Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 23(11): 2995-3007, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24436303

RESUMEN

Protein acetylation, which is central to transcriptional control as well as other cellular processes, is disrupted in Huntington's disease (HD). Treatments that restore global acetylation levels, such as inhibiting histone deacetylases (HDACs), are effective in suppressing HD pathology in model organisms. However, agents that selectively target the disease-relevant HDACs have not been available. SirT1 (Sir2 in Drosophila melanogaster) deacetylates histones and other proteins including transcription factors. Genetically reducing, but not eliminating, Sir2 has been shown to suppress HD pathology in model organisms. To date, small molecule inhibitors of sirtuins have exhibited low potency and unattractive pharmacological and biopharmaceutical properties. Here, we show that highly selective pharmacological inhibition of Drosophila Sir2 and mammalian SirT1 using the novel inhibitor selisistat (selisistat; 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide) can suppress HD pathology caused by mutant huntingtin exon 1 fragments in Drosophila, mammalian cells and mice. We have validated Sir2 as the in vivo target of selisistat by showing that genetic elimination of Sir2 eradicates the effect of this inhibitor in Drosophila. The specificity of selisistat is shown by its effect on recombinant sirtuins in mammalian cells. Reduction of HD pathology by selisistat in Drosophila, mammalian cells and mouse models of HD suggests that this inhibitor has potential as an effective therapeutic treatment for human disease and may also serve as a tool to better understand the downstream pathways of SirT1/Sir2 that may be critical for HD.


Asunto(s)
Carbazoles/administración & dosificación , Proteínas de Drosophila/antagonistas & inhibidores , Inhibidores Enzimáticos/administración & dosificación , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/enzimología , Sirtuina 1/antagonistas & inhibidores , Sirtuinas/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Células PC12 , Ratas , Ratas Sprague-Dawley , Sirtuina 1/genética , Sirtuina 1/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo
2.
PLoS Biol ; 11(11): e1001717, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24302884

RESUMEN

Histone deacetylase (HDAC) 4 is a transcriptional repressor that contains a glutamine-rich domain. We hypothesised that it may be involved in the molecular pathogenesis of Huntington's disease (HD), a protein-folding neurodegenerative disorder caused by an aggregation-prone polyglutamine expansion in the huntingtin protein. We found that HDAC4 associates with huntingtin in a polyglutamine-length-dependent manner and co-localises with cytoplasmic inclusions. We show that HDAC4 reduction delayed cytoplasmic aggregate formation, restored Bdnf transcript levels, and rescued neuronal and cortico-striatal synaptic function in HD mouse models. This was accompanied by an improvement in motor coordination, neurological phenotypes, and increased lifespan. Surprisingly, HDAC4 reduction had no effect on global transcriptional dysfunction and did not modulate nuclear huntingtin aggregation. Our results define a crucial role for the cytoplasmic aggregation process in the molecular pathology of HD. HDAC4 reduction presents a novel strategy for targeting huntingtin aggregation, which may be amenable to small-molecule therapeutics.


Asunto(s)
Histona Desacetilasas/genética , Enfermedad de Huntington/enzimología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/enzimología , Corteza Cerebral/patología , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/metabolismo , Proteína Huntingtina , Enfermedad de Huntington/fisiopatología , Enfermedad de Huntington/terapia , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Neuronas/fisiología , Fenotipo , Prueba de Desempeño de Rotación con Aceleración Constante , Transmisión Sináptica , Transcripción Genética
3.
Brain ; 137(Pt 3): 819-33, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24459107

RESUMEN

Huntington's disease is an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. The peripheral innate immune system contributes to Huntington's disease pathogenesis and has been targeted successfully to modulate disease progression, but mechanistic understanding relating this to mutant huntingtin expression in immune cells has been lacking. Here we demonstrate that human Huntington's disease myeloid cells produce excessive inflammatory cytokines as a result of the cell-intrinsic effects of mutant huntingtin expression. A direct effect of mutant huntingtin on the NFκB pathway, whereby it interacts with IKKγ, leads to increased degradation of IκB and subsequent nuclear translocation of RelA. Transcriptional alterations in intracellular immune signalling pathways are also observed. Using a novel method of small interfering RNA delivery to lower huntingtin expression, we show reversal of disease-associated alterations in cellular function-the first time this has been demonstrated in primary human cells. Glucan-encapsulated small interfering RNA particles were used to lower huntingtin levels in human Huntington's disease monocytes/macrophages, resulting in a reversal of huntingtin-induced elevated cytokine production and transcriptional changes. These findings improve our understanding of the role of innate immunity in neurodegeneration, introduce glucan-encapsulated small interfering RNA particles as tool for studying cellular pathogenesis ex vivo in human cells and raise the prospect of immune cell-directed HTT-lowering as a therapeutic in Huntington's disease.


Asunto(s)
Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Células Mieloides/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Transducción de Señal/genética , Regulación de la Expresión Génica/inmunología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Inmunidad Innata/genética , Células Mieloides/inmunología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , ARN Interferente Pequeño/uso terapéutico , Transducción de Señal/inmunología , Células U937
4.
J Neurosci ; 33(12): 5127-37, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23516279

RESUMEN

MicroRNAs (miRNAs) have been shown to play important roles in both brain development and the regulation of adult neural cell functions. However, a systematic analysis of brain miRNA functions has been hindered by a lack of comprehensive information regarding the distribution of miRNAs in neuronal versus glial cells. To address this issue, we performed microarray analyses of miRNA expression in the four principal cell types of the CNS (neurons, astrocytes, oligodendrocytes, and microglia) using primary cultures from postnatal d 1 rat cortex. These analyses revealed that neural miRNA expression is highly cell-type specific, with 116 of the 351 miRNAs examined being differentially expressed fivefold or more across the four cell types. We also demonstrate that individual neuron-enriched or neuron-diminished RNAs had a significant impact on the specification of neuronal phenotype: overexpression of the neuron-enriched miRNAs miR-376a and miR-434 increased the differentiation of neural stem cells into neurons, whereas the opposite effect was observed for the glia-enriched miRNAs miR-223, miR-146a, miR-19, and miR-32. In addition, glia-enriched miRNAs were shown to inhibit aberrant glial expression of neuronal proteins and phenotypes, as exemplified by miR-146a, which inhibited neuroligin 1-dependent synaptogenesis. This study identifies new nervous system functions of specific miRNAs, reveals the global extent to which the brain may use differential miRNA expression to regulate neural cell-type-specific phenotypes, and provides an important data resource that defines the compartmentalization of brain miRNAs across different cell types.


Asunto(s)
MicroARNs/fisiología , Células-Madre Neurales/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Transcriptoma , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/fisiología , Diferenciación Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/fisiología , Femenino , Masculino , MicroARNs/genética , Microglía/citología , Microglía/fisiología , Células-Madre Neurales/citología , Neuroglía/citología , Neuronas/citología , Oligodendroglía/citología , Oligodendroglía/fisiología , Fenotipo , Cultivo Primario de Células , Ratas , Sinapsis/fisiología
5.
J Neurosci ; 33(6): 2313-25, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23392662

RESUMEN

We previously demonstrated that sodium butyrate is neuroprotective in Huntington's disease (HD) mice and that this therapeutic effect is associated with increased expression of mitogen-activated protein kinase/dual-specificity phosphatase 1 (MKP-1/DUSP1). Here we show that enhancing MKP-1 expression is sufficient to achieve neuroprotection in lentiviral models of HD. Wild-type MKP-1 overexpression inhibited apoptosis in primary striatal neurons exposed to an N-terminal fragment of polyglutamine-expanded huntingtin (Htt171-82Q), blocking caspase-3 activation and significantly reducing neuronal cell death. This neuroprotective effect of MKP-1 was demonstrated to be dependent on its enzymatic activity, being ablated by mutation of its phosphatase domain and being attributed to inhibition of specific MAP kinases (MAPKs). Overexpression of MKP-1 prevented the polyglutamine-expanded huntingtin-induced activation of c-Jun N-terminal kinases (JNKs) and p38 MAPKs, whereas extracellular signal-regulated kinase (ERK) 1/2 activation was not altered by either polyglutamine-expanded Htt or MKP-1. Moreover, mutants of MKP-1 that selectively prevented p38 or JNK binding confirmed the important dual contributions of p38 and JNK regulation to MKP-1-mediated neuroprotection. These results demonstrate additive effects of p38 and JNK MAPK inhibition by MKP-1 without consequence to ERK activation in this striatal neuron-based paradigm. MKP-1 also provided neuroprotection in vivo in a lentiviral model of HD neuropathology in rat striatum. Together, these data extend previous evidence that JNK- and p38-mediated pathways contribute to HD pathogenesis and, importantly, show that therapies simultaneously inhibiting both JNK and p38 signaling pathways may lead to improved neuroprotective outcomes.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/biosíntesis , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/prevención & control , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Fármacos Neuroprotectores/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Células Cultivadas , Femenino , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratas , Ratas Wistar , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Nat Methods ; 8(11): 945-7, 2011 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-21983921

RESUMEN

Human diseases are often accompanied by histological changes that confound interpretation of molecular analyses and identification of disease-related effects. We developed population-specific expression analysis (PSEA), a computational method of analyzing gene expression in samples of varying composition that can improve analyses of quantitative molecular data in many biological contexts. PSEA of brains from individuals with Huntington's disease revealed myelin-related abnormalities that were undetected using standard differential expression analysis.


Asunto(s)
Encefalopatías/genética , Perfilación de la Expresión Génica , Humanos
7.
Hum Mol Genet ; 20(6): 1049-60, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21177255

RESUMEN

Huntington's disease (HD), caused by a CAG repeat expansion in the huntingtin (HTT) gene, is characterized by abnormal protein aggregates and motor and cognitive dysfunction. Htt protein is ubiquitously expressed, but the striatal medium spiny neuron (MSN) is most susceptible to dysfunction and death. Abnormal gene expression represents a core pathogenic feature of HD, but the relative roles of cell-autonomous and non-cell-autonomous effects on transcription remain unclear. To determine the extent of cell-autonomous dysregulation in the striatum in vivo, we examined genome-wide RNA expression in symptomatic D9-N171-98Q (a.k.a. DE5) transgenic mice in which the forebrain expression of the first 171 amino acids of human Htt with a 98Q repeat expansion is limited to MSNs. Microarray data generated from these mice were compared with those generated on the identical array platform from a pan-neuronal HD mouse model, R6/2, carrying two different CAG repeat lengths, and a relatively high degree of overlap of changes in gene expression was revealed. We further focused on known canonical pathways associated with excitotoxicity, oxidative stress, mitochondrial dysfunction, dopamine signaling and trophic support. While genes related to excitotoxicity, dopamine signaling and trophic support were altered in both DE5 and R6/2 mice, which may be either cell autonomous or non-cell autonomous, genes related to mitochondrial dysfunction, oxidative stress and the peroxisome proliferator-activated receptor are primarily affected in DE5 transgenic mice, indicating cell-autonomous mechanisms. Overall, HD-induced dysregulation of the striatal transcriptome can be largely attributed to intrinsic effects of mutant Htt, in the absence of expression in cortical neurons.


Asunto(s)
Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Enfermedad de Huntington/genética , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transcripción Genética , Animales , Células Cultivadas , Corteza Cerebral/citología , Cuerpo Estriado/citología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Masculino , Ratones , Ratones Transgénicos , Especificidad de Órganos , Expansión de Repetición de Trinucleótido
8.
Proc Natl Acad Sci U S A ; 107(17): 7927-32, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20378838

RESUMEN

Huntington's disease (HD), an incurable neurodegenerative disorder, has a complex pathogenesis including protein aggregation and the dysregulation of neuronal transcription and metabolism. Here, we demonstrate that inhibition of sirtuin 2 (SIRT2) achieves neuroprotection in cellular and invertebrate models of HD. Genetic or pharmacologic inhibition of SIRT2 in a striatal neuron model of HD resulted in gene expression changes including significant down-regulation of RNAs responsible for sterol biosynthesis. Whereas mutant huntingtin fragments increased sterols in neuronal cells, SIRT2 inhibition reduced sterol levels via decreased nuclear trafficking of SREBP-2. Importantly, manipulation of sterol biosynthesis at the transcriptional level mimicked SIRT2 inhibition, demonstrating that the metabolic effects of SIRT2 inhibition are sufficient to diminish mutant huntingtin toxicity. These data identify SIRT2 inhibition as a promising avenue for HD therapy and elucidate a unique mechanism of SIRT2-inhibitor-mediated neuroprotection. Furthermore, the ascertainment of SIRT2's role in regulating cellular metabolism demonstrates a central function shared with other sirtuin proteins.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedad de Huntington/prevención & control , Fármacos Neuroprotectores/farmacología , Sirtuina 2/antagonistas & inhibidores , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Esteroles/biosíntesis , Análisis de Varianza , Animales , Western Blotting , Caenorhabditis elegans , Drosophila , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratones , Microscopía Confocal
9.
Nat Commun ; 14(1): 6750, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37891183

RESUMEN

A positron emission tomography (PET) tracer detecting α-synuclein pathology will improve the diagnosis, and ultimately the treatment of α-synuclein-related diseases. Here we show that the PET ligand, [18F]ACI-12589, displays good in vitro affinity and specificity for pathological α-synuclein in tissues from patients with different α-synuclein-related disorders including Parkinson's disease (PD) and Multiple-System Atrophy (MSA) using autoradiography and radiobinding techniques. In the initial clinical evaluation we include 23 participants with α-synuclein related disorders, 11 with other neurodegenerative disorders and eight controls. In vivo [18F]ACI-12589 demonstrates clear binding in the cerebellar white matter and middle cerebellar peduncles of MSA patients, regions known to be highly affected by α-synuclein pathology, but shows limited binding in PD. The binding statistically separates MSA patients from healthy controls and subjects with other neurodegenerative disorders, including other synucleinopathies. Our results indicate that α-synuclein pathology in MSA can be identified using [18F]ACI-12589 PET imaging, potentially improving the diagnostic work-up of MSA and allowing for detection of drug target engagement in vivo of novel α-synuclein targeting therapies.


Asunto(s)
Atrofia de Múltiples Sistemas , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/metabolismo , Atrofia de Múltiples Sistemas/metabolismo , Enfermedad de Parkinson/metabolismo , Tomografía de Emisión de Positrones
10.
Brain Commun ; 5(6): fcad306, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38025276

RESUMEN

In amyotrophic lateral sclerosis, a disease driven by abnormal transactive response DNA-binding protein of 43 kDa aggregation, CSF may contain pathological species of transactive response DNA-binding protein of 43 kDa contributing to the propagation of pathology and neuronal toxicity. These species, released in part by degenerating neurons, would act as a template for the aggregation of physiological protein contributing to the spread of pathology in the brain and spinal cord. In this study, a robust seed amplification assay was established to assess the presence of seeding-competent transactive response DNA-binding protein of 43 kDa species in CSF of apparently sporadic amyotrophic lateral sclerosis patients. These samples resulted in a significant acceleration of substrate aggregation differentiating the kinetics from healthy controls. In parallel, a second assay was developed to determine the level of target engagement that would be necessary to neutralize such species in human CSF by a therapeutic monoclonal antibody targeting transactive response DNA-binding protein of 43 kDa. For this, evaluation of the pharmacokinetic/pharmacodynamic effect for the monoclonal antibody, ACI-5891.9, in vivo and in vitro confirmed that a CSF concentration of ≍1100 ng/mL would be sufficient for sustained target saturation. Using this concentration in the seed amplification assay, ACI-5891.9 was able to neutralize the transactive response DNA-binding protein of 43 kDa pathogenic seeds derived from amyotrophic lateral sclerosis patient CSF. This translational work adds to the evidence of transmission of transactive response DNA-binding protein of 43 kDa pathology via CSF that could contribute to the non-contiguous pattern of clinical manifestations observed in amyotrophic lateral sclerosis and demonstrates the ability of a therapeutic monoclonal antibody to neutralize the toxic, extracellular seeding-competent transactive response DNA-binding protein of 43 kDa species in the CSF of apparently sporadic amyotrophic lateral sclerosis patients.

11.
Neurobiol Dis ; 45(1): 83-98, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21820514

RESUMEN

The availability of many high-quality genome-wide expression datasets has provided an exciting and unique opportunity to better understand the molecular etiology of Huntington's disease. Combining this knowledge with other aspects of huntingtin biology and disease modification screens is yielding important new insights into disease-mitigating therapeutic strategies. Having followed this line of inquiry for some time, we note that there have been a number of surprises regarding the subsequently confirmed relationships between gene expression and disease etiology. Moreover, the complexity and sheer number of proposed mechanisms by which huntingtin can perturb gene expression continues to expand. Nonetheless, ongoing efforts to enthusiastically and critically evaluate the relationships between HD pathobiology and gene expression promise to deliver accurate predictions as to which therapeutic strategies will be most effective. An exciting new arm of this research also demonstrates the power of pharmacogenomics to detect (and rule out) important neuroprotective gene expression effects.


Asunto(s)
Encéfalo/metabolismo , Perfilación de la Expresión Génica , Enfermedad de Huntington/genética , Animales , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Enfermedad de Huntington/metabolismo
12.
Hum Mol Genet ; 19(4): 609-22, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19934114

RESUMEN

Selective degeneration of striatal neurons is a pathologic hallmark of Huntington disease (HD). The exact mechanism(s) behind this specific neurodegeneration is still unknown. Expression studies of diseased human post-mortem brain, as well as different mouse models exhibiting striatal degeneration, have demonstrated changes in the expression of many important genes with a large proportion of changes being observed in the striatal-enriched genes. These investigations have raised questions about how enrichment of particular transcripts in the striatum can lead to its selective vulnerability to neurodegeneration. Monitoring the expression changes of striatal-enriched genes during the course of the disease may be informative about their potential involvement in selective degeneration. In this study, we analyzed a Serial Analysis of Gene Expression (SAGE) database (www.mouseatlas.org) and compared the mouse striatum to 18 other brain regions to generate a novel list of striatal-enriched transcripts. These novel striatal-enriched transcripts were subsequently evaluated for expression changes in the YAC128 mouse model of HD, and differentially expressed transcripts were further examined in human post-mortem caudate samples. We identified transcripts with altered expression in YAC128 mice, which also showed consistent expression changes in human post-mortem tissue. The identification of novel striatal-enriched genes with altered expression in HD offers new avenues of study, leading towards a better understanding of specific pathways involved in the selective degeneration of striatal neurons in HD.


Asunto(s)
Cuerpo Estriado/metabolismo , Perfilación de la Expresión Génica , Enfermedad de Huntington/genética , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedad de Huntington/metabolismo , Ratones
13.
Hum Mol Genet ; 19(8): 1438-52, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20089533

RESUMEN

Evaluation of transcriptional changes in the striatum may be an effective approach to understanding the natural history of changes in expression contributing to the pathogenesis of Huntington disease (HD). We have performed genome-wide expression profiling of the YAC128 transgenic mouse model of HD at 12 and 24 months of age using two platforms in parallel: Affymetrix and Illumina. The data from these two powerful platforms were integrated to create a combined rank list, thereby revealing the identity of additional genes that proved to be differentially expressed between YAC128 and control mice. Using this approach, we identified 13 genes to be differentially expressed between YAC128 and controls which were validated by quantitative real-time PCR in independent cohorts of animals. In addition, we analyzed additional time points relevant to disease pathology: 3, 6 and 9 months of age. Here we present data showing the evolution of changes in the expression of selected genes: Wt1, Pcdh20 and Actn2 RNA levels change as early as 3 months of age, whereas Gsg1l, Sfmbt2, Acy3, Polr2a and Ppp1r9a RNA expression levels are affected later, at 12 and 24 months of age. We also analyzed the expression of these 13 genes in human HD and control brain, thereby revealing changes in SLC45A3, PCDH20, ACTN2, DDAH1 and PPP1R9A RNA expression. Further study of these genes may unravel novel pathways contributing to HD pathogenesis. DDBJ/EMBL/GenBank accession no: GSE19677.


Asunto(s)
Perfilación de la Expresión Génica , Genoma , Enfermedad de Huntington/genética , Transcripción Genética , Factores de Edad , Animales , Modelos Animales de Enfermedad , Genoma Humano , Humanos , Proteína Huntingtina , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos
14.
J Alzheimers Dis Rep ; 6(1): 739-748, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36606207

RESUMEN

Background: Vascular dementia (VaD) and Alzheimer's disease (AD) are the two most common forms of dementia. Although these two types of dementia have different etiologies, they share some similarities in their pathophysiology, such as neuronal loss and decreased levels of tau protein. We hypothesize that these can have an impact upon the molecular changes in tubulin, precede the neuronal cell loss, and lead to changes in cytoskeletal associated proteins, as documented in both VaD and AD. Objective: We characterized different isotypes of tubulin together with their posttranslational modifications, as well as several microtubule associated proteins (MAPs), such as tau protein, MAP2 and MAP6, all together known as the tubulin code. Methods: We performed western blotting in human brain homogenates of controls and AD and VaD subjects. Results: We report that the levels of different tubulin isotypes differ depending on the dementia type and the brain area being studied: whereas α-tubulin is increased in the temporal lobe of VaD patients, it is decreased in the frontal lobe of AD patients. In VaD patients, the frontal lobe had a decrease in tyrosinated tubulin, which was accompanied by a decrease in tau protein and a tendency for lower levels of MAP2. Conclusion: Our findings highlight distinct changes in the tubulin code in VaD and AD, suggesting a therapeutic opportunity for different dementia subtypes in the future.

15.
Neurobiol Dis ; 42(3): 459-67, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21334439

RESUMEN

R6/2 transgenic mice with expanded CAG repeats (>300) have a surprisingly prolonged disease progression and longer lifespan than prototypical parent R6/2 mice (carrying 150 CAGs); however, the mechanism of this phenotype amelioration is unknown. We compared gene expression profiles in the striatum of R6/2 transgenic mice carrying ~300 CAG repeats (R6/2(Q300) transgenic mice) to those carrying ~150 CAG repeats (R6/2(Q150) transgenic mice) and littermate wildtype controls in order to identify genes that may play determinant roles in the time course of phenotypic expression in these mice. Of the top genes showing concordant expression changes in the striatum of both R6/2 lines, 85% were decreased in expression, while discordant expression changes were observed mostly for genes upregulated in R6/2(Q300) transgenic mice. Upregulated genes in the R6/2(Q300) mice were associated with the ubiquitin ligase complex, cell adhesion, protein folding, and establishment of protein localization. We qPCR-validated increases in expression of genes related to the latter category, including Lrsam1, Erp29, Nasp, Tap1, Rab9b, and Pfdn5 in R6/2(Q300) mice, changes that were not observed in R6/2 mice with shorter CAG repeats, even in late stages (i.e., 12 weeks of age). We further tested Lrsam1 and Erp29, the two genes showing the greatest upregulation in R6/2(Q300) transgenic mice, for potential neuroprotective effects in primary striatal cultures overexpressing a mutated human huntingtin (htt) fragment. Overexpression of Lrsam1 prevented the loss of NeuN-positive cell bodies in htt171-82Q cultures, concomitant with a reduction of nuclear htt aggregates. Erp29 showed no significant effects in this model. This is consistent with the distinct pattern of htt inclusion localization observed in R6/2(Q300) transgenic mice, in which smaller cytoplasmic inclusions represent the major form of insoluble htt in the cell, as opposed to large nuclear inclusions observed in R6/2(Q150) transgenic mice. We suggest that the prolonged onset and disease course observed in R6/2 mice with greatly expanded CAG repeats might result from differential upregulation of genes related to protein localization and clearance. Such genes may represent novel therapeutic avenues to decrease htt aggregate toxicity and cell death in HD patients, with Lrsam1 being a promising, novel candidate disease modifier.


Asunto(s)
Cuerpo Estriado/metabolismo , Progresión de la Enfermedad , Enfermedad de Huntington/genética , Neuronas/metabolismo , Repeticiones de Trinucleótidos/genética , Animales , Muerte Celular/genética , Células Cultivadas , Cuerpo Estriado/citología , Perfilación de la Expresión Génica , Enfermedad de Huntington/metabolismo , Inmunohistoquímica , Ratones , Ratones Transgénicos , Neuronas/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Anal Chem ; 83(20): 7712-20, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21888376

RESUMEN

Huntington's disease (HD), caused by a mutation of the corresponding gene encoding the protein huntingtin (htt), is characterized by progressive deterioration of cognitive and motor functions, paralleled by extensive loss of striatal neurons. At the cellular level, pathogenesis involves an early and prolonged period of neuronal dysfunction followed by neuronal death. Understanding the molecular events driving these deleterious processes is critical to the successful development of therapies to slow down or halt the progression of the disease. Here, we examined biochemical processes in a HD ex vivo rat model, as well as in a HD model for cultured neurons using synchrotron-assisted Fourier transform infrared microspectroscopy (S-FTIRM). The model, based on lentiviral-mediated delivery of a fragment of the HD gene, expresses a mutant htt fragment in one brain hemisphere and a wild-type htt fragment in the control hemisphere. S-FTIRM allowed for high spatial resolution and distinction between spectral features occurring in gray and white matter. We measured a higher content of ß-sheet protein in the striatal gray matter exposed to mutant htt as early as 4 weeks following the initiation of mutant htt exposure. In contrast, white matter tracts did not exhibit any changes in protein structure but surprisingly showed reduced content of unsaturated lipids and a significant increase in spectral features associated with phosphorylation. The former is reminiscent of changes consistent with a myelination deficiency, while the latter is characteristic of early pro-apoptotic events. These findings point to the utility of the label-free FTIRM method to follow mutant htt's ß-sheet-rich transformation in striatal neurons ex vivo, provide further evidence for mutant htt amyloidogenesis in vivo, and demonstrate novel chemical features indicative of white matter changes in HD. Parallel studies in cultured neurons expressing the same htt fragments showed similar changes.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Espectroscopía Infrarroja por Transformada de Fourier , Amiloidosis , Animales , Encéfalo/patología , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Análisis Discriminante , Femenino , Proteína Huntingtina , Enfermedad de Huntington/genética , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Estructura Secundaria de Proteína , Ratas , Ratas Wistar , Sincrotrones
17.
FASEB J ; 24(7): 2464-74, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20237298

RESUMEN

Gamma-secretase is an intramembrane-cleaving protease responsible for the final proteolytic event in the production of the amyloid-beta peptides (Abeta) implicated in Alzheimer's disease (AD). Inhibition of gamma-secretase activity is thus an attractive therapeutic strategy to slow down the pathogenesis of AD. Drugs often target more than one biomolecule because of conserved 3-dimensional structures in prospective protein binding sites. We have capitalized on this phenomenon of nature to identify new gamma-secretase inhibitors. Here we show that 2-hydroxy naphthyl derivatives, a previously identified subclass of NAD(+) analog inhibitors of sirtuin 2 (SIRT2), are direct gamma-secretase inhibitors. Subsequent structure-activity relationship studies further showed that 2-hydroxy-1-naphthaldehyde is the minimal pharmacophore for gamma-secretase inhibition. In evaluating target protein determinants of inhibition, we identified a common GXG signature nucleotide-binding site (NBS) shared by the gamma-secretase subunit presenilin-1 C-terminal fragment (PS1-CTF), SIRT2, and Janus kinase 3 (JAK3). Because a detailed 3-dimensional structure of gamma-secretase is beyond our knowledge, we took advantage of the known crystal structure of human JAK3 to model the NBS of the PS1-CTF, which includes the catalytic residue D385. Our results suggest that the flexible PS1-CTF (381)LGLG(384) loop comprises a substrate-docking site capable of recognizing specifically different gamma-secretase substrates.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Descubrimiento de Drogas/métodos , Janus Quinasa 3/química , Nucleótidos/metabolismo , Presenilina-1/química , Sirtuina 2/química , Sitios de Unión , Inhibidores Enzimáticos/química , Humanos , Naftalenos , Relación Estructura-Actividad , Especificidad por Sustrato
18.
Brain ; 133(Pt 4): 1094-110, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20375136

RESUMEN

Huntington's disease is an autosomal dominant inherited neurodegenerative disease with motor symptoms that are variably co-expressed with mood and cognitive symptoms, and in which variable neuronal degeneration is also observed in the basal ganglia and the cerebral cortex. We have recently shown that the variable symptomatology in Huntington's disease correlates with the variable compartmental pattern of GABAA receptor and cell loss in the striatum. To determine whether the phenotypic variability in Huntington's disease is also related to variable neuronal degeneration in the cerebral cortex, we undertook a double-blind study using unbiased stereological cell counting methods to determine the pattern of cell loss in the primary motor and anterior cingulate cortices in the brains of 12 cases of Huntington's disease and 15 controls, and collected detailed data on the clinical symptomatology of the patients with Huntington's disease from family members and clinical records. The results showed a significant association between: (i) pronounced motor dysfunction and cell loss in the primary motor cortex; and (ii) major mood symptomatology and cell loss in the anterior cingulate cortex. This association held for both total neuronal loss (neuronal N staining) and pyramidal cell loss (SMI32 staining), and also correlated with marked dystrophic changes in the remaining cortical neurons. There was also an association between cortical cell loss and striatal neuropathological grade, but no significant association with CAG repeat length in the Huntington's disease gene. These findings suggest that the heterogeneity in clinical symptomatology that characterizes Huntington's disease is associated with variation in the extent of cell loss in the corresponding functional regions of the cerebral cortex whereby motor dysfunction correlates with primary motor cortex cell loss and mood symptomatology is associated with cell loss in the cingulate cortex.


Asunto(s)
Giro del Cíngulo/patología , Enfermedad de Huntington/diagnóstico , Enfermedad de Huntington/patología , Corteza Motora/patología , Neuronas/patología , Adulto , Anciano , Recuento de Células/métodos , Femenino , Giro del Cíngulo/citología , Humanos , Masculino , Persona de Mediana Edad , Corteza Motora/citología , Neuronas/citología , Células Piramidales/citología , Células Piramidales/patología , Estudios Retrospectivos
19.
Mol Cell Proteomics ; 8(4): 720-34, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19043139

RESUMEN

Huntington disease (HD) is fatal in humans within 15-20 years of symptomatic disease. Although late stage HD has been studied extensively, protein expression changes that occur at the early stages of disease and during disease progression have not been reported. In this study, we used a large two-dimensional gel/mass spectrometry-based proteomics approach to investigate HD-induced protein expression alterations and their kinetics at very early stages and during the course of disease. The murine HD model R6/2 was investigated at 2, 4, 6, 8, and 12 weeks of age, corresponding to absence of disease and early, intermediate, and late stage HD. Unexpectedly the most HD stage-specific protein changes (71-100%) as well as a drastic alteration (almost 6% of the proteome) in protein expression occurred already as early as 2 weeks of age. Early changes included mainly the up-regulation of proteins involved in glycolysis/gluconeogenesis and the down-regulation of the actin cytoskeleton. This suggests a period of highly variable protein expression that precedes the onset of HD phenotypes. Although an up-regulation of glycolysis/gluconeogenesis-related protein alterations remained dominant during HD progression, late stage alterations at 12 weeks showed an up-regulation of proteins involved in proteasomal function. The early changes in HD coincide with a peak in protein alteration during normal mouse development at 2 weeks of age that may be responsible for these massive changes. Protein and mRNA data sets showed a large overlap on the level of affected pathways but not single proteins/mRNAs. Our observations suggest that HD is characterized by a highly dynamic disease pathology not represented by linear protein concentration alterations over the course of disease.


Asunto(s)
Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Proteínas/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Desarrollo Embrionario , Metabolismo Energético , Femenino , Regulación de la Expresión Génica , Enfermedad de Huntington/genética , Cinética , Masculino , Redes y Vías Metabólicas , Ratones , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Extractos de Tejidos
20.
Mol Cell Neurosci ; 44(4): 374-85, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20470891

RESUMEN

The large glutamatergic calyx of Held synapse in the auditory brainstem has become a powerful model for studying transmitter release mechanisms, but the molecular bases of presynaptic function at this synapse are not well known. Here, we have used single-cell quantitative PCR (qPCR) to study the developmental expression of all major Synaptotagmin (Syt) isoforms in putative calyx of Held-generating neurons (globular bushy cells) of the ventral cochlear nucleus. Using electrophysiological criteria and the expression of marker genes including VGluTs (vesicular glutamate transporters), Ca(2+) binding proteins, and the transcription factor Math5, we identified a subset of the recorded neurons as putative calyx of Held-generating bushy cells. At postnatal days 12-15 these neurons expressed Syt-2 and Syt-11, and also Syt-3, -4, -7 and -13 at lower levels, whereas Syt-1 and -9 were absent. Interestingly, early in development (at P3-P6), immature bushy cells expressed a larger number of Syt-isoforms, with Syt-1, Syt-5, Syt-9 and Syt-13 detected in a significantly higher percentage of neurons. Our study sheds light on the molecular properties of putative calyx of Held-generating neurons and shows the developmental regulation of the Syt-isoform expression profile in a single neuron type.


Asunto(s)
Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Sinaptotagminas/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Vías Auditivas/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Tronco Encefálico/citología , Proteínas de Unión al Calcio/análisis , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Neuronas/citología , Terminales Presinápticos/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Sinaptotagminas/genética , Proteínas de Transporte Vesicular de Glutamato/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA