Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
RNA ; 21(7): 1306-12, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26001796

RESUMEN

The scavenger decapping enzyme DcpS is a multifunctional protein initially identified by its property to hydrolyze the resulting cap structure following 3' end mRNA decay. In Saccharomyces cerevisiae, the DcpS homolog Dcs1 is an obligate cofactor for the 5'-3' exoribonuclease Xrn1 while the Caenorhabditis elegans homolog Dcs-1, facilitates Xrn1 mediated microRNA turnover. In both cases, this function is independent of the decapping activity. Whether DcpS and its decapping activity can affect mRNA steady state or stability in mammalian cells remains unknown. We sought to determine DcpS target genes in mammalian cells using a cell-permeable DcpS inhibitor compound, RG3039 initially developed for therapeutic treatment of spinal muscular atrophy. Global mRNA levels were examined following DcpS decapping inhibition with RG3039. The steady-state levels of 222 RNAs were altered upon RG3039 treatment. Of a subset selected for validation, two transcripts that appear to be long noncoding RNAs HS370762 and BC011766, were dependent on DcpS and its scavenger decapping catalytic activity and referred to as DcpS-responsive noncoding transcripts (DRNT) 1 and 2, respectively. Interestingly, only the increase in DRNT1 transcript was accompanied with an increase of its RNA stability and this increase was dependent on both DcpS and Xrn1. Importantly, unlike in yeast where the DcpS homolog is an obligate cofactor for Xrn1, stability of additional Xrn1 dependent RNAs were not altered by a reduction in DcpS levels. Collectively, our data demonstrate that DcpS in conjunction with Xrn1 has the potential to regulate RNA stability in a transcript-selective manner in mammalian cells.


Asunto(s)
Endorribonucleasas/fisiología , ARN Mensajero/genética , Secuencia de Bases , Línea Celular , Cartilla de ADN , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/genética , Humanos , Quinazolinas/farmacología , Estabilidad del ARN , ARN Mensajero/metabolismo , ARN no Traducido/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
2.
Blood ; 126(21): 2392-403, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26447190

RESUMEN

Histone deacetylase (HDAC) inhibitors (HDACis) have demonstrated activity in hematological and solid malignancies. Vorinostat, romidepsin, belinostat, and panobinostat are Food and Drug Administration-approved for hematological malignancies and inhibit class II and/or class I HDACs, including HDAC1, 2, 3, and 6. We combined genetic and pharmacological approaches to investigate whether suppression of individual or multiple Hdacs phenocopied broad-acting HDACis in 3 genetically distinct leukemias and lymphomas. Individual Hdacs were depleted in murine acute myeloid leukemias (MLL-AF9;Nras(G12D); PML-RARα acute promyelocytic leukemia [APL] cells) and Eµ-Myc lymphoma in vitro and in vivo. Strikingly, Hdac3-depleted cells were selected against in competitive assays for all 3 tumor types. Decreased proliferation following Hdac3 knockdown was not prevented by BCL-2 overexpression, caspase inhibition, or knockout of Cdkn1a in Eµ-Myc lymphoma, and depletion of Hdac3 in vivo significantly reduced tumor burden. Interestingly, APL cells depleted of Hdac3 demonstrated a more differentiated phenotype. Consistent with these genetic studies, the HDAC3 inhibitor RGFP966 reduced proliferation of Eµ-Myc lymphoma and induced differentiation in APL. Genetic codepletion of Hdac1 with Hdac2 was pro-apoptotic in Eµ-Myc lymphoma in vitro and in vivo and was phenocopied by the HDAC1/2-specific agent RGFP233. This study demonstrates the importance of HDAC3 for the proliferation of leukemia and lymphoma cells, suggesting that HDAC3-selective inhibitors could prove useful for the treatment of hematological malignancies. Moreover, our results demonstrate that codepletion of Hdac1 with Hdac2 mediates a robust pro-apoptotic response. Our integrated genetic and pharmacological approach provides important insights into the individual or combinations of HDACs that could be prioritized for targeting in a range of hematological malignancies.


Asunto(s)
Histona Desacetilasas/metabolismo , Leucemia Promielocítica Aguda/enzimología , Leucemia Promielocítica Aguda/genética , Linfoma/enzimología , Linfoma/genética , Animales , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/patología , Linfoma/tratamiento farmacológico , Linfoma/patología , Ratones , Células 3T3 NIH , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
3.
J Neurosci ; 35(38): 13124-32, 2015 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-26400942

RESUMEN

Research over the past decade indicates a novel role for epigenetic mechanisms in memory formation. Of particular interest is chromatin modification by histone deacetylases (HDACs), which, in general, negatively regulate transcription. HDAC deletion or inhibition facilitates transcription during memory consolidation and enhances long-lasting forms of synaptic plasticity and long-term memory. A key open question remains: How does blocking HDAC activity lead to memory enhancements? To address this question, we tested whether a normal function of HDACs is to gate information processing during memory formation. We used a class I HDAC inhibitor, RGFP966 (C21H19FN4O), to test the role of HDAC inhibition for information processing in an auditory memory model of learning-induced cortical plasticity. HDAC inhibition may act beyond memory enhancement per se to instead regulate information in ways that lead to encoding more vivid sensory details into memory. Indeed, we found that RGFP966 controls memory induction for acoustic details of sound-to-reward learning. Rats treated with RGFP966 while learning to associate sound with reward had stronger memory and additional information encoded into memory for highly specific features of sounds associated with reward. Moreover, behavioral effects occurred with unusually specific plasticity in primary auditory cortex (A1). Class I HDAC inhibition appears to engage A1 plasticity that enables additional acoustic features to become encoded in memory. Thus, epigenetic mechanisms act to regulate sensory cortical plasticity, which offers an information processing mechanism for gating what and how much is encoded to produce exceptionally persistent and vivid memories. Significance statement: Here we provide evidence of an epigenetic mechanism for information processing. The study reveals that a class I HDAC inhibitor (Malvaez et al., 2013; Rumbaugh et al., 2015; RGFP966, chemical formula C21H19FN4O) alters the formation of auditory memory by enabling more acoustic information to become encoded into memory. Moreover, RGFP966 appears to affect cortical plasticity: the primary auditory cortex reorganized in a manner that was unusually "tuned-in" to the specific sound cues and acoustic features that were related to reward and subsequently remembered. We propose that HDACs control "informational capture" at a systems level for what and how much information is encoded by gating sensory cortical plasticity that underlies the sensory richness of newly formed memories.


Asunto(s)
Corteza Auditiva/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Memoria/efectos de los fármacos , Acrilamidas/farmacología , Animales , Corteza Auditiva/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Potenciales Evocados/efectos de los fármacos , Masculino , Plasticidad Neuronal/efectos de los fármacos , Fenilendiaminas/farmacología , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Estadísticas no Paramétricas , Factores de Tiempo , Privación de Agua
4.
Proc Natl Acad Sci U S A ; 110(7): 2647-52, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23297220

RESUMEN

Nonspecific histone deacetylase (HDAC) inhibition has been shown to facilitate the extinction of drug-seeking behavior in a manner resistant to reinstatement. A key open question is which specific HDAC is involved in the extinction of drug-seeking behavior. Using the selective HDAC3 inhibitor RGFP966, we investigated the role of HDAC3 in extinction and found that systemic treatment with RGFP966 facilitates extinction in mice in a manner resistant to reinstatement. We also investigated whether the facilitated extinction is related to the enhancement of extinction consolidation during extinction learning or to negative effects on performance or reconsolidation. These are key distinctions with regard to any compound being used to modulate extinction, because a more rapid decrease in a defined behavior is interpreted as facilitated extinction. Using an innovative combination of behavioral paradigms, we found that a single treatment of RGFP966 enhances extinction of a previously established cocaine-conditioned place preference, while simultaneously enhancing long-term object-location memory within subjects. During extinction consolidation, HDAC3 inhibition promotes a distinct pattern of histone acetylation linked to gene expression within the infralimbic cortex, hippocampus, and nucleus accumbens. Thus, the facilitated extinction of drug-seeking cannot be explained by adverse effects on performance. These results demonstrate that HDAC3 inhibition enhances the memory processes involved in extinction of drug-seeking behavior.


Asunto(s)
Acrilamidas/farmacología , Cocaína , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Fenilendiaminas/farmacología , Acetilación/efectos de los fármacos , Acrilamidas/sangre , Acrilamidas/farmacocinética , Análisis de Varianza , Animales , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Inhibidores de Histona Desacetilasas/sangre , Inhibidores de Histona Desacetilasas/farmacocinética , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenilendiaminas/sangre , Fenilendiaminas/farmacocinética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo
5.
Hum Mol Genet ; 22(20): 4084-101, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23736298

RESUMEN

Spinal muscular atrophy (SMA) is caused by insufficient levels of the survival motor neuron (SMN) protein due to the functional loss of the SMN1 gene and the inability of its paralog, SMN2, to fully compensate due to reduced exon 7 splicing efficiency. Since SMA patients have at least one copy of SMN2, drug discovery campaigns have sought to identify SMN2 inducers. C5-substituted quinazolines increase SMN2 promoter activity in cell-based assays and a derivative, RG3039, has progressed to clinical testing. It is orally bioavailable, brain-penetrant and has been shown to be an inhibitor of the mRNA decapping enzyme, DcpS. Our pharmacological characterization of RG3039, reported here, demonstrates that RG3039 can extend survival and improve function in two SMA mouse models of varying disease severity (Taiwanese 5058 Hemi and 2B/- SMA mice), and positively impacts neuromuscular pathologies. In 2B/- SMA mice, RG3039 provided a >600% survival benefit (median 18 days to >112 days) when dosing began at P4, highlighting the importance of early intervention. We determined the minimum effective dose and the associated pharmacokinetic (PK) and exposure relationship of RG3039 and DcpS inhibition ex vivo. These data support the long PK half-life with extended pharmacodynamic outcome of RG3039 in 2B/- SMA mice. In motor neurons, RG3039 significantly increased both the average number of cells with gems and average number of gems per cell, which is used as an indirect measure of SMN levels. These studies contribute to dose selection and exposure estimates for the first studies with RG3039 in human subjects.


Asunto(s)
Endorribonucleasas/antagonistas & inhibidores , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Atrofia Muscular Espinal/fisiopatología , Quinazolinas/farmacología , Quinazolinas/farmacocinética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Administración Oral , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Endorribonucleasas/metabolismo , Femenino , Humanos , Ratones , Ratones Transgénicos , Atrofia Muscular Espinal/tratamiento farmacológico , Quinazolinas/uso terapéutico , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
6.
Hum Mol Genet ; 22(20): 4074-83, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23727836

RESUMEN

Spinal muscular atrophy (SMA) is caused by mutations of the survival motor neuron 1 (SMN1) gene, retention of the survival motor neuron 2 (SMN2) gene and insufficient expression of full-length survival motor neuron (SMN) protein. Quinazolines increase SMN2 promoter activity and inhibit the ribonucleic acid scavenger enzyme DcpS. The quinazoline derivative RG3039 has advanced to early phase clinical trials. In preparation for efficacy studies in SMA patients, we investigated the effects of RG3039 in severe SMA mice. Here, we show that RG3039 distributed to central nervous system tissues where it robustly inhibited DcpS enzyme activity, but minimally activated SMN expression or the assembly of small nuclear ribonucleoproteins. Nonetheless, treated SMA mice showed a dose-dependent increase in survival, weight and motor function. This was associated with improved motor neuron somal and neuromuscular junction synaptic innervation and function and increased muscle size. RG3039 also enhanced survival of conditional SMA mice in which SMN had been genetically restored to motor neurons. As this systemically delivered drug may have therapeutic benefits that extend beyond motor neurons, it could act additively with SMN-restoring therapies delivered directly to the central nervous system such as antisense oligonucleotides or gene therapy.


Asunto(s)
Endorribonucleasas/antagonistas & inhibidores , Neuronas Motoras/efectos de los fármacos , Atrofia Muscular Espinal/fisiopatología , Quinazolinas/farmacología , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Ratones Transgénicos , Neuronas Motoras/fisiología , Músculos/efectos de los fármacos , Músculos/metabolismo , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/fisiología , Quinazolinas/administración & dosificación , Quinazolinas/farmacocinética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Transmisión Sináptica
7.
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
8.
Neurobiol Learn Mem ; 114: 193-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24956240

RESUMEN

Non-selective inhibition of histone deacetylases (HDACs), enzymes that remove acetyl groups from histone core proteins, enhances cognition and NMDAR-dependent long-term potentiation at hippocampal CA3-CA1 synapses. It is not known whether this is a general mechanism by which HDACs modulate plasticity at other hippocampal synapses. Furthermore, it has yet to be tested whether HDAC inhibition can reverse deficits in synaptic plasticity in disease models. Here, we investigated whether inhibition of HDACs, and specifically HDAC3, a class I HDAC isoform known to negatively regulate hippocampus-dependent learning and memory, enhances LTP at medial perforant path-dentate granule cell (MPP-DGC) synapses in wild-type and Fragile X (Fmr1-/y) mice, a model with known LTP deficits at this synapse. The non-selective HDAC inhibitor trichostatin A (TSA) significantly increased the magnitude of LTP at MPP-DGC synapses in wild-type mice, similar to reports at CA3-CA1 synapses. The enhancement of LTP was mimicked by selective HDAC3 inhibition, implicating a role for this isoform in the negative regulation of synaptic plasticity. However, HDAC3 inhibition was completely ineffective in reversing the deficit in LTP at MPP-DGC synapses in slices from Fmr1-/y mice, and in fact, HDAC3 inhibition was unable to induce any improvement whatsoever. These findings indicate that the enhancing effect of HDAC3 inhibition on LTP in wild-type mice requires FMRP, revealing a novel role for FMRP in hippocampal plasticity.


Asunto(s)
Giro Dentado/fisiología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Inhibidores de Histona Desacetilasas/farmacología , Potenciación a Largo Plazo/fisiología , Vía Perforante/fisiología , Sinapsis/fisiología , Animales , Giro Dentado/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Histona Desacetilasas/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/fisiología , Vía Perforante/efectos de los fármacos , Sinapsis/efectos de los fármacos
9.
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
10.
J Neurosci ; 31(2): 764-74, 2011 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-21228185

RESUMEN

Gene expression is dynamically regulated by chromatin modifications on histone tails, such as acetylation. In general, histone acetylation promotes transcription, whereas histone deacetylation negatively regulates transcription. The interplay between histone acetyltranserases and histone deacetylases (HDACs) is pivotal for the regulation of gene expression required for long-term memory processes. Currently, very little is known about the role of individual HDACs in learning and memory. We examined the role of HDAC3 in long-term memory using a combined genetic and pharmacologic approach. We used HDAC3-FLOX genetically modified mice in combination with adeno-associated virus-expressing Cre recombinase to generate focal homozygous deletions of Hdac3 in area CA1 of the dorsal hippocampus. To complement this approach, we also used a selective inhibitor of HDAC3, RGFP136 [N-(6-(2-amino-4-fluorophenylamino)-6-oxohexyl)-4-methylbenzamide]. Immunohistochemistry showed that focal deletion or intrahippocampal delivery of RGFP136 resulted in increased histone acetylation. Both the focal deletion of HDAC3 as well as HDAC3 inhibition via RGFP136 significantly enhanced long-term memory in a persistent manner. Next we examined expression of genes implicated in long-term memory from dorsal hippocampal punches using quantitative reverse transcription-PCR. Expression of nuclear receptor subfamily 4 group A, member 2 (Nr4a2) and c-fos was significantly increased in the hippocampus of HDAC3-FLOX mice compared with wild-type controls. Memory enhancements observed in HDAC3-FLOX mice were abolished by intrahippocampal delivery of Nr4a2 small interfering RNA, suggesting a mechanism by which HDAC3 negatively regulates memory formation. Together, these findings demonstrate a critical role for HDAC3 in the molecular mechanisms underlying long-term memory formation.


Asunto(s)
Benzamidas/farmacología , Histona Desacetilasas/fisiología , Memoria a Largo Plazo/fisiología , Acetilación , Animales , Hipocampo/enzimología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Histonas/metabolismo , Memoria a Largo Plazo/efectos de los fármacos , Ratones , Ratones Mutantes , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/biosíntesis , Eliminación de Secuencia , Percepción Espacial/efectos de los fármacos , Percepción Espacial/fisiología
11.
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
12.
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
13.
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
14.
Amyotroph Lateral Scler ; 11(6): 520-30, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20565334

RESUMEN

There is strong evidence from studies in humans and animal models to suggest the involvement of energy metabolism defects in neurodegenerative diseases. Uridine, a pyrimidine nucleoside, has been suggested to be neuroprotective in neurological disorders by improving bioenergetic effects, increasing ATP levels and enhancing glycolytic energy production. We assessed whether uridine treatment extended survival and improved the behavioral and neuropathological phenotype observed in G93A-ALS mice. In vitro and in vivo pharmacokinetic analyses in mutant SOD models provided optimal dose and assurance that uridine entered the brain. A dose-ranging efficacy trial in G93A mice was performed using survival, body weight, open-field analysis, and neuropathology as outcome measures. Urinary levels of 8-hydroxy-2'-deoxyguanosine, identifying DNA oxidative damage, were measured and used as a pharmacodynamic biomarker. Uridine administration significantly extended survival in a dose-dependent manner in G93A mice, while improving the behavioral and neuropathological phenotype. Uridine increased survival by 17.4%, ameliorated body weight loss, enhanced motor performance, reduced gross lumbar and ventral horn atrophy, attenuated lumbar ventral horn neuronal cell death, and decreased reactive astrogliosis. Consistent with a therapeutic effect, uridine significantly reduced urinary 8-hydroxy-2'-deoxyguanosine in G93A mice. These data suggest that uridine may be a therapeutic candidate in ALS patients.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/fisiopatología , Fármacos Neuroprotectores/uso terapéutico , Superóxido Dismutasa/metabolismo , Uridina/uso terapéutico , 8-Hidroxi-2'-Desoxicoguanosina , Esclerosis Amiotrófica Lateral/genética , Animales , Células del Asta Anterior/efectos de los fármacos , Células del Asta Anterior/metabolismo , Células del Asta Anterior/patología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Peso Corporal/efectos de los fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/orina , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Metabolismo Energético/fisiología , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fármacos Neuroprotectores/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Superóxido Dismutasa/genética , Tasa de Supervivencia , Uridina/farmacología
15.
Epilepsy Behav ; 13(1): 47-51, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18321784

RESUMEN

The anticonvulsant effect of the nucleoside uridine has been studied for several decades with controversial results. One of its attractive properties is that as a natural endogenous molecule, it lacks the serious side effects of common antiepileptic drugs used today. In the current study, we examined the potential antiepileptogenic effect of uridine in the hippocampal kindling model, using once-daily stimulations. Uridine was administered once or three times daily; levetiracetam was administered as a positive control; and normal saline was used as a negative control. Rats receiving uridine or levetiracetam had slower kindling rates and shorter afterdischarge durations than the normal saline controls. These results are consistent with previous work using a rapid kindling model and suggest that uridine has antiepileptogenic properties. Because of its combination of low toxicity and efficacy, uridine is a possible candidate for the treatment of epilepsy.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Excitación Neurológica/efectos de los fármacos , Uridina/administración & dosificación , Análisis de Varianza , Animales , Estimulación Eléctrica/efectos adversos , Electroencefalografía/efectos de los fármacos , Electroencefalografía/efectos de la radiación , Hipocampo/efectos de los fármacos , Hipocampo/efectos de la radiación , Levetiracetam , Masculino , Piracetam/administración & dosificación , Piracetam/análogos & derivados , Ratas , Ratas Sprague-Dawley
16.
Exp Clin Psychopharmacol ; 16(3): 191-8, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18540778

RESUMEN

It has recently been reported that secretin activates gene expression in the central nucleus of the amygdala in rats. To examine the neurophysiological effects of secretin on amygdalar activation in humans, the authors measured Blood Oxygen Level Dependent functional magnetic resonance imaging signal change during facial affect processing in a placebo-controlled double-blind study. The authors studied 12 healthy male subjects who were presented with three stimulus conditions: viewing happy, fearful, and neutral faces, before and after infusion with either secretin or placebo. To test whether treatment was associated with distinct patterns of activation, the two conditions (Pre and Post) were subjected to a subtraction analyses in SPM99 and hypotheses regarding the activation of the left and right amygdala were tested using a region-of-interest approach. Subtraction of treatment minus baseline activation during the fear condition yielded significant (p=.001) activation in the right amygdala and a nonsignificant increase in activation in the left amygdala. No significant differences were seen between the treatment conditions for the amygdala when viewing happy or neutral faces. These preliminary findings indicate that secretin may alter responsivity to affective stimuli. The presence of increased activation of the amygdala during the viewing of fearful faces is consistent with findings from animal studies and suggests a mechanism by which secretin may modulate social behavior.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Secretina/farmacología , Adulto , Emociones/fisiología , Lateralidad Funcional/efectos de los fármacos , Lateralidad Funcional/fisiología , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Neuronas/fisiología , Oxígeno/sangre , Percepción Social , Estimulación Química
17.
Exp Clin Psychopharmacol ; 16(3): 199-206, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18540779

RESUMEN

Eleven patients with bipolar depression were given doses of up to 18 g per day of triacetyluridine (TAU) over 6 weeks to test the effect of uridine on symptoms of depression via Montgomery-Asberg Depression Rating Scale (MADRS; Asberg, Montgomery, Perris, Schalling, & Sedvall, 1978) scores and on cellular bioenergetics using phosphorus magnetic resonance spectroscopic imaging (31P-MRSI). All patients and comparison participants (n = 9) completed baseline 31P-MRSI scans, and 9 patients completed posttherapy scans. The percentage changes for MADRS scores (Week 2, -23.8; Week 3, -34.9; Week 4, -42.5) and the time effects of TAU on MADRS scores (Week 2, z = -2.07, p = .039; Week 3, z = -4.28, p < .001; Week 4, z = -4.54, p < .001) may reflect TAU effects on early symptom improvement. TAU responders (patients who had a 50% or greater reduction in MADRS scores from baseline at any time) demonstrated a significant difference from nonresponders in pH changes from baseline (effect size = 150). These results suggest that TAU treatment may decrease symptoms of depression and improve mitochondrial functioning.


Asunto(s)
Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/psicología , Química Encefálica/efectos de los fármacos , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/psicología , Uridina/análogos & derivados , Acetatos , Adulto , Trastorno Bipolar/metabolismo , Trastorno Depresivo/metabolismo , Femenino , Humanos , Concentración de Iones de Hidrógeno , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Masculino , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Pruebas Neuropsicológicas , Fosfocreatina/metabolismo , Escalas de Valoración Psiquiátrica , Uridina/farmacología , Uridina/uso terapéutico
18.
Epilepsy Res ; 70(1): 73-82, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16621451

RESUMEN

Due to the limited efficacy and side effects of current antiepileptic drugs (AEDs), the search for new therapeutic agents is critical. Uridine, a possible endogenous antiepileptic modulator, has been demonstrated to have anticonvulsant effects in some models of epilepsy, but not others. In this study, we examined possible neuroprotective effects of uridine by administering the agent following lithium-pilocarpine induced status epilepticus. The effects of uridine were assessed on EEG patterns, visual-spatial memory in the water maze and histopathology. There was a trend for reduced EEG spike frequency, improved visual spatial memory and better histology score in rats receiving uridine. The antiepileptogenic and anticonvulsant effects of uridine were studied by administering uridine to rats undergoing rapid kindling or following full kindling. In the rapid kindling models, uridine had a moderate antiepileptogenic and anticonvulsant effect. These results suggest uridine may have potential to aid in the prevention and treatment of epilepsy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Hipocampo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Estado Epiléptico/tratamiento farmacológico , Uridina/uso terapéutico , Animales , Anticonvulsivantes/efectos adversos , Anticonvulsivantes/farmacología , Conducta Animal , Modelos Animales de Enfermedad , Electroencefalografía/efectos de los fármacos , Hipocampo/patología , Excitación Neurológica , Litio , Masculino , Pilocarpina , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Uridina/efectos adversos , Uridina/farmacología , Agua
19.
PLoS One ; 11(3): e0152498, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27031333

RESUMEN

An important epigenetic modification in Huntington's disease (HD) research is histone acetylation, which is regulated by histone acetyltransferase and histone deacetylase (HDAC) enzymes. HDAC inhibitors have proven effective in HD model systems, and recent work is now focused on functional dissection of the individual HDAC enzymes in these effects. Histone deacetylase 3 (HDAC3), a member of the class I subfamily of HDACs, has previously been implicated in neuronal toxicity and huntingtin-induced cell death. Hence, we tested the effects of RGFP966 ((E)-N-(2-amino-4-fluorophenyl)-3-(1-cinnamyl-1H-pyrazol-4-yl)acrylamide), a benzamide-type HDAC inhibitor that selectively targets HDAC3, in the N171-82Q transgenic mouse model of HD. We found that RGFP966 at doses of 10 and 25 mg/kg improves motor deficits on rotarod and in open field exploration, accompanied by neuroprotective effects on striatal volume. In light of previous studies implicating HDAC3 in immune function, we measured gene expression changes for 84 immune-related genes elicited by RGFP966 using quantitative PCR arrays. RGFP966 treatment did not cause widespread changes in cytokine/chemokine gene expression patterns, but did significantly alter the striatal expression of macrophage migration inhibitory factor (Mif), a hormone immune modulator associated with glial cell activation, in N171-82Q transgenic mice, but not WT mice. Accordingly, RGFP966-treated mice showed decreased glial fibrillary acidic protein (GFAP) immunoreactivity, a marker of astrocyte activation, in the striatum of N171-82Q transgenic mice compared to vehicle-treated mice. These findings suggest that the beneficial actions of HDAC3 inhibition could be related, in part, with lowered Mif levels and its associated downstream effects.


Asunto(s)
Histona Desacetilasas/química , Enfermedad de Huntington/patología , Acrilamidas/farmacología , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Enfermedad de Huntington/metabolismo , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fenilendiaminas/farmacología , ARN Mensajero/metabolismo
20.
Biol Psychiatry ; 58(1): 67-73, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15992525

RESUMEN

BACKGROUND: Secretin is a "gut-brain" peptide whose neural function is as yet poorly understood. Several clinical studies have reported modestly increased social interaction in autistic children following intravenous secretin administration. Very recently secretin also was administered to schizophrenic patients and found to increase social interaction in some individuals. METHODS: In light of this finding, we assessed the ability of secretin to reverse phencyclidine- (PCP) induced impairment in prepulse inhibition (PPI), a leading animal model of sensorimotor gating deficits in schizophrenia. RESULTS: Similar to atypical antipsychotics, secretin (1, 3, 10, 30, and 100 microg/kg) partially and dose-dependently reversed the PCP-induced deficit in PPI without significantly affecting baseline startle when administered intraperitoneally (IP) 10 minutes following IP administration of PCP (3 mg/kg). CONCLUSIONS: This finding may be relevant to observations of antipsychotic efficacy of secretin in schizophrenic patients as well as our previous report that systemically administered secretin is capable of modulating conditioned fear, even at quite low doses.


Asunto(s)
Conducta Animal/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Fenciclidina/farmacología , Reflejo de Sobresalto/efectos de los fármacos , Secretina/farmacología , Estimulación Acústica , Animales , Percepción Auditiva/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Miedo/efectos de los fármacos , Miedo/fisiología , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Esquizofrenia/tratamiento farmacológico , Psicología del Esquizofrénico , Secretina/administración & dosificación , Secretina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA