Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cancer Lett ; 93(1): 121-8, 1995 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-7600539

RESUMO

We have shown previously that approximately 1 in 10,000 primary hepatocytes isolated from untreated rats undergo clonal growth in soft agar in vitro in response to the synergistic action of nafenopin, a peroxisome proliferator (PP) and epidermal growth factor (EGF), a naturally occurring liver growth regulator. Here, we demonstrate that prior treatment of the animals with the genotoxic hepatocarcinogen diethylnitrosamine (DEN) caused a dose-dependent increase in soft agar colony numbers formed in vitro. These data suggest that the colony assay may offer a method of detecting in vitro hepatocytes transformed in vivo by DEN. It is known that rats treated with DEN develop enzyme altered foci prior to the development of tumours. The majority of these foci express high levels of gamma-glutamyl transpeptidase (GGT). However, foci promoted by PPs do not show this increased enzyme activity. In the present study, the colonies we have generated in vitro mimicked this pattern since the majority (approximately 80%) of the spontaneous colonies expressed GGT whereas colonies promoted by the synergistic action of nafenopin and EGF were mainly (75%) GGT negative. The proportion of colonies positive for GGT were similar using either hepatocytes isolated from control or from DEN-initiated rats. Further studies are required to assess if the hepatocytes selected for clonal expansion by this EGF/nafenopin regime reflect the presumed pre-neoplastic cells induced by genotoxin in vivo and associated with an increased propensity to cancer.


Assuntos
Testes de Carcinogenicidade/métodos , Carcinoma Hepatocelular/induzido quimicamente , Inibição de Contato/efeitos dos fármacos , Neoplasias Hepáticas/induzido quimicamente , Ensaio Tumoral de Célula-Tronco/métodos , Ágar , Animais , Células Cultivadas , Dietilnitrosamina , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Fator de Crescimento Epidérmico/farmacologia , Regulação Neoplásica da Expressão Gênica , Masculino , Nafenopina/farmacologia , Ratos , Ratos Wistar , gama-Glutamiltransferase
2.
Toxicology ; 181-182: 167-70, 2002 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-12505304

RESUMO

Peroxisome proliferators (PPs) such as the hypolipidaemic drug, nafenopin and the phthalate plasticiser 2-diethylhexylphthalate induce rodent hepatocyte cell proliferation and suppress apoptosis leading to tumours. PPs act via the nuclear hormone receptor peroxisome proliferator activated receptor alpha (PPAR alpha) which directly regulates genes implicated in the response to PPs such as the peroxisomal gene acyl CoA oxidase. As expected for xenobiotics that perturb proliferation, PPs alter expression of cell cycle regulatory proteins. However, the ability to alter expression of cyclins and cyclin-dependent kinases is shared by physiological hepatic mitogens such as epidermal growth factor and is thus unlikely to be specific to the PP-induced aberrant growth associated with hepatocarcinogenesis. Recent evidence suggests that the response of hepatocytes to PPs is not only dependent upon PPAR alpha but also on the trophic environment provided by nonparenchymal cells and by cytokines such as tumour necrosis factor alpha. Additionally, the ability of PPs to suppress apoptosis and induce proliferation depends upon survival signalling mediated by p38 mitogen activated protein kinase. The cross talk between PPAR alpha-mediated transcription, survival signalling and cell cycle will be discussed with particular emphasis on relevance to toxicology.


Assuntos
Apoptose/fisiologia , Divisão Celular/fisiologia , Proliferadores de Peroxissomos/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Antígenos CD/genética , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Citocinas/fisiologia , Humanos , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral
3.
Toxicol Lett ; 120(1-3): 301-6, 2001 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-11323188

RESUMO

Chemicals with the potential to cause cancer through damaging DNA can be readily identified in a range of in vitro screens that detect genotoxicity. However, many carcinogens are non-genotoxic yet cause rodent tumours, particularly in the liver. Some non-genotoxic carcinogens such as the peroxisome proliferators (PPs) act directly to cause liver growth and proliferation, whereas others such as carbon tetrachloride cause liver damage, followed by regenerative hyperplasia. Current data support a role for cytokines such as tumour necrosis factor alpha (TNFalpha) and interleukin 1 (IL1) in hepatocarcinogenesis. However, these data give rise to conflicting hypotheses; in some experimental models, TNFalpha appears to mediate damage, whereas in others it is postulated to play a role in tissue repair. Recently, we have shown that TNFalpha acting via TNFalpha receptor 1 and p38 MAP kinase suppresses hepatocyte apoptosis. However, when new protein synthesis is disabled, TNFalpha becomes a death signal. An understanding of the role of cytokines in rodent hepatocarcinogenesis will allow the development of markers that can be used to identify, at an early stage, those chemicals with the potential to induce rodent tumours.


Assuntos
Citocinas/fisiologia , Neoplasias Hepáticas/induzido quimicamente , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Humanos , Proliferadores de Peroxissomos/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno
4.
Toxicol Lett ; 112-113: 49-57, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10720712

RESUMO

Peroxisome proliferators (PPs) are nongenotoxic rodent hepatocarcinogens that cause liver enlargement and hepatocarcinogenesis associated with peroxisome proliferation, induction of hepatocyte DNA synthesis and suppression of apoptosis. Acyl CoA oxidase (ACO) is a key enzyme of peroxisomal beta-oxidation and its transcriptional activation by PPs is often used as marker for the rodent response. PPs activate the peroxisome proliferator activated receptor-alpha, PPARalpha. Recent data suggest a role for tumour necrosis factor alpha (TNFalpha). This cytokine appears to be permissive for a PPARalpha-dependent growth response to PPs. Humans and guinea pigs appear to be nonresponsive to the adverse effects of PPs noted in rodents. These species differences can be attributed to reduced quantity of full length functional PPARalpha in human liver and evidence supports the presence of a truncated form of PPARalpha, hPPARalpha8/14 in human liver. In addition, species differences could be attributed to qualitative differences in the PPARalpha-mediated response because the promoter for human ACO differs in sequence and activity from the rat equivalent. These data contribute to our understanding of how chemicals may cause tumours in rodents and how this response may differ in humans.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Proliferadores de Peroxissomos/toxicidade , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Fatores de Transcrição/efeitos dos fármacos , Animais , Sequência de Bases , Genoma Humano , Cobaias , Humanos , Fígado/metabolismo , Reação em Cadeia da Polimerase , Ratos , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Especificidade da Espécie , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
5.
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
6.
Mutat Res ; 448(2): 193-200, 2000 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-10725472

RESUMO

Peroxisome proliferators (PPs) are a diverse group of nongenotoxic chemicals that in rodents cause hepatic peroxisome proliferation, liver enlargement, increased replicative DNA synthesis and suppression of apoptosis. The effects of PPs in vivo can be reproduced in vitro where PPs can induce mouse hepatocyte DNA synthesis and suppress both spontaneous apoptosis and that induced by transforming growth factor beta (TGFbeta). In vitro, high concentrations (>500 U/ml) of exogenous tumour necrosis factor (TNFalpha) [M. Rolfe, N.H. James, R.A. Roberts, TNF suppresses apoptosis and induces S-phase in rodent hepatocytes: a mediator of the hepatocarcinogenicity of peroxisome proliferators?, Carcinogenesis 18 (1997) 2277-2280] are also able to stimulate hepatocyte DNA synthesis and suppress apoptosis, implicating TNFalpha in mediating or permitting the liver growth response to PPs. Here, using cultured mouse hepatocytes isolated from PPARalpha null mice, we have examined the role of the peroxisome proliferator activated receptor alpha (PPARalpha) in mediating the suppression of apoptosis caused by PPs. In addition we have investigated further the role of TNFalpha in mediating the rodent response to PPs. The PP nafenopin (50 microM) was unable to stimulate DNA synthesis measured by bromodeoxyuridine incorporation in these PPARalpha null mouse hepatocytes (96% of control), unlike epidermal growth factor, a growth factor used as a positive control. In assays of apoptosis using H33258 staining of chromatin condensation, nafenopin was unable to suppress either spontaneous or TGFbeta1-induced apoptosis. In contrast, high concentrations of TNFalpha (>500 U/ml) were able to both stimulate DNA synthesis (204% of control) and suppress apoptosis in PPARalpha null hepatocytes (40% and 38% of control for spontaneous and TGFbeta1-induced apoptosis respectively). However, TNFalpha could not stimulate beta-oxidation of palmitoyl CoA in either PPARalpha null mouse or B6C3F1 (PPARalpha wild type) mouse hepatocytes. These data confirm the dependence of the response to PPs on PPARalpha by demonstrating that PPARalpha mediates the suppression of hepatocyte apoptosis in response to PPs. In addition, the data provide evidence that high concentrations of TNFalpha can modulate DNA synthesis and apoptosis in the absence of PPs and PPARalpha. Thus, in vivo, physiological levels of TNFalpha may be permissive for a PPARalpha-dependent growth response to PPs.


Assuntos
Apoptose/efeitos dos fármacos , Fígado/efeitos dos fármacos , Nafenopina/toxicidade , Proliferadores de Peroxissomos/toxicidade , Receptores Citoplasmáticos e Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , DNA/biossíntese , DNA/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Peroxissomos/efeitos dos fármacos , Fenobarbital/farmacologia , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/farmacologia
7.
Fundam Appl Toxicol ; 26(1): 143-9, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7657058

RESUMO

Peroxisome proliferators are members of the nongenotoxic family of rodent hepatocarcinogens. There exist substantial species differences in response to peroxisome proliferators among mammalian species. We have reported previously that peroxisome proliferators can synergize with epidermal growth factor (EGF) to promote the clonal expansion of rat hepatocytes associated with the early stages of hepatocarcinogenesis. The aim of the present study was to determine whether responsiveness in this in vitro assay reflected the known species differences in response to peroxisome proliferators. The process of tumorigenicity was modeled in the soft agar cloning assay since growth in soft agar is thought to reflect the early stages of tumorigenesis. This is because clonal expansion under these conditions requires the cells to survive, to undergo mitosis, and to escape from the contact-dependent growth associated with normal cell behavior. The data presented here show that mouse hepatocytes are able to undergo clonal expansion in soft agar in response to nafenopin and EGF giving a three- to fourfold increase in colony numbers over control. This result is comparable to the fivefold increase in rat hepatocyte colony numbers that we have reported previously. In contrast, hamster, guinea pig, and human hepatocytes did not respond to the concerted action of EGF and nafenopin despite their ability to respond to EGF as a mitogen in monolayer culture. These data demonstrate that the clonal expansion of rodent hepatocytes in soft agar in response to peroxisome proliferators and EGF displays the same species differences as other pleiotropic responses to these compounds and is likely therefore to be relevant to the process of hepatocarcinogenesis.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Fígado/citologia , Nafenopina/farmacologia , Animais , Células Clonais/efeitos dos fármacos , Cricetinae , DNA/biossíntese , Sinergismo Farmacológico , Cobaias , Humanos , Masculino , Camundongos , Mitógenos/farmacologia , Especificidade da Espécie
8.
Carcinogenesis ; 15(12): 2687-94, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8001222

RESUMO

The mechanisms by which the peroxisome proliferator (PP) class of non-genotoxic carcinogens perturb growth regulation and cause rodent liver cancer are unknown. Using a soft agar cloning assay, we have demonstrated that PPs synergize with the physiological liver mitogen epidermal growth factor (EGF) to cause the clonal expansion of rat hepatocytes associated with the early stages of tumourigenesis. In the presence of EGF (25 ng/ml), the PP nafenopin (100 microM) was able to stimulate a 5-fold increase in the number of colonies (35 colonies/50,000 hepatocytes compared to seven in the control). EGF alone or nafenopin alone gave 11 and 14 colonies respectively. TGF alpha, which acts through the EGF receptor, also synergized with nafenopin, whereas HGF was inactive, despite its potency as an hepatocyte growth factor. The ability to promote colony formation was shared by the potent PP Wyeth-14,643 but not by the less potent compounds methylclofenapate or trichloroacetic acid. TGF beta, a physiological negative regulator of liver growth, was able to inhibit the nafenopin/EGF-stimulated colony formation at 0.25 ng/ml, a concentration below that required for TGF beta-induced hepatocyte apoptosis. The colonies formed are derived from and consist of hepatocytes, since they express the hepatocyte-specific marker albumin, although the majority are negative for the PP-induced cytochrome, P4504A1. Pre-treatment in vivo with the genotoxic carcinogen dimethylhydrazine hydrochloride (150 mg/kg) caused a doubling in the number of colonies from 35 to 75/50,000 hepatocytes. Taken together, these data suggest that some PPs act as hepatocarcinogens by synergizing with EGF and/or TGF alpha to promote the clonal expansion of spontaneously initiated hepatocytes. This clonal expansion may be inhibited by TGF beta. Such a synergy may provide a mechanistic basis for the hepatocarcinogenicity of this class of non-genotoxic carcinogens.


Assuntos
Carcinógenos/farmacologia , Cocarcinogênese , Fator de Crescimento Epidérmico/farmacologia , Neoplasias Hepáticas Experimentais/induzido quimicamente , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Nafenopina/farmacologia , Albuminas/biossíntese , Animais , Biomarcadores , Carcinógenos/classificação , Divisão Celular/efeitos dos fármacos , Transformação Celular Neoplásica/induzido quimicamente , Células Cultivadas , Células Clonais/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Citocromo P-450 CYP4A , Sistema Enzimático do Citocromo P-450/biossíntese , Replicação do DNA/efeitos dos fármacos , Sinergismo Farmacológico , Fator de Crescimento Epidérmico/toxicidade , Hiperplasia , Fígado/patologia , Neoplasias Hepáticas Experimentais/patologia , Masculino , Oxigenases de Função Mista/biossíntese , Nafenopina/toxicidade , Ratos , Ratos Wistar , Fator de Crescimento Transformador alfa/farmacologia
9.
Carcinogenesis ; 17(8): 1623-32, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8761418

RESUMO

Tumorigenesis caused by the peroxisome proliferator (PP) class of non-genotoxic hepatocarcinogens is species restricted; rat and mouse are considered responsive whereas the available evidence suggests that humans, non-human primates, dogs, hamsters and guinea pigs are non-responsive. We have demonstrated previously that the PP, nafenopin can suppress rat hepatocyte apoptosis both in vitro and in vivo. Here we describe the ability of nafenopin to suppress apoptosis in mouse, hamster, guinea pig and rat hepatocytes and induce S-phase in mouse and rat hepatocytes. Hepatocyte monolayers from all species examined degenerated rapidly in culture. However, nafenopin (50 microM) reversibly maintained the viability of both rat and mouse hepatocytes. This maintenance was associated with a decrease (P < or = 0.01) in the number of hepatocytes displaying chromatin condensation patterns characteristic of apoptosis. Treatment of rat and mouse monolayers with 5 ng/ml transforming growth factor-beta 1 (TGF beta 1) induced high levels of apoptosis (P < or = 0.01); co-addition of nafenopin suppressed this induced apoptosis (P < or = 0.01). TGF beta 1 also induced apoptosis in hamster and guinea pig hepatocytes (P < or = 0.01) and unexpectedly nafenopin was able to suppress this induced apoptosis (P < or = 0.01) as well as reversibly maintaining the viability of hamster and guinea pig hepatocyte monolayers. Thus, all the species examined responded to nafenopin by a suppression of both spontaneous and TGF beta 1-induced apoptosis. In contrast, only rat and mouse hepatocytes showed an induction of S-phase in response to nafenopin (P < or = 0.01). Certain key experiments were repeated using the PPs methyl clofenapate (MCP) (100 microM) and Wy-14, 643 (10 microM). Both were able to suppress spontaneous and TGF beta 1-induced apoptosis in rat and guinea pig hepatocytes although the effects of MCP were weak (P < or = 0.05) compared with nafenopin or Wy-14 643 (P < or = 0.01). The rat and mouse liver tumour promoter, phenobarbitone (PB) was assessed also. Rat hepatocytes responded to PB with a suppression of apoptosis and an induction of S-phase (P < or = 0.01). Hamster and guinea pig cells gave no response in the S-phase assay and exhibited no suppression of either spontaneous or TGF beta 1-induced apoptosis. Interestingly, nafenopin suppressed the apoptosis induced by the DNA damaging drugs, etoposide and hydroxyurea (P < or = 0.01) suggesting that PPs can impact on diverse apoptosis signalling pathways. Overall, species differences in response to the non-genotoxic hepatocarcinogens studied, correlate with induction of DNA synthesis rather than with suppression of apoptosis. The data extend our knowledge of the mechanisms of species differences in non-genotoxic hepatocarcinogenesis, posing interesting questions on the relative roles of apoptosis and DNA synthesis in carcinogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Animais , Sangue , Clofenapato/farmacologia , Cricetinae , Meios de Cultura , Dano ao DNA , Cães , Cobaias , Homeostase , Masculino , Camundongos , Nafenopina/farmacologia , Fenobarbital/farmacologia , Pirimidinas/farmacologia , Ratos , Especificidade da Espécie , Fatores de Crescimento Transformadores/antagonistas & inibidores , Fatores de Crescimento Transformadores/farmacologia
10.
Arch Toxicol ; 72(12): 784-90, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9950075

RESUMO

Nongenotoxic rodent hepatocarcinogens do not damage DNA but cause liver tumours in the rat and mouse, associated with the induction of hepatic DNA synthesis. Previously, we have demonstrated that nongenotoxic hepatocarcinogens such as phenobarbitone and the peroxisome proliferator (PP), nafenopin, also suppress rat hepatocyte apoptosis. The nongenotoxic chemicals 1,4-dichlorobenzene (DCB) and the PP, diethylhexyl phthalate (DEHP), both induce high levels of DNA synthesis in rat liver in vivo, but only DEHP is hepatocarcinogenic in this species. Here, we investigate whether the difference in rat carcinogenicity of these two hepatic mitogens may be due to differences in their ability to suppress hepatocyte apoptosis. In rat hepatocytes in vitro, MEHP (the active metabolite of DEHP) induced DNA synthesis 2.5-fold (P = 0.001) and suppressed 10- and 4-fold, respectively both spontaneous (P = 0.0008) and transforming growth factor beta1 (TGFbeta1)-induced (P = 0.0001) apoptosis. DCB gave a small (1.7-fold) increase in DNA synthesis (P = 0.03) and a small (1.7- to 2-fold) suppression of both spontaneous (P = 0.022) and TGFbeta1-induced (P = 0.015) apoptosis. We next analysed the induction of DNA synthesis and the suppression of apoptosis in rat liver in vivo. Both DEHP and DCB were able to induce DNA synthesis although, as seen in vitro, the induction by DCB (4.2-fold; P = 0.023) was less marked than that with DEHP (13.4-fold; P = 0.007). Similarly, DEHP and DCB were both able to suppress rat hepatocyte apoptosis in vivo but the magnitude of the suppression was comparable; apoptosis was reduced to undetectable levels in four out of five animals with DCB and three out of five with DEHP. Since both chemicals suppressed apoptosis and induced DNA synthesis in rat liver but, overall, DCB was less potent, the disparate hepatocarcinogenic potential of these two chemicals could arise from differences in the magnitude of growth perturbation. To test this hypothesis, we repeated the studies in mouse, a species where both DCB and DEHP are hepatocarcinogenic. Both in vitro and in vivo, DCB and DEHP/MEHP were able to suppress apoptosis and induce hepatocyte DNA synthesis in the mouse with comparable potencies. The data support the hypothesis that the carcinogenicity of nongenotoxic hepatocarcinogens is associated strongly with the ability to perturb hepatocyte growth regulation. However, the ability to effect such changes is not unique to nongenotoxic carcinogens and is common to some noncarcinogenic chemicals, such as DCB, suggesting that the growth perturbation may need to exceed a threshold for carcinogenesis.


Assuntos
Carcinógenos/toxicidade , Clorobenzenos/toxicidade , DNA/biossíntese , Dietilexilftalato/toxicidade , Fígado/efeitos dos fármacos , Animais , Disponibilidade Biológica , Depressão Química , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Ratos , Ratos Endogâmicos F344 , Especificidade da Espécie
11.
Carcinogenesis ; 18(11): 2277-80, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9395232

RESUMO

Peroxisome proliferators (PPs) are a class of non-genotoxic rodent hepatocarcinogens that cause increased hepatocyte DNA synthesis, peroxisome proliferation and liver enlargement. We have demonstrated previously that PPs suppress both spontaneous rat hepatocyte apoptosis and that induced by the physiological negative regulator of liver growth, transforming growth factor beta (TGF beta1). Evidence suggests that the suppression of apoptosis by PPs is mediated via activation of the peroxisome proliferator activated receptor-alpha (PPAR alpha), a member of the nuclear hormone receptor superfamily. Here, we investigate the effects of tumour necrosis factor alpha (TNF alpha) on cultured rat or mouse hepatocytes to determine whether TNF alpha influences hepatocyte growth in a manner analogous to that seen with PPs. Rat recombinant TNF alpha was found to stimulate DNA synthesis and suppress apoptosis in isolated rat hepatocyte monolayers (P < or = 0.01). These effects were seen in the range of 500-5000 U/ml with a maximum effect at 5000 U/ml. Similarly, mouse recombinant TNF alpha was able to stimulate DNA synthesis in mouse hepatocyte monolayers (P < or = 0.01) with a maximal effect at 1000 U/ml. Suppression of mouse hepatocyte apoptosis by TNF alpha was not detected, possibly because of the low levels of apoptosis under control conditions. However, when the levels of mouse hepatocyte apoptosis were augmented using TGF beta1, TNF alpha caused a significant suppression (P < or = 0.01). The neutralization of TNF alpha using anti-TNF alpha antibodies abrogated significantly (P < or = 0.01) the suppression of apoptosis by the PP, nafenopin. These data that suggest TNF alpha may mediate, at least in part, the growth perturbation, liver enlargement and hepatocarcinogenesis seen in response to the PP class of non-genotoxic hepatocarcinogens.


Assuntos
Apoptose/efeitos dos fármacos , DNA/biossíntese , Neoplasias Hepáticas Experimentais/etiologia , Fígado/efeitos dos fármacos , Microcorpos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Fígado/patologia , Camundongos , Ratos , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Proteínas Recombinantes/farmacologia , Fatores de Transcrição/fisiologia
12.
Hepatology ; 31(2): 420-7, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10655266

RESUMO

Perturbation of hepatocyte growth regulation is associated with a number of liver diseases such as fibrosis and cancer. These diseases are mediated by a network of growth factors and cytokines that regulate the induction of hepatocyte proliferation and apoptosis. In this study, we have investigated the role of signaling pathways activated by tumor necrosis factor alpha (TNF-alpha) and epidermal growth factor (EGF) in the regulation of apoptosis induced by transforming growth factor beta(1) (TGF-beta(1)), because this physiological factor is believed to regulate spontaneous apoptosis in the liver. We show that pretreatment with (10 ng/mL) EGF or (25 ng/mL) TNF-alpha can suppress TGF-beta(1)-induced apoptosis by 73% and 50%, respectively, in isolated rat hepatocytes. However, suppression of TGF-beta(1)-induced apoptosis by EGF and TNF-alpha occurs via different protein kinase signaling pathways. Using specific inhibitors, we show that suppression of apoptosis by EGF is dependent on activation of phosphoinositide 3-kinase (PI 3-kinase) and the extracellular signal regulated kinase (ERK) mitogen-activated protein (MAP) kinase pathways, but not p38 MAP kinase. In contrast, suppression of TGF-beta(1)-induced apoptosis by TNF-alpha does not require PI 3-kinase and protein kinase B (PKB or Akt)-mediated pathways, but is dependent on ERK and p38 MAP kinase activity. These data contribute to our understanding of the intracellular survival signals that play a role in normal liver homeostasis and in diverse pathological conditions.


Assuntos
Apoptose/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Fígado/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática , Fígado/citologia , Fígado/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno
13.
Arch Toxicol ; 72(12): 777-83, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9950074

RESUMO

In rats and mice, nafenopin is a nongenotoxic hepatocarcinogen, which induces hepatic DNA synthesis and enzyme induction both in vivo and in hepatocyte cultures in vitro. However, humans and guinea-pigs are considered to be non-responsive to the liver growth effects of peroxisome proliferators (PPs). The ability to stimulate cell replication coupled with the ability to suppress apoptosis is thought to underpin the carcinogenicity of nongenotoxic carcinogens such as PPs. Previous studies in this laboratory have shown that in rats in vivo and in vitro nafenopin suppressed spontaneous hepatocyte apoptosis and that induced by the physiological negative growth regulator transforming growth factors beta1 (TGFbeta1). In addition nafenopin suppressed apoptosis in cultured hepatocytes from guinea-pig and hamster. The effects of PPs on apoptosis in human hepatocyte cultures is not known. To correlate these previous in vitro findings to the known species differences in hepatocarcinogenicity of PPs we have investigated the effects of nafenopin on guinea-pig liver growth in vivo. Also, we have examined the effects of nafenopin on apoptosis in cultures of human hepatocytes, a valuable model for human risk assessment. Nafenopin did not inhibit either spontaneous or TGFbeta1 induced apoptosis in human hepatocytes in vitro. Administration of nafenopin to guinea-pigs in vivo produced none of the changes seen previously in responsive species, such as rats and mice. There was no change in liver/body weight ratio, peroxisomal volume of hepatocytes or DNA synthesis as determined by incorporation of bromodeoxyuridine and there was no suppression of apoptosis. The lack of response to nafenopin in guinea-pigs in vivo and human hepatocytes in vitro provides further evidence that these species may be refractory to the liver growth effects of PPs despite the ability of guinea-pigs and humans to respond to PPs by alterations in lipid metabolism. The data presented add to our overall understanding of species differences in response to the PP class of rodent nongenotoxic carcinogens.


Assuntos
Apoptose/efeitos dos fármacos , Fígado/efeitos dos fármacos , Nafenopina/farmacologia , Proliferadores de Peroxissomos/farmacologia , Animais , Células Cultivadas , DNA/biossíntese , Depressão Química , Avaliação Pré-Clínica de Medicamentos , Cobaias , Humanos , Fígado/patologia , Masculino , Medição de Risco , Especificidade da Espécie
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
15.
Carcinogenesis ; 19(2): 299-304, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9498280

RESUMO

The suppression of apoptosis may contribute to the carcinogenicity of the peroxisome proliferators (PPs), a class of non-genotoxic rodent hepatocarcinogens. Our previous work demonstrated that the PP nafenopin suppressed both spontaneous and transforming growth factor beta1 (TGFbeta1)-induced hepatocyte apoptosis both in vivo and in vitro. Here, we extend these observations by demonstrating the ability of nafenopin to suppress apoptosis induced by other major candidates for the signalling of cell death in the liver. Treatment of rat or mouse hepatocyte monolayers with TGFbeta1 or the DNA damaging drugs etoposide or hydroxyurea induced high levels of apoptosis. Western blot analysis did not support a role for either p53 or p21waf1 in etoposide-induced apoptosis in rat hepatocytes. Treatment of mouse hepatocytes with an agonistic anti-Fas antibody also resulted in an induction of high levels of apoptosis. Pre-addition and continued exposure to nafenopin suppressed apoptosis induced by all three stimuli. Overall, our studies demonstrate that the ability of nafenopin to protect hepatocytes from apoptosis is not restricted to species or apoptotic stimulus. It is possible, therefore, that the PPs may suppress apoptosis by acting on diverse signalling pathways. However, it seems more likely that nafenopin suppresses hepatocyte apoptosis elicited by each death stimulus by impinging on a core apoptotic mechanism.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos/farmacologia , Dano ao DNA/efeitos dos fármacos , Ácido Graxo Sintases/fisiologia , Nafenopina/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Receptor fas/fisiologia , Animais , Anticorpos/farmacologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Relação Dose-Resposta Imunológica , Ácido Graxo Sintases/imunologia , Camundongos , Ratos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Receptor fas/imunologia
16.
Arch Toxicol ; 74(2): 85-91, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10839475

RESUMO

Diethylhexylphthalate (DEHP) is a phthalate plasticizer that belongs to the peroxisome proliferator (PP) class of rodent nongenotoxic hepatocarcinogens. Previously, we have shown that MEHP (a principal metabolite of DEHP and the proximal PP) induced DNA synthesis and suppressed apoptosis in rat but not in human hepatocytes in vitro. Here, we present further studies of species differences in response to DEHP. In rats, 4 days of exposure to DEHP (950 mg/kg per day by gavage) induced peroxisomal beta-oxidation, DNA synthesis and suppressed apoptosis. In contrast, there was no response of guinea pig liver to DEHP. In rat hepatocytes in vitro, MEHP (250, 500 and 750 microM) induced peroxisomal beta-oxidation, DNA synthesis and suppressed apoptosis. In contrast to the pleiotropic response noted in rat hepatocytes, there was no response of human hepatocytes to 250, 500 or 750 microM MEHP. PPs activate the peroxisome proliferator activated receptor alpha (PPARalpha) that binds to DNA at peroxisome proliferator response elements (PPREs) within the promoters of PP-responsive genes such as rat acyl CoA oxidase (ACO). However, the human ACO gene promoter differs at three bases within the PPRE from the rat ACO promoter and appears refractory to PPs. To address species differences in response to DEHP at the molecular level, we used promoter-reporter gene assays to compare the ability of MEHP to induce gene expression from the rat or the human ACO promoter. MEHP gave a concentration-dependent increase in reporter gene expression from the rat ACO gene promoter with either mouse or human PPARalpha. In contrast, the human ACO promoter was unable to drive MEHP-induced gene transcription irrespective of the species origin of PPARalpha. These data provide further weight of evidence at the cellular and molecular levels for a lack of risk to human health from the phthalate DEHP.


Assuntos
Apoptose/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA/biossíntese , Dietilexilftalato/farmacologia , Microcorpos/genética , Proliferadores de Peroxissomos/farmacologia , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Células 3T3 , Animais , DNA/efeitos dos fármacos , Dietilexilftalato/análogos & derivados , Fator de Crescimento Epidérmico/farmacologia , Expressão Gênica/efeitos dos fármacos , Genes Reporter , Cobaias , Humanos , Marcação In Situ das Extremidades Cortadas , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Microcorpos/efeitos dos fármacos , Microcorpos/enzimologia , Nafenopina/farmacologia , Oxirredução , Ratos , Ratos Endogâmicos F344 , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Especificidade da Espécie , Fatores de Transcrição/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , beta-Galactosidase/genética
17.
Exp Cell Res ; 203(2): 407-19, 1992 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1281111

RESUMO

An in vitro model of liver in which rat hepatocytes are maintained as cocultures with nonparenchymal epithelial cells (NPC) derived from liver has been developed and characterized with respect to maintenance of hepatocyte viability and differentiated function. The system was then evaluated as a model for studying peroxisome proliferator-induced rodent liver nongenotoxic carcinogenesis. Within the coculture model, hepatocyte viability and morphology were maintained for 1 month or more within a system that is both easily accessible for microscopic examination and is free of any additives that may lead to artifacts. Even after 1 month or more, hepatocyte cocultures retained expression of the constitutive liver marker albumin. In addition, they maintained the ability to show induction of the peroxisome proliferator-inducible enzymes peroxisomal bifunctional enzyme (PBE) and cytochrome P450IVA1 in response to the peroxisome proliferator nafenopin. After 4 weeks, NPC cocultures showed a six- and a fourfold induction of PBE and cytochrome P450IVA1 expression, respectively, which compared well with the three- and fivefold induction seen in freshly isolated cells. This was paralleled by an increase in the cytoplasmic volume fraction of peroxisomes averaging eightfold. Interestingly, great heterogeneity was exhibited between adjacent hepatocytes in terms of the degree of peroxisome proliferation, a finding reflected by immunocytochemical staining which indicated heterogeneity in the level of expression of the peroxisome proliferator-inducible enzymes. Other cell lines representing different tissue types, morphologies, and species were also examined for their ability to support hepatocyte survival but were found to be ineffective, with the exception of a bovine corneal endothelial cell line. This line supported hepatocyte survival and maintenance of differentiated function but to a lesser extent than that observed with NPC. Ultrastructural examination of NPC cocultures revealed extensive interhepatocyte junctional complexes and interdigitation of adjacent membranes together with the presence of bile canalicular structures. There were no junctional complexes between the hepatocytes and the supporting feeder cells with any contact being limited to a close association of the hepatocytes with the extracellular matrix presumably produced by the NPC. The data demonstrate that hepatocytes maintained in vitro within an NPC coculture system retain differentiated function and the ability to respond to the peroxisome proliferator class of nongenotoxic carcinogens. Cocultures will provide us with a model system for the study of changes in hepatocyte growth regulation during rodent liver nongenotoxic carcinogenesis.


Assuntos
Neoplasias Hepáticas/induzido quimicamente , Fígado/citologia , Nafenopina/farmacologia , 3-Hidroxiacil-CoA Desidrogenases/biossíntese , Albuminas/biossíntese , Animais , Bovinos , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Citocromo P-450 CYP4A , Sistema Enzimático do Citocromo P-450/biossíntese , Endotélio Corneano/citologia , Enoil-CoA Hidratase/biossíntese , Indução Enzimática , Isomerases/biossíntese , Queratinas/análise , Fígado/efeitos dos fármacos , Fígado/ultraestrutura , Microcorpos/efeitos dos fármacos , Microcorpos/ultraestrutura , Microscopia Eletrônica , Oxigenases de Função Mista/biossíntese , Complexos Multienzimáticos/biossíntese , Enzima Bifuncional do Peroxissomo , Ratos , Vimentina/análise
18.
Toxicol Appl Pharmacol ; 135(2): 192-9, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8545827

RESUMO

It has been proposed that several nongenotoxic compounds act as hepatocarcinogens by suppressing the apoptosis that would normally act to remove damaged or potentially initiated cells from the liver. During our investigations of this hypothesis using a widely applied protocol, we have found that the stress induced by the process of gavage dosing can induce massive apoptosis in livers uniquely primed by withdrawal of the hepatomitogen cyproterone acetate from the hyperplastic rat liver. This effect of gavage dosing was not seen in livers of naive animals. Apoptosis was measured by both in situ end labeling (ISEL) of the DNA damage associated with programmed cell death and conventional hematoxylin and eosin (H&E) staining of apoptotic morphology. Apoptotic rates measured by H&E increased significantly from 0.005 +/- 0.010% on Day 11 to 0.657 +/- 0.315% of hepatocytes on Day 15, 4 days after cessation of 10 days dosing with CPA (120 mg/kg). The readministration of CPA suppressed > 89% of this Day 15 apoptosis. However, the readministration of vehicle alone (corn oil) caused a 390% increase in apoptosis to 2.56 +/- 1.31% of hepatocytes. Similar results were obtained using ISEL. Measurements of liver to body weight ratios and total DNA per liver reflected these changes in cell loss by apoptosis. In a second experiment, CPA was administered for 10 days as before then animals were subjected to readministration of CPA in corn oil, CPA in saline, corn oil, saline, or sham dosed. Again, apoptosis was dramatically suppressed by the readministration of CPA in either vehicle but was dramatically increased to around 2% of hepatocytes in all other groups, including the sham dosed group. Data on food consumption provided no evidence for a reduction in food intake as a causative agent but rather pointed to a less efficient usage of food in the stressed animals. The ability of stress to induce liver apoptosis should be borne in mind in the design and interpretation of future toxicological studies aimed at understanding the putative suppression of apoptosis by liver nongenotoxic carcinogens and other toxicants.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos/toxicidade , Acetato de Ciproterona/toxicidade , Intubação Gastrointestinal/efeitos adversos , Fígado/citologia , Estresse Fisiológico/patologia , Animais , Carcinógenos/administração & dosagem , Acetato de Ciproterona/administração & dosagem , DNA/análise , Dano ao DNA , Ingestão de Alimentos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar , Projetos de Pesquisa
19.
Toxicol Pathol ; 23(3): 410-5, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7659962

RESUMO

Nongenotoxic hepatocarcinogens have been proposed to inhibit hepatocyte apoptosis, preventing the removal of DNA-damaged cells. Conclusive proof of this hypothesis has been hindered by the lack of a marker or stain for apoptosis suitable for high throughput counting. A method is described for the detection of apoptotic bodies (AB) in paraffin sections of rat liver using an in situ end labeling (ISEL) technique that detects DNA damage. Results of this AB quantitation were compared with routine hematoxylin-eosin (H&E)-stained sections. The number of apoptotic bodies was enhanced by the withdrawal of the rodent nongenotoxic hepatocarcinogens and hepatomitogens nafenopin (naf) or cyproterone acetate (CPA) after 7 days of daily dosing. In rat livers 24-96 hr after cessation of dosing with naf, and in control livers, an AB index of approximately 0.1% of hepatocyte nuclei was seen when stained by H&E or ISEL. However, livers examined 48 hr after cessation of 7 days of daily dosing with CPA had an AB index of approximately 1% hepatocyte nuclei when stained with H&E, but only approximately 0.4% hepatocyte nuclei using the ISEL technique. Thus, CPA withdrawal from the hyperplastic liver generated a wave of apoptosis in contrast to naf withdrawal where little was seen up to 96 hr after withdrawal. The differing kinetics of CPA and naf clearance may explain this discrepancy. Less apoptosis was detected by the ISEL method following CPA withdrawal; this could arise if the stage of apoptosis labeled by ISEL is shorter than the morphologically recognizable stages (using H&E). The ISEL method for the evaluation of AB indices is useful in parallel with H&E, although more validation is required before it can be used routinely for the quantitation of AB in tissue sections.


Assuntos
Apoptose/genética , Dano ao DNA/genética , Fígado/patologia , Animais , DNA Polimerase I , DNA de Cadeia Simples , Feminino , Masculino , Ratos , Ratos Endogâmicos , Transcrição Gênica/genética
20.
Carcinogenesis ; 16(8): 1693-8, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7634391

RESUMO

Non-genotoxic hepatocarcinogenesis may involve suppression of the hepatocyte apoptosis that would normally remove damaged or initiated cells. These protected hepatocytes could then remain as preferential targets for promotion by this class of compounds. Here we demonstrate clearly that the non-genotoxic liver carcinogens and hepatomitogens cyproterone acetate (CPA) and nafenopin, a peroxisome proliferator, both suppress the basal level of rat liver apoptosis in vivo. After 10 days of dosing with CPA (120 mg/kg/day) or nafenopin (25 mg/kg/day) there were 0.005 +/- 0.010 and 0.002 +/- 0.021 apoptotic bodies/100 hepatocytes respectively, compared with 0.031 +/- 0.008 per 100 in controls. Concomitant with this suppression of apoptosis, bromodeoxyuridine (BrdU) labelling indices and mitotic figures rose, confirming a perturbation of both sides of the growth equation between cell death and replication. Withdrawal of CPA or nafenopin resulted in a 100- to 200-fold elevation in apoptosis. This was inhibited by the re-administration of either compound. To investigate if cells protected from apoptosis by non-genotoxic carcinogens are targets for replication, we examined the replicative history of the apoptotic bodies generated upon withdrawal of CPA or nafenopin. Rats were administered BrdU during the hyperplastic phase of compound administration (0-10 days). Livers were examined 5 days after compound withdrawal. With both CPA and nafenopin, apoptotic bodies and S phase were predominantly in the periportal region. However, despite this zonal co-localization, very few (< 10%) of the apoptotic bodies were labelled with BrdU. Overall, our data provide in vivo evidence to support the hypothesis that non-genotoxic hepatocarcinogens such as CPA and the peroxisome proliferators suppress apoptosis. Surprisingly, the majority of the hepatocytes generated during compound-induced hyperplasia were protected from apoptosis during liver regression. These data contribute to our understanding of clonal selection and promotion during non-genotoxic hepatocarcinogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Acetato de Ciproterona/toxicidade , DNA/biossíntese , Neoplasias Hepáticas Experimentais/induzido quimicamente , Fígado/efeitos dos fármacos , Nafenopina/toxicidade , Animais , Masculino , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA