Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Neurosci ; 19(Suppl 1): 13, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29745864

RESUMO

BACKGROUND: The dominant hypothesis about the pathogenesis of Alzheimer's disease (AD) is the "amyloid cascade" concept and modulating the expression of proteins involved in the metabolism of amyloid-beta (Aß) is proposed as an effective strategy for the prevention and therapy of AD. Recently, we found that an antibiotic ceftriaxone (CEF), which possesses neuroprotective activity, reduced cognitive deficits and neurodegenerative changes in OXYS rats, a model of sporadic AD. The molecular mechanisms of this effect are not completely clear, we suggested that the drug might serve as the regulator of the expression of the genes involved in the metabolism of Aß and the pathogenesis of AD. The study was aimed to determine the effects of CEF on mRNA levels of Bace1 (encoding ß-secretase BACE1 involved in Aß production), Mme, Ide, Ece1, Ace2 (encoding enzymes involved in Aß degradation), Epo (encoding erythropoietin related to endothelial function and clearance of Aß across the blood brain barrier) in the frontal cortex, hippocampus, striatum, hypothalamus, and amygdala of OXYS and Wistar (control strain) male rats. Starting from the age of 14 weeks, animals received CEF (100 mg/kg/day, i.p., 36 days) or saline. mRNA levels were evaluated with RT-qPCR method. Biochemical parameters of plasma were measured for control of system effects of the treatment. RESULTS: To better understand strain variations studied here, we compared the gene expression between untreated OXYS and Wistar rats. This comparison showed a significant decrease in mRNA levels of Ace2 in the frontal cortex and hypothalamus, and of Actb in the amygdala of untreated OXYS rats. Analysis of potential effects of CEF revealed its novel targets. In the compound-treated OXYS cohort, CEF diminished mRNA levels of Bace1 and Ace2 in the hypothalamus, and Aktb in the frontal cortex. Furthermore, CEF augmented Mme, Ide, and Epo mRNA levels in the amygdala as well as the levels of Ece1 and Aktb in the striatum. Finally, CEF also attenuated the activity of ALT and AST in plasma of OXYS rats. CONCLUSION: Those findings disclosed novel targets for CEF action that might be involved into neuroprotective mechanisms at early, pre-plaque stages of AD-like pathology development.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ceftriaxona/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Doença de Alzheimer/tratamento farmacológico , Animais , Modelos Animais de Doenças , Masculino , RNA Mensageiro/metabolismo , Ratos Wistar
2.
Neurochem Res ; 41(3): 620-30, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26376806

RESUMO

Currently, deficit of amyloid ß-peptide (Aß) clearance from the brain is considered as one of the possible causes of amyloid accumulation and neuronal death in the sporadic form of Alzheimer's disease (AD). Aß clearance can involve either specific proteases present in the brain or Aß-binding/transport proteins. Among amyloid-degrading enzymes the most intensively studied are neprilysin (NEP) and insulin-degrading enzyme (IDE). Since ageing and development of brain pathologies is often accompanied by a deficit in the levels of expression and activity of these enzymes in the brain, there is an urgent need to understand the mechanisms involved in their regulation. We have recently reported that NEP and also an Aß-transport protein, transthyretin are epigenetically co-regulated by the APP intracellular domain (AICD) and this regulation depends on the cell type and APP695 isoform expression in a process that can be regulated by the tyrosine kinase inhibitor, Gleevec. We have now extended our work and shown that, unlike NEP, another amyloid-degrading enzyme, IDE, is not related to over-expression of APP695 in neuroblastoma SH-SY5Y cells but is up-regulated by APP751 and APP770 isoforms independently of AICD but correlating with reduced HDAC1 binding to its promoter. Studying the effect of the nuclear retinoid X receptor agonist, bexarotene, on NEP and IDE expression, we have found that both enzymes can be up-regulated by this compound but this mechanism is not APP-isoform specific and does not involve AICD but, on the contrary, affects HDAC1 occupancy on the NEP gene promoter. These new insights into the mechanisms of NEP and IDE regulation suggest possible pharmacological targets in developing AD therapies.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Epigênese Genética , Insulisina/metabolismo , Neprilisina/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Bexaroteno , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Humanos , Insulisina/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Receptores X de Retinoides/antagonistas & inibidores , Tetra-Hidronaftalenos/farmacologia
3.
Front Aging Neurosci ; 6: 235, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25278875

RESUMO

Abnormal elevation of amyloid ß-peptide (Aß) levels in the brain is the primary trigger for neuronal cell death specific to Alzheimer's disease (AD). It is now evident that Aß levels in the brain are manipulable due to a dynamic equilibrium between its production from the amyloid precursor protein (APP) and removal by amyloid clearance proteins. Clearance can be either enzymic or non-enzymic (binding/transport proteins). Intriguingly several of the main amyloid-degrading enzymes (ADEs) are members of the M13 peptidase family (neprilysin (NEP), NEP2 and the endothelin converting enzymes (ECE-1 and -2)). A distinct metallopeptidase, insulin-degrading enzyme (IDE), also contributes to Aß degradation in the brain. The ADE family currently embraces more than 20 members, both membrane-bound and soluble, and of differing cellular locations. NEP plays an important role in brain function terminating neuropeptide signals. Its decrease in specific brain areas with age or after hypoxia, ischaemia or stroke contribute significantly to the development of AD pathology. The recently discovered mechanism of epigenetic regulation of NEP (and other genes) by the APP intracellular domain (AICD) and its dependence on the cell type and APP isoform expression suggest possibilities for selective manipulation of NEP gene expression in neuronal cells. We have also observed that another amyloid-clearing protein, namely transthyretin (TTR), is also regulated in the neuronal cell by a mechanism similar to NEP. Dependence of amyloid clearance proteins on histone deacetylases and the ability of HDAC inhibitors to up-regulate their expression in the brain opens new avenues for developing preventive strategies in AD.

4.
J Neurochem ; 130(3): 419-31, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24528201

RESUMO

Proteolytic cleavage of the amyloid precursor protein (APP) by the successive actions of ß- and γ-secretases generates several biologically active metabolites including the amyloid ß-peptide (Aß) and the APP intracellular domain (AICD). By analogy with the Notch signalling pathway, AICD has been proposed to play a role in transcriptional regulation. Among the cohort of genes regulated by AICD is the Aß-degrading enzyme neprilysin (NEP). AICD binds to the NEP promoter causing transcriptional activation by competitive replacement with histone deacetylases (HDACs) leading to increased levels of NEP activity and hence increased Aß clearance. We now show that the Aß-clearance protein transthyretin (TTR) is also epigenetically up-regulated by AICD. Like NEP regulation, AICD derived specifically from the neuronal APP isoform, APP695 , binds directly to the TTR promoter displacing HDAC1 and HDAC3. Cell treatment with the tyrosine kinase inhibitor Gleevec (imatinib) or with the alkalizing agent NH4 Cl causes an accumulation of 'functional' AICD capable of up-regulating both TTR and NEP, leading to a reduction in total cellular Aß levels. Pharmacological regulation of both NEP and TTR might represent a viable therapeutic target in Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Epigênese Genética/genética , Neprilisina/genética , Pré-Albumina/genética , Cloreto de Amônio/farmacologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Benzamidas/farmacologia , Western Blotting , Imunoprecipitação da Cromatina , Citidina Desaminase/metabolismo , Eletroforese em Gel de Poliacrilamida , Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Mesilato de Imatinib , Imuno-Histoquímica , Neprilisina/fisiologia , Piperazinas/farmacologia , Pré-Albumina/fisiologia , Pirimidinas/farmacologia
5.
Clin Sci (Lond) ; 126(7): 507-16, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24147777

RESUMO

ACE2 (angiotensin-converting enzyme 2) counterbalances the actions of ACE (angiotensin-converting enzyme) by metabolizing its catalytic product, the vasoactive and fibrogenic peptide AngII (angiotensin II), into Ang-(1-7) [angiotensin-(1-7)]. Enhanced ACE2 expression may be protective in diabetes, cardiovascular disease and cancer. However, relatively little is known about the specific physiological factors regulating ACE2 expression. In the present paper, we show, by Western blotting and qPCR (quantitative real-time PCR), that ACE2 expression is increased under conditions of cell stress, including hypoxic conditions, IL (interleukin)-1ß treatment and treatment with the AMP mimic AICAR (5-amino-4-imidazolecarboxamide riboside). The NAD+-dependent deacetylase SIRT1 (silent information regulator T1) was found to be up-regulated after AICAR treatment but, conversely, was down-regulated after IL-1ß treatment. ChIP analysis demonstrated that SIRT1 bound to the ACE2 promoter and that binding was increased after AICAR treatment, but decreased after IL-1ß treatment. Inhibition of SIRT1 activity ablated the AICAR-induced increase in ACE2. In conclusion, we have established that the expression of the ACE2 transcript is controlled by the activity of SIRT1 under conditions of energy stress.


Assuntos
Metabolismo Energético , Epigênese Genética/fisiologia , Peptidil Dipeptidase A/genética , Sirtuína 1/fisiologia , Aminoimidazol Carboxamida/análogos & derivados , Enzima de Conversão de Angiotensina 2 , Western Blotting , Hipóxia Celular , Linhagem Celular Tumoral , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metformina/farmacologia , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real , Ribonucleotídeos/fisiologia
6.
Cell Signal ; 24(2): 402-409, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22024280

RESUMO

Alzheimer's disease (AD) is a neurodegenerative illness and the leading cause of dementia in the elderly. The accumulation of amyloid-ß peptide (Aß) is a well-known pathological hallmark associated with the disease. However, Aß is only one of several metabolites produced by ß- and γ-secretase actions on the transmembrane protein, the amyloid precursor protein (APP). A proteolytic fragment termed the APP intracellular domain (AICD) is also produced. By analogy with the Notch signalling pathway, AICD has been proposed as a transcriptional regulator although its mechanism of action and the complement of genes regulated remain controversial. This review will focus on the contributions that studies of APP processing have brought to the understanding of a novel nuclear signalling pathway that may contribute to the pathology of AD and may provide new therapeutic opportunities.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Transdução de Sinais , Idoso , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Comunicação Celular , Membrana Celular/química , Membrana Celular/genética , Núcleo Celular/genética , Citoplasma/genética , Humanos , Camundongos , Camundongos Transgênicos , Neprilisina/genética , Neprilisina/metabolismo , Estrutura Terciária de Proteína , Proteólise , Receptores Notch/genética , Receptores Notch/metabolismo
7.
J Neurochem ; 120 Suppl 1: 167-185, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22122230

RESUMO

: The amyloid cascade hypothesis of Alzheimer's disease envisages that the initial elevation of amyloid ß-peptide (Aß) levels, especially of Aß(1-42) , is the primary trigger for the neuronal cell death specific to onset of Alzheimer's disease. There is now substantial evidence that brain amyloid levels are manipulable because of a dynamic equilibrium between their synthesis from the amyloid precursor protein and their removal by amyloid-degrading enzymes (ADEs) providing a potential therapeutic strategy. Since the initial reports over a decade ago that two zinc metallopeptidases, insulin-degrading enzyme and neprilysin (NEP), contributed to amyloid degradation in the brain, there is now an embarras de richesses in relation to this category of enzymes, which currently number almost 20. These now include serine and cysteine proteinases, as well as numerous zinc peptidases. The experimental validation for each of these enzymes, and which to target, varies enormously but up-regulation of several of them individually in mouse models of Alzheimer's disease has proved effective in amyloid and plaque clearance, as well as cognitive enhancement. The relative status of each of these enzymes will be critically evaluated. NEP and its homologues, as well as insulin-degrading enzyme, remain as principal ADEs and recently discovered mechanisms of epigenetic regulation of NEP expression potentially open new avenues in manipulation of AD-related genes, including ADEs.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/prevenção & controle , Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Placa Amiloide/patologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Sistemas de Liberação de Medicamentos/métodos , Humanos , Placa Amiloide/metabolismo , Proteólise/efeitos dos fármacos
8.
J Mol Neurosci ; 46(3): 569-77, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21932040

RESUMO

Alzheimer's disease (AD) is accompanied by memory loss due to neuronal cell death caused by toxic amyloid ß-peptide (Aß) aggregates. In the healthy brain, a group of amyloid-degrading enzymes including neprilysin (NEP) maintain Aß levels at physiologically low concentrations but, with age and under some pathological conditions, expression and activity of these enzymes decline predisposing to late-onset AD. Hence, up-regulation of NEP might be a viable strategy for prevention of Aß accumulation and development of the disease. As we have recently shown, inhibitors of histone deacetylases, in particular, valproic acid (VA), were capable of up-regulating NEP expression and activity in human neuroblastoma SH-SY5Y cell lines characterised by very low levels of NEP. In the present study, analysing the effect of i.p. injections of VA to rats, we have observed up-regulation of expression and activity of NEP in rat brain structures, in particular, in the hippocampus. This effect was brain region- and age-specific. Administration of VA has also restored NEP activity and memory deficit in adult rats caused by prenatal hypoxia. This suggests that VA and more specific HDAC inhibitors can be considered as potential pharmaceutical agents for up-regulation of NEP activity and improvement of cognitive functions of ageing brain.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Memória/efeitos dos fármacos , Neprilisina/genética , Ácido Valproico/farmacologia , Doença de Alzheimer/psicologia , Animais , Anticonvulsivantes/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Memória/fisiologia , Transtornos da Memória/psicologia , Neprilisina/metabolismo , Neuroblastoma/química , Neuroblastoma/patologia , Neuroblastoma/psicologia , Ratos , Ratos Wistar
9.
Int J Cancer ; 130(4): 775-86, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21365649

RESUMO

The zinc metallopeptidase, neprilysin (NEP), is an endothelin-1 degrading enzyme whose expression is extensively downregulated in prostate cancer. The expression of NEP in neuronal cells is regulated by intramembrane proteolysis of the amyloid precursor protein (APP) through its intracellular domain (AICD) facilitating histone acetylation of the NEP promoter and gene transcription. The present study has examined whether similar mechanisms operate in prostate cell lines. The expression of APP and its processing enzymes (ß- and γ-secretases) was examined in a number of prostate cell lines, and the effect of γ-secretase inhibition was explored on NEP expression and activity. The potential interaction of AICD with the NEP promoter was examined by chromatin immunoprecipitation. Our results indicated that all key components involved in APP processing were expressed in prostate cancer cell lines but suppression of AICD production using a γ-secretase inhibitor did not decrease NEP expression and activity, and no direct AICD-NEP promoter interaction could be detected. However, histone deacetylase inhibitors (valproate and trichostatin A) caused a 2- to 3-fold increase in NEP expression in PC-3 cells, and combinatorial treatment with the DNA demethylating agent, AzaC, further increased NEP expression levels. Although AICD is detectable in prostate cell lines, it does not appear to regulate NEP by AICD-mediated signalling. Apart from promoter de-methylation, the data suggest that histone acetylation may facilitate partial re-activation of NEP expression in advanced prostate cancer cells. Upregulation of this tumour-suppressing protein may provide a novel therapeutic strategy in prostate cancer.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Regulação Neoplásica da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Neprilisina/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais/fisiologia , Secretases da Proteína Precursora do Amiloide/análise , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/análise , Precursor de Proteína beta-Amiloide/química , Linhagem Celular Tumoral , Humanos , Insulisina/metabolismo , Masculino , Regiões Promotoras Genéticas , Neoplasias da Próstata/patologia , Estrutura Terciária de Proteína
11.
J Biol Chem ; 285(53): 41443-54, 2010 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-20961856

RESUMO

Amyloidogenic processing of the amyloid precursor protein (APP) by ß- and γ-secretases generates several biologically active products, including amyloid-ß (Aß) and the APP intracellular domain (AICD). AICD regulates transcription of several neuronal genes, especially the Aß-degrading enzyme, neprilysin (NEP). APP exists in several alternatively spliced isoforms, APP(695), APP(751), and APP(770). We have examined whether each isoform can contribute to AICD generation and hence up-regulation of NEP expression. Using SH-SY5Y neuronal cells stably expressing each of the APP isoforms, we observed that only APP(695) up-regulated nuclear AICD levels (9-fold) and NEP expression (6-fold). Increased NEP expression was abolished by a ß- or γ-secretase inhibitor but not an α-secretase inhibitor. This correlated with a marked increase in both Aß(1-40) and Aß(1-42) in APP(695) cells as compared with APP(751) or APP(770) cells. Similar phenomena were observed in Neuro2a but not HEK293 cells. SH-SY5Y cells expressing the Swedish mutant of APP(695) also showed an increase in Aß levels and NEP expression as compared with wild-type APP(695) cells. Chromatin immunoprecipitation revealed that AICD was associated with the NEP promoter in APP(695), Neuro2a, and APP(Swe) cells but not APP(751) nor APP(770) cells where AICD was replaced by histone deacetylase 1 (HDAC1). AICD occupancy of the NEP promoter was replaced by HDAC1 after treatment of the APP(695) cells with a ß- but not an α-secretase inhibitor. The increased AICD and NEP levels were significantly reduced in cholesterol-depleted APP(695) cells. In conclusion, Aß and functional AICD appear to be preferentially synthesized through ß-secretase action on APP(695).


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Regulação da Expressão Gênica , Doença de Alzheimer/metabolismo , Animais , Linhagem Celular Tumoral , Colesterol/química , Imunoprecipitação da Cromatina , Histona Desacetilases/metabolismo , Humanos , Ligantes , Camundongos , Neprilisina/biossíntese , Doenças Neurodegenerativas/metabolismo , Isoformas de Proteínas , Estrutura Terciária de Proteína
12.
Trends Pharmacol Sci ; 30(10): 509-14, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19762089

RESUMO

Sodium valproate, or Epilim, has been widely used as a broad spectrum, anticonvulsant drug for over 40 years and exhibits a good safety profile. Some of the actions of valproate arise from its more recently described histone deacetylase (HDAC) inhibitory properties and hence it can specifically modulate gene expression. There is now accumulating evidence that HDAC inhibitors may have potential in the treatment of CNS disorders and, in this context, valproate has much potential as a brain-penetrant, clinically available and well tested drug. This article reviews the pharmacology of this remarkable molecule, focusing on its actions as a neuroprotectant and hence with new potential in the treatment of neurodegenerative diseases.


Assuntos
Inibidores de Histona Desacetilases/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Ácido Valproico/uso terapêutico , Animais , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Inibidores de Histona Desacetilases/farmacologia , Humanos , Doenças Neurodegenerativas/metabolismo , Fármacos Neuroprotetores/farmacologia , Ácido Valproico/farmacologia
13.
EMBO Rep ; 10(1): 94-100, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19057576

RESUMO

Amyloid beta-peptide (Abeta) accumulation leads to neurodegeneration and Alzheimer disease; however, amyloid metabolism is a dynamic process and enzymic mechanisms exist for Abeta removal. Considerable controversy surrounds whether the intracellular domain of the amyloid precursor protein (AICD) regulates expression of the Abeta-degrading metalloprotease, neprilysin (NEP). By comparing two neuroblastoma cell lines differing substantially in NEP expression, we show by chromatin immunoprecipitation (ChIP) that AICD is bound directly to the NEP promoter in high NEP-expresser (NB7) cells but not in low-expresser (SH-SY5Y) cells. The methylation status of the NEP promoter does not regulate expression in these cells, whereas the histone deacetylase inhibitors trichostatin A and valproate partly restore NEP expression and activity in SH-SY5Y cells. ChIP analysis also reveals AICD binding to the NEP promoter in rat primary neurons but not in HUVEC cells. Chromatin remodelling of crucial Alzheimer disease-related genes by valproate could provide a new therapeutic strategy.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Regulação da Expressão Gênica/genética , Neprilisina/metabolismo , Regiões Promotoras Genéticas/genética , Doença de Alzheimer/genética , Animais , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Neprilisina/genética , Ligação Proteica , Ratos
14.
Curr Alzheimer Res ; 5(2): 212-24, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18393806

RESUMO

The steady state concentration of the Alzheimer's amyloid-beta peptide in the brain represents a balance between its biosynthesis from the transmembrane amyloid precursor protein (APP), its oligomerisation into neurotoxic and stable species and its degradation by a variety of amyloid-degrading enzymes, principally metallopeptidases. These include, among others, neprilysin (NEP) and its homologue endothelin-converting enzyme (ECE), insulysin (IDE), angiotensin-converting enzyme (ACE) and matrix metalloproteinase-9 (MMP-9). In addition, the serine proteinase, plasmin, may participate in extracellular metabolism of the amyloid peptide under regulation of the plasminogen-activator inhibitor. These various amyloid-degrading enzymes have distinct subcellular localizations, and differential responses to aging, oxidative stress and pharmacological agents and their upregulation may provide a novel and viable therapeutic strategy for prevention and treatment of Alzheimer's disease. Potential approaches to manipulate expression levels of the key amyloid-degrading enzymes are highlighted.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Amiloide/metabolismo , Inibidores Enzimáticos/uso terapêutico , Animais , Ácido Aspártico Endopeptidases/metabolismo , Enzimas Conversoras de Endotelina , Fibrinolisina/antagonistas & inibidores , Fibrinolisina/metabolismo , Humanos , Insulisina/antagonistas & inibidores , Insulisina/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Metaloendopeptidases/metabolismo , Neprilisina/antagonistas & inibidores , Neprilisina/metabolismo , Peptidil Dipeptidase A/metabolismo
15.
Neurobiol Dis ; 29(3): 438-45, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18082412

RESUMO

Huntington's disease (HD) is a dominantly-inherited neurodegenerative disorder which is incurable and ultimately fatal. HD is characterised by widespread mRNA dysregulation, particularly in neurons of the forebrain, by mechanisms which are not fully understood. Such dysregulation has been demonstrated to result, in part, from aberrant nuclear localisation of the transcriptional repressor, REST. Here, we show that expression of a number of neuronal-specific microRNAs is also dysregulated in HD tissues, probably as a result of increased repression by REST. This phenomenon is observed in both murine models of HD and in the brains of human HD sufferers. MicroRNA loss is reflected in increased levels of a number of target messenger RNAs. These data are the first to demonstrate a role for microRNAs in HD, and indicate that the molecular aetiology of HD is reflected in a loss of neuronal identity, caused in part by dysregulation of both transcriptional and post-transcriptional mechanisms.


Assuntos
Doença de Huntington/genética , Doença de Huntington/metabolismo , MicroRNAs/fisiologia , Transdução de Sinais/genética , Animais , Encéfalo/patologia , Encéfalo/fisiologia , Células Cultivadas , Marcação de Genes/métodos , Humanos , Doença de Huntington/patologia , Camundongos , Camundongos Transgênicos , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
16.
J Biol Chem ; 281(51): 38974-80, 2006 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-17023429

RESUMO

Chromatin remodeling enzymes such as SWI/SNF use the hydrolysis of ATP to power the movement of nucleosomes with respect to DNA. BRG1, one of the ATPases of the SWI/SNF complex, can be recruited by both activators and repressors, although the precise role of BRG1 in mechanisms of repression has thus far remained unclear. One transcription factor that recruits BRG1 as a corepressor is the repressor element 1-silencing transcription factor (REST). Here we address for the first time the mechanism of BRG1 activity in gene repression. We found that BRG1 enhanced REST-mediated repression at some REST target genes by increasing the interaction of REST with the local chromatin at its binding sites. Furthermore, REST-chromatin interactions, mediated by BRG1, were enhanced following an increase in histone acetylation in a manner dependent on the BRG1 bromodomain. Our data suggest that BRG1 facilitates REST repression by increasing the interaction between REST and chromatin. Such a mechanism may be applicable to other transcriptional repressors that utilize BRG1.


Assuntos
Cromatina/química , DNA Helicases/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Repressoras/química , Proteínas Repressoras/fisiologia , Fatores de Transcrição/química , Fatores de Transcrição/fisiologia , Acetilação , Trifosfato de Adenosina/química , Sítios de Ligação , Linhagem Celular , Núcleo Celular/metabolismo , DNA Helicases/química , Histonas/química , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteínas Nucleares/química , Plasmídeos/metabolismo , Ligação Proteica , Transcrição Gênica , Transfecção
17.
Mol Biol Cell ; 16(12): 5630-8, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16195345

RESUMO

Differentiation of pluripotent embryonic stem (ES) cells through multipotent neural stem (NS) cells into differentiated neurons is accompanied by wholesale changes in transcriptional programs. One factor that is present at all three stages and a key to neuronal differentiation is the RE1-silencing transcription factor (REST/NRSF). Here, we have used a novel chromatin immunoprecipitation-based cloning strategy (SACHI) to identify 89 REST target genes in ES cells, embryonic hippocampal NS cells and mature hippocampus. The gene products are involved in all aspects of neuronal function, especially neuronal differentiation, axonal growth, vesicular transport and release, and ionic conductance. Most target genes are silent or expressed at low levels in ES and NS cells, but are expressed at much higher levels in hippocampus. These data indicate that the REST regulon is specific to each developmental stage and support the notion that REST plays distinct roles in regulating gene expression in pluripotent ES cells, multipotent NS cells, and mature neurons.


Assuntos
Neurônios/fisiologia , Proteínas Repressoras/metabolismo , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/fisiologia , Cromatina/fisiologia , Cromatina/ultraestrutura , Clonagem Molecular , Sequência Consenso , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/genética
18.
J Biol Chem ; 279(1): 556-61, 2004 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-14561745

RESUMO

Establishment of neuronal identity requires co-ordinated expression of specific batteries of genes. These programs of gene expression are executed by activation of neuron-specific genes in neuronal cells and their repression in non-neuronal cells. Such co-ordinate regulation requires that individual activators and repressors regulate transcription from specific subsets of their potential target genes, yet we know little of the mechanisms that underlie this selective process. The RE-1 silencing transcription factor (REST) is a repressor that is proposed to silence transcription of numerous neuron-specific genes in non-neuronal cells via recruitment of two independent histone deacetylase (HDAC)-containing co-repressor complexes. However, in vivo, REST appears to be an obligate silencer for only a minority of RE-1-bearing genes. Here we examine the interaction of REST, Co-REST, Sin3A, HDAC1, and HDAC2 with two archetypical endogenous target genes, the M4 muscarinic receptor and the sodium type II channel (NaV1.2) genes. We find that these genes are present in distinct chromosomal domains. The NaV1.2 gene is actively transcribed but repressed by REST independently of histone deacetylation or DNA methylation and does not co-localize with epigenetic markers of silence, including dimethylation of H3K9 and HP1. In contrast, the M4 gene is maintained in a silent state independently of REST and co-localizes with dimethylated H3K9 and HP1alpha and HP1gamma, characteristic of silenced or senescent euchromatic DNA. This contrasts with the coordinate REST-dependent regulation of this locus reported previously. Taken together, we infer that distinct repressor complexes and mechanisms are operative at particular loci even in cell lines derived from a common embryological origin.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Inativação Gênica/fisiologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Regulação da Expressão Gênica , Genes Reporter , Humanos , Canal de Sódio Disparado por Voltagem NAV1.2 , Proteínas do Tecido Nervoso/genética , Ratos , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Sódio/genética , Fatores de Transcrição/genética , Transfecção
19.
J Mol Biol ; 334(5): 863-74, 2003 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-14643653

RESUMO

The repressor element 1-silencing transcription factor (REST) has been proposed to restrict expression of repressor element 1 (RE1) bearing genes to differentiated neurons by silencing their expression in non-neural tissue. Here, we have examined the interaction of REST with the M(4) muscarinic acetylcholine receptor gene. We show that REST binds to the RE1 of the M(4) gene in those cell lines and brain regions where the M(4) gene is expressed but not in those where the M(4) is not expressed. Furthermore, in cells that express M(4), the presence of REST represses but is insufficient to silence transcription of M(4). In non-neural cells REST is absent from the RE1 of the silent M(4) gene and perturbation of REST function fails to induce M(4) expression. We propose that REST acts to regulate expression levels of some RE1-bearing genes in neural cells, thereby playing an important role in defining neuronal activity.


Assuntos
Receptores Muscarínicos/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Bases , Linhagem Celular , Primers do DNA , Desoxirribonuclease I/metabolismo
20.
Clin Sci (Lond) ; 105(4): 467-72, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12773093

RESUMO

Plasma concentrations of HDL (high-density lipoprotein) cholesterol are low in the Saudi Arabian population. A B polymorphism at the CETP (cholesteryl ester protein transfer) locus that is detectable with the restriction enzyme Taq I is a genetic determinant of the plasma HDL cholesterol concentration. We assessed the relationship between the Taq I B CETP polymorphism and lipid and apolipoprotein concentrations in a study sample of 335 Saudi residents. The Taq I B1 and B2 allele frequencies were 0.54 and 0.46 respectively, similar to those in other populations. HDL cholesterol levels in B2B2 homozygotes were significantly higher than in B1B1 homozygotes [1.01 (0.3) compared with 0.92 (0.2) mmol/l; mean (S.D.); P=0.03]. There was also a significant difference between the B2B2 and B1B1 homozygotes with regard to apolipoprotein AI concentration [123.6 (16.4) compared with 113.7 (13.9) mg/dl; P=0.04]. This genetic variation was independent of metabolic risk factors known to influence HDL cholesterol levels. The allele frequency of the Taq I B CETP polymorphism and its relatively modest impact on HDL cholesterol concentrations argue against an important role for this allele, or for strongly linked loci, in determining the low levels of HDL cholesterol seen in the Saudi population.


Assuntos
Proteínas de Transporte/genética , HDL-Colesterol/sangue , Glicoproteínas , Polimorfismo Genético , Adulto , Análise de Variância , Apolipoproteína A-I/análise , Proteínas de Transferência de Ésteres de Colesterol , Frequência do Gene , Homozigoto , Humanos , Masculino , Arábia Saudita
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...