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












Base de datos
Intervalo de año de publicación
1.
Stem Cell Res ; 78: 103462, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38851031

RESUMEN

Spinocerebellar Ataxia Type 7 (SCA7) is an autosomal dominantly inherited disorder, primarily characterized by cerebellar ataxia and visual loss. SCA7 is caused by a CAG repeat expansion in exon 3 of the ATXN7 gene. We generated human induced pluripotent stem cells (hiPSCs) from peripheral blood-derived erythroblasts from two SCA7 patients (LUMCi051-A,B and LUMCi052-A,B,C) using integration-free episomal vectors. All hiPSC clones express pluripotency factors, show a normal karyotype, and can differentiate into the three germ layers. These lines can be used for in vitro disease modeling and therapy testing.


Asunto(s)
Células Madre Pluripotentes Inducidas , Ataxias Espinocerebelosas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/genética , Línea Celular , Masculino , Diferenciación Celular , Femenino , Adulto
2.
Mov Disord ; 38(8): 1428-1442, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37278528

RESUMEN

BACKGROUND: Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by a polyglutamine expansion in the ataxin-1 protein resulting in neuropathology including mutant ataxin-1 protein aggregation, aberrant neurodevelopment, and mitochondrial dysfunction. OBJECTIVES: Identify SCA1-relevant phenotypes in patient-specific fibroblasts and SCA1 induced pluripotent stem cells (iPSCs) neuronal cultures. METHODS: SCA1 iPSCs were generated and differentiated into neuronal cultures. Protein aggregation and neuronal morphology were evaluated using fluorescent microscopy. Mitochondrial respiration was measured using the Seahorse Analyzer. The multi-electrode array (MEA) was used to identify network activity. Finally, gene expression changes were studied using RNA-seq to identify disease-specific mechanisms. RESULTS: Bioenergetics deficits in patient-derived fibroblasts and SCA1 neuronal cultures showed altered oxygen consumption rate, suggesting involvement of mitochondrial dysfunction in SCA1. In SCA1 hiPSC-derived neuronal cells, nuclear and cytoplasmic aggregates were identified similar in localization as aggregates in SCA1 postmortem brain tissue. SCA1 hiPSC-derived neuronal cells showed reduced dendrite length and number of branching points while MEA recordings identified delayed development in network activity in SCA1 hiPSC-derived neuronal cells. Transcriptome analysis identified 1050 differentially expressed genes in SCA1 hiPSC-derived neuronal cells associated with synapse organization and neuron projection guidance, where a subgroup of 151 genes was highly associated with SCA1 phenotypes and linked to SCA1 relevant signaling pathways. CONCLUSIONS: Patient-derived cells recapitulate key pathological features of SCA1 pathogenesis providing a valuable tool for the identification of novel disease-specific processes. This model can be used for high throughput screenings to identify compounds, which may prevent or rescue neurodegeneration in this devastating disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Células Madre Pluripotentes Inducidas , Ataxias Espinocerebelosas , Ratones , Animales , Ataxinas/metabolismo , Agregado de Proteínas , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Ratones Transgénicos , Células de Purkinje/metabolismo , Células de Purkinje/patología , Ataxias Espinocerebelosas/metabolismo , Fibroblastos/metabolismo
3.
Front Aging Neurosci ; 14: 1048584, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36733499

RESUMEN

Introduction: ADutch-type cerebral amyloid angiopathy (D-CAA) is a hereditary brain disorder caused by a point mutation in the amyloid precursor protein (APP) gene. The mutation is located within the amyloid beta (Aß) domain of APP and leads to Aß peptide accumulation in and around the cerebral vasculature. There lack of disease models to study the cellular and molecular pathological mechanisms of D-CAA together with the absence of a disease phenotype in vitro in overexpression cell models, as well as the limited availability of D-CAA animal models indicates the need for a D-CAA patient-derived model. Methods: We generated cerebral organoids from four D-CAA patients and four controls, cultured them up to 110 days and performed immunofluorescent and targeted gene expression analyses at two time points (D52 and D110). Results: D-CAA cerebral organoids exhibited Aß accumulations, showed enhanced neuronal and astrocytic gene expression and TGFß pathway de-regulation. Conclusions: These results illustrate the potential of cerebral organoids as in vitro disease model of D-CAA that can be used to understand disease mechanisms of D-CAA and can serve as therapeutic intervention platform for various Aß-related disorders.

4.
Nucleic Acid Ther ; 31(5): 351-363, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34061681

RESUMEN

Dutch-type cerebral amyloid angiopathy (D-CAA) is a monogenic form of cerebral amyloid angiopathy and is inherited in an autosomal dominant manner. The disease is caused by a point mutation in exon 17 of the amyloid precursor protein (APP) gene that leads to an amino acid substitution at codon 693. The mutation is located within the amyloid beta (Aß) domain of APP, and leads to accumulation of toxic Aß peptide in and around the cerebral vasculature. We have designed an antisense oligonucleotide (AON) approach that results in skipping of exon 17, generating a shorter APP isoform that lacks part of the Aß domain and the D-CAA mutation. We demonstrate efficient AON-induced skipping of exon 17 at RNA level and the occurrence of a shorter APP protein isoform in three different cell types. This resulted in a reduction of Aß40 in neuronally differentiated, patient-derived induced pluripotent stem cells. AON-treated wild-type mice showed successful exon skipping on RNA and protein levels throughout the brain. These results illustrate APP splice modulation as a promising therapeutic approach for D-CAA.


Asunto(s)
Precursor de Proteína beta-Amiloide , Angiopatía Amiloide Cerebral , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Angiopatía Amiloide Cerebral/genética , Angiopatía Amiloide Cerebral/terapia , Humanos , Ratones , Oligonucleótidos Antisentido/genética
5.
Stem Cell Res ; 49: 102055, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33128956

RESUMEN

Propionic acidemia (PA) is an inherited metabolic disease caused by mutations in the PCCA and PCCB genes. We have previously generated an induced pluripotent stem cell (iPSC) line (UAMi004-A) from a PA patient with the c.1218_1231del14ins12 (p.Gly407Argfs*14) homozygous mutation in the PCCB gene. Here, we report the generation of the isogenic control in which the mutation was genetically corrected using CRISPR/Cas9 technology. Off-target editing presence was excluded and the iPSCs had typical embryonic stem cell-like morphology and normal karyotype that expressed pluripotency markers and maintained their in vitro differentiation potential.


Asunto(s)
Células Madre Pluripotentes Inducidas , Acidemia Propiónica , Sistemas CRISPR-Cas/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Metilmalonil-CoA Descarboxilasa/genética , Mutación/genética , Acidemia Propiónica/genética , Tecnología
6.
Stem Cell Res ; 39: 101498, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31326748

RESUMEN

Huntington disease (HD) is an autosomal dominant, neurodegenerative disease caused by a CAG repeat expansion within the coding sequence of the HTT gene, resulting in a highly toxic protein with an expanded polyglutamine stretch that forms typical protein aggregates throughout the brain. We generated human induced pluripotent stem cells (hiPSCs) from two HD patients using non-integrating Sendai virus (SeV). The hiPSCs display a normal karyotype, express all pluripotency markers, have the same CAG repeat expansion as the original fibroblasts and are able to differentiate into the three germ layers in vitro.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Cultivadas , Femenino , Humanos , Enfermedad de Huntington/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipo , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sendai/genética
7.
Stem Cell Res ; 34: 101359, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30611017

RESUMEN

Hereditary Cerebral Hemorrhage with Amyloidosis-Dutch type (HCHWA-D) is an autosomal dominant hereditary disease caused by a point mutation in exon 17 of the APP gene. We generated human induced pluripotent stem cells (hiPSCs) from a symptomatic HCHWA-D patient by using non-integrating Sendai virus (SeV). The newly generated hiPSCs express all pluripotency markers, have a normal karyotype, carry the Dutch mutation, can differentiate in the three germ layers in vitro and are SeV free.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Angiopatía Amiloide Cerebral Familiar/patología , Células Madre Pluripotentes Inducidas/patología , Secuencia de Bases , Línea Celular , Femenino , Humanos , Persona de Mediana Edad
8.
Stem Cell Res ; 29: 125-128, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29656178

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is a hereditary neurodegenerative disease caused by a CAG repeat expansion in exon 8 of the ATXN1 gene. We generated induced pluripotent stem cells (hiPSCs) from a SCA1 patient and his non-affected sister by using non-integrating Sendai Viruses (SeV). The resulting hiPSCs are SeVfree, express pluripotency markers, display a normal karyotype, retain the mutation (length of the CAG repeat expansion in the ATXN1 gene) and are able to differentiate into the three germ layers in vitro.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Ataxias Espinocerebelosas/metabolismo , Adulto , Diferenciación Celular , Línea Celular , Femenino , Humanos , Masculino , Hermanos
9.
PLoS One ; 12(6): e0178556, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28570578

RESUMEN

Huntington disease is associated with elongation of a CAG repeat in the HTT gene that results in a mutant huntingtin protein. Several studies have implicated N-terminal huntingtin protein fragments in Huntington disease pathogenesis. Ideally, these fragments are studied in human brain tissue. However, the use of human brain tissue comes with certain unavoidable variables such as post mortem delay, artefacts from freeze-thaw cycles and subject-to-subject variation. Knowledge on how these variables might affect N-terminal huntingtin protein fragments in post mortem human brain is important for a proper interpretation of study results. The effect of post mortem delay on protein in human brain is known to vary depending on the protein of interest. In the present study, we have assessed the effect of post mortem delay on N-terminal huntingtin protein fragments using western blot. We mimicked post mortem delay in one individual control case and one individual Huntington disease case with low initial post mortem delay. The influence of subject-to-subject variation on N-terminal huntingtin fragments was assessed in human cortex and human striatum using two cohorts of control and Huntington disease subjects. Our results show that effects of post mortem delay on N-terminal huntingtin protein fragments are minor in our individual subjects. Additionally, one freeze-thaw cycle decreases the huntingtin western blot signal intensity in the cortex control subject, but does not introduce additional N-terminal huntingtin fragments. Our results suggest that subject-to-subject variation contributes more to variability in N-terminal huntingtin fragments than post mortem delay.


Asunto(s)
Encéfalo/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/patología , Regiones no Traducidas 3' , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Encéfalo/patología , Estudios de Casos y Controles , Línea Celular , Femenino , Células HEK293 , Humanos , Enfermedad de Huntington/metabolismo , Masculino , Persona de Mediana Edad , Sistemas de Lectura Abierta , Cambios Post Mortem , Homología de Secuencia de Aminoácido
10.
Mol Neurodegener ; 10: 21, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25928884

RESUMEN

BACKGROUND: Huntington disease (HD) is an autosomal dominant neurodegenerative disorder, characterized by motor, psychiatric and cognitive symptoms. HD is caused by a CAG repeat expansion in the first exon of the HTT gene, resulting in an expanded polyglutamine tract at the N-terminus of the huntingtin protein. Typical disease onset is around mid-life (adult-onset HD) whereas onset below 21 years is classified as juvenile HD. While much research has been done on the underlying HD disease mechanisms, little is known about regulation and expression levels of huntingtin RNA and protein. RESULTS: In this study we used 15 human post-mortem HD brain samples to investigate the expression of wild-type and mutant huntingtin mRNA and protein. In adult-onset HD brain samples, there was a small but significantly lower expression of mutant huntingtin mRNA compared to wild-type huntingtin mRNA, while wild-type and mutant huntingtin protein expression levels did not differ significantly. Juvenile HD subjects did show a lower expression of mutant huntingtin protein compared to wild-type huntingtin protein. Our results in HD brain and fibroblasts suggest that protein aggregation does not affect levels of huntingtin RNA and protein. Additionally, we did not find any evidence for a reduced expression of huntingtin antisense in fibroblasts derived from a homozygous HD patient. CONCLUSIONS: We found small differences in allelic huntingtin mRNA levels in adult-onset HD brain, with significantly lower mutant huntingtin mRNA levels. Wild-type and mutant huntingtin protein were not significantly different in adult-onset HD brain samples. Conversely, in juvenile HD brain samples mutant huntingtin protein levels were lower compared with wild-type huntingtin, showing subtle differences between juvenile HD and adult-onset HD. Since most HD model systems harbor juvenile repeat expansions, our results suggest caution with the interpretation of huntingtin mRNA and protein studies using HD cell and animal models with such long repeats. Furthermore, our huntingtin antisense results in homozygous HD cells do not support reduced huntingtin antisense expression due to an expanded CAG repeat.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/metabolismo , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Autopsia , Encéfalo/patología , Fibroblastos/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Proteínas Mutantes/genética , ARN Mensajero/metabolismo
11.
Neurol Sci ; 36(3): 429-34, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25294428

RESUMEN

Huntington disease is caused by expansion of a CAG repeat in the huntingtin gene that is translated into an elongated polyglutamine stretch within the N-terminal domain of the huntingtin protein. The mutation is thought to introduce a gain-of-toxic function in the mutant huntingtin protein, and blocking this toxicity by antibody binding could alleviate Huntington disease pathology. Llama single domain antibodies (VHH) directed against mutant huntingtin are interesting candidates as therapeutic agents or research tools in Huntington disease because of their small size, high thermostability, low cost of production, possibility of intracellular expression, and potency of blood-brain barrier passage. We have selected VHH from llama phage display libraries that specifically target the N-terminal domain of the huntingtin protein. Our VHH are capable of binding wild-type and mutant human huntingtin under native and denatured conditions and can be used in Huntington disease studies as a novel antibody that is easy to produce and manipulate.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Enfermedad de Huntington/terapia , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/inmunología , Secuencia de Aminoácidos , Especificidad de Anticuerpos , Epítopos/inmunología , Escherichia coli , Humanos , Proteína Huntingtina , Enfermedad de Huntington/inmunología , Datos de Secuencia Molecular , Unión Proteica
12.
Ann Rheum Dis ; 74(8): 1571-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24695009

RESUMEN

OBJECTIVES: To investigate how the genetic susceptibility gene DIO2 confers risk to osteoarthritis (OA) onset in humans and to explore whether counteracting the deleterious effect could contribute to novel therapeutic approaches. METHODS: Epigenetically regulated expression of DIO2 was explored by assessing methylation of positional CpG-dinucleotides and the respective DIO2 expression in OA-affected and macroscopically preserved articular cartilage from end-stage OA patients. In a human in vitro chondrogenesis model, we measured the effects when thyroid signalling during culturing was either enhanced (excess T3 or lentiviral induced DIO2 overexpression) or decreased (iopanoic acid). RESULTS: OA-related changes in methylation at a specific CpG dinucleotide upstream of DIO2 caused significant upregulation of its expression (ß=4.96; p=0.0016). This effect was enhanced and appeared driven specifically by DIO2 rs225014 risk allele carriers (ß=5.58, p=0.0006). During in vitro chondrogenesis, DIO2 overexpression resulted in a significant reduced capacity of chondrocytes to deposit extracellular matrix (ECM) components, concurrent with significant induction of ECM degrading enzymes (ADAMTS5, MMP13) and markers of mineralisation (ALPL, COL1A1). Given their concurrent and significant upregulation of expression, this process is likely mediated via HIF-2α/RUNX2 signalling. In contrast, we showed that inhibiting deiodinases during in vitro chondrogenesis contributed to prolonged cartilage homeostasis as reflected by significant increased deposition of ECM components and attenuated upregulation of matrix degrading enzymes. CONCLUSIONS: Our findings show how genetic variation at DIO2 could confer risk to OA and raised the possibility that counteracting thyroid signalling may be a novel therapeutic approach.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Yoduro Peroxidasa/genética , Osteoartritis/genética , Cartílago Articular/enzimología , Cartílago Articular/fisiopatología , Condrogénesis/genética , Metilación de ADN , Epigénesis Genética , Regulación de la Expresión Génica , Silenciador del Gen/fisiología , Humanos , Pérdida de Heterocigocidad , Osteoartritis/fisiopatología , Osteoartritis de la Cadera/genética , Osteoartritis de la Rodilla/genética , Hormonas Tiroideas/fisiología , Regulación hacia Arriba/fisiología , Yodotironina Deyodinasa Tipo II
13.
Neurobiol Dis ; 58: 49-56, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23659897

RESUMEN

Spinocerebellar ataxia type 3 is caused by a polyglutamine expansion in the ataxin-3 protein, resulting in gain of toxic function of the mutant protein. The expanded glutamine stretch in the protein is the result of a CAG triplet repeat expansion in the penultimate exon of the ATXN3 gene. Several gene silencing approaches to reduce mutant ataxin-3 toxicity in this disease aim to lower ataxin-3 protein levels, but since this protein is involved in deubiquitination and proteasomal protein degradation, its long-term silencing might not be desirable. Here, we propose a novel protein modification approach to reduce mutant ataxin-3 toxicity by removing the toxic polyglutamine repeat from the ataxin-3 protein through antisense oligonucleotide-mediated exon skipping while maintaining important wild type functions of the protein. In vitro studies showed that exon skipping did not negatively impact the ubiquitin binding capacity of ataxin-3. Our in vivo studies showed no toxic properties of the novel truncated ataxin-3 protein. These results suggest that exon skipping may be a novel therapeutic approach to reduce polyglutamine-induced toxicity in spinocerebellar ataxia type 3.


Asunto(s)
Enfermedad de Machado-Joseph/patología , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Repeticiones de Trinucleótidos/genética , Animales , Ataxina-3 , Células Cultivadas , Análisis Mutacional de ADN , Relación Dosis-Respuesta a Droga , Exones/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Enfermedad de Machado-Joseph/tratamiento farmacológico , Enfermedad de Machado-Joseph/genética , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Oligonucleótidos Antisentido/farmacología , Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Transfección , Ubiquitina/metabolismo
14.
PLoS One ; 6(9): e24308, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21909428

RESUMEN

To date there are 9 known diseases caused by an expanded polyglutamine repeat, with the most prevalent being Huntington's disease. Huntington's disease is a progressive autosomal dominant neurodegenerative disorder for which currently no therapy is available. It is caused by a CAG repeat expansion in the HTT gene, which results in an expansion of a glutamine stretch at the N-terminal end of the huntingtin protein. This polyglutamine expansion plays a central role in the disease and results in the accumulation of cytoplasmic and nuclear aggregates. Here, we make use of modified 2'-O-methyl phosphorothioate (CUG)n triplet-repeat antisense oligonucleotides to effectively reduce mutant huntingtin transcript and protein levels in patient-derived Huntington's disease fibroblasts and lymphoblasts. The most effective antisense oligonucleotide, (CUG)(7), also reduced mutant ataxin-1 and ataxin-3 mRNA levels in spinocerebellar ataxia 1 and 3, respectively, and atrophin-1 in dentatorubral-pallidoluysian atrophy patient derived fibroblasts. This antisense oligonucleotide is not only a promising therapeutic tool to reduce mutant huntingtin levels in Huntington's disease but our results in spinocerebellar ataxia and dentatorubral-pallidoluysian atrophy cells suggest that this could also be applicable to other polyglutamine expansion disorders as well.


Asunto(s)
Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/terapia , Oligonucleótidos Antisentido/farmacología , Expansión de Repetición de Trinucleótido/genética , Ataxina-1 , Ataxina-3 , Ataxinas , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Huntingtina , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Epilepsias Mioclónicas Progresivas/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Péptidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Ataxias Espinocerebelosas/genética
15.
BMC Biotechnol ; 9: 50, 2009 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-19463169

RESUMEN

BACKGROUND: Methodologies like phage display selection, in vitro mutagenesis and the determination of allelic expression differences include steps where large numbers of clones need to be compared and characterised. In the current study we show that high-resolution melt curve analysis (HRMA) is a simple, cost-saving tool to quickly study clonal variation without prior nucleotide sequence knowledge. RESULTS: HRMA results nicely matched those obtained with ELISA and compared favourably to DNA fingerprinting of restriction digested clone insert-PCR. DNA sequence analysis confirmed that HRMA-clustered clones contained identical inserts. CONCLUSION: Using HRMA, analysis of up to 384 samples can be done simultaneously and will take approximately 30 minutes. Clustering of clones can be largely automated using the system's software within 2 hours. Applied to the analysis of clones obtained after phage display antibody selection, HRMA facilitated a quick overview of the overall success as well as the identification of identical clones. Our approach can be used to characterize any clone set prior to sequencing, thereby reducing sequencing costs significantly.


Asunto(s)
Biblioteca de Péptidos , Análisis de Secuencia de ADN/métodos , Análisis por Conglomerados , Dermatoglifia del ADN , Ensayo de Inmunoadsorción Enzimática , Reacción en Cadena de la Polimerasa , Programas Informáticos
16.
BMC Mol Biol ; 9: 84, 2008 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-18844975

RESUMEN

BACKGROUND: Huntington's disease is a progressive autosomal dominant neurodegenerative disorder that is caused by a CAG repeat expansion in the HD or Huntington's disease gene. Although micro array studies on patient and animal tissue provide valuable information, the primary effect of mutant huntingtin will inevitably be masked by secondary processes in advanced stages of the disease. Thus, cell models are instrumental to study early, direct effects of mutant huntingtin. mRNA changes were studied in an inducible PC12 model of Huntington's disease, before and after aggregates became visible, to identify groups of genes that could play a role in the early pathology of Huntington's disease. RESULTS: Before aggregation, up-regulation of gene expression predominated, while after aggregates became visible, down-regulation and up-regulation occurred to the same extent. After aggregates became visible there was a down-regulation of dopamine biosynthesis genes accompanied by down-regulation of dopamine levels in culture, indicating the utility of this model to identify functionally relevant pathways. Furthermore, genes of the anti-oxidant Nrf2-ARE pathway were up-regulated, possibly as a protective mechanism. In parallel, we discovered alterations in genes which may result in increased oxidative stress and damage. CONCLUSION: Up-regulation of gene expression may be more important in HD pathology than previously appreciated. In addition, given the pathogenic impact of oxidative stress and neuroinflammation, the Nrf2-ARE signaling pathway constitutes a new attractive therapeutic target for HD.


Asunto(s)
Dopamina/biosíntesis , Enfermedad de Huntington/genética , Mutación , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Animales , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Factor 2 Relacionado con NF-E2/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Estrés Oxidativo , Células PC12 , ARN Mensajero/metabolismo , Ratas , Regulación hacia Arriba
17.
Exp Neurol ; 199(2): 257-64, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16380118

RESUMEN

N-terminal fragments of huntingtin containing an expanded polyglutamine stretch play an important role in the molecular pathogenesis of Huntington's disease. Their ultimate accumulation in insoluble protein aggregates constitutes an important pathological hallmark of Huntington's disease. We report on systematic biochemical comparison studies of soluble wild type and mutant N-terminal huntingtin fragments. The results show that soluble wild type exon 1 fragments are predominantly present in higher molecular weight complexes with a molecular size of approximately 300 kDa, while their mutant counterparts are mainly present in their monomeric form. In contrast, longer N-terminal fragments corresponding to peptides produced by caspase cleavage do not display these differential properties. These findings suggest that especially an increased amount of monomeric form of small N-terminal mutant huntingtin fragments may facilitate aberrant interactions both with itself via the polyglutamine stretch and with other proteins and thereby contribute to molecular pathogenesis.


Asunto(s)
Mutagénesis/fisiología , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/química , Péptidos/genética , Péptidos/metabolismo , Animales , Western Blotting/métodos , Cromatografía en Gel/métodos , Electroforesis en Gel de Poliacrilamida/métodos , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/biosíntesis , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Peso Molecular , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Células PC12 , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Péptidos/química , Ratas , Factores de Tiempo , Transfección/métodos
18.
Hybridoma (Larchmt) ; 24(5): 231-5, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16225422

RESUMEN

Huntington's disease is a dominantly inherited, devastating neurodegenerative disorder, caused by a polyglutamine expansion (>37) in the N-terminal region of huntingtin, a protein of unknown function. In patients and normal individuals, N-terminal fragments of huntingtin are found, and the N-terminal fragments of mutant huntingtin are cytotoxic. The functions of wild-type huntingtin and the mechanisms underlying the toxic effects of mutant huntingtin are still ill defined. To get more insight into these topics, monoclonal antibodies (MAbs) are indispensable tools. Antibodies raised against the N-terminus are especially important. Among these, the 4C8 mouse MAb has been extensively used in various approaches. In this study, we have mapped the epitope of 4C8 to a 15-amino acid (aa) region spanning from aa 443 to 457 of the human protein, and found that mutation of three consecutive glutamic acids present in this region disrupts the recognition by 4C8. These results allow a more accurate interpretation of the results obtained by usage of the 4C8 antibody and broaden the utility of this antibody.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Mapeo Epitopo , Proteínas del Tejido Nervioso/inmunología , Proteínas Nucleares/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Células Cultivadas , Ácido Glutámico/genética , Humanos , Proteína Huntingtina , Ratones , Datos de Secuencia Molecular , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína
19.
Mol Cell Neurosci ; 30(1): 12-23, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15994095

RESUMEN

Huntington's disease can be used as a model to study neurodegenerative disorders caused by aggregation-prone proteins. It has been proposed that the entrapment of transcription factors in aggregates plays an important role in pathogenesis. We now report that the transcriptional activity of CBP is already repressed in the early time points by soluble mutant huntingtin, whereas the histone acetylase activity of CBP/p300 is gradually diminished over time. Mutant huntingtin bound much stronger to CBP than normal huntingtin, possibly contributing to repression. Especially at the later time points, CBP protein level was gradually reduced via the proteasome pathway. In sharp contrast, p300 was unaffected by mutant huntingtin. This selective degradation of CBP was absent in spinocerebellar ataxia 3. Thus, mutant huntingtin specifically affects CBP and not p300 both at the early and later time points, via multiple mechanisms. In addition to the reduction of CBP, also the altered ratio of these closely related histone acetyltransferases may affect chromatin structure and transcription and thus contribute to neurodegeneration.


Asunto(s)
Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Animales , Proteína de Unión a CREB , Proteína p300 Asociada a E1A , Regulación de la Expresión Génica , Proteína Huntingtina , Enfermedad de Huntington/fisiopatología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Células PC12 , Unión Proteica , Ratas , Transcripción Genética , Transfección
20.
Mol Cell Neurosci ; 30(4): 560-71, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16456924

RESUMEN

Huntington's disease can be used as a model to study neurodegenerative disorders caused by aggregation-prone proteins. It has been proposed that the entrapment of transcription factors in aggregates plays an important role in pathogenesis. We now report that the transcriptional activity of CBP is already repressed in the early time points by soluble mutant huntingtin, whereas the histone acetylase activity of CBP/p300 is gradually diminished over time. Mutant huntingtin bound much stronger to CBP than normal huntingtin, possibly contributing to repression. Especially at the later time points, CBP protein level was gradually reduced via the proteasome pathway. In sharp contrast, p300 was unaffected by mutant huntingtin. This selective degradation of CBP was absent in spinocerebellar ataxia 3. Thus, mutant huntingtin specifically affects CBP and not p300 both at the early and later time points, via multiple mechanisms. In addition to the reduction of CBP, also the altered ratio of these closely related histone acetyl transferases may affect chromatin structure and transcription and thus contribute to neurodegeneration.


Asunto(s)
Encéfalo/metabolismo , Proteína de Unión a CREB/metabolismo , Regulación hacia Abajo/fisiología , Mutación/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Animales , Encéfalo/fisiopatología , Histona Acetiltransferasas/metabolismo , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Células PC12 , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/fisiología , Ratas , Elementos Reguladores de la Transcripción/fisiología , Proteínas Represoras/genética , Factores de Tiempo , Factores de Transcripción p300-CBP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...