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29162692

RESUMO

Posttranslational modifications can have profound effects on the biological and biophysical properties of proteins associated with misfolding and aggregation. However, their detection and quantification in clinical samples and an understanding of the mechanisms underlying the pathological properties of misfolding- and aggregation-prone proteins remain a challenge for diagnostics and therapeutics development. We have applied an ultrasensitive immunoassay platform to develop and validate a quantitative assay for detecting a posttranslational modification (phosphorylation at residue T3) of a protein associated with polyglutamine repeat expansion, namely Huntingtin, and characterized its presence in a variety of preclinical and clinical samples. We find that T3 phosphorylation is greatly reduced in samples from Huntington's disease models and in Huntington's disease patients, and we provide evidence that bona-fide T3 phosphorylation alters Huntingtin exon 1 protein conformation and aggregation properties. These findings have significant implications for both mechanisms of disease pathogenesis and the development of therapeutics and diagnostics for Huntington's disease.


Assuntos
Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Imunoensaio/métodos , Animais , Células Cultivadas , Éxons , Células HEK293 , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilação , Conformação Proteica , Processamento de Proteína Pós-Traducional , Sensibilidade e Especificidade
18.
Mol Cell Neurosci ; 82: 46-57, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28476540

RESUMO

DNA methylation (DNAm) changes are of increasing relevance to neurodegenerative disorders, including Huntington's disease (HD). We performed genome-wide screening of possible DNAm changes occurring during striatal differentiation in human induced pluripotent stem cells derived from a HD patient (HD-hiPSCs) as cellular model. We identified 240 differentially methylated regions (DMRs) at promoters in fully differentiated HD-hiPSCs. Subsequently, we focused on the methylation differences in a subcluster of genes related to Jumonji Domain Containing 3 (JMJD3), a demethylase that epigenetically regulates neuronal differentiation and activates neuronal progenitor associated genes, which are indispensable for neuronal fate acquisition. Noticeably among these genes, WD repeat-containing protein 5 (WDR5) promoter was found hypermethylated in HD-hiPSCs, resulting in a significant down-modulation in its expression and of the encoded protein. A similar WDR5 expression decrease was seen in a small series of HD-hiPSC lines characterized by different CAG length. The decrease in WDR5 expression was particularly evident in HD-hiPSCs compared to hESCs and control-hiPSCs from healthy subjects. WDR5 is a core component of the MLL/SET1 chromatin remodeling complexes essential for H3K4me3, previously reported to play an important role in stem cells self-renewal and differentiation. These results suggest the existence of epigenetic mechanisms in HD and the identification of genes, which are able to modulate HD phenotype, is important both for biomarker discovery and therapeutic interventions.


Assuntos
Diferenciação Celular/genética , Epigênese Genética/genética , Histona-Lisina N-Metiltransferase/genética , Doença de Huntington/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Linhagem Celular , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina/genética , Humanos , Doença de Huntington/genética , Peptídeos e Proteínas de Sinalização Intracelular , Neurônios/metabolismo
19.
Stem Cells Dev ; 24(20): 2366-77, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26132372

RESUMO

The potential use of human embryonic stem cells (hESCs) in cell-based therapies points out the critical importance of epigenomic evaluation for cell-based therapies. Specifically, DNA methylation appears to be a crucial player in establishing cell fate commitment and lineage choices. In this study, we report the global changes observed on the CpG islands distributed in promoters, gene bodies, and intergenic regions and the major biochemical pathways and genes involved in methylation changes as H9-hESCs turn into a neuronal culture containing medium-sized spiny striatal neurons (MSNs). Using an ontogeny-recapitulating protocol of striatal neuron differentiation, we analyzed DNA methylation profiles during the conversion from pluripotency to neuropotency up to the acquisition of a mature neuronal phenotype. H9-hESCs changed the methylation pattern both through de novo methylation and hypomethylation of specific gene promoters. Bioinformatic analysis allowed us to identify a panel of striatal-associated genes, which were regulated by DNA methylation and differentially expressed during striatal commitment. Importantly, DNA methylation analysis revealed that H9-hESCs did not acquire methylation-based oncogenic properties after differentiation. Indeed, hypermethylation of cancer-associated genes that characterize transformed cells, such as Polycomb repressive complex-associated genes, was not detected in the neuronal cultures. However, the oncosuppressor gene, BCL2L11, became hypermethylated in H9-hESC-derived mature neurons. Whole-genome DNA methylation profiling could become a technological platform to predict the differentiative potential of hESC-derived cultures and establish further biosafety assessment quality control tools of the cell-based products.


Assuntos
Diferenciação Celular/genética , Metilação de DNA , Células-Tronco Embrionárias Humanas/citologia , Neurônios/citologia , Linhagem Celular , Corpo Estriado/citologia , Humanos , Fenótipo , Regiões Promotoras Genéticas/genética
20.
Neuropathol Appl Neurobiol ; 40(7): 899-910, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24634989

RESUMO

AIMS: The repressor element-1 silencing transcription factor/neurone-restrictive silencer factor (REST/NRSF) is a master regulator of neuronal gene expression. REST/NRSF functions by recruiting other cofactors to genomic loci that contain the repressor element 1/neurone restrictive silencer element (RE1/NRSE) binding motif. In brain, demonstration of REST protein presence in neurones has remained controversial. However, RE1/NRSE containing neuronal genes are actively modulated and REST dysregulation is implicated in Huntington's disease (HD). We aimed to investigate REST distribution in autopsy brain from control and HD patients. METHODS: Brain tissues from six controls and six HD cases (Vonsattel grade 3 and 4) were investigated using immunohistochemical analysis. RESULTS: REST was present in neurones and glial cells of the cortex, caudate nucleus, hippocampus and cerebellum. REST labelling was mainly cytoplasmic in neurones while preferential nuclear staining of REST was found in glial cells. We also found that REST and huntingtin (HTT) colocalize in human neurones. Low levels of cytoplasmic REST were detected in neurones of the HD cortex and caudate but no direct relationship between decreased neuronal REST expression and disease grade was observed. CONCLUSIONS: These data support the notion of REST presence in human brain neurones and glial cells and indicate the importance of developing compounds able to restore REST-regulated transcription of neuronal genes in HD.


Assuntos
Encéfalo/metabolismo , Doença de Huntington/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/análise , Adulto , Idoso , Feminino , Humanos , Proteína Huntingtina , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...