Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Invest New Drugs ; 29(1): 41-51, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19851711

RESUMO

The nonsteroidal anti-inflammatory drug (NSAID), tolfenamic acid (TA) is emerging as a new anti-cancer agent. TA induces the degradation of specific Specificity protein (Sp) transcription factors, Sp1, Sp3 and Sp4 which are associated with tumor growth and metastasis. In this study we have evaluated the effect of TA on lung cancer using both in vitro and in vivo models. TA in a dose dependent manner inhibited proliferation and cell viability of two different lung cancer cells, A549 and CRL5803. TA treatment for 48 h significantly decreased the expression of Sp1, Sp3 and Sp4. The hepatocyte growth factor receptor, c-Met is overexpressed in a variety of cancers including lung cancer and Sp proteins mediate the regulation of c-Met. TA diminished the expression of c-Met protein and modulates its downstream signaling pathway. Furthermore, TA treatment significantly increased the number of apoptotic cells and pro-apoptotic markers c-PARP and Bax confirming the activation of apoptotic pathways. In vivo studies using the orthotopic mice model for lung cancer showed that TA (25 mg/kg/2 days and 50 mg/kg/2 days) resulted in a dose dependent decrease in tumor formation. The immunohistochemical staining of lung tissue showed high expression of Sp1, Sp3, Sp4, c-Met and phospho Met in control group and a dose dependent decrease in TA treated groups. The crucial findings of this study support that targeting c-Met with a potent inhibitor of Sp proteins is a robust strategy for the implications in lung cancer treatment and TA can serve as a therapeutic agent for this devastating disease.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Fatores de Transcrição Sp/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , ortoaminobenzoatos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-met/genética , Transcrição Gênica/efeitos dos fármacos , ortoaminobenzoatos/uso terapêutico
2.
Mol Pharmacol ; 78(2): 226-36, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20488920

RESUMO

The anticancer agent 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) and its methyl ester (CDDO-Me) typically induce a broad spectrum of growth-inhibitory, proapoptotic, and antiangiogenic responses. Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). RNA interference studies indicate that these repressed genes are regulated by specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and Western blot analysis of lysates from pancreatic cancer cells treated with CDDO and CDDO-Me shows for the first time that both compounds decreased the expression of Sp1, Sp3, and Sp4. Moreover, CDDO-Me (7.5 mg/kg/day) also inhibited pancreatic human L3.6pL tumor growth and down-regulated Sp1, Sp3, and Sp4 in tumors using an orthotopic pancreatic cancer model. CDDO-Me also induced reactive oxygen species (ROS) and decreased mitochondrial membrane potential (MMP) in Panc1 and L3.6pL cells, and cotreatment with antioxidants (glutathione and dithiothreitol) blocked the formation of ROS, reversed the loss of MMP, and inhibited down-regulation of Sp1, Sp3, and Sp4. Repression of Sp and Sp-dependent genes by CDDO-Me was due to the down-regulation of microRNA-27a and induction of zinc finger and BTB domain containing 10 (ZBTB10), an Sp repressor, and these responses were also reversed by antioxidants. Thus, the anticancer activity of CDDO-Me is due, in part, to activation of ROS, which in turn targets the microRNA-27a:ZBTB10-Sp transcription factor axis. This results in decreased expression of Sp-regulated genes, growth inhibition, induction of apoptosis, and antiangiogenic responses.


Assuntos
MicroRNAs/metabolismo , Ácido Oleanólico/análogos & derivados , Neoplasias Pancreáticas/patologia , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Primers do DNA , Humanos , Masculino , Camundongos , Camundongos Nus , Ácido Oleanólico/farmacologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Reação em Cadeia da Polimerase , Espécies Reativas de Oxigênio/metabolismo
3.
J Alzheimers Dis ; 18(1): 211-30, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19584442

RESUMO

Alzheimer's disease (AD) is characterized by plaques of amyloid-beta (Abeta) peptide, cleaved from amyloid-beta protein precursor (AbetaPP). Our hypothesis is that lifespan profiles of AD-associated mRNA and protein levels in monkeys would differ from mice and that differential lifespan expression profiles would be useful to understand human AD pathogenesis. We compared profiles of AbetaPP mRNA, AbetaPP protein, and Abeta levels in rodents and primates. We also tracked a transcriptional regulator of the AbetaPP gene, specificity protein 1 (SP1), and the beta amyloid precursor cleaving enzyme (BACE1). In mice, AbetaPP and SP1 mRNA and their protein products were elevated late in life; Abeta levels declined in old age. In monkeys, SP1, AbetaPP, and BACE1 mRNA declined in old age, while protein products and Abeta levels rose. Proteolytic processing in both species did not match production of Abeta. In primates, AbetaPP and SP1 mRNA levels coordinate, but an inverse relationship exists with corresponding protein products as well as Abeta levels. Comparison of human DNA and mRNA sequences to monkey and mouse counterparts revealed structural features that may explain differences in transcriptional and translational processing. These findings are important for selecting appropriate models for AD and other age-related diseases.


Assuntos
Doença de Alzheimer/genética , Longevidade/genética , Fatores Etários , Animais , Haplorrinos , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Especificidade da Espécie , Fatores de Transcrição/genética
4.
Mol Cancer Ther ; 8(3): 533-42, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19258429

RESUMO

Survivin is overexpressed in most human cancers, including pancreatic adenocarcinoma. Expression of survivin is regulated by specificity protein (Sp) proteins and related to resistance to radiation therapy. Tolfenamic acid induces Sp protein degradation in several cancer cell lines. The purpose of this study is to investigate whether tolfenamic acid inhibits survivin expression and sensitizes pancreatic cancer cells/tumor to radiotherapy. Panc1 and L3.6pl cells have been used to study the effect of radiation on survivin expression and to investigate the efficacy of tolfenamic acid in enhancing the response to radiation therapy. In addition, an orthotopic model for human pancreatic cancer has been used to confirm the efficacy of tolfenamic acid to enhance tumor response to radiation in vivo. Pancreatic cancer cell lines express variable levels of survivin mRNA/protein, which correlate with their radiosensitivity. Radiation increased survivin promoter activity and protein expression in Panc1 and L3.6pl cells and tolfenamic acid inhibited both constitutive and radiation-induced survivin protein expression and enhanced the response of pancreatic cancer cells to radiation therapy. In vivo studies show that tolfenamic acid enhanced the radiation-induced apoptosis associated with decreased survivin expression in tumors and this correlates with the enhanced response of these tumors to the radiation. Thus, tolfenamic acid significantly enhances pancreatic cancer cells/tumor response to radiation therapy. The underlying mechanism includes tolfenamic acid-induced degradation of Sp proteins, which in tumor decreases expression of the Sp-dependent antiapoptotic protein survivin. These preclinical data suggest that tolfenamic acid has the potential to increase the response of pancreatic adenocarcinoma to radiation therapy.


Assuntos
Adenocarcinoma/radioterapia , Proteínas Associadas aos Microtúbulos/genética , Neoplasias Pancreáticas/radioterapia , Tolerância a Radiação/efeitos dos fármacos , ortoaminobenzoatos/farmacologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Proteínas Inibidoras de Apoptose , Masculino , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Survivina , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , ortoaminobenzoatos/uso terapêutico
5.
J Neurosci ; 28(1): 3-9, 2008 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18171917

RESUMO

The sporadic nature of Alzheimer's disease (AD) argues for an environmental link that may drive AD pathogenesis; however, the triggering factors and the period of their action are unknown. Recent studies in rodents have shown that exposure to lead (Pb) during brain development predetermined the expression and regulation of the amyloid precursor protein (APP) and its amyloidogenic beta-amyloid (Abeta) product in old age. Here, we report that the expression of AD-related genes [APP, BACE1 (beta-site APP cleaving enzyme 1)] as well as their transcriptional regulator (Sp1) were elevated in aged (23-year-old) monkeys exposed to Pb as infants. Furthermore, developmental exposure to Pb altered the levels, characteristics, and intracellular distribution of Abeta staining and amyloid plaques in the frontal association cortex. These latent effects were accompanied by a decrease in DNA methyltransferase activity and higher levels of oxidative damage to DNA, indicating that epigenetic imprinting in early life influenced the expression of AD-related genes and promoted DNA damage and pathogenesis. These data suggest that AD pathogenesis is influenced by early life exposures and argue for both an environmental trigger and a developmental origin of AD.


Assuntos
Envelhecimento , Doença de Alzheimer , Exposição Ambiental , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Chumbo/toxicidade , Fatores Etários , Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides/análise , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , Córtex Cerebral , Modelos Animais de Doenças , Embrião de Mamíferos , Epigênese Genética , Feminino , Imunoglobulinas/metabolismo , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Neurônios , Fragmentos de Peptídeos/análise
6.
J Mol Neurosci ; 34(1): 1-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18157652

RESUMO

Alzheimer's Disease (AD) is a progressive, irreversible neurodegenerative disease. Despite several genetic mutations (Haass et al., J. Biol. Chem. 269:17741-17748, 1994; Ancolio et al., Proc. Natl. Acad. Sci. USA 96:4119-4124, 1999; Munoz and Feldman, CMAJ 162:65-72, 2000; Gatz et al., Neurobiol. Aging 26:439-447, 2005) found in AD patients, more than 90% of AD cases are sporadic (Bertram and Tanzi, Hum. Mol. Genet. 13:R135-R141, 2004). Therefore, it is plausible that environmental exposure may be an etiologic factor in the pathogenesis of AD. The AD brain is characterized by extracellular beta-amyloid (Abeta) deposition and intracellular hyperphosphorylated tau protein. Our lab has demonstrated that developmental exposure of rodents to the heavy metal lead (Pb) increases APP (amyloid precursor protein) and Abeta production later in the aging brain (Basha et al., J. Neurosci. 25:823-829, 2005a). We also found elevations in the oxidative marker 8-oxo-dG in older animals that had been developmentally exposed to Pb (Bolin et al., FASEB J. 20:788-790, 2006) as well as promotion of amyloidogenic histopathology in primates. These findings indicate that early life experiences contribute to amyloidogenesis in old age perhaps through epigenetic pathways. Here we explore the role of epigenetics as the underlying mechanism that mediates this early exposure-latent pathogenesis with a special emphasis on alterations in the methylation profiles of CpG dinucleotides in the promoters of genes and their influence on both gene transcription and oxidative DNA damage.


Assuntos
Doença de Alzheimer/genética , Exposição Ambiental , Epigênese Genética/genética , Placa Amiloide/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/biossíntese , Animais , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Metilação de DNA/efeitos dos fármacos , Feminino , Humanos , Intoxicação do Sistema Nervoso por Chumbo/complicações , Intoxicação do Sistema Nervoso por Chumbo/genética , Intoxicação do Sistema Nervoso por Chumbo/metabolismo , Placa Amiloide/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
7.
Toxicol Appl Pharmacol ; 225(1): 1-27, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17904601

RESUMO

Lead (Pb) is a xenobiotic metal with no known essential function in cellular growth, proliferation, or signaling. Decades of research characterizing the toxicology of Pb have shown it to be a potent neurotoxicant, especially during nervous system development. New concepts in the neurotoxicology of Pb include advances in understanding the mechanisms and cellular specificity of Pb. Experimental studies have shown that stress can significantly alter the effects of Pb, effects that could potentially be mediated through alterations in the interactions of glucocorticoids with the mesocorticolimbic dopamine system of the brain. Elevated stress, with corresponding elevated glucocorticoid levels, has been postulated to contribute to the increased levels of many diseases and dysfunctions in low socioeconomic status populations. Cellular models of learning and memory have been utilized to investigate the potential mechanisms of Pb-induced cognitive deficits. Examination of long-term potentiation in the rodent hippocampus has revealed Pb-induced increases in threshold, decreases in magnitude, and shorter retention times of synaptic plasticity. Structural plasticity in the form of adult neurogenesis in the hippocampus is also impacted by Pb exposure. The action of Pb on glutamate release, NMDA receptor function, or structural plasticity may underlie perturbations in synaptic plasticity and contribute to learning impairments. In addition to providing insight into potential mechanisms of Pb-induced cognitive deficits, cellular models offer an opportunity to investigate direct effects of Pb on isolated biological substrates. A target of interest is the 78-kDa molecular chaperone glucose-regulated protein (GRP78). GRP78 chaperones the secretion of the cytokine interleukin-6 (IL-6) by astrocytes. In vitro evidence shows that Pb strongly binds to GRP78, induces GRP78 aggregation, and blocks IL-6 secretion in astroglial cells. These findings provide evidence for a significant chaperone deficiency in Pb-exposed astrocytes in culture. In the long term, chaperone deficiency could underlie protein conformational diseases such as Alzheimer's Disease (AD). Lead exposure in early life has been implicated in subsequent progression of amyloidogenesis in rodents during old age. This exposure resulted in an increase in proteins associated with AD pathology viz., beta-amyloid precursor protein (beta-APP), and beta-amyloid (Abeta). These four new lines of research comprise compelling evidence that exposures to Pb have adverse effects on the nervous system, that environmental factors increase nervous system susceptibility to Pb, and that exposures in early life may cause neurodegeneration in later life.


Assuntos
Intoxicação do Sistema Nervoso por Chumbo/fisiopatologia , Chumbo/toxicidade , Estresse Fisiológico/complicações , Doença de Alzheimer/etiologia , Doença de Alzheimer/fisiopatologia , Animais , Chaperona BiP do Retículo Endoplasmático , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fatores de Tempo
8.
Expert Rev Neurother ; 7(7): 887-900, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17610395

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects millions in the aging population worldwide and will affect millions more in the next 20 years. Over 90% of all cases are sporadic, with genetics playing a minor role in the etiology of AD. Therefore, it is crucial to investigate the environment and diet as primary risk factors in AD pathology. This review considers epidemiologic case control studies, and in vitro and in vivo research to investigate the potential of environmental exposure to metals, air pollution and pesticides as well as diet as risk factors for AD. In some cases, the role of genetic mutations and environmental risk is discussed. The evidence examined in this review provides a brief overview of the current literature on selected, significant risk factors in promoting amyloid-beta accumulation and aggregation, thus contributing to neurodegeneration.


Assuntos
Doença de Alzheimer/etiologia , Dieta/efeitos adversos , Meio Ambiente , Poluição do Ar/efeitos adversos , Animais , Exposição Ambiental , Humanos , Metais/efeitos adversos , Praguicidas/efeitos adversos , Fatores de Risco
9.
Curr Alzheimer Res ; 4(2): 219-28, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17430250

RESUMO

Alzheimer's disease (AD) is currently the most prominent form of dementia among the elderly. Although AD manifests in late adult life, it is not clear when the disease actually starts and how long the neuropathological processes take to develop AD. The major unresolved question is the timing and the nature of triggering leading to AD. Is it an early or developmental and/or late phenomenon and what are the factors that trigger the cascade of pathobiochemical processes? To explain the etiology of AD one should consider the neuropathological features, such as neuronal cell death, tau tangles, and amyloid plaque, and environmental factors associated with AD, such as diet, toxicological exposure, and hormonal factors. Current dominant theories of AD etiology are "protein-only", they attribute the cause of the disease directly to the activities of associated proteins once they have been produced; the major limitation is that protein aggregations occur "late in the game". Development and progression of AD has not been explained by protein-only models. In view of this limitation, we propose a "Latent Early-Life Associated Regulation" (LEARn) model, which postulates a latent expression of specific genes triggered at the developmental stage. According to this model, environmental agents (e.g., heavy metals), intrinsic factors (e.g., cytokines), and dietary factors (e.g., cholesterol) perturb gene regulation in a long-term fashion, beginning at early developmental stages; however, these perturbations do not have pathological results until significantly later in life. For example, such actions would perturb APP gene regulation at very early stage via its transcriptional machinery, leading to delayed overexpression of APP and subsequently of Abeta deposition. This model operates on the regulatory region (promoter) of the gene and by the effect of methylation at certain sites within the promoter of specific genes. Promoters tend to have both positive and negative regulatory elements, and promoter activity can be altered by changes in the primary DNA sequence and by epigenetic changes through mechanisms such as DNA methylation at CpG dinucleotides or oxidation of guanosine residues. The basis of the LEARn model is that environmental factors, including metals and dietary factors, operate by interfering the interaction of methylated CpG clusters with binding proteins, such as MeCP2 and SP1. The LEARn model may explain the etiology of AD and other neuropsychiatric and developmental disorders.


Assuntos
Doença de Alzheimer/etiologia , Dieta/efeitos adversos , Meio Ambiente , Idade de Início , Doença de Alzheimer/genética , Animais , Humanos , Modelos Biológicos , Fatores de Risco , Fatores de Tempo
10.
Toxicol Sci ; 95(2): 436-42, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17093203

RESUMO

Lead is a highly neurotoxic metal, and the developing central nervous system is particularly vulnerable to the effects of lead. In this study, transcription factors (TFs) that are altered due to lead exposure were identified using macroarray analysis. Rat pups were lactationally exposed to 0.2% lead acetate from birth through weaning. Changes in the developmental profiles of 30 TFs were screened in hippocampal tissue on postnatal day (PND) 5, 15, and 30. The temporal patterns of some TFs were transiently upregulated or repressed following lead exposure in a stage-specific manner; however, Oct-2, which is involved in the regulation of key developmental processes, exhibited sustained elevations during the entire period of study. Lead-induced elevation of Oct-2 was validated by reverse transcriptase-polymerase chain reaction analysis; however, significant elevation of Oct-2 mRNA expression was detected only on PND 5. The DNA-binding activity and protein levels of Oct-2 were further evaluated and found to be consistently induced on PND 5. The elevations observed in Oct-2 mRNA and protein levels as well as DNA-binding activity on PND 5 suggest that developmental maintenance of Oct-2 DNA binding could be impacted through de novo synthesis. These findings identify Oct-2 as a potential molecular target for Pb and suggest that Oct-2 may be associated with lead-induced disturbances in gene expression.


Assuntos
Hipocampo , Chumbo/toxicidade , Fator 2 de Transcrição de Octâmero , RNA Mensageiro , Animais , Animais Recém-Nascidos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Chumbo/sangue , Masculino , Fator 2 de Transcrição de Octâmero/biossíntese , Fator 2 de Transcrição de Octâmero/metabolismo , Compostos Organometálicos/toxicidade , RNA Mensageiro/biossíntese , RNA Mensageiro/metabolismo , Ratos , Ratos Long-Evans , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima , Desmame
11.
Neurotoxicology ; 27(1): 118-24, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16159668

RESUMO

Polychlorinated biphenyls (PCBs) are prevalent in the environment despite the ban of their use for decades and offer a model system to understand developmental neurotoxicity of persistent pollutants. Disturbances in brain development and cognition are among the neurotoxic manifestations of PCBs. The cellular and molecular basis for PCB-induced developmental neurotoxicity is still unclear; however, a series of in vitro and some in vivo studies have revealed that the disruption of Ca(2+) homeostasis and Ca(2+)-mediated signal transduction play a significant role. The culminating event in a variety of signal transduction pathways is the regulation of gene expression. Therefore, we examined the DNA-binding of prototypical transcription factors in order to identify those that are involved in signal transduction-transcription coupling in the cerebellum and hippocampus of developing rat brains following exposure to a commercial PCB mixture Aroclor 1254. Pregnant rats (Long Evans) were exposed perinatally to 0 or 6 mg/kg/day of Aroclor 1254 (Accu Standard Inc., Lot # 124-191) from gestational day 6 through postnatal day (PND) 21. On specific time points such as days 7, 14, 21, and 60, the DNA-binding of various transcription factors (Sp1, AP-1, NF-kappaB and CREB) was monitored in the cerebellum and hippocampus by a gel mobility shift assay. The induction of DNA-binding of transcription factors was more pronounced during the first 2 weeks after birth than at later periods. Sp1, AP1, and NF-kappaB exhibited a significant increase in DNA-binding following PCB exposure and peaked between 7 and 14 days after birth. However, the DNA-binding activity of CREB remained unchanged. These distinct changes delineate the involvement of specific transduction-transcription coupling pathways that may mediate PCB-induced perturbations in developmental gene expression.


Assuntos
Cerebelo/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Feminino , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Masculino , Exposição Materna , NF-kappa B/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Long-Evans , Fator de Transcrição Sp1/metabolismo , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo
12.
FASEB J ; 19(14): 2083-4, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16230335

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder with clinical manifestations appearing in old age, however, the initial stages of this disease may begin early in life. AD is characterized by the presence of excessive deposits of aggregated beta-amyloid (Abeta) peptides, which are derived from the beta-amyloid precursor protein (APP) following processing by beta-secretase and gamma-secretase. Recently, we have reported that developmental exposure of rats to Pb resulted in latent elevation of APP mRNA, APP, and Abeta in old age. Here we examined whether latent up-regulation in APP expression and Abeta levels is exacerbated by concurrent disturbances in APP processing or Abeta aggregation. Among the environmental metals tested, only Abeta solutions containing Pb promoted the formation of Abeta aggregates at nanomolar concentrations. The lifetime profiles of alpha-, beta-, and gamma-secretases remained constant in adult and aging animals, and developmental exposure to Pb did not alter them. Furthermore, the addition of various concentrations of Pb (0.1 to 50 microM) to cerebral cortical extracts derived from control animals also did not affect the proteolytic activities of these enzymes. Therefore, we propose that amyloidogenesis is promoted by a latent response to developmental reprogramming of the expression of the APP gene by early exposure to Pb, as well as enhancement of Abeta aggregation in old age. In rodents, these events occur without Pb-induced disturbances to the enzymatic processing of APP. The aforementioned results provide further evidence for the developmental basis of amyloidogenesis and late-life disturbances in AD-associated proteins by environmental agents.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Intoxicação por Chumbo/patologia , Naftalenossulfonato de Anilina/farmacologia , Animais , Bário/farmacologia , Relação Dose-Resposta a Droga , Meio Ambiente , Feminino , Corantes Fluorescentes/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Chumbo/farmacologia , Masculino , Mercúrio/farmacologia , Microscopia de Fluorescência , Modelos Biológicos , Peptídeos/química , Ligação Proteica , RNA Mensageiro/metabolismo , Ratos , Ratos Long-Evans , Fatores de Tempo , Regulação para Cima , Zinco/farmacologia
13.
Curr Protoc Toxicol ; Chapter 12: Unit12.11, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23045098

RESUMO

Environmental metals are potentially toxic and can interfere with the metal-binding motifs of various critical proteins. This unit describes an electrophoretic method that can be used to measure the ability of a xenobiotic metal to bind a zinc-finger motif. Information gained using this protocol can lead to the identification of protein targets of metals and shed better light on their mechanisms of action.


Assuntos
Eletroforese em Gel de Poliacrilamida/métodos , Metais/metabolismo , Proteínas/metabolismo , Xenobióticos/metabolismo , Sequência de Aminoácidos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas/química
14.
Int J Dev Neurosci ; 21(6): 347-52, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12927583

RESUMO

Exposure to low-levels of lead (Pb) during early development has been implicated in behavioral abnormalities and cognitive deficits in children. The present study is focused on developmental changes in hippocampus and cerebellum of rats following perinatal exposure to Pb. Pregnant rats were exposed to 0.2% Pb-acetate from gestation day 6 (GD 6) through postnatal day (PND) 21 and the activity levels of acetylcholinesterase (AChE) were estimated in cerebellum and hippocampus of pups at specific time points for 5 weeks. In both the brain regions, Pb-exposure decreased AChE activity with an increase in age. Histochemical observations conducted in 35 days old rat brain showed decreased AChE activity conspicuously in stratum oriens and dentate gyrus of hippocampus, and molecular and granule cell layers of cerebellum. In vitro studies conducted in 35 days old rat brain showed a considerable decrease in the specific activity of AChE at high concentrations (50-100 microM) of Pb in a concentration-dependent manner. However, at low concentrations (5-20 microM), Pb failed to produce such changes. In the presence of eserine (physostigmine), the specific inhibitor of AChE, the inhibitory effect of Pb was potentiated and this was more pronounced at low-concentrations of Pb. The behavioral responses in open-field also showed a significant decrease in both Pb exposed as well as eserine administered rats. These data suggest that low-level perinatal Pb-exposure induces alterations in cholinergic system in the cerebellum and hippocampus of developing brain even after the withdrawal of Pb-exposure, that may contribute to behavioral and learning deficits.


Assuntos
Acetilcolinesterase/biossíntese , Acetilcolinesterase/efeitos dos fármacos , Cerebelo/enzimologia , Hipocampo/enzimologia , Chumbo/toxicidade , Troca Materno-Fetal , Efeitos Tardios da Exposição Pré-Natal , Administração Oral , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Cerebelo/crescimento & desenvolvimento , Inibidores da Colinesterase , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/crescimento & desenvolvimento , Chumbo/administração & dosagem , Neurônios/enzimologia , Gravidez , Ratos , Ratos Sprague-Dawley
15.
Int J Dev Neurosci ; 21(5): 235-44, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12850056

RESUMO

It has been well established that exposure to Pb during critical periods of brain development results in both cognitive and behavioral deficits. Although the mechanism by which Pb induces developmental neurotoxicity is unknown, it may involve alterations in transcription of genes that are essential for growth and differentiation. Recent studies reveal that Pb interferes with growth and differentiation by acting on the transcription factor Sp1. Pb-induced changes in the activity of Sp1 may be consequent to alterations in intermediates in signal transduction pathways. This study examines both in vivo and in vitro the role of signaling factors in mediating the effects of Pb on Sp1 DNA-binding. Hippocampal developmental profiles of Sp1 DNA-binding, PKC, and MAPK protein levels were monitored in Pb-exposed rats. Pb exposure resulted in an induction of Sp1 DNA-binding during PND 5-10 followed by a subsequent decline on PND 15-20. The protein expression profiles for PKCalpha and MAPK followed a relatively similar pattern. To examine the interdependence between Sp1 DNA-binding, PKCalpha, and MAPK, PC12 cells were exposed to Pb and/or NGF. Pb or NGF exposure increased Sp1 DNA-binding. Addition of the PKC inhibitor (staurosporine) diminished NGF and Pb-induced Sp1 DNA-binding, while the MAPK inhibitor (PD 98059), completely abolished both basal and induced Sp1 DNA-binding. These findings demonstrate that Sp1 DNA-binding is regulated by PKC and MAPK, which may serve as mediators through which Pb may indirectly modulate Sp1 DNA-binding.


Assuntos
Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Chumbo/toxicidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Fator de Transcrição Sp1/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Western Blotting , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Regulação da Expressão Gênica , Chumbo/administração & dosagem , Masculino , Fator de Crescimento Neural/farmacologia , Células PC12/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Long-Evans , Transdução de Sinais , Frações Subcelulares
16.
Int J Dev Neurosci ; 21(1): 1-12, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12565691

RESUMO

Zinc finger protein (ZFP) transcription factors are essential for regulation of gene expression in the developing brain. We previously reported that Pb exposure perturbed the DNA-binding of ZFP such as Sp1 and Egr-1 in the cerebellum, which play critical role in CNS development. In this study, we focused on hippocampal Sp1 DNA-binding and mRNA expression in neonatal Pb-exposed animals. The expression pattern of an Sp1 target (NMDAR1) gene was also monitored. To study in vivo and in vitro competition between Pb and Zn, we supplemented animals with Zn, and examined the effects of both metals on hippocampal Sp1 DNA-binding and the DNA-binding of a recombinant Sp1 protein (rhSp1). Tissue metal analysis revealed that only the disposition of Pb in the brain but not its distribution in the blood was influenced by the presence of Zn. The developmental profile of Sp1 DNA-binding exhibited a peak on PND 15 which subsequently declined to adult levels. Consistent with earlier studies, Pb exposure produced premature peaks of Sp1 DNA-binding on PND 5 which later returned to adult levels. The basal and Pb-induced developmental patterns of Sp1 mRNA departed from its DNA-binding profiles. However, the expression patterns of the NMDAR1 gene were relative to Sp1 DNA-binding. Supplementation with zinc provided a protective effect on Pb-induced changes in Sp1 DNA-binding. Moreover, Pb and Zn directly interfered with the DNA-binding of rhSp1 in vitro. These data suggest that Pb and Zn can compete both in vivo and in vitro at the zinc finger domain of Sp1 with a consequential effect on Sp1 DNA-binding, subsequent gene expression and brain development.


Assuntos
Ligação Competitiva/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Chumbo/toxicidade , Fator de Transcrição Sp1/metabolismo , Zinco/administração & dosagem , Administração Oral , Animais , Animais Recém-Nascidos , Química Encefálica/efeitos dos fármacos , Química Encefálica/genética , DNA/metabolismo , Hipocampo/efeitos dos fármacos , Intoxicação do Sistema Nervoso por Chumbo/tratamento farmacológico , Intoxicação do Sistema Nervoso por Chumbo/etiologia , Intoxicação do Sistema Nervoso por Chumbo/metabolismo , Intoxicação do Sistema Nervoso por Chumbo/prevenção & controle , Masculino , Ratos , Valores de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Zinco/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA