Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(32): e2204779119, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35914128

RESUMEN

Earlier work has shown that siRNA-mediated reduction of the SUPT4H or SUPT5H proteins, which interact to form the DSIF complex and facilitate transcript elongation by RNA polymerase II (RNAPII), can decrease expression of mutant gene alleles containing nucleotide repeat expansions differentially. Using luminescence and fluorescence assays, we identified chemical compounds that interfere with the SUPT4H-SUPT5H interaction and then investigated their effects on synthesis of mRNA and protein encoded by mutant alleles containing repeat expansions in the huntingtin gene (HTT), which causes the inherited neurodegenerative disorder, Huntington's Disease (HD). Here we report that such chemical interference can differentially affect expression of HTT mutant alleles, and that a prototypical chemical, 6-azauridine (6-AZA), that targets the SUPT4H-SUPT5H interaction can modify the biological response to mutant HTT gene expression. Selective and dose-dependent effects of 6-AZA on expression of HTT alleles containing nucleotide repeat expansions were seen in multiple types of cells cultured in vitro, and in a Drosophila melanogaster animal model for HD. Lowering of mutant HD protein and mitigation of the Drosophila "rough eye" phenotype associated with degeneration of photoreceptor neurons in vivo were observed. Our findings indicate that chemical interference with DSIF complex formation can decrease biochemical and phenotypic effects of nucleotide repeat expansions.


Asunto(s)
Azauridina , Proteína Huntingtina , Enfermedad de Huntington , Proteínas Mutantes , Mutación , Proteínas Nucleares , Fenotipo , Proteínas Represoras , Factores de Elongación Transcripcional , Alelos , Animales , Azauridina/farmacología , Células Cultivadas , Expansión de las Repeticiones de ADN , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Mediciones Luminiscentes , Proteínas Mutantes/biosíntesis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Células Fotorreceptoras de Invertebrados/efectos de los fármacos , Proteínas Represoras/metabolismo , Factores de Elongación Transcripcional/metabolismo
2.
Nucleic Acids Res ; 49(20): 11883-11899, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34718744

RESUMEN

In neurodegenerative diseases, including pathologies with well-known causative alleles, genetic factors that modify severity or age of onset are not entirely understood. We recently documented the unexpected prevalence of transfer RNA (tRNA) mutants in the human population, including variants that cause amino acid mis-incorporation. We hypothesized that a mistranslating tRNA will exacerbate toxicity and modify the molecular pathology of Huntington's disease-causing alleles. We characterized a tRNAPro mutant that mistranslates proline codons with alanine, and tRNASer mutants, including a tRNASerAGA G35A variant with a phenylalanine anticodon (tRNASerAAA) found in ∼2% of the population. The tRNAPro mutant caused synthetic toxicity with a deleterious huntingtin poly-glutamine (polyQ) allele in neuronal cells. The tRNASerAAA variant showed synthetic toxicity with proteasome inhibition but did not enhance toxicity of the huntingtin allele. Cells mistranslating phenylalanine or proline codons with serine had significantly reduced rates of protein synthesis. Mistranslating cells were slow but effective in forming insoluble polyQ aggregates, defective in protein and aggregate degradation, and resistant to the neuroprotective integrated stress response inhibitor (ISRIB). Our findings identify mistranslating tRNA variants as genetic factors that slow protein aggregation kinetics, inhibit aggregate clearance, and increase drug resistance in cellular models of neurodegenerative disease.


Asunto(s)
Proteína Huntingtina/biosíntesis , Enfermedad de Huntington/genética , ARN de Transferencia de Prolina/genética , Acetamidas/farmacología , Animales , Línea Celular Tumoral , Codón/genética , Ciclohexilaminas/farmacología , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Mutación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Células PC12 , Péptidos/toxicidad , Proteolisis , ARN de Transferencia de Prolina/metabolismo , Ratas
3.
Mol Neurobiol ; 58(8): 3992-4006, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33904021

RESUMEN

Impairment of proteostasis network is one of the characteristic features of many age-related neurodegenerative disorders including autosomal dominantly inherited Huntington's disease (HD). In HD, N-terminal portion of mutant huntingtin protein containing expanded polyglutamine repeats accumulates as inclusion bodies and leads to progressive deterioration of various cellular functioning including proteostasis network. Here we report that Withaferin A (a small bioactive molecule derived from Indian medicinal plant, Withania somnifera) partially rescues defective proteostasis by activating heat shock response (HSR) and delays the disease progression in a HD mouse model. Exposure of Withaferin A activates HSF1 and induces the expression of HSP70 chaperones in an in vitro cell culture system and also suppresses mutant huntingtin aggregation in a cellular model of HD. Withaferin A treatment to HD mice considerably increased their lifespan as well as restored progressive motor behavioral deficits and declined body weight. Biochemical studies confirmed the activation of HSR and global decrease in mutant huntingtin aggregates load accompanied with improvement of striatal function in Withaferin A-treated HD mouse brain. Withaferin A-treated HD mice also exhibit significant decrease in inflammatory processes as evident from the decreased microglial activation. These results indicate immense potential of Withaferin A for the treatment of HD and related neurodegenerative disorders involving protein misfolding and aggregation.


Asunto(s)
Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas HSP70 de Choque Térmico/biosíntesis , Enfermedad de Huntington/metabolismo , Witanólidos/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Proteínas HSP70 de Choque Térmico/genética , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Ratones , Ratones Endogámicos CBA , Ratones Transgénicos , Witanólidos/farmacología
4.
J Med Chem ; 64(2): 941-957, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33185430

RESUMEN

GPR52 is an orphan G protein-coupled receptor (GPCR) that has been recently implicated as a potential drug target of Huntington's disease (HD), an incurable monogenic neurodegenerative disorder. In this research, we found that striatal knockdown of GPR52 reduces mHTT levels in adult HdhQ140 mice, validating GPR52 as an HD target. In addition, we discovered a highly potent and specific GPR52 antagonist Comp-43 with an IC50 value of 0.63 µM by a structure-activity relationship (SAR) study. Further studies showed that Comp-43 reduces mHTT levels by targeting GPR52 and promotes survival of mouse primary striatal neurons. Moreover, in vivo study showed that Comp-43 not only reduces mHTT levels but also rescues HD-related phenotypes in HdhQ140 mice. Taken together, our study confirms that inhibition of GPR52 is a promising strategy for HD therapy, and the GPR52 antagonist Comp-43 might serve as a lead compound for further investigation.


Asunto(s)
Proteína Huntingtina/biosíntesis , Enfermedad de Huntington/tratamiento farmacológico , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Diseño de Fármacos , Humanos , Enfermedad de Huntington/fisiopatología , Enfermedad de Huntington/psicología , Ratones , Modelos Moleculares , Neuronas/efectos de los fármacos , Relación Estructura-Actividad
5.
J Neuroinflammation ; 17(1): 290, 2020 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-33023623

RESUMEN

BACKGROUND: Evidence shows significant heterogeneity in astrocyte gene expression and function. We previously demonstrated that brain-derived neurotrophic factor (BDNF) exerts protective effects on whole brain primary cultured rat astrocytes treated with 3-nitropropionic acid (3NP), a mitochondrial toxin widely used as an in vitro model of Huntington's disease (HD). Therefore, we now investigated 3NP and BDNF effects on astrocytes from two areas involved in HD: the striatum and the entire cortex, and their involvement in neuron survival. METHODS: We prepared primary cultured rat cortical or striatal astrocytes and treated them with BDNF and/or 3NP for 24 h. In these cells, we assessed expression of astrocyte markers, BDNF receptor, and glutamate transporters, and cytokine release. We prepared astrocyte-conditioned medium (ACM) from cortical and striatal astrocytes and tested its effect on a cellular model of HD. RESULTS: BDNF protected astrocytes from 3NP-induced death, increased expression of its own receptor, and activation of ERK in both cortical and striatal astrocytes. However, BDNF modulated glutamate transporter expression differently by increasing GLT1 and GLAST expression in cortical astrocytes but only GLT1 expression in striatal astrocytes. Striatal astrocytes released higher amounts of tumor necrosis factor-α than cortical astrocytes in response to 3NP but BDNF decreased this effect in both populations. 3NP decreased transforming growth factor-ß release only in cortical astrocytes, whereas BDNF treatment increased its release only in striatal astrocytes. Finally, we evaluated ACM effect on a cellular model of HD: the rat striatal neuron cell line ST14A expressing mutant human huntingtin (Q120) or in ST14A cells expressing normal human huntingtin (Q15). Neither striatal nor cortical ACM modified the viability of Q15 cells. Only ACM from striatal astrocytes treated with BDNF and ACM from 3NP + BDNF-treated striatal astrocytes protected Q120 cells, whereas ACM from cortical astrocytes did not. CONCLUSIONS: Data suggest that cortical and striatal astrocytes respond differently to mitochondrial toxin 3NP and BDNF. Moreover, striatal astrocytes secrete soluble neuroprotective factors in response to BDNF that selectively protect neurons expressing mutant huntingtin implicating that BDNF modulation of striatal astrocyte function has therapeutic potential against neurodegeneration.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/toxicidad , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Proteína Huntingtina/biosíntesis , Nitrocompuestos/toxicidad , Propionatos/toxicidad , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Proteína Huntingtina/genética , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación/efectos de los fármacos , Mutación/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuroprotección/efectos de los fármacos , Neuroprotección/fisiología , Ratas , Ratas Wistar
6.
PLoS One ; 15(7): e0233583, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32735619

RESUMEN

Mutations that cause Huntington's Disease involve a polyglutamine (polyQ) sequence expansion beyond 35 repeats in exon 1 of Huntingtin. Intracellular inclusion bodies of mutant Huntingtin protein are a key feature of Huntington's disease brain pathology. We previously showed that in cell culture the formation of inclusions involved the assembly of disordered structures of mHtt exon 1 fragments (Httex1) and they were enriched with translational machinery when first formed. We hypothesized that nascent mutant Httex1 chains co-aggregate during translation by phase separation into liquid-like disordered aggregates and then convert to more rigid, amyloid structures. Here we further examined the mechanisms of inclusion assembly in a human epithelial kidney (AD293) cell culture model. We found mHttex1 did not appear to stall translation of its own nascent chain, or at best was marginal. We also found the inclusions appeared to recruit low levels of RNA but there was no difference in enrichment between early formed and mature inclusions. Proteins involved in translation or ribosome quality control were co-recruited to the inclusions (Ltn1 Rack1) compared to a protein not anticipated to be involved (NACAD), but there was no major specificity of enrichment in the early formed inclusions compared to mature inclusions. Furthermore, we observed co-aggregation with other proteins previously identified in inclusions, including Upf1 and chaperone-like proteins Sgta and Hspb1, which also suppressed aggregation at high co-expression levels. The newly formed inclusions also contained immobile mHttex1 molecules which points to the disordered aggregates being mechanically rigid prior to amyloid formation. Collectively our findings show little evidence that inclusion assembly arises by a discrete clustering of stalled nascent chains and associated quality control machinery. Instead, the machinery appear to be recruited continuously, or secondarily, to the nucleation of inclusion formation.


Asunto(s)
Exones/genética , Proteína Huntingtina/genética , Extensión de la Cadena Peptídica de Translación , Agregado de Proteínas/genética , ARN Mensajero/genética , Ribosomas/metabolismo , Secuencia de Bases , Células Epiteliales , Genes Reporteros , Células HEK293 , Humanos , Proteína Huntingtina/biosíntesis , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Repeticiones de Minisatélite , Péptidos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
7.
Nanomedicine ; 24: 102119, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31666200

RESUMEN

Therapies to lower gene expression in brain disease currently require chronic administration into the cerebrospinal fluid (CSF) by intrathecal infusions or direct intracerebral injections. Though well-tolerated in the short-term, this approach is not tenable for a life-time of administration. Nose-to-brain delivery of enriched chitosan-based nanoparticles loaded with anti-HTT siRNA was studied in a transgenic YAC128 mouse model of Huntington's Disease (HD). A series of chitosan-based nanoparticle (NP) formulations encapsulating anti-HTT small interfering RNA (siRNA) was designed to protect the payload from degradation "en route" to the target. Factors to improve production of effective nanocarriers of anti-HTT siRNA were identified and tested in a YAC128 mouse model of Huntington's disease. Four formulations of nanocarriers were identified to be effective in lowering HTT mRNA expression by at least 50%. Intranasal administration of nanoparticles carrying siRNA is a promising therapeutic alternative for safe and effective lowering of mutant HTT expression.


Asunto(s)
Quitosano , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Huntingtina , Enfermedad de Huntington , Nanopartículas , ARN Interferente Pequeño , Administración Intranasal , Animales , Quitosano/química , Quitosano/farmacología , Modelos Animales de Enfermedad , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Nanopartículas/química , Nanopartículas/uso terapéutico , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
8.
Sci Rep ; 9(1): 16137, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31695145

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a CAG repeat expansion within exon 1 of the huntingtin (HTT) gene. HTT mRNA contains 67 exons and does not always splice between exon 1 and exon 2 leading to the production of a small polyadenylated HTTexon1 transcript, and the full-length HTT mRNA has three 3'UTR isoforms. We have developed a QuantiGene multiplex panel for the simultaneous detection of all of these mouse Htt transcripts directly from tissue lysates and demonstrate that this can replace the more work-intensive Taqman qPCR assays. We have applied this to the analysis of brain regions from the zQ175 HD mouse model and wild type littermates at two months of age. We show that the incomplete splicing of Htt occurs throughout the brain and confirm that this originates from the mutant and not endogenous Htt allele. Given that HTTexon1 encodes the highly pathogenic exon 1 HTT protein, it is essential that the levels of all Htt transcripts can be monitored when evaluating HTT lowering approaches. Our QuantiGene panel will allow the rapid comparative assessment of all Htt transcripts in cell lysates and mouse tissues without the need to first extract RNA.


Asunto(s)
Encéfalo/metabolismo , Ensayo de Amplificación de Señal de ADN Ramificado/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteína Huntingtina/genética , Proteínas del Tejido Nervioso/genética , Empalme del ARN , Regiones no Traducidas 3'/genética , Animales , Modelos Animales de Enfermedad , Exones/genética , Proteína Huntingtina/biosíntesis , Intrones/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Proteínas del Tejido Nervioso/biosíntesis , Especificidad de Órganos , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Mol Cell ; 76(4): 617-631.e4, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31564557

RESUMEN

Spt5 is a conserved and essential transcription elongation factor that promotes promoter-proximal pausing, promoter escape, elongation, and mRNA processing. Spt5 plays specific roles in the transcription of inflammation and stress-induced genes and tri-nucleotide expanded-repeat genes involved in inherited neurological pathologies. Here, we report the identification of Spt5-Pol II small-molecule inhibitors (SPIs). SPIs faithfully reproduced Spt5 knockdown effects on promoter-proximal pausing, NF-κB activation, and expanded-repeat huntingtin gene transcription. Using SPIs, we identified Spt5 target genes that responded with profoundly diverse kinetics. SPIs uncovered the regulatory role of Spt5 in metabolism via GDF15, a food intake- and body weight-inhibitory hormone. SPIs further unveiled a role for Spt5 in promoting the 3' end processing of histone genes. While several SPIs affect all Spt5 functions, a few inhibit a single one, implying uncoupling and selective targeting of Spt5 activities. SPIs expand the understanding of Spt5-Pol II functions and are potential drugs against metabolic and neurodegenerative diseases.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , ARN Polimerasa II/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Factores de Elongación Transcripcional/antagonistas & inhibidores , Regiones no Traducidas 3' , Animales , Núcleo Celular/enzimología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Descubrimiento de Drogas/métodos , Metabolismo Energético/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Histonas/genética , Histonas/metabolismo , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Células Jurkat , Células MCF-7 , Ratones Transgénicos , Mutación , FN-kappa B/biosíntesis , FN-kappa B/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Polimerasa II/genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
10.
J Neurochem ; 147(4): 454-476, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30182387

RESUMEN

As the peripheral sympathoadrenal axis is tightly controlled by the cortex via hypothalamus and brain stem, the central pathological features of Hunting's disease, (HD) that is, deposition of mutated huntingtin and synaptic dysfunctions, could also be expressed in adrenal chromaffin cells. To test this hypothesis we here present a thorough investigation on the pathological and functional changes undergone by chromaffin cells (CCs) from 2-month (2 m) to 7-month (7 m) aged wild-type (WT) and R6/1 mouse model of Huntington's disease (HD), stimulated with acetylcholine (ACh) or high [K+ ] (K+ ). In order to do this, we used different techniques such as inmunohistochemistry, patch-clamp, and amperometric recording. With respect to WT cells, some of the changes next summarized were already observed in HD mice at a pre-disease stage (2 m); however, they were more pronounced at 7 m when motor deficits were clearly established, as follows: (i) huntingtin over-expression as nuclear aggregates in CCs; (ii) smaller CC size with decreased dopamine ß-hydroxylase expression, indicating lesser number of chromaffin secretory vesicles; (iii) reduced adrenal tissue catecholamine content; (iv) reduced Na+ currents with (v) membrane hyperpolarization and reduced ACh-evoked action potentials; (v) reduced [Ca2+ ]c transients with faster Ca2+ clearance; (vi) diminished quantal secretion with smaller vesicle quantal size; (vii) faster kinetics of the exocytotic fusion pore, pore expansion, and closure. On the basis of these data, the hypothesis is here raised in the sense that nuclear deposition of mutated huntingtin in adrenal CCs of R6/1 mice could be primarily responsible for poorer Na+ channel expression and function, giving rise to profound depression of cell excitability, altered Ca2+ handling and exocytosis. OPEN PRACTICES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14201.


Asunto(s)
Células Cromafines/metabolismo , Células Cromafines/patología , Exocitosis , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Médula Suprarrenal/metabolismo , Médula Suprarrenal/patología , Animales , Catecolaminas/metabolismo , Humanos , Enfermedad de Huntington/psicología , Cinética , Masculino , Potenciales de la Membrana , Ratones , Ratones Transgénicos , Trastornos del Movimiento/etiología , Trastornos del Movimiento/fisiopatología , Mutación/genética , Desempeño Psicomotor , Canales de Sodio/biosíntesis , Vesículas Sinápticas/patología
12.
Cell Rep ; 23(4): 1020-1033, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29694882

RESUMEN

Huntington's disease (HD) is a dominantly inherited neurological disorder caused by CAG-repeat expansion in exon 1 of Huntingtin (HTT). But in addition to the neurological disease, mutant HTT (mHTT), which is ubiquitously expressed, impairs other organ systems. Indeed, epidemiological and animal model studies suggest higher incidence of and mortality from heart disease in HD. Here, we show that the protein complex mTORC1 is dysregulated in two HD mouse models through a mechanism that requires intrinsic mHTT expression. Moreover, restoring cardiac mTORC1 activity with constitutively active Rheb prevents mortality and relieves the mHTT-induced block to hypertrophic adaptation to cardiac stress. Finally, we show that chronic mTORC1 dysregulation is due in part to mislocalization of endogenous Rheb. These data provide insight into the increased cardiac-related mortality of HD patients, with cardiac mHTT expression inhibiting mTORC1 activity, limiting heart growth, and decreasing the heart's ability to compensate to chronic stress.


Asunto(s)
Proteína Huntingtina/biosíntesis , Enfermedad de Huntington/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Miocardio/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Animales , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Mutantes , Miocardio/patología , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética
14.
Nat Neurosci ; 21(3): 341-352, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29403030

RESUMEN

In Huntington's disease (HD), expansion of CAG codons in the huntingtin gene (HTT) leads to the aberrant formation of protein aggregates and the differential degeneration of striatal medium spiny neurons (MSNs). Modeling HD using patient-specific MSNs has been challenging, as neurons differentiated from induced pluripotent stem cells are free of aggregates and lack an overt cell death phenotype. Here we generated MSNs from HD patient fibroblasts through microRNA-based direct neuronal conversion, bypassing the induction of pluripotency and retaining age signatures of the original fibroblasts. We found that patient MSNs consistently exhibited mutant HTT (mHTT) aggregates, mHTT-dependent DNA damage, mitochondrial dysfunction and spontaneous degeneration in culture over time. We further provide evidence that erasure of age stored in starting fibroblasts or neuronal conversion of presymptomatic HD patient fibroblasts results in differential manifestation of cellular phenotypes associated with HD, highlighting the importance of age in modeling late-onset neurological disorders.


Asunto(s)
Envejecimiento/patología , Fibroblastos/patología , Enfermedad de Huntington/patología , Neostriado/patología , Neuronas/patología , Daño del ADN , Potenciales Postsinápticos Excitadores/genética , Perfilación de la Expresión Génica , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , MicroARNs/genética , Enfermedades Mitocondriales/patología , Enfermedades Neurodegenerativas/patología , Estrés Oxidativo , Fenotipo , Células Madre Pluripotentes
15.
Neurochem Int ; 117: 174-187, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28532681

RESUMEN

Parkinsonian-like motor deficits in Huntington's Disease (HD) patients are associated with abnormal dopamine neurotransmission in the striatum. Dopamine metabolism leads to the formation of oxidized dopamine quinones that exacerbates mitochondrial dysfunction with production of reactive oxygen species (ROS) that eventually lead to neuronal cell death. We have previously shown that dopamine-induced oxidative stress triggers apoptotic cell death in dopaminergic neuroblastoma SH-SY5Y cells hyper-expressing the mutant polyQ Huntingtin (polyQ-Htt) protein. Dopamine toxicity was paralleled by impaired autophagy clearance of the polyQ-Htt aggregates. In this study, we found that Dopamine affects the stability and function of ATG4, a redox-sensitive cysteine-protein involved in the processing of LC3, a key step in the formation of autophagosomes. Resveratrol, a dietary polyphenol with anti-oxidant and pro-autophagic properties, has shown neuroprotective potential in HD. Yet the molecular mechanism through which Resveratrol can protect HD cells against DA is not known. Here, we show that Resveratrol prevents the generation of ROS, restores the level of ATG4, allows the lipidation of LC3, facilitates the degradation of polyQ-Htt aggregates and protects the cells from Dopamine toxicity. The present findings provide a mechanistic explanation of the neuroprotective activity of Resveratrol and support its inclusion in a therapeutic regimen to slow down HD progression.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Dopamina/toxicidad , Proteína Huntingtina/biosíntesis , Fármacos Neuroprotectores/farmacología , Fagosomas/efectos de los fármacos , Resveratrol/farmacología , Antioxidantes/farmacología , Autofagia/fisiología , Línea Celular Tumoral , Humanos , Proteína Huntingtina/genética , Mutación/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fagosomas/metabolismo , Fagosomas/patología
16.
Ann Neurol ; 82(6): 981-994, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29171910

RESUMEN

OBJECTIVE: The aim of the study was to assess the distribution, frequency, and specific location of mutant huntingtin protein (mHTT) aggregates-the pathological hallmark of Huntington disease (HD)-within the various compartments of the spinal cord and their potential impact on the local vasculature and blood-spinal cord barrier (BSCB). METHODS: We performed a series of postmortem immunohistochemical and immunofluorescent stainings, as well as Western blot analyses, on cervical and lumbar sections of the spinal cord in patients diagnosed with HD (n = 11 of all grades of disease severity) along with sex- and age-matched healthy controls (n = 9). RESULTS: We observed that mHTT was preferably expressed within the anterior horn of the gray matter, in both cervical and lumbar sections. At the cellular level, mHTT aggregates were more often encountered in the extracellular matrix but could also be observed within cell bodies and neurites as well as within the endothelium of blood vessels with an increase in the density of small blood vessels in cervical sections of HD cases. These vasculature changes were accompanied with features of BSCB leakage, as assessed by the presence of increased levels of fibrinogen in the surrounding parenchyma and enhanced leukocyte infiltration. INTERPRETATION: This alteration in BSCB integrity may be explained, in part, by the dysregulation we found in some of the main proteins associated with it such as junctional adhesion molecule-1 and vascular endothelial cadherin. These observations have important implications for our understanding of HD pathology and may also have significant therapeutic implications. Ann Neurol 2017;82:981-994.


Asunto(s)
Barrera Hematoencefálica/patología , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas Mutantes/genética , Médula Espinal/patología , Adulto , Anciano , Anciano de 80 o más Años , Barrera Hematoencefálica/metabolismo , Femenino , Expresión Génica , Humanos , Proteína Huntingtina/biosíntesis , Enfermedad de Huntington/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Mutantes/biosíntesis , Médula Espinal/metabolismo
17.
J Clin Invest ; 127(7): 2719-2724, 2017 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-28628038

RESUMEN

Huntington's disease is a neurodegenerative disorder caused by a polyglutamine repeat in the Huntingtin gene (HTT). Although suppressing the expression of mutant HTT (mHTT) has been explored as a therapeutic strategy to treat Huntington's disease, considerable efforts have gone into developing allele-specific suppression of mHTT expression, given that loss of Htt in mice can lead to embryonic lethality. It remains unknown whether depletion of HTT in the adult brain, regardless of its allele, could be a safe therapy. Here, we report that permanent suppression of endogenous mHTT expression in the striatum of mHTT-expressing mice (HD140Q-knockin mice) using CRISPR/Cas9-mediated inactivation effectively depleted HTT aggregates and attenuated early neuropathology. The reduction of mHTT expression in striatal neuronal cells in adult HD140Q-knockin mice did not affect viability, but alleviated motor deficits. Our studies suggest that non-allele-specific CRISPR/Cas9-mediated gene editing could be used to efficiently and permanently eliminate polyglutamine expansion-mediated neuronal toxicity in the adult brain.


Asunto(s)
Alelos , Sistemas CRISPR-Cas , Edición Génica , Regulación de la Expresión Génica , Proteína Huntingtina , Enfermedad de Huntington , Animales , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/terapia , Ratones , Ratones Mutantes , Péptidos/genética , Péptidos/metabolismo
18.
Hum Mol Genet ; 26(5): 913-922, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334820

RESUMEN

Huntington's disease is a dominantly inherited neurodegenerative disease caused by the expansion of a CAG repeat in the HTT gene. In addition to the length of the CAG expansion, factors such as genetic background have been shown to contribute to the age at onset of neurological symptoms. A central challenge in understanding the disease progression that leads from the HD mutation to massive cell death in the striatum is the ability to characterize the subtle and early functional consequences of the CAG expansion longitudinally. We used dense time course sampling between 4 and 20 postnatal weeks to characterize early transcriptomic, molecular and cellular phenotypes in the striatum of six distinct knock-in mouse models of the HD mutation. We studied the effects of the HttQ111 allele on the C57BL/6J, CD-1, FVB/NCr1, and 129S2/SvPasCrl genetic backgrounds, and of two additional alleles, HttQ92 and HttQ50, on the C57BL/6J background. We describe the emergence of a transcriptomic signature in HttQ111/+ mice involving hundreds of differentially expressed genes and changes in diverse molecular pathways. We also show that this time course spanned the onset of mutant huntingtin nuclear localization phenotypes and somatic CAG-length instability in the striatum. Genetic background strongly influenced the magnitude and age at onset of these effects. This work provides a foundation for understanding the earliest transcriptional and molecular changes contributing to HD pathogenesis.


Asunto(s)
Cuerpo Estriado/metabolismo , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Antecedentes Genéticos , Inestabilidad Genómica/genética , Humanos , Proteína Huntingtina/biosíntesis , Enfermedad de Huntington/patología , Ratones , Mutación/genética , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Transcriptoma/genética
19.
Hum Mol Genet ; 26(7): 1258-1267, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28165127

RESUMEN

Huntington's disease (HD) reflects dominant consequences of a CAG repeat expansion mutation in HTT. Expanded CAG repeat size is the primary determinant of age at onset and age at death in HD. Although HD pathogenesis is driven by the expanded CAG repeat, whether the mutation influences the expression levels of mRNA and protein from the disease allele is not clear due to the lack of sensitive allele-specific quantification methods and the presence of confounding factors. To determine the impact of CAG expansion at the molecular level, we have developed novel allele-specific HTT mRNA and protein quantification methods based on principles of multiplex ligation-dependent probe amplification and targeted MS/MS parallel reaction monitoring, respectively. These assays, exhibiting high levels of specificity and sensitivity, were designed to distinguish allelic products based upon expressed polymorphic variants in HTT, including rs149 109 767. To control for other cis-haplotype variations, we applied allele-specific quantification assays to a panel of HD lymphoblastoid cell lines, each carrying the major European disease haplotype (i.e. hap.01) on the mutant chromosome. We found that steady state levels of HTT mRNA and protein were not associated with expanded CAG repeat length. Rather, the products of mutant and normal alleles, both mRNA and protein, were balanced, thereby arguing that a cis-regulatory effect of the expanded CAG repeat is not a critical component of the underlying mechanism of HD. These robust allele-specific assays could prove valuable for monitoring the impact of allele-specific gene silencing strategies currently being explored as therapeutic interventions in HD.


Asunto(s)
Encéfalo/metabolismo , Proteína Huntingtina/biosíntesis , Enfermedad de Huntington/genética , Expansión de Repetición de Trinucleótido/genética , Adulto , Edad de Inicio , Alelos , Autopsia , Encéfalo/patología , Femenino , Regulación de la Expresión Génica , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Masculino , ARN Mensajero/biosíntesis
20.
Neurochem Int ; 109: 24-33, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28062223

RESUMEN

The mechanisms implicated in the pathology of Huntington's disease (HD) remain not completely understood, although dysfunction of mitochondrial oxidative metabolism and Ca2+ handling have been suggested as contributing factors. However, in our previous studies with mitochondria isolated from the whole brains of HD mice, we found no evidence for defects in mitochondrial respiration and Ca2+ handling. In the present study, we used the YAC128 mouse model of HD to evaluate the effect of mHtt on respiratory activity and Ca2+ uptake capacity of mitochondria isolated from the striatum, the most vulnerable brain region in HD. Isolated, Percoll-gradient purified striatal mitochondria from YAC128 mice were free of cytosolic and ER contaminations, but retained attached mHtt. Both nonsynaptic and synaptic striatal mitochondria isolated from early symptomatic 2-month-old YAC128 mice had similar respiratory rates and Ca2+ uptake capacities compared with mitochondria from wild-type FVB/NJ mice. Consistent with the lack of difference in mitochondrial respiration, we found that the expression of several nuclear-encoded proteins in striatal mitochondria was similar between wild-type and YAC128 mice. Taken together, our data demonstrate that mHtt does not alter respiration and Ca2+ uptake capacity in striatal mitochondria isolated from YAC128 mice, suggesting that respiratory defect and Ca2+ uptake deficiency most likely do not contribute to striatal pathology associated with HD.


Asunto(s)
Señalización del Calcio/fisiología , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Huntington/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Animales , Femenino , Humanos , Proteína Huntingtina/biosíntesis , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA