Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 29(23): 3818-3829, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33432325

RESUMEN

Friedreich ataxia (FRDA) is typically caused by homozygosity for an expanded GAA triplet-repeat in intron 1 of the FXN gene, which results in transcriptional deficiency via epigenetic silencing. Most patients are homozygous for alleles containing > 500 triplets, but a subset (~20%) have at least one expanded allele with < 500 triplets and a distinctly milder phenotype. We show that in FRDA DNA methylation spreads upstream from the expanded repeat, further than previously recognized, and establishes an FRDA-specific region of hypermethylation in intron 1 (~90% in FRDA versus < 10% in non-FRDA) as a novel epigenetic signature. The hypermethylation of this differentially methylated region (FRDA-DMR) was observed in a variety of patient-derived cells; it significantly correlated with FXN transcriptional deficiency and age of onset, and it reverted to the non-disease state in isogenically corrected induced pluripotent stem cell (iPSC)-derived neurons. Bisulfite deep sequencing of the FRDA-DMR in peripheral blood mononuclear cells from 73 FRDA patients revealed considerable intra-individual epiallelic variability, including fully methylated, partially methylated, and unmethylated epialleles. Although unmethylated epialleles were rare (median = 0.33%) in typical patients homozygous for long GAA alleles with > 500 triplets, a significantly higher prevalence of unmethylated epialleles (median = 9.8%) was observed in patients with at least one allele containing < 500 triplets, less severe FXN deficiency (>20%) and later onset (>15 years). The higher prevalence in mild FRDA of somatic FXN epialleles devoid of DNA methylation is consistent with variegated epigenetic silencing mediated by expanded triplet-repeats. The proportion of unsilenced somatic FXN genes is an unrecognized phenotypic determinant in FRDA and has implications for the deployment of effective therapies.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Ataxia de Friedreich/patología , Silenciador del Gen , Leucocitos Mononucleares/patología , Fenotipo , Adolescente , Adulto , Alelos , Niño , Preescolar , Femenino , Ataxia de Friedreich/genética , Humanos , Lactante , Leucocitos Mononucleares/metabolismo , Masculino , Adulto Joven
2.
J Biol Chem ; 294(5): 1652-1660, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30710013

RESUMEN

During Herbert Tabor's tenure as Editor-in-Chief from 1971 to 2010, JBC has published many seminal papers in the fields of chromatin structure, epigenetics, and regulation of transcription in eukaryotes. As of this writing, more than 21,000 studies on gene transcription at the molecular level have been published in JBC since 1971. This brief review will attempt to highlight some of these ground-breaking discoveries and show how early studies published in JBC have influenced current research. Papers published in the Journal have reported the initial discovery of multiple forms of RNA polymerase in eukaryotes, identification and purification of essential components of the transcription machinery, and identification and mechanistic characterization of various transcriptional activators and repressors and include studies on chromatin structure and post-translational modifications of the histone proteins. The large body of literature published in the Journal has inspired current research on how chromatin organization and epigenetics impact regulation of gene expression.


Asunto(s)
Bioquímica/historia , Cromatina/genética , Cromatina/metabolismo , Histonas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Epigénesis Genética , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Publicaciones Periódicas como Asunto , Procesamiento Proteico-Postraduccional
3.
J Biol Chem ; 294(6): 1846-1859, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30552117

RESUMEN

Friedreich ataxia (FRDA) is a neurodegenerative disorder caused by transcriptional silencing of the frataxin (FXN) gene, resulting in loss of the essential mitochondrial protein frataxin. Based on the knowledge that a GAA·TTC repeat expansion in the first intron of FXN induces heterochromatin, we previously showed that 2-aminobenzamide-type histone deacetylase inhibitors (HDACi) increase FXN mRNA levels in induced pluripotent stem cell (iPSC)-derived FRDA neurons and in circulating lymphocytes from patients after HDACi oral administration. How the reduced expression of frataxin leads to neurological and other systemic symptoms in FRDA patients remains unclear. Similar to other triplet-repeat disorders, it is unknown why FRDA affects only specific cell types, primarily the large sensory neurons of the dorsal root ganglia and cardiomyocytes. The combination of iPSC technology and genome-editing techniques offers the unique possibility to address these questions in a relevant cell model of FRDA, obviating confounding effects of variable genetic backgrounds. Here, using "scarless" gene-editing methods, we created isogenic iPSC lines that differ only in the length of the GAA·TTC repeats. To uncover the gene expression signatures due to the GAA·TTC repeat expansion in FRDA neuronal cells and the effect of HDACi on these changes, we performed RNA-seq-based transcriptomic analysis of iPSC-derived central nervous system (CNS) and isogenic sensory neurons. We found that cellular pathways related to neuronal function, regulation of transcription, extracellular matrix organization, and apoptosis are affected by frataxin loss in neurons of the CNS and peripheral nervous system and that these changes are partially restored by HDACi treatment.


Asunto(s)
Ataxia de Friedreich/genética , Inhibidores de Histona Desacetilasas/farmacología , Neuronas/patología , Transcriptoma , Células Cultivadas , Ataxia de Friedreich/patología , Edición Génica/métodos , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/química , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Neuronas/química , Expansión de Repetición de Trinucleótido/genética , Frataxina
4.
J Biol Chem ; 293(36): 13775-13777, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30068547

RESUMEN

Regulation of transcription in eukaryotic cells is a dynamic interplay between chromatin structure and recruitment of a plethora of transcription factors to enhancers, upstream activator sequences, and proximal promoter elements. These factors serve to recruit RNA polymerase to the core promoter for productive transcription. In this Thematic Minireview Series on chromatin and transcription, five reviews summarize current knowledge of diverse aspects of transcriptional regulation and the role of chromatin structure in transcription and development.


Asunto(s)
Cromatina/genética , Transcripción Genética , Animales , Regulación de la Expresión Génica , Humanos
5.
Bioorg Med Chem Lett ; 27(15): 3289-3293, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28648462

RESUMEN

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder that is caused by inactivating mutations in the Survival of motor neuron 1 (SMN1) gene, resulting in decreased SMN protein expression. Humans possess a paralog gene, SMN2, which contains a splicing defect in exon 7 leading to diminished expression of full-length, fully functional SMN protein. Increasing SMN2 expression has been a focus of therapeutic development for SMA. Multiple studies have reported the efficacy of histone deacetylase inhibitors (HDACi) in this regard. However, clinical trials involving HDACi have been unsatisfactory, possibly because previous efforts to identify HDACi to treat SMA have employed non-neuronal cells as the screening platform. To address this issue, we generated an SMA-patient specific, induced pluripotent stem cell (iPSC) derived neuronal cell line that contains homogenous Tuj1+neurons. We screened a small library of cyclic tetrapeptide HDACi using this SMA neuronal platform and discovered compounds that elevate SMN2 expression by an impressive twofold or higher. These candidates are also capable of forming gems intranuclearly in SMA neurons, demonstrating biological activity. Our study identifies new potential HDACi therapeutics for SMA screened using a disease-relevant SMA neuronal cellular model.


Asunto(s)
Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Atrofia Muscular Espinal/tratamiento farmacológico , Neuronas/efectos de los fármacos , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Células Cultivadas , Evaluación Preclínica de Medicamentos , Humanos , Células Madre Pluripotentes Inducidas/citología , Atrofia Muscular Espinal/genética , Neurogénesis , Neuronas/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Regulación hacia Arriba/efectos de los fármacos
6.
J Biol Chem ; 290(10): 5979-90, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25593321

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is an inherited degenerative disease that affects the internal endothelial cell monolayer of the cornea and can result in corneal edema and vision loss in severe cases. FECD affects ∼5% of middle-aged Caucasians in the United States and accounts for >14,000 corneal transplantations annually. Among the several genes and loci associated with FECD, the strongest association is with an intronic (CTG·CAG)n trinucleotide repeat expansion in the TCF4 gene, which is found in the majority of affected patients. Corneal endothelial cells from FECD patients harbor a poly(CUG)n RNA that can be visualized as RNA foci containing this condensed RNA and associated proteins. Similar to myotonic dystrophy type 1, the poly(CUG)n RNA co-localizes with and sequesters the mRNA-splicing factor MBNL1, leading to missplicing of essential MBNL1-regulated mRNAs. Such foci and missplicing are not observed in similar cells from FECD patients who lack the repeat expansion. RNA-Seq splicing data from the corneal endothelia of FECD patients and controls reveal hundreds of differential alternative splicing events. These include events previously characterized in the context of myotonic dystrophy type 1 and epithelial-to-mesenchymal transition, as well as splicing changes in genes related to proposed mechanisms of FECD pathogenesis. We report the first instance of RNA toxicity and missplicing in a common non-neurological/neuromuscular disease associated with a repeat expansion. The FECD patient population with this (CTG·CAG)n trinucleotide repeat expansion exceeds that of the combined number of patients in all other microsatellite expansion disorders.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Distrofia Endotelial de Fuchs/genética , ARN Mensajero/genética , Factores de Transcripción/genética , Expansión de Repetición de Trinucleótido/genética , Córnea/metabolismo , Córnea/patología , Distrofia Endotelial de Fuchs/patología , Humanos , Empalme del ARN/genética , Proteínas de Unión al ARN/genética , Factor de Transcripción 4
7.
J Biol Chem ; 289(8): 4553-4, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24362035

RESUMEN

With the advent of human induced pluripotent stem cell (hiPSC) technology, it is now possible to derive patient-specific cell lines that are of great potential in both basic research and the development of new therapeutics for human diseases. Not only do hiPSCs offer unprecedented opportunities to study cellular differentiation and model human diseases, but the differentiated cell types obtained from iPSCs may become therapeutics themselves. These cells can also be used in the screening of therapeutics and in toxicology assays for potential liabilities of therapeutic agents. The remarkable achievement of transcription factor reprogramming to generate iPSCs was recognized by the award of the Nobel Prize in Medicine to Shinya Yamanaka in 2012, just 6 years after the first publication of reprogramming methods to generate hiPSCs (Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., and Yamanaka, S. (2007) Cell 131, 861-872). This minireview series highlights both the promises and challenges of using iPSC technology for disease modeling, drug screening, and the development of stem cell therapeutics.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Trasplante de Células Madre , Evaluación Preclínica de Medicamentos , Humanos
8.
Hum Mol Genet ; 22(25): 5276-87, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23933738

RESUMEN

Myotonic dystrophy type 1 (DM1) is an inherited dominant muscular dystrophy caused by expanded CTG·CAG triplet repeats in the 3' untranslated region of the DMPK1 gene, which produces a toxic gain-of-function CUG RNA. It has been shown that the severity of disease symptoms, age of onset and progression are related to the length of the triplet repeats. However, the mechanism(s) of CTG·CAG triplet-repeat instability is not fully understood. Herein, induced pluripotent stem cells (iPSCs) were generated from DM1 and Huntington's disease patient fibroblasts. We isolated 41 iPSC clones from DM1 fibroblasts, all showing different CTG·CAG repeat lengths, thus demonstrating somatic instability within the initial fibroblast population. During propagation of the iPSCs, the repeats expanded in a manner analogous to the expansion seen in somatic cells from DM1 patients. The correlation between repeat length and expansion rate identified the interval between 57 and 126 repeats as being an important length threshold where expansion rates dramatically increased. Moreover, longer repeats showed faster triplet-repeat expansion. However, the overall tendency of triplet repeats to expand ceased on differentiation into differentiated embryoid body or neurospheres. The mismatch repair components MSH2, MSH3 and MSH6 were highly expressed in iPSCs compared with fibroblasts, and only occupied the DMPK1 gene harboring longer CTG·CAG triplet repeats. In addition, shRNA silencing of MSH2 impeded CTG·CAG triplet-repeat expansion. The information gained from these studies provides new insight into a general mechanism of triplet-repeat expansion in iPSCs.


Asunto(s)
Distrofia Miotónica/genética , Células Madre Pluripotentes/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Expansión de Repetición de Trinucleótido/genética , Regiones no Traducidas 3'/genética , Técnicas de Cultivo de Célula , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteína 2 Homóloga a MutS/biosíntesis , Proteína 2 Homóloga a MutS/genética , Proteína 3 Homóloga de MutS , Distrofia Miotónica/patología , Proteína Quinasa de Distrofia Miotónica , Células Madre Pluripotentes/patología
9.
Ann Neurol ; 76(4): 489-508, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25159818

RESUMEN

OBJECTIVE: To investigate whether a histone deacetylase inhibitor (HDACi) would be effective in an in vitro model for the neurodegenerative disease Friedreich ataxia (FRDA) and to evaluate safety and surrogate markers of efficacy in a phase I clinical trial in patients. METHODS: We used a human FRDA neuronal cell model, derived from patient induced pluripotent stem cells, to determine the efficacy of a 2-aminobenzamide HDACi (109) as a modulator of FXN gene expression and chromatin histone modifications. FRDA patients were dosed in 4 cohorts, ranging from 30mg/day to 240mg/day of the formulated drug product of HDACi 109, RG2833. Patients were monitored for adverse effects as well as for increases in FXN mRNA, frataxin protein, and chromatin modification in blood cells. RESULTS: In the neuronal cell model, HDACi 109/RG2833 increases FXN mRNA levels and frataxin protein, with concomitant changes in the epigenetic state of the gene. Chromatin signatures indicate that histone H3 lysine 9 is a key residue for gene silencing through methylation and reactivation through acetylation, mediated by the HDACi. Drug treatment in FRDA patients demonstrated increased FXN mRNA and H3 lysine 9 acetylation in peripheral blood mononuclear cells. No safety issues were encountered. INTERPRETATION: Drug exposure inducing epigenetic changes in neurons in vitro is comparable to the exposure required in patients to see epigenetic changes in circulating lymphoid cells and increases in gene expression. These findings provide a proof of concept for the development of an epigenetic therapy for this fatal neurological disease.


Asunto(s)
Ataxia de Friedreich/tratamiento farmacológico , Ataxia de Friedreich/genética , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Proteínas de Unión a Hierro/genética , Administración Oral , Adolescente , Adulto , Aminocaproatos/farmacología , Aminocaproatos/uso terapéutico , Área Bajo la Curva , Benzamidas/farmacología , Benzamidas/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Transformada , Inmunoprecipitación de Cromatina , Estudios de Cohortes , Estudios Transversales , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Ataxia de Friedreich/patología , Regulación de la Expresión Génica/genética , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Células Madre Pluripotentes , Expansión de Repetición de Trinucleótido/genética , Adulto Joven , Frataxina
10.
J Proteome Res ; 13(11): 4558-66, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24933366

RESUMEN

Members of the 2-aminobenzamide class of histone deacetylase (HDAC) inhibitors show promise as therapeutics for the neurodegenerative diseases Friedreich's ataxia (FRDA) and Huntington's disease (HD). While it is clear that HDAC3 is one of the important targets of the 2-aminobenzamide HDAC inhibitors, inhibition of other class I HDACs (HDACs 1 and 2) may also be involved in the beneficial effects of these compounds in FRDA and HD, and other HDAC interacting proteins may be impacted by the compound. To this end, we synthesized activity-based profiling probe (ABPP) versions of one of our HDAC inhibitors (compound 106), and in the present study we used a quantitative proteomic method coupled with multidimensional protein identification technology (MudPIT) to identify the proteins captured by the ABPP 106 probe. Nuclear proteins were extracted from FRDA patient iPSC-derived neural stem cells, and then were reacted with control and ABPP 106 probe. After reaction, the bound proteins were digested on the beads, and the peptides were modified using stable isotope-labeled formaldehyde to form dimethyl amine. The selectively bound proteins determined by mass spectrometry were subjected to functional and pathway analysis. Our findings suggest that the targets of compound 106 are involved not only in transcriptional regulation but also in posttranscriptional processing of mRNA.


Asunto(s)
Ataxia de Friedreich/metabolismo , Inhibidores de Histona Desacetilasas/metabolismo , Células-Madre Neurales/metabolismo , Proteómica/métodos , Pirazoles/farmacología , Compuestos de Piridinio/farmacología , Procesamiento Postranscripcional del ARN/fisiología , ortoaminobenzoatos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Espectrometría de Masas , Sondas Moleculares/metabolismo , Sondas Moleculares/farmacología , Proteínas Nucleares/metabolismo , Pirazoles/metabolismo , Compuestos de Piridinio/metabolismo
11.
J Biol Chem ; 287(35): 29861-72, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22798143

RESUMEN

The genetic mutation in Friedreich ataxia (FRDA) is a hyperexpansion of the triplet-repeat sequence GAA·TTC within the first intron of the FXN gene. Although yeast and reporter construct models for GAA·TTC triplet-repeat expansion have been reported, studies on FRDA pathogenesis and therapeutic development are limited by the availability of an appropriate cell model in which to study the mechanism of instability of the GAA·TTC triplet repeats in the human genome. Herein, induced pluripotent stem cells (iPSCs) were generated from FRDA patient fibroblasts after transduction with the four transcription factors Oct4, Sox2, Klf4, and c-Myc. These cells were differentiated into neurospheres and neuronal precursors in vitro, providing a valuable cell model for FRDA. During propagation of the iPSCs, GAA·TTC triplet repeats expanded at a rate of about two GAA·TTC triplet repeats/replication. However, GAA·TTC triplet repeats were stable in FRDA fibroblasts and neuronal stem cells. The mismatch repair enzymes MSH2, MSH3, and MSH6, implicated in repeat instability in other triplet-repeat diseases, were highly expressed in pluripotent stem cells compared with fibroblasts and neuronal stem cells and occupied FXN intron 1. In addition, shRNA silencing of MSH2 and MSH6 impeded GAA·TTC triplet-repeat expansion. A specific pyrrole-imidazole polyamide targeting GAA·TTC triplet-repeat DNA partially blocked repeat expansion by displacing MSH2 from FXN intron 1 in FRDA iPSCs. These studies suggest that in FRDA, GAA·TTC triplet-repeat instability occurs in embryonic cells and involves the highly active mismatch repair system.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Ataxia de Friedreich/metabolismo , Genoma Humano , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de Unión a Hierro/metabolismo , Modelos Biológicos , Proteína 2 Homóloga a MutS/metabolismo , Expansión de Repetición de Trinucleótido , Animales , Diferenciación Celular/genética , Línea Celular , Fibroblastos/metabolismo , Fibroblastos/patología , Ataxia de Friedreich/genética , Ataxia de Friedreich/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Intrones/genética , Proteínas de Unión a Hierro/genética , Factor 4 Similar a Kruppel , Ratones , Proteína 2 Homóloga a MutS/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Frataxina
12.
J Neurochem ; 126 Suppl 1: 147-54, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23859350

RESUMEN

The genetic defect in Friedreich's ataxia (FRDA) is the expansion of a GAA·TCC triplet in the first intron of the FXN gene, which encodes the mitochondrial protein frataxin. Previous studies have established that the repeats reduce transcription of this essential gene, with a concomitant decrease in frataxin protein in affected individuals. As the repeats do not alter the FXN protein coding sequence, one therapeutic approach would be to increase transcription of pathogenic FXN genes. Histone posttranslational modifications near the expanded repeats are consistent with heterochromatin formation and FXN gene silencing. In an effort to find small molecules that would reactivate this silent gene, histone deacetylase inhibitors were screened for their ability to up-regulate FXN gene expression in patient cells and members of the pimelic 2-aminobenzamide family of class I histone deacetylase inhibitors were identified as potent inducers of FXN gene expression and frataxin protein. Importantly, these molecules up-regulate FXN expression in human neuronal cells derived from patient-induced pluripotent stem cells and in two mouse models for the disease. Preclinical studies of safety and toxicity have been completed for one such compound and a phase I clinical trial in FRDA patients has been initiated. Furthermore, medicinal chemistry efforts have identified improved compounds with superior pharmacological properties.


Asunto(s)
Expresión Génica/fisiología , Inhibidores de Histona Desacetilasas/uso terapéutico , Proteínas de Unión a Hierro/biosíntesis , Proteínas de Unión a Hierro/genética , Animales , Ensayos Clínicos como Asunto , Silenciador del Gen , Heterocromatina/metabolismo , Inhibidores de Histona Desacetilasas/química , Humanos , Proteínas de Unión a Hierro/efectos de los fármacos , Ratones , Relación Estructura-Actividad , Frataxina
13.
Neurobiol Dis ; 46(2): 351-61, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22590724

RESUMEN

We have previously demonstrated amelioration of Huntington's disease (HD)-related phenotypes in R6/2 transgenic mice in response to treatment with the novel histone deacetylase (HDAC) inhibitor 4b. Here we have measured the selectivity profiles of 4b and related compounds against class I and class II HDACs and have tested their ability to restore altered expression of genes related to HD pathology in mice and to rescue disease effects in cell culture and Drosophila models of HD. R6/2 transgenic and wild-type (wt) mice received daily injections of HDAC inhibitors for 3 days followed by real-time PCR analysis to detect expression differences for 13 HD-related genes. We find that HDACi 4b and 136, two compounds showing high potency for inhibiting HDAC3 were most effective in reversing the expression of genes relevant to HD, including Ppp1r1b, which encodes DARPP-32, a marker for medium spiny striatal neurons. In contrast, compounds targeting HDAC1 were less effective at correcting gene expression abnormalities in R6/2 transgenic mice, but did cause significant increases in the expression of selected genes. An additional panel of 4b-related compounds was tested in a Drosophila model of HD and in STHdhQ111 striatal cells to further distinguish HDAC selectivity. Significant improvement in huntingtin-elicited Drosophila eye neurodegeneration in the fly was observed in response to treatment with compounds targeting human HDAC1 and/or HDAC3. In STHdhQ111 striatal cells, the ability of HDAC inhibitors to improve huntingtin-elicited metabolic deficits correlated with the potency at inhibiting HDAC1 and HDAC3, although the IC50 values for HDAC1 inhibition were typically 10-fold higher than for inhibition of HDAC3. Assessment of HDAC protein localization in brain tissue by Western blot analysis revealed accumulation of HDAC1 and HDAC3 in the nucleus of HD transgenic mice compared to wt mice, with a concurrent decrease in cytoplasmic localization, suggesting that these HDACs contribute to a repressive chromatin environment in HD. No differences were detected in the localization of HDAC2, HDAC4 or HDAC7. These results suggest that inhibition of HDACs 1 and 3 can relieve HD-like phenotypes in model systems and that HDAC inhibitors targeting these isotypes might show therapeutic benefit in human HD.


Asunto(s)
Modelos Animales de Enfermedad , Histona Desacetilasa 1/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/administración & dosificación , Histona Desacetilasas/metabolismo , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/genética , Péptidos/fisiología , Fenotipo , Animales , Células Cultivadas , Drosophila melanogaster , Sistemas de Liberación de Medicamentos/métodos , Células HCT116 , Histona Desacetilasa 1/metabolismo , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos
14.
Ann Neurol ; 70(5): 790-804, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22162061

RESUMEN

OBJECTIVE: Gene expression studies in peripheral tissues from patients with neurodegenerative disorders can provide insights into disease pathogenesis, and identify potential biomarkers, an important goal of translational research in neurodegeneration. Friedreich Ataxia (FRDA) is a chronic neurodegenerative disease caused by reduced transcription of frataxin, a ubiquitously expressed protein. We studied in vitro lymphocytes from FRDA patients and carriers to identify a peripheral gene expression phenotype. Peripheral biomarkers related to disease status would be extremely valuable for assessing drug efficacy and could provide new pathophysiological insights. METHODS: We characterized the gene expression profiles in peripheral blood mononuclear cells (PBMCs) from FRDA patients, compared with controls and related carriers. Cells were studied both before and after in vitro treatment with compounds that increase frataxin levels. Quantitative real-time polymerase chain reaction and additional microarrays were used to confirm a core set of genes in multiple independent series. RESULTS: We identified a subset of genes changed in cells from patients with pathological frataxin deficiency, and a core set of these genes were confirmed in independent series. Changes in gene expression were related to the mitochondria, lipid metabolism, cell cycle, and DNA repair, consistent with FRDA's known pathophysiology. We evaluated the in vitro effect of multiple compounds (histone deacetylase inhibitors) on this putative biomarker set, and found that this biochemical phenotype was ameliorated in accordance with drug efficacy. INTERPRETATION: Frataxin downregulation is associated with robust changes in gene expression in PBMCs, providing pathogenetic insights and a core subset of genes that, if verified in vivo, could be used as a peripheral biomarker.


Asunto(s)
Ataxia de Friedreich/metabolismo , Proteínas de Unión a Hierro/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Adulto , Técnicas de Cultivo de Célula , Femenino , Ataxia de Friedreich/genética , Expresión Génica , Humanos , Proteínas de Unión a Hierro/genética , Leucocitos Mononucleares/citología , Masculino , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Frataxina
15.
Nat Chem Biol ; 6(1): 25-33, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19966789

RESUMEN

Chemical modulation of histone deacetylase (HDAC) activity by HDAC inhibitors (HDACi) is an increasingly important approach for modifying the etiology of human disease. Loss-of-function diseases arise as a consequence of protein misfolding and degradation, which lead to system failures. The DeltaF508 mutation in cystic fibrosis transmembrane conductance regulator (CFTR) results in the absence of the cell surface chloride channel and a loss of airway hydration, leading to the premature lung failure and reduced lifespan responsible for cystic fibrosis. We now show that the HDACi suberoylanilide hydroxamic acid (SAHA) restores surface channel activity in human primary airway epithelia to levels that are 28% of those of wild-type CFTR. Biological silencing of all known class I and II HDACs reveals that HDAC7 plays a central role in restoration of DeltaF508 function. We suggest that the tunable capacity of HDACs can be manipulated by chemical biology to counter the onset of cystic fibrosis and other human misfolding disorders.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Histona Desacetilasas/metabolismo , Mutación , Animales , Bronquios/metabolismo , Membrana Celular/metabolismo , Cricetinae , Células Epiteliales/metabolismo , Silenciador del Gen , Humanos , Ácidos Hidroxámicos/química , Desnaturalización Proteica , Pliegue de Proteína , ARN Interferente Pequeño/metabolismo , Vorinostat
16.
Bioorg Med Chem Lett ; 22(12): 4068-71, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22607671

RESUMEN

We have shown that a specific pyrrole-imidazole polyamide-DNA alkylator (chlorambucil) conjugate, 1R-Chl, alters the growth characteristics of various cancer cell lines in culture, and causes these cells to arrest in the G2/M stage of the cell cycle, without apparent cytotoxicity. This molecule has also shown efficacy in several mouse xenograft models, preventing tumor growth. Previous microarray studies have suggested that members of the histone H4 gene family, H4c and H4j/k, are the primary targets of this molecule, leading to reduced histone mRNA synthesis and growth arrest in cancer cells. In the present study, we examine the effects of 1R-Chl on transcription of other members of the H4 gene family, with the result that mRNA transcription of most genomic copies of H4 are down-regulated by 1R-Chl in a human pancreatic cancer cell line (MIA PaCa-2), but not in a cell line of non-cancerous origin (HEK293 cells). The basis for this differential effect is likely an open chromatin conformation within the H4 genes in cancer cells. Chromatin immunoprecipitation experiments show increased histone acetylation on the histone H4 genes in cancer cells, compared to HEK293 cells, explaining the differential activity of this molecule in cancer versus non-cancer cells.


Asunto(s)
Antineoplásicos Alquilantes/química , Clorambucilo/química , Cromatina/metabolismo , Histonas/genética , Nylons/química , Acetilación , Antineoplásicos Alquilantes/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Clorambucilo/farmacología , Cromatina/ultraestructura , Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Células HEK293 , Humanos , Imidazoles/química , Nylons/farmacología , Especificidad de Órganos , Neoplasias Pancreáticas , Pirroles/química , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Transcripción Genética/efectos de los fármacos
17.
Front Mol Biosci ; 9: 933788, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36133907

RESUMEN

Introduction: Friedreich ataxia (FRDA) is a recessive neurodegenerative disease characterized by progressive ataxia, dyscoordination, and loss of vision. The variable length of the pathogenic GAA triplet repeat expansion in the FXN gene in part explains the interindividual variability in the severity of disease. The GAA repeat expansion leads to epigenetic silencing of FXN; therefore, variability in properties of epigenetic effector proteins could also regulate the severity of FRDA. Methods: In an exploratory analysis, DNA from 88 individuals with FRDA was analyzed to determine if any of five non-synonymous SNPs in HDACs/SIRTs predicted FRDA disease severity. Results suggested the need for a full analysis at the rs352493 locus in SIRT6 (p.Asn46Ser). In a cohort of 569 subjects with FRDA, disease features were compared between subjects homozygous for the common thymine SIRT6 variant (TT) and those with the less common cytosine variant on one allele and thymine on the other (CT). The biochemical properties of both variants of SIRT6 were analyzed and compared. Results: Linear regression in the exploratory cohort suggested that an SNP (rs352493) in SIRT6 correlated with neurological severity in FRDA. The follow-up analysis in a larger cohort agreed with the initial result that the genotype of SIRT6 at the locus rs352493 predicted the severity of disease features of FRDA. Those in the CT SIRT6 group performed better on measures of neurological and visual function over time than those in the more common TT SIRT6 group. The Asn to Ser amino acid change resulting from the SNP in SIRT6 did not alter the expression or enzymatic activity of SIRT6 or frataxin, but iPSC-derived neurons from people with FRDA in the CT SIRT6 group showed whole transcriptome differences compared to those in the TT SIRT6 group. Conclusion: People with FRDA in the CT SIRT6 group have less severe neurological and visual dysfunction than those in the TT SIRT6 group. Biochemical analyses indicate that the benefit conferred by T to C SNP in SIRT6 does not come from altered expression or enzymatic activity of SIRT6 or frataxin but is associated with changes in the transcriptome.

18.
Neurobiol Dis ; 42(3): 496-505, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21397024

RESUMEN

Friedreich ataxia (FRDA) is an inherited neurodegenerative disorder caused by GAA repeat expansion within the FXN gene, leading to epigenetic changes and heterochromatin-mediated gene silencing that result in a frataxin protein deficit. Histone deacetylase (HDAC) inhibitors, including pimelic o-aminobenzamide compounds 106, 109 and 136, have previously been shown to reverse FXN gene silencing in short-term studies of FRDA patient cells and a knock-in mouse model, but the functional consequences of such therapeutic intervention have thus far not been described. We have now investigated the long-term therapeutic effects of 106, 109 and 136 in our GAA repeat expansion mutation-containing YG8R FRDA mouse model. We show that there is no overt toxicity up to 5 months of treatment and there is amelioration of the FRDA-like disease phenotype. Thus, while the neurological deficits of this model are mild, 109 and 106 both produced an improvement of motor coordination, whereas 109 and 136 produced increased locomotor activity. All three compounds increased global histone H3 and H4 acetylation of brain tissue, but only 109 significantly increased acetylation of specific histone residues at the FXN locus. Effects on FXN mRNA expression in CNS tissues were modest, but 109 significantly increased frataxin protein expression in brain tissue. 109 also produced significant increases in brain aconitase enzyme activity, together with reduction of neuronal pathology of the dorsal root ganglia (DRG). Overall, these results support further assessment of HDAC inhibitors for treatment of Friedreich ataxia.


Asunto(s)
Ataxia de Friedreich/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Actividad Motora/efectos de los fármacos , Aconitato Hidratasa/metabolismo , Análisis de Varianza , Animales , Benzamidas/farmacología , Benzamidas/uso terapéutico , Western Blotting , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Ataxia de Friedreich/genética , Ataxia de Friedreich/fisiopatología , Inhibidores de Histona Desacetilasas/farmacología , Ratones , Actividad Motora/fisiología , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Prueba de Desempeño de Rotación con Aceleración Constante
19.
Proc Natl Acad Sci U S A ; 105(40): 15564-9, 2008 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-18829438

RESUMEN

Transcriptional dysregulation has emerged as a core pathologic feature of Huntington's disease (HD), one of several triplet-repeat disorders characterized by movement deficits and cognitive dysfunction. Although the mechanisms contributing to the gene expression deficits remain unknown, therapeutic strategies have aimed to improve transcriptional output via modulation of chromatin structure. Recent studies have demonstrated therapeutic effects of commercially available histone deacetylase (HDAC) inhibitors in several HD models; however, the therapeutic value of these compounds is limited by their toxic effects. Here, beneficial effects of a novel pimelic diphenylamide HDAC inhibitor, HDACi 4b, in an HD mouse model are reported. Chronic oral administration of HDACi 4b, beginning after the onset of motor deficits, significantly improved motor performance, overall appearance, and body weight of symptomatic R6/2(300Q) transgenic mice. These effects were associated with significant attenuation of gross brain-size decline and striatal atrophy. Microarray studies revealed that HDACi 4b treatment ameliorated, in part, alterations in gene expression caused by the presence of mutant huntingtin protein in the striatum, cortex, and cerebellum of R6/2(300Q) transgenic mice. For selected genes, HDACi 4b treatment reversed histone H3 hypoacetylation observed in the presence of mutant huntingtin, in association with correction of mRNA expression levels. These findings suggest that HDACi 4b, and possibly related HDAC inhibitors, may offer clinical benefit for HD patients and provide a novel set of potential biomarkers for clinical assessment.


Asunto(s)
Anilidas/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Enfermedad de Huntington/genética , Transcripción Genética , Anilidas/administración & dosificación , Anilidas/síntesis química , Animales , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/síntesis química , Histona Desacetilasas/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Masculino , Ratones , Ratones Transgénicos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/farmacología , Fenotipo , Transcripción Genética/efectos de los fármacos
20.
Bioorg Med Chem ; 18(1): 168-74, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19962902

RESUMEN

We designed and synthesized conjugates between pyrrole-imidazole polyamides and seco-CBI that alkylate within the coding regions of the histone H4 genes. DNA alkylating activity on the histone H4 fragment and cellular effects against K562 chronic myelogenous leukemia cells were investigated. One of the conjugates, 5-CBI, showed strong DNA alkylation activity and good sequence specificity on a histone H4 gene fragment. K562 cells treated with 5-CBI down-regulated the histone H4 gene and induced apoptosis efficiently. Global gene expression data revealed that a number of histone H4 genes were down-regulated by 5-CBI treatment. These results suggest that sequence-specific DNA alkylating agents may have the potential of targeting specific genes for cancer chemotherapy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Genes/efectos de los fármacos , Histonas/genética , Imidazoles/farmacología , Nylons/farmacología , Pirroles/farmacología , Antineoplásicos Alquilantes/síntesis química , Antineoplásicos Alquilantes/química , Apoptosis/efectos de los fármacos , ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/síntesis química , Imidazoles/química , Células K562 , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Nylons/síntesis química , Nylons/química , Pirroles/síntesis química , Pirroles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA