Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Ann Oncol ; 30(7): 1114-1120, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31020334

RESUMO

BACKGROUND: The clinical significance of circulating tumour cells (CTCs) in limited-stage small-cell lung cancer (LS-SCLC) is not well defined. We report a planned exploratory analysis of the prevalence and prognostic value of CTCs in LS-SCLC patients enrolled within the phase III randomised CONVERT (concurrent once-daily versus twice-daily chemoradiotherapy) trial. PATIENTS AND METHODS: Baseline blood samples were enumerated for CTCs using CellSearch in 75 patients with LS-SCLC who were enrolled in the CONVERT trial and randomised between twice- and once-daily concurrent chemoradiation. Standard statistical methods were used for correlations of CTCs with clinical factors. Log-rank test and Cox regression analyses were applied to establish the associations of 2, 15 and 50 CTC thresholds with progression-free survival (PFS) and overall survival (OS). An optimal CTC count threshold for LS-SCLC was established. RESULTS: CTCs were detected in 60% (45/75) of patients (range 0-3750). CTC count thresholds of 2, 15 and 50 CTCs all significantly correlate with PFS and OS. An optimal CTC count threshold in LS-SCLC was established at 15 CTCs, defining 'favourable' and 'unfavourable' prognostic risk groups. The median OS in <15 versus ≥15 CTCs was 26.7 versus 5.9 m (P = 0.001). The presence of ≥15 CTCs at baseline independently predicted ≤1 year survival in 70% and ≤2 years survival in 100% of patients. CONCLUSION: We report the prognostic value of baseline CTC count in an exclusive LS-SCLC population at thresholds of 2, 15 and 50 CTCs. Specific to LS-SCLC, ≥15 CTCs was associated with worse PFS and OS independent of all other factors and predicted ≤2 years survival. These results may improve disease stratification in future clinical trial designs and aid clinical decision making. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00433563.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/terapia , Células Neoplásicas Circulantes/patologia , Carcinoma de Pequenas Células do Pulmão/sangue , Carcinoma de Pequenas Células do Pulmão/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimiorradioterapia , Progressão da Doença , Fracionamento da Dose de Radiação , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Células Neoplásicas Circulantes/efeitos dos fármacos , Células Neoplásicas Circulantes/efeitos da radiação , Carcinoma de Pequenas Células do Pulmão/patologia , Taxa de Sobrevida
2.
Br J Pharmacol ; 172(4): 957-74, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25302413

RESUMO

The incidence of drug-induced structural cardiotoxicity, which may lead to heart failure, has been recognized in association with the use of anthracycline anti-cancer drugs for many years, but has also been shown to occur following treatment with the new generation of targeted anti-cancer agents that inhibit one or more receptor or non-receptor tyrosine kinases, serine/threonine kinases as well as several classes of non-oncology agents. A workshop organized by the Medical Research Council Centre for Drug Safety Science (University of Liverpool) on 5 September 2013 and attended by industry, academia and regulatory representatives, was designed to gain a better understanding of the gaps in the field of structural cardiotoxicity that can be addressed through collaborative efforts. Specific recommendations from the workshop for future collaborative activities included: greater efforts to identify predictive (i) preclinical; and (ii) clinical biomarkers of early cardiovascular injury; (iii) improved understanding of comparative physiology/pathophysiology and the clinical predictivity of current preclinical in vivo models; (iv) the identification and use of a set of cardiotoxic reference compounds for comparative profiling in improved animal and human cellular models; (v) more sharing of data (through publication/consortia arrangements) on target-related toxicities; (vi) strategies to develop cardio-protective agents; and (vii) closer interactions between preclinical scientists and clinicians to help ensure best translational efforts.


Assuntos
Cardiotoxicidade/etiologia , Cardiotoxinas/efeitos adversos , Doenças Cardiovasculares/etiologia , Animais , Antineoplásicos/efeitos adversos , Cardiotoxicidade/metabolismo , Cardiotoxicidade/patologia , Cardiotoxicidade/fisiopatologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Humanos
3.
Breast ; 23(5): 552-60, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25124235

RESUMO

Haematogenous spread of circulating tumour cells (CTCs) is the principle mechanism for development of metastases. Research into the enumeration and characterisation of CTCs, particularly in the last decade, has allowed the introduction of semi-automated CTC assessment in the clinical setting. In breast cancer, CTC enumeration is being used as a prognostic biomarker, a predictive biomarker of treatment response and is being assessed to guide treatment in both the early and metastatic setting. CTC characterisation has the potential to direct targeted therapies, such as HER2 therapies in HER2 negative primary breast tumour patients. However, CTC assessment has considerable challenges. Capture and identification of these very rare cells is currently largely dependent on a presumed homogeneity of phenotype. In addition, high throughput assays are lacking. The clinical significance of CTCs is incompletely understood. A large proportion of CTC positive patients have no evidence of metastases, raising the issue of either inconsequential tumour dormancy or non-viable CTCs. CTCs may have additional clinical sequelae such as promoting venous thrombosis. However CTCs provide a real-time liquid biopsy of the tumour and represent an exciting, minimally invasive method of assessing disease status and also a novel therapeutic target for malignancy.


Assuntos
Neoplasias da Mama/patologia , Células Neoplásicas Circulantes , Biomarcadores Tumorais , Contagem de Células , Feminino , Humanos , Fenótipo , Prognóstico
4.
Xenobiotica ; 36(10-11): 1122-39, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17118920

RESUMO

With the aim of evaluating the usefulness of an in vitro system for assessing the potential hepatotoxicity of compounds, the paper describes several methods of obtaining mathematical models for the prediction of compound-induced toxicity in vivo. These models are based on data derived from treating rat primary hepatocytes with various compounds, and thereafter using microarrays to obtain gene expression 'profiles' for each compound. Predictive models were constructed so as to reduce the number of 'probesets' (genes) required, and subjected to rigorous cross-validation. Since there are a number of possible approaches to derive predictive models, several distinct modelling strategies were applied to the same data set, and the outcomes were compared and contrasted. While all the strategies tested showed significant predictive capability, it was interesting to note that the different approaches generated models based on widely disparate probesets. This implies that while these models may be useful in ascribing relative potential toxicity to compounds, they are unlikely to provide significant information on underlying toxicity mechanisms. Improved predictivity will be obtained through the generation of more comprehensive gene expression databases, covering more 'toxicity space', and by the development of models that maximize the observation, and combination, of individual differences between compounds.


Assuntos
Expressão Gênica , Hepatócitos/metabolismo , Modelos Biológicos , Toxicogenética , Animais , Análise por Conglomerados , Análise dos Mínimos Quadrados , Análise de Componente Principal , Ratos
5.
Hum Exp Toxicol ; 21(8): 429-37, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12412636

RESUMO

The peroxisome proliferator activated receptor-gamma (PPARgamma) is an attractive target for therapeutic intervention, as modulation of PPARgamma-regulated pathways is potentially beneficial in a number of disease areas. This review provides an overview of what is known about the biology of PPARgamma, and an indication of what progress has been made towards drug development in several therapy areas. As well as efficacy, the safety of drugs is of course an important issue, and a substantial volume of preclinical and clinical information has already accumulated for PPARgamma agonists. Here we discuss some of the major toxicology issues with PPARgamma agonists, and give a perspective on likely issues concerning the development of PPARgamma modulators in the future.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Hiperlipidemias/fisiopatologia , Hipoglicemiantes/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Tiazóis/farmacologia , Tiazolidinedionas , Fatores de Transcrição/fisiologia , Proteínas de Ligação a DNA , Humanos , Inflamação , Resistência à Insulina , Ligantes , Microcorpos , Proteínas Nucleares , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/química , Proteínas Repressoras , Fatores de Transcrição/agonistas , Fatores de Transcrição/química , Dedos de Zinco
7.
IDrugs ; 3(2): 179-81, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16107935

RESUMO

This meeting focused on the problems facing the pharmaceutical industry, given the need for faster and higher throughput toxicity testing, and how emerging technologies are being applied to address this need. Inevitably, the emphasis was on in vitro approaches, in particular, genomics-based technology such as transcript profiling, but due attention was paid to the necessity of relating in vitro-derived data to toxic endpoints in vivo. A number of technology-providing companies were given the opportunity to draw attention to their proprietary systems and explain how they might be applied to drug safety assessment. Finally, a session was devoted to a discussion of available 'in silico' systems and how these are being developed and refined as the need for computer-based screening increases.

8.
Carcinogenesis ; 20(3): 369-72, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10190548

RESUMO

Peroxisome proliferators (PP) cause peroxisome proliferation, associated with rodent hepatocyte growth perturbation and hepatocarcinogenesis. However, in humans this class of non-genotoxic carcinogens does not appear to have the same adverse effects. The peroxisome proliferator-activated receptor alpha (PPARalpha) mediates the effects of PPs in rodents via peroxisome proliferator response elements (PPREs) upstream of PP-responsive genes such as acyl coenzyme A oxidase (ACO). When the human ACO promoter was cloned previously, it was found to be active and to contain a consensus PPRE (-1918 AGGTCA C TGGTCA -1906). To confirm and extend those original findings, we isolated a 2 kb genomic fragment of the ACO gene promoter from a human liver biopsy and used it to create a beta-galactosidase reporter gene plasmid. The human ACO promoter reporter plasmid was added to both Hepalclc7 and NIH 3T3 cells together with a plasmid expressing mPPARa and assessed for its ability to drive PP-mediated gene transcription. The human ACO promoter fragment was inactive, unlike the equivalent rat ACO promoter fragment used as a positive control. The PPRE within our cloned fragment of the human ACO promoter differed at three positions (5'-AGGTCA G CTGTCA-3') from the previously published active human ACO promoter. Next, we studied the frequency of the inactive versus the active human PPRE within the human population. Using a PCR strategy, we isolated and analysed genomic DNA fragments from 22 unrelated human individuals and from the human hepatoma cell line HepG2. In each case, the PPRE contained the inactive sequence. These data show that the human ACO gene promoter found in a sample human population is inactive. This may explain at the genomic level the lack of response of humans to some of the adverse effects of the PP class of non-genotoxic hepatocarcinogens.


Assuntos
Oxirredutases/genética , Proliferadores de Peroxissomos/toxicidade , Células 3T3 , Acil-CoA Oxidase , Animais , Sequência de Bases , Linhagem Celular , DNA , Primers do DNA , Humanos , Neoplasias Hepáticas Experimentais/induzido quimicamente , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ratos , Especificidade da Espécie
9.
J Mol Endocrinol ; 22(1): 1-8, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9924174

RESUMO

Peroxisome proliferators (PPs) are chemicals of industrial and pharmaceutical importance that elicit liver carcinogenesis by a non-genotoxic mechanism. One of the intriguing properties of PPs is that the pleiotropic effects of these compounds (including increased DNA synthesis and peroxisome proliferation) are seen in rats and mice only, but not humans. It is important to determine the risks to humans of environmental and therapeutic exposure to these compounds by understanding the mechanisms of non-genotoxic hepatocarcinogenesis in rodents. To understand this apparent lack of human susceptibility, attention has focused on the peroxisome proliferator-activated receptor alpha (PPARalpha), which appears to mediate the effects of PPs in rodents. It is also known to mediate the hypolipidaemic effects that fibrate drugs exert on humans with elevated plasma cholesterol and triglyceride levels. Human PPARalphas share many functional characteristics with the rodent receptors, in that they can be transcriptionally activated by PPs and regulate specific gene expression. However, one key difference is that PPARalpha is less abundant in human than in rodent liver, which has led to the suggestion that species differences result from quantitative differences in gene expression. In this review we describe the effects of PPs and what is known of the molecular mechanisms of action and species differences with respect to rodents and man. Attention will be given to differences in the amounts of PPARalpha between species as well as the 'qualitative' aspects of PPARalpha-mediated gene regulation which might also explain the activation of some genes and not of others in human liver by PPs.


Assuntos
Neoplasias Hepáticas Experimentais/fisiopatologia , Proliferadores de Peroxissomos/toxicidade , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Anticolesterolemiantes/efeitos adversos , Anticolesterolemiantes/toxicidade , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fator I de Transcrição COUP , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito , Humanos , Metabolismo dos Lipídeos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Modelos Biológicos , Fosfoproteínas/metabolismo , Ratos , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides , Especificidade da Espécie , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Toxicol Appl Pharmacol ; 152(1): 107-18, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9772206

RESUMO

The implementation of a rat hepatocyte model system and differential display-polymerase chain reaction resulted in the isolation of ZFP-37 as a peroxisome proliferator-responsive gene. In addition to being responsive to peroxisome proliferators, rat ZFP-37 (rZFP-37) mRNA accumulates rapidly after treating cells with several other hepatic tumor promoters, serum, and cycloheximide, indicating that this gene belongs to the immediate-early growth responsive gene family. Although rZFP-37 and mouse ZFP-37 (mZFP-37) are both members of the Krüppel-associated box and C2H2 zinc finger superfamily of proteins, there are several features that distinguish the two proteins. The primary protein sequences of rat and mouse ZFP-37 are highly conserved, especially within the region encoding the 12 C2H2 zinc finger motifs; however, a region believed to be involved in DNA binding in mZFP-37 is divergent in rZFP-37. Mouse ZFP-37 mRNA is expressed almost exclusively in testes and brain, whereas rZFP-37 mRNA is expressed in testes, brain, kidney, spleen, thymus, lung, and at low levels in liver. A major difference between regulation of ZFP-37 in the two species exists as rZFP-37 is induced, while mZFP-37 is repressed, in liver by the administration of the potent peroxisome proliferator Wy 14,643. Despite the fact that mZFP-37 is believed to be important in cell growth and differentiation in testes and brain, the pronounced differences in regulation of this gene in two closely related species preclude an extrapolation to rZFP-37's biological role. Nonetheless, the effects of tumor promoters and mitogens on its expression and the inclusion of rZFP-37 into the immediate-early growth gene families raise the possibility that this gene plays a role in hepatocyte proliferation and/or differentiation.


Assuntos
DNA Complementar/genética , Proteínas de Ligação a DNA/genética , Fígado/efeitos dos fármacos , Microcorpos/metabolismo , Proliferadores de Peroxissomos/farmacologia , Pirimidinas/farmacologia , Dedos de Zinco/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Primers do DNA/química , Proteínas de Ligação a DNA/isolamento & purificação , Proteínas de Ligação a DNA/metabolismo , Dactinomicina/farmacologia , Expressão Gênica/efeitos dos fármacos , Genes Precoces/genética , Fígado/química , Masculino , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Distribuição Tecidual , Fatores de Transcrição
11.
Regul Toxicol Pharmacol ; 27(1 Pt 1): 47-60, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9629596

RESUMO

The purpose of the workshop "Do Peroxisome Proliferating Compounds Pose a Hepatocarcinogenic Hazard to Humans?" was to provide a review of the current state of the science on the relationship between peroxisome proliferation and hepatocarcinogenesis. There has been much debate regarding the mechanism by which peroxisome proliferators may induce liver tumors in rats and mice and whether these events occur in humans. A primary goal of the workshop was to determine where consensus might be reached regarding the interpretation of these data relative to the assessment of potential human risks. A core set of biochemical and cellular events has been identified in the rodent strains that are susceptible to the hepatocarcinogenic effects of peroxisome proliferators, including peroxisome proliferation, increases in fatty acyl-CoA oxidase levels, microsomal fatty acid oxidation, excess production of hydrogen peroxide, increases in rates of cell proliferation, and expression and activation of the alpha subtype of the peroxisome proliferator-activated receptor (PPAR-alpha). Such effects have not been identified clinically in liver biopsies from humans exposed to peroxisome proliferators or in in vitro studies with human hepatocytes, although PPAR-alpha is expressed at a very low level in human liver. Consensus was reached regarding the significant intermediary roles of cell proliferation and PPAR-alpha receptor expression and activation in tumor formation. Information considered necessary for characterizing a compound as a peroxisome proliferating hepatocarcinogen include hepatomegaly, enhanced cell proliferation, and an increase in hepatic acyl-CoA oxidase and/or palmitoyl-CoA oxidation levels. Given the lack of genotoxic potential of most peroxisome proliferating agents, and since humans appear likely to be refractive or insensitive to the tumorigenic response, risk assessments based on tumor data may not be appropriate. However, nontumor data on intermediate endpoints would provide appropriate toxicological endpoints to determine a point of departure such as the LED10 or NOAEL which would be the basis for a margin-of-exposure (MOE) risk assessment approach. Pertinent factors to be considered in the MOE evaluation would include the slope of the dose-response curve at the point of departure, the background exposure levels, and variability in the human response.


Assuntos
Carcinógenos/toxicidade , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Animais , Testes de Carcinogenicidade , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cobaias , Humanos , Fígado/patologia , Mesocricetus , Camundongos , Ratos , Medição de Risco , Especificidade da Espécie , Estados Unidos , United States Environmental Protection Agency , United States Food and Drug Administration
12.
Regul Toxicol Pharmacol ; 27(1 Pt 2): 47-60, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9618323

RESUMO

The purpose of the workshop "Do Peroxisome Proliferating Compounds Pose a Hepatocarcinogenic Hazard to Humans?" was to provide a review of the current state of the science on the relationship between peroxisome proliferation and hepatocarcinogenesis. There has been much debate regarding the mechanism by which peroxisome proliferators may induce liver tumors in rats and mice and whether these events occur in humans. A primary goal of the workshop was to determine where consensus might be reached regarding the interpretation of these data relative to the assessment of potential human risks. A core set of biochemical and cellular events has been identified in the rodent strains that are susceptible to the hepatocarcinogenic effects of peroxisome proliferators, including peroxisome proliferation, increases in fatty acyl-CoA oxidase levels, microsomal fatty acid oxidation, excess production of hydrogen peroxide, increases in rates of cell proliferation, and expression and activation of the alpha subtype of the peroxisome proliferator-activated receptor (PPAR-alpha). Such effects have not been identified clinically in liver biopsies from humans exposed to peroxisome proliferators or in in vitro studies with human hepatocytes, although PPAR-alpha is expressed at a very low level in human liver. Consensus was reached regarding the significant intermediary roles of cell proliferation and PPAR-alpha receptor expression and activation in tumor formation. Information considered necessary for characterizing a compound as a peroxisome proliferating hepatocarcinogen include hepatomegaly, enhanced cell proliferation, and an increase in hepatic acyl-CoA oxidase and/or palmitoyl-CoA oxidation levels. Given the lack of genotoxic potential of most peroxisome proliferating agents, and since humans appear likely to be refractive or insensitive to the tumorigenic response, risk assessments based on tumor data may not be appropriate. However, nontumor data on intermediate endpoints would provide appropriate toxicological endpoints to determine a point of departure such as the LED10 or NOAEL which would be the basis for a margin-of-exposure (MOE) risk assessment approach. Pertinent factors to be considered in the MOE evaluation would include the slope of the dose-response curve at the point of departure, the background exposure levels, and variability in the human response. Copyright 1998 Academic Press.

13.
Arch Toxicol ; 72(3): 169-77, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9520140

RESUMO

The peroxisome proliferator class of non-genotoxic rodent hepatocarcinogens cause hepatocyte DNA synthesis, peroxisome proliferation and liver tumours when administered to rats and mice, but fail to induce S-phase or peroxisome proliferation in hepatocytes from other species including guinea-pigs, dogs, and primates including humans. There are compelling data that implicate a nuclear receptor, the peroxisome proliferator-activated receptor-alpha (PPARalpha) as an important mediator of the toxic and carcinogenic effects of peroxisome proliferators (PPs). We were interested to consider the guinea-pig as a possible model for human responses to these compounds. This manuscript describes the isolation of a full-length cDNA encoding PPARalpha from guinea-pig liver that is closely related to receptors identified previously in mouse, rat and human. RNA hybridisation experiments suggested that the livers of the PP-responsive rat and mouse contained relatively high levels of PPARalpha transcripts, whereas in human and guinea-pig liver PPARalpha mRNA was much less abundant. Functional analyses suggested that the guinea-pig PPARalpha was able to be activated by PPs. DNA binding studies using in vitro translated proteins showed that the guinea-pig receptor was able to bind specifically to DNA in the presence of the retinoid X receptor (RXR), and transient transfection assays showed that the guinea-pig PPARalpha was capable of being transcriptionally activated in a concentration-dependent fashion by the PPs Wy-14,643 and nafenopin. Also, in guinea-pig primary hepatocyte cultures, a dominant negative repressor of PPARalpha ablated the suppression of spontaneous apoptosis by PPs. Taken together, these data show that the 'non-responsive' guinea-pig expresses active PPARalpha in the liver at reduced levels, and may be a useful model for exploring the mechanisms underlying the human response to PPs.


Assuntos
Fígado/química , Receptores Citoplasmáticos e Nucleares/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Cobaias , Humanos , Fígado/efeitos dos fármacos , Fígado/fisiologia , Camundongos , Dados de Sequência Molecular , Ratos , Receptores Citoplasmáticos e Nucleares/genética , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Fatores de Transcrição/genética
14.
Carcinogenesis ; 19(1): 43-8, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9472691

RESUMO

Peroxisome proliferators (PPs) are a class of nongenotoxic rodent hepatocarcinogens. We have demonstrated previously that PPs suppress both spontaneous rat hepatocyte apoptosis and that induced by exogenous stimuli such as transforming growth factor-beta1 (TGFbeta1). PPs transcriptionally activate the peroxisome proliferator activated receptor-alpha (PPAR alpha), a member of the nuclear hormone receptor superfamily. Here, we investigate whether activation of PPAR alpha mediates the suppression of rat hepatocyte apoptosis induced by PPs. We isolated a naturally occurring variant form of PPAR alpha (hPPAR alpha-6/29) from human liver by PCR cloning. Electrophoretic mobility shift assays (EMSA) demonstrated that hPPAR alpha-6/29 shared the ability of mPPAR alpha to heterodimerise with the retinoid X receptor (RXR) and bind to DNA. When hPPAR alpha-6/29 was transfected into Hepa1c1c7 cells together with a reporter plasmid containing a PPAR response element (PPRE), hPPAR alpha-6/29, unlike mPPAR alpha, could not be activated by PPs. Furthermore, hPPAR alpha-6/29 could act as a dominant negative regulator of PPAR-mediated gene transcription since increasing concentrations of hPPAR alpha-6/29 abrogated the activation of co-transfected mPPAR alpha. When introduced into primary rat liver cell cultures by transient transfection, hPPAR alpha-6/29 prevented the suppression of hepatocyte apoptosis by the PP nafenopin, but not that seen in response to phenobarbitone (PB), a nongenotoxic carcinogen whose action does not involve PPAR alpha. The suppression of hepatocyte apoptosis was abrogated completely even though only 30% of hepatocytes were transfected, suggesting the involvement of a soluble factor. These data indicate that activation of rat liver PPAR alpha provides a survival signal for hepatocytes, preventing their death in response to apoptotic stimuli.


Assuntos
Apoptose , Fígado/fisiologia , Linfotoxina-alfa/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Carcinoma Hepatocelular , Células Cultivadas , Primers do DNA , Genes Reporter , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Neoplasias Hepáticas , Masculino , Camundongos , Proteínas Nucleares/fisiologia , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/biossíntese , Proteínas Recombinantes/biossíntese , Alinhamento de Sequência , Fatores de Transcrição/biossíntese , Transfecção , Células Tumorais Cultivadas
16.
J Biochem Mol Toxicol ; 12(2): 79-82, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9443064

RESUMO

The nongenotoxic carcinogens phenobarbitone (PB) and methyl clofenapate (MCP) and the hepatomitogen pregnenolone 16 alpha carbonitrile (PCN) are direct inducers of hepatic S-phase in rats, whereas the S-phase seen after partial hepatectomy is regenerative. We have investigated S-phase and immediate-early gene expression (c-myc and c-jun) in rat liver following these treatments to study the differences in gene expression associated with direct vs. regenerative responses. Both partial hepatectomy (one- and two-thirds) and mitogen treatment caused an increase in hepatic S-phase that peaked around 36 hours. Two-thirds partial hepatectomy caused the greatest increase in S-phase followed by one-third partial hepatectomy, then the mitogens PCN, MCP, and PB in that order. This order of response was also seen with c-jun and to a lesser degree with c-myc expression, suggesting that immediate-early gene expression might be linked not only to regenerative S-phase but also to direct mitogen-induced responses.


Assuntos
Genes Precoces , Regeneração Hepática , Fígado/crescimento & desenvolvimento , Mitógenos/farmacologia , Animais , Expressão Gênica/efeitos dos fármacos , Genes jun , Genes myc , Fígado/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos F344 , Fase S
17.
Toxicol Lett ; 102-103: 91-6, 1998 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-10022238

RESUMO

Peroxisome proliferators (PPs) are a class of non-genotoxic rodent hepatocarcinogens that act by perturbing liver growth regulation. We have demonstrated previously that PPs suppress both spontaneous rat hepatocyte apoptosis and that induced by exogenous stimuli such as transforming growth factor-beta1 (TGF beta1). More recently, we have demonstrated that PPs can suppress apoptosis induced by more diverse stimuli such as DNA damage or ligation of Fas, a receptor related to the tumour necrosis factor alpha (TNF alpha) family of cell surface receptors. PPs transcriptionally activate the peroxisome proliferator activated receptor-alpha, PPAR alpha, a member of the nuclear hormone receptor superfamily. We investigated whether activation of PPAR alpha mediates the suppression of rat hepatocyte apoptosis induced by PPs. We isolated a naturally occurring variant form of PPAR alpha (hPPAR alpha-6/29) from human liver by PCR cloning. hPPAR alpha-6/29 shared the ability of mPPAR alpha to bind to DNA but, unlike mPPAR alpha, could not be activated by PPs. Furthermore, hPPAR alpha-6/29 could act as a dominant negative regulator of PPAR-mediated gene transcription. When introduced into primary rat liver cell cultures by transient transfection, hPPAR alpha-6/29 prevented the suppression of hepatocyte apoptosis by the PP nafenopin, but not that seen in response to phenobarbitone (PB), a non-genotoxic carcinogen whose action does not involve PPAR alpha. The suppression of hepatocyte apoptosis was abrogated completely even though only 30% of hepatocytes were transfected, suggesting the involvement of a soluble factor. Recent data have suggested that TNF alpha, perhaps released by liver Kupffer cells in response to PPs, may play a key role in mediating the effects of PPs on hepatocyte growth regulation.


Assuntos
Neoplasias Hepáticas/induzido quimicamente , Proliferadores de Peroxissomos/toxicidade , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Citocinas/fisiologia , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Camundongos , Ratos
18.
Biochem Biophys Res Commun ; 239(2): 522-6, 1997 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-9344863

RESUMO

We have investigated the basis of the lack of activity of a natural variant human peroxisome proliferator-activated receptor alpha, hPPARalpha6/29. A subcloning approach was used to change the four variant amino acids in the hPPARalpha6/29 sequence, individually and in combination, to those found in an active human PPARalpha. Individual amino acid "back mutations" were unable to confer on hPPARalpha6/29 the ability to be activated by peroxisome proliferators in a transient transfection assay. Although hPPARalpha6/29 was able to bind specifically to DNA in the presence of the retinoid X receptor alpha (RXRalpha), the complete restoration of receptor transcriptional activity required two separate back mutations of the hPPARalpha6/29 sequence, namely amino acid 123 in the DNA binding domain, and amino acid 444 close to the C-terminus. This suggests that sequences in the PPARalpha DNA binding domain influence other receptor functions besides DNA binding.


Assuntos
Aminoácidos/genética , DNA/metabolismo , Microcorpos/genética , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Transcrição Gênica , Alanina/genética , Alanina/fisiologia , Substituição de Aminoácidos/genética , Aminoácidos/fisiologia , Humanos , Ligantes , Metionina/genética , Metionina/fisiologia , Ligação Proteica/genética , Estrutura Terciária de Proteína
19.
Eur J Drug Metab Pharmacokinet ; 22(4): 325-8, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9512929

RESUMO

Peroxisome proliferators (PPs) are a group of compounds which cause peroxisome proliferation and hepatocellular carcinomas in rodents, and form a class of non-genotoxic carcinogens. It is thought that PPs act via a receptor similar to members of the nuclear hormone superfamily termed the peroxisome proliferator activated receptor (PPAR). Multiple subtypes (alpha, beta, delta and gamma) of the receptor exist and are differentially expressed between tissues and species. PPAR alpha has been shown to activate transcription by binding to response elements upstream of peroxisome proliferator responsive genes. However, despite the isolation of transcriptionally active human subtypes of the receptor, hPPAR alpha and hNUC1, humans are thought to be non-responsive to PPs. This is possibly due to regulation of PPAR, and it has been recently reported that PPAR alpha is a phosphoprotein in vivo and insulin regulates its phosphorylation. A system employing epitope-tagged receptors has been developed to study this further, with the aim of establishing stably transfected cell lines expressing high levels of epitope-tagged mouse and human PPAR alpha. Our experiments clearly demonstrate that an epitope-tagged mPPAR alpha receptor has an equal ability to modulate transcription as the native receptor in transactivation assays and will be further used to examine the molecular mechanisms of peroxisome proliferation.


Assuntos
Epitopos/genética , Regulação da Expressão Gênica/genética , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Animais , Cálcio/fisiologia , Genes Reporter , Vetores Genéticos , Humanos , Neoplasias Hepáticas Experimentais/genética , Camundongos , Mutagênicos/farmacologia , Plasmídeos/genética , Pirimidinas/farmacologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Fatores de Transcrição/biossíntese , Ativação Transcricional/genética , Transfecção/genética
20.
Ann N Y Acad Sci ; 804: 252-65, 1996 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-8993548

RESUMO

We have been attempting to elucidate the molecular mechanisms through which peroxisome proliferators exert their pleiotropic effects, with particular emphasis on understanding why humans appear unresponsive to these compounds. There is a wealth of data to implicate the peroxisome proliferator-activated receptor alpha (PPAR alpha) in mediating these effects in rodent species; PPAR alpha is expressed in tissues that show physiological changes in response to PPs, is transcriptionally activated in vitro by a variety of PPs, and it has been recently demonstrated that mice lacking this receptor are refractory to the effects of clofibrate and Wy-14,643, at least in the short term. It is conceivable that differences in PPAR alpha between responsive rodent and unresponsive human subjects may provide the key to understanding the basis of this species variation in response, and with this in mind we have been studying the biology of PPAR alpha in humans and looking at interindividual variation. There is already published evidence, albeit on only two sequences, for structural and functional polymorphism in human PPAR alphas. We have extended these findings, and shown that: There is considerable variation in hPPAR alpha cDNAs obtained from different individuals, both at the gross structural level (lack of a coding exon) and of a more subtle nature (single base changes leading to amino acid substitutions). One such cDNA, the sequence of which differs at only three amino acids from that published, encodes a receptor that is incapable of transcriptional activation by potent PPs. The degree to which hPPAR alpha transcripts are expressed in human livers can vary by up to an order of magnitude between individuals. The tissue-specific expression profile of PPAR alpha in humans is very different from that in rat and mouse. In particular, the human liver contains generally low levels of PPAR alpha in contrast to the responsive rodents, in which potent PPs cause liver tumors. Taken together, these data suggest first that human and rodent PPAR alphas differ according to a number of molecular and biochemical criteria, and secondly that there is a degree of interindividual variation in PPAR alpha structure and function. Studies are ongoing to clarify this further, but human polymorphism may go some way towards explaining the apparent paradox that active PPAR alpha receptors can be isolated from an "unresponsive" species.


Assuntos
Microcorpos/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Acil-CoA Oxidase , Animais , DNA Complementar/genética , Proteínas de Ligação a DNA/fisiologia , Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Humanos , Camundongos , Oxirredutases/genética , Regiões Promotoras Genéticas , Ratos , Especificidade da Espécie , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...