Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Br J Cancer ; 109(10): 2654-64, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24136149

RESUMEN

BACKGROUND: The DNA-repair gene DNA-dependent kinase catalytic subunit (DNA-PKcs) favours or inhibits carcinogenesis, depending on the cancer type. Its role in human hepatocellular carcinoma (HCC) is unknown. METHODS: DNA-dependent protein kinase catalytic subunit, H2A histone family member X (H2AFX) and heat shock transcription factor-1 (HSF1) levels were assessed by immunohistochemistry and/or immunoblotting and qRT-PCR in a collection of human HCC. Rates of proliferation, apoptosis, microvessel density and genomic instability were also determined. Heat shock factor-1 cDNA or DNA-PKcs-specific siRNA were used to explore the role of both genes in HCC. Activator protein 1 (AP-1) binding to DNA-PKcs promoter was evaluated by chromatin immunoprecipitation. Kaplan-Meier curves and multivariate Cox model were used to study the impact on clinical outcome. RESULTS: Total and phosphorylated DNA-PKcs and H2AFX were upregulated in HCC. Activated DNA-PKcs positively correlated with HCC proliferation, genomic instability and microvessel density, and negatively with apoptosis and patient's survival. Proliferation decline and massive apoptosis followed DNA-PKcs silencing in HCC cell lines. Total and phosphorylated HSF1 protein, mRNA and activity were upregulated in HCC. Mechanistically, we demonstrated that HSF1 induces DNA-PKcs upregulation through the activation of the MAPK/JNK/AP-1 axis. CONCLUSION: DNA-dependent protein kinase catalytic subunit transduces HSF1 effects in HCC cells, and might represent a novel target and prognostic factor in human HCC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Proteína Quinasa Activada por ADN/genética , Neoplasias Hepáticas/patología , Proteínas Nucleares/genética , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Proteínas de Unión al ADN/fisiología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción del Choque Térmico , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Valor Predictivo de las Pruebas , Pronóstico , Factores de Transcripción/fisiología
2.
Gut ; 58(5): 679-87, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19136513

RESUMEN

BACKGROUND AND AIMS: Previous studies indicate unrestrained cell cycle progression in liver lesions from hepatocarcinogenesis-susceptible Fisher 344 (F344) rats and a block of G(1)-S transition in corresponding lesions from resistant Brown Norway (BN) rats. Here, the role of the Forkhead box M1B (FOXM1) gene during hepatocarcinogenesis in both rat models and human hepatocellular carcinoma (HCC) was assessed. METHODS AND RESULTS: Levels of FOXM1 and its targets were determined by immunoprecipitation and real-time PCR analyses in rat and human samples. FOXM1 function was investigated by either FOXM1 silencing or overexpression in human HCC cell lines. Activation of FOXM1 and its targets (Aurora Kinose A, Cdc2, cyclin B1, Nek2) occurred earlier and was most pronounced in liver lesions from F344 than BN rats, leading to the highest number of Cdc2-cyclin B1 complexes (implying the highest G(2)-M transition) in F344 rats. In human HCC, the level of FOXM1 progressively increased from surrounding non-tumorous livers to HCC, reaching the highest levels in tumours with poorer prognosis (as defined by patients' length of survival). Furthermore, expression levels of FOXM1 directly correlated with the proliferation index, genomic instability rate and microvessel density, and inversely with apoptosis. FOXM1 upregulation was due to extracellular signal-regulated kinase (ERK) and glioblastoma-associated oncogene 1 (GLI1) combined activity, and its overexpression resulted in increased proliferation and angiogenesis and reduced apoptosis in human HCC cell lines. Conversely, FOXM1 suppression led to decreased ERK activity, reduced proliferation and angiogenesis, and massive apoptosis of human HCC cell lines. CONCLUSIONS: FOXM1 upregulation is associated with the acquisition of a susceptible phenotype in rats and influences human HCC development and prognosis.


Asunto(s)
Carcinoma Hepatocelular/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Factores de Transcripción Forkhead/genética , Neoplasias Hepáticas/genética , Animales , Carcinoma Hepatocelular/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/fisiología , Regulación Neoplásica de la Expresión Génica/genética , Genes cdc , Predisposición Genética a la Enfermedad/genética , Hígado/patología , Neoplasias Hepáticas/patología , Neovascularización Patológica/etiología , Ratas , Ratas Endogámicas F344 , Activación Transcripcional , Regulación hacia Arriba
3.
Biochim Biophys Acta ; 1765(2): 126-47, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16216419

RESUMEN

The different frequency of hepatocellular carcinoma (HCC) in humans at risk suggests a polygenic predisposition. However, detection of genetic variants is difficult in genetically heterogeneous human population. Studies on mouse and rat models identified 7 hepatocarcinogenesis susceptibility (Hcs) and 2 resistance (Hcr) loci in mice, and 7 Hcs and 9 Hcr loci in rats, controlling multiplicity and size of neoplastic liver lesions. Six liver neoplastic nodule remodeling (Lnnr) loci control number and volume of re-differentiating lesions in rat. A Hcs locus, with high phenotypic effects, and various epistatic gene-gene interactions were identified in rats, suggesting a genetic model of predisposition to hepatocarcinogenesis with different subset of low-penetrance genes, at play in different subsets of population, and a major locus. This model is in keeping with human HCC epidemiology. Several putative modifier genes in rodents, deregulated in HCC, are located in chromosomal segments syntenic to sites of chromosomal aberrations in humans, suggesting possible location of predisposing loci. Resistance to HCC is associated with lower genomic instability and downregulation of cell cycle key genes in preneoplastic and neoplastic lesions. p16(INK4A) upregulation occurs in susceptible and resistant rat lesions. p16(INK4A)-induced growth restraint was circumvented by Hsp90/Cdc37 chaperons and E2f4 nuclear export by Crm1 in susceptible, but not in resistant rats and human HCCs with better prognosis. Thus, protective mechanisms seem to be modulated by HCC modifiers, and differences in their efficiency influence the susceptibility to hepatocarcinogenesis and probably the prognosis of human HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Predisposición Genética a la Enfermedad , Neoplasias Hepáticas/genética , Animales , Transformación Celular Neoplásica/genética , Humanos
4.
Cancer Res ; 52(18): 4979-86, 1992 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-1516054

RESUMEN

Previous work has shown a consistent fall in S-adenosyl-L-methionine (SAM) in the liver of diethylnitrosamine-initiated rats, during the development of preneoplastic lesions, in persistent nodules (PNs), and hepatocellular carcinomas. The injection of SAM into rats causes the reconstitution of the SAM pool, coupled with growth restraint, remodeling, and apoptosis of preneoplastic cells, and inhibits the development of PNs and hepatocellular carcinomas. To evaluate if SAM treatment causes a long-term prevention of preneoplastic and neoplastic liver lesions or merely causes a delay in their development, we evaluated the effect of a relatively short SAM treatment on the development of preneoplastic and neoplastic lesions in a long-term study. Male Wistar rats were subjected to initiation with diethylnitrosamine, followed by selection and then by the administration of phenobarbital for 16 weeks. After selection, the rats were given i.m. injections of a purified SAM preparation (384 mumol/kg/day) for 24 weeks. In SAM-treated rats, a decrease in the incidence of PNs was found 6, 14, and 24-28 months after initiation. At the end of SAM treatment the number of PNs per rat liver, nodule diameter, and labeling and mitotic indices of nodular cells decreased considerably in control rats. Nodule diameter started to increase rapidly again only 8 months after arresting SAM treatment, when complete recovery of DNA synthesis in nodular cells occurred. The majority of nodules present in the liver 6-28 months after initiation belonged to the clear and acidophilic cell types, with lower percentages of mixed cell and basophilic cell types. A decrease in basophilic nodules occurred in SAM-treated rats. Fourteen and 24-28 months after initiation hepatocellular carcinoma incidence was 11 of 12 and 10 of 10 in control rats, respectively, and only 1 of 12 and 3 of 11 in SAM-treated rats. At the 24th-28th month all control rats had tumors identified as 2 poorly differentiated carcinomas, 6 trabecular carcinomas, or 3 adenocarcinomas, while only 2 relatively small trabecular carcinomas and 1 small glandular tumor developed in SAM-treated rats. In 3 of 11 SAM-treated rats, but in none of the control rats, leukemic infiltration of liver occurred 24-28 months after initiation. Leukemic infiltration of the spleen occurred in 5 and 3 control and SAM-treated rats, respectively.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Neoplasias Hepáticas Experimentales/prevención & control , Neoplasias Hepáticas/prevención & control , Lesiones Precancerosas/prevención & control , S-Adenosilmetionina/uso terapéutico , Animales , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/patología , Masculino , Índice Mitótico/efectos de los fármacos , Lesiones Precancerosas/patología , Ratas , Ratas Endogámicas , Factores de Tiempo
5.
Cancer Res ; 59(18): 4651-7, 1999 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10493520

RESUMEN

Neoplastic liver nodules and hepatocellular carcinomas (HCCs) were induced, by "resistant hepatocyte" model, 32 and 70 weeks after initiation with diethylnitrosamine, respectively, in F344 Brown Norway (BN), and (BNxF344)F1 rats. Nodule number/liver (N) did not significantly differ among rat strains, whereas nodule mean volume (V) and nodule volume fraction (VF) were higher in susceptible F344 than in resistant BN and BFF1 strains and were predictive of subsequent development of HCCs. Genomic scanning of 157 backcross BFF1xF344 rats with 190 polymorphic microsatellites, and linkage analysis, revealed two quantitative trait loci (QTL) on chromosomes 7 and 10, which showed significant linkage with VF, and two QTL on chromosomes 4 and 8, which showed suggestive linkage with V and VF. On the basis of phenotypic patterns of homozygous and heterozygous backcross progeny and of allelic distribution pattern, QTL on chromosomes 10, 8, and 4 were tentatively identified as resistance loci, and QTL on chromosome 7 was identified as susceptibility locus for rat hepatocarcinogenesis. An analysis of interactions allowed us to identify additional putative QTL on chromosomes 5 and 8 and suggested an additive effect of loci on chromosomes 10, 8, and 4 for VF and V. These data are the first to identify chromosomal regions containing putative susceptibility/resistance loci for rat hepatocarcinogenesis, which seems to be highly complex in terms of the number of genetic factors involved.


Asunto(s)
Mapeo Cromosómico , Neoplasias Hepáticas Experimentales/genética , Hígado/patología , Animales , Carcinógenos/toxicidad , Cruzamientos Genéticos , Dietilnitrosamina/toxicidad , Femenino , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Genotipo , Hígado/efectos de los fármacos , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/patología , Masculino , Fenotipo , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344
6.
Oncogenesis ; 5(12): e274, 2016 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-27918553

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is a rare yet deadly malignancy with limited treatment options. Activation of the Notch signalling cascade has been implicated in cholangiocarcinogenesis. However, while several studies focused on the Notch receptors required for ICC development, little is known about the upstream inducers responsible for their activation. Here, we show that the Jagged 1 (Jag1) ligand is almost ubiquitously upregulated in human ICC samples when compared with corresponding non-tumorous counterparts. Furthermore, we found that while overexpression of Jag1 alone does not lead to liver tumour development, overexpression of Jag1 synergizes with activated AKT signalling to promote liver carcinogenesis in AKT/Jag1 mice. Histologically, tumours consisted exclusively of ICC, with hepatocellular tumours not occurring in AKT/Jag1 mice. Furthermore, tumours from AKT/Jag1 mice exhibited extensive desmoplastic reaction, an important feature of human ICC. At the molecular level, we found that both AKT/mTOR and Notch cascades are activated in AKT/Jag1 ICC tissues, and that the Notch signalling is necessary for ICC development in AKT/Jag1 mice. In human ICC cell lines, silencing of Jag1 via specific small interfering RNA reduces proliferation and increases apoptosis. Finally, combined inhibition of AKT and Notch pathways is highly detrimental for the in vitro growth of ICC cell lines. In summary, our study demonstrates that Jag1 is an important upstream inducer of the Notch signalling in human and mouse ICC. Targeting Jag1 might represent a novel therapeutic strategy for the treatment of this deadly disease.

7.
Anticancer Res ; 13(5A): 1341-56, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8239505

RESUMEN

Carcinogenesis is a complex process characterized by the cumulative activation of various oncogenes and the inactivation of suppressor genes. Epigenetic mechanisms are also involved. Mutational activation of ras family genes occurs in most spontaneous or carcinogen-induced liver tumors, in susceptible mice, and less frequently in preneoplastic lesions. This suggests a pathogenetic role of these changes in hepatic carcinogenesis, in the mouse. Overexpression of various growth-related genes occurs in preneoplastic tissue during rat liver carcinogenesis, but mutational activation of protooncogenes, notably of ras family genes, seems to be a late and rare event, while c-myc amplification is a late but frequent event in both rodent and human carcinogenesis. However, mutation of the suppressor p53 gene has been found in relatively early preneoplastic lesions in rat liver, and it may be frequently seen in human hepatocellular carcinomas. The possibility that this mutation is involved in the initiation stage of liver carcinogenesis is an attractive hypothesis which needs further evaluation. DNA hypomethylation is involved in carcinogenesis, but the mechanisms underlying this effect are still elusive. Hypomethylation of growth-related genes is associated with their overexpression and this could favor overgrowth of preneoplastic liver tissue. Decrease in S-adenosyl methionine/S-adenosylhomocysteine (SAM/SAH) ratio occurs in the liver of rats fed a methyl deficient diet, which is a carcinogenic treatment, and in preneoplastic liver tissue, developing in initiated/promoted rats fed an adequate diet. The role of low SAM/SAH ratio in carcinogenesis is substantiated by the tumor chemopreventive effect of lipotropic compounds. Treatment with exogenous SAM prevents the development of preneoplastic and neoplastic lesions in rat liver. This is associated with recovery of SAM/SAH ratio, DNA methylation and inhibition of growth-related gene expression. SAM effect on prenoplastic cell growth is abolished by 5-azacytidine, a hypomethylating agent, indicating the involvement of DNA methylation. The possibility that in SAM-treated rats, methylation and inhibition of the expression of growth-related genes is implicated in growth restraint is attractive and should be further evaluated. Modulation of rat liver carcinogenesis by influencing gene expression through DNA methylation or other epigenetic mechanisms could be a new approach to chemoprevention of these tumors.


Asunto(s)
Adenoma/genética , Carcinoma Hepatocelular/genética , ADN de Neoplasias/metabolismo , Desoxiadenosinas/antagonistas & inhibidores , Genes ras/genética , Neoplasias Hepáticas/genética , Mutación Puntual/genética , Lesiones Precancerosas/genética , S-Adenosilmetionina/farmacología , Tionucleósidos/antagonistas & inhibidores , Adenoma/prevención & control , Animales , Carcinoma Hepatocelular/prevención & control , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes myc/genética , Genes p53/genética , Neoplasias Hepáticas/prevención & control , Metilación , Ratones , Ratones Endogámicos , Lesiones Precancerosas/prevención & control , Ratas
8.
Anticancer Res ; 13(6A): 1965-72, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8297102

RESUMEN

gamma-Glutamyltranspeptidase (GGT)-positive foci and glutathione-S-transferase, placental (GST-P)-positive lesions occupied 36% and 54% of liver parenchyma, respectively, in Wistar rats 8 weeks after initiation with diethylnitrosamine, followed by selection. The administration of S-adenosyl-L-methionine (SAM, 384 mumol/kg/day) caused 77% and 42% falls in the percentage of GGT-positive and GST-P-positive lesions, respectively. There also occurred a 46% decrease in labeling index of GGT-positive foci, in SAM-treated rats. These changes were associated with decrease in liver pyruvate kinase (PK), lactate dehydrogenase and glycerol-3-phosphate dehydrogenase. SAM did not affect these enzymatic activities in normal and uninitiated controls, but it caused a consistent increase in initiated rats. Enolase, fructose-biphosphatase and malic enzyme (ME) activities increased in the liver of initiated rats. SAM did not modify significantly these enzymatic activities, either in control or in initiated rats. Glucose-6-phosphate dehydrogenase (G6PDH) was 113% higher in the liver of initiated rats than in uninitiated controls. SAM treatment did not significantly affect this enzymatic activity in uninitiated rats, but caused a great decrease in initiated ones. As expected, there occurred a marked rise in GGT activity in the liver of initiated rats, with respect to controls. SAM caused an increase in GGT activity in normal and uninitiated controls, but it caused a 77% fall in GGT activity in initiated rats, coupled with a 380% rise in remodeling of GGT-positive lesions. Histochemical determination of G6PDH and ME activities showed that in the absence of SAM many preneoplastic lesions expressed higher G6PDH and ME activities than surrounding liver. SAM did not affect ME-positive lesions, while it caused a decrease in the number of G6PDH-positive lesions. Immunohistochemical determination of PK activity, isoenzyme L, showed a decrease in GST-P-positive lesions. Many of these lesions were no longer recognizable as lesions expressing a low PK activity, in SAM-treated rats. However, a relatively small number of GST-P-positive lesions expressing a low PK activity were still present in these rats. These data suggest that glucose channelled into triacylglycerol and pyruvate synthesis decreases in rat liver, during the development of preneoplastic foci, while the production of reducing equivalents and pentose phosphates increases, thus favoring DNA synthesis and detoxification reactions. Decrease in DNA synthesis, in SAM-treated rats, is paralleled by a partial reversion of carbohydrate metabolic features to those present in normal liver.


Asunto(s)
Neoplasias Hepáticas/inducido químicamente , Lesiones Precancerosas/patología , S-Adenosilmetionina/farmacología , Animales , Metabolismo de los Hidratos de Carbono , Dietilnitrosamina/toxicidad , Glutatión Transferasa/análisis , Inmunohistoquímica , Isoenzimas/análisis , L-Lactato Deshidrogenasa/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Masculino , Modelos Biológicos , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/enzimología , Piruvato Quinasa/metabolismo , Ratas , Ratas Wistar
9.
Anticancer Res ; 11(4): 1617-24, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1684097

RESUMEN

Male Wistar rats, initiated with diethylnitrosamine (DENA), were subjected to a selection treatment, according to the "resistant hepatocyte" model, followed or not followed by phenobarbital (PB). Rats received, for 3 weeks after selection, 4 i.m. doses (96 mmol/kg) of L-methionine, S-adenosyl-L-methionine (SAM), or 5'-methylthioadenosine (MTA), a SAM catabolite formed during polyamine synthesis or by spontaneous splitting of SAM at physiologic temperature and pH. They were then killed. In some rats, SAM and MTA treatments were started 20 weeks after initiation. The animals were killed 3 weeks later and persistent (neoplastic) nodules (PN) were collected. Some rat groups received 1/2 and 1/4 of the above SAM and MTA doses, or 1/8 of the above MTA dose. SAM and MTA, but not methionine, caused a dose-dependent decrease in number and surface area of gamma-glutamyltranspeptidase (GGT)-positive foci, and in labeling index (LI) of focal cells, coupled with remodeling. SAM and MTA liver contents, SAM/S-adenosylhomocysteine (SAH) ratio and overall methylation of liver DNA were low during the development of GGT-positive foci. SAM, but not methionine, caused a dose-dependent recovery of SAM content and DNA methylation, and a partial reconstitution of liver MTA pool. Exogenous MTA only induced the reconstitution of MTA pool, without affecting SAM level and DNA methylation. Recovery of SAM and MTA pool and DNA methylation was found in the rats subjected to SAM plus MTA, indicating the absence of inhibition of DNA methyltransferases in vivo by MTA. MTA also inhibited liver reparative growth in partially hepatectomized rats, without modifying SAM content and DNA methylation of regenerating liver (RL). A high activity of ornithine decarboxylase (ODC) was found in the liver, during the development of preneoplastic foci, and in PN. This activity was inhibited by SAM and MTA treatments. Although MTA was more effective than SAM, the decrease in ODC activity was coupled with a larger fall in DNA synthesis in SAM-treated than in MTA-treated rats. Thus the antipromotion effect of SAM could not merely depend on its (spontaneous) transformation into MTA. Although MTA production may play a role in the SAM antipromotion effect, other mechanisms could be involved. A role of DNA methylation in the inhibition of growth by SAM is suggested. MTA is a potential chemopreventive agent for liver carcinogenesis.


Asunto(s)
Adenosina/análogos & derivados , ADN/metabolismo , Desoxiadenosinas , Neoplasias Hepáticas Experimentales/patología , Hígado/patología , Metionina/farmacología , Lesiones Precancerosas/patología , S-Adenosilmetionina/farmacología , Tionucleósidos/farmacología , Adenosina/farmacología , Animales , Biomarcadores de Tumor/análisis , Carcinógenos , División Celular/efectos de los fármacos , ADN/efectos de los fármacos , Hígado/efectos de los fármacos , Neoplasias Hepáticas Experimentales/inducido químicamente , Regeneración Hepática , Masculino , Lesiones Precancerosas/inducido químicamente , Ratas , Ratas Endogámicas , Análisis de Regresión , gamma-Glutamiltransferasa/análisis
10.
Carcinogenesis ; 14(6): 1095-9, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8508494

RESUMEN

The study was designed to understand the effect of orotic acid (OA) on the expression of beta 1,4-galactosyltransferase (GalTase), an enzyme involved in the transfer of galactose from UDP-galactose to a non-reducing terminal N-acetylglucosamine of a glycoprotein or glycolipid. Rats were fed a semisynthetic diet containing 1% OA for 2 weeks and the livers were stimulated to regenerate by two-thirds partial hepatectomy (PH). The level of activity of the enzyme and the steady-state level of hepatic mRNA transcripts of GalTase were determined prior to PH and at 12, 24, 48, 72 h and 10 days after the surgery. The data show that the hepatic activity of GalTase is unaltered in both the control and OA-fed groups until 12 h following surgery, but begins to increase after this time period. In the control group a progressive increase was seen throughout the experimental period following PH. On the other hand in the OA-fed group 24 h after PH the initial increase seen up to 24 h was arrested later on and the activity remained inhibited throughout the rest of the experimental period. The supplementation of 1% OA diet with 0.3% adenine, which is known to reverse the OA-induced imbalance in the nucleotide pool sizes, relieved the inhibition of GalTase activity. The steady-state level of hepatic mRNA paralleled the activity of GalTase at all the time points studied during liver regeneration. The reduction in the level of mRNA transcripts of GalTase in the OA-fed group may not be due to either a general inhibition of synthesis and/or degradation of mRNAs as revealed by a comparison of the expression of beta-galactoside 2,6-sialyltransferase in both the control and OA-fed groups. The study thus suggests an imbalance in nucleotide pools, such as the one induced by OA, may play a role in the regulation of glycosylation by modulating the glycosyltransferases.


Asunto(s)
Galactosiltransferasas/metabolismo , Regeneración Hepática , Ácido Orótico/farmacología , Adenina/farmacología , Animales , Galactosiltransferasas/genética , Hepatectomía , Cinética , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344
11.
Res Commun Chem Pathol Pharmacol ; 81(2): 251-4, 1993 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8210704

RESUMEN

Highly purified 5-l-methyltetrahydrofolate (m-THF) and 5-l-formyl-THF (f-THF) preparations were compared for rescuing from methotrexate (MTX) toxicity in DBA2 mice transplanted with L1210 leukemia. Mice received two doses of reduced folates (2 mg/kg, s.c.) 16 and 24 h after a single s.c. MTX dose. f-THF was 1.8 time more effective than m-THF in protecting tumor cells from MTX (800 mg/kg). This MTX dose caused a 57% fall in circulating polymorphonucleates, which was prevented by both reduced folates. Treatment with 800 mg/kg of MTX plus m-THF was 1.5 fold more effective than the same MTX dose plus f-THF in increasing survival time of tumor-bearing mice. These data suggest a higher selectivity and efficacy of l-m-THF with respect to l-f-THF in rescuing from MTX toxicity.


Asunto(s)
Leucovorina/uso terapéutico , Leucemia L1210/tratamiento farmacológico , Metotrexato/toxicidad , Tetrahidrofolatos/uso terapéutico , Animales , Leucovorina/administración & dosificación , Metotrexato/administración & dosificación , Ratones , Ratones Endogámicos DBA , Trasplante de Neoplasias , Tetrahidrofolatos/administración & dosificación
12.
Crit Rev Oncog ; 11(1): 19-62, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10795626

RESUMEN

In this review, genetic changes known to occur in human and experimental animal hepatocarcinogenesis are evaluated comparatively, with the aim of identifying genes that could potentially be targets of new preventive and therapeutic strategies, albeit the fact that although a step-by-step analysis of the premalignant stages has been largely accomplished in experimental hepatocarcinogenesis, this goal is still elusive in the case of humans. Overexpression of several of the genes implicated in the MAPK signaling cascade and cell cycle control appears to be most likely responsible for initiated cells acquiring a proliferating phenotype that facilitates the accumulation of structural changes in additional genes, resulting in the generation of autonomously growing preneoplastic and neoplastic lesions. Several gene abnormalities seen in precancerous lesions of rodents also occur in human hepatocellular carcinomas, suggesting that at least some of them could be present also in human precancerous lesions. Furthermore, there are reports that epigenetic events, such as abnormal DNA methylation, may be critical in hepatocarcinogenesis. DNA hypomethylation is an early event, both in human and experimental hepatocarcinogenesis, and its role in the activation of various genes, has been postulated. In recent years, linkage analysis studies have led to the identification of susceptibility/resistance loci that influence the progression stage of hepatocarcinogenesis in mice and rats. The relevance of these findings, though, will depend on the identification of the genes, and on whether in humans there are genes ortholog with rodent's susceptibility/resistance genes. It is proposed that rodent hepatocarcinogenesis represents a promising model for the identification of genes implicated in the early stages of the process, and that many of these genes may represent key targets for the application of gene therapy in the prevention and treatment of liver cancer.


Asunto(s)
Metilación de ADN , Genes Supresores de Tumor , Neoplasias Hepáticas/genética , Oncogenes , Animales , División Celular , Aberraciones Cromosómicas , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/terapia , Pérdida de Heterocigocidad , Ratones , Ratas
13.
Int J Cancer ; 62(2): 210-5, 1995 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-7622298

RESUMEN

Dehydroepiandrosterone (DHEA) inhibits the development of early pre-neoplastic lesions and prevents tumor development in various tissues when given to animals during the initiation/promotion stages of carcinogenesis. Our purpose was to evaluate whether DHEA can also arrest the growth and progression of late lesions, such as persistent nodules (PNs) of rat liver. Male F344 rats were subjected to initiation by diethylnitrosamine followed by selection according to the "resistant hepatocyte" (RH) protocol. Fifteen weeks after initiation, when PNs were present in the liver, the rats were fed a diet with/without 0.6% DHEA for a maximum of 15 weeks. Glucose-6-phosphate dehydrogenase (G6PD) activity was 17- to 20-fold higher in PNs than in normal liver 15-30 weeks after initiation. It significantly decreased, in both liver and PNs, 16 hr after starting DHEA feeding. Further DHEA feeding for 3-15 weeks decreased G6PD activity by 55-58% in both tissues. Eight weeks after starting DHEA, a fall in the proportion of labeled cells, after continuous contact with 3H thymidine for 7 days, was found in nodules. Treatment for 15 weeks with DHEA caused a marked decrease in the number of nodules per liver, as well as in the incidence of PNs with diameters of 3-6 and > 6 mm, respectively, while it did not affect PNs with diameters < 3 mm. Nodules showing patterns of malignant transformation were present in 40% of rats not treated with DHEA, but not in DHEA-treated rats. All of 8 surviving rats not treated with DHEA had carcinomas at the 56th week, while only 1 out of 4 surviving rats treated with DHEA had carcinoma. These data indicate that DHEA inhibits G6PD activity in rat liver and in PNs in vivo. This is associated with growth restraint of PNs and results in inhibition of their progression to malignancy.


Asunto(s)
Deshidroepiandrosterona/farmacología , Hepatopatías/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Hígado/efectos de los fármacos , Lesiones Precancerosas/tratamiento farmacológico , Animales , Peso Corporal/efectos de los fármacos , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Hepatopatías/patología , Neoplasias Hepáticas/patología , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas
14.
Carcinogenesis ; 16(7): 1537-42, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7614686

RESUMEN

Previous work has demonstrated that dehydroepiandrosterone (DHEA) strongly inhibits growth and de novo cholesterol (CH) biosynthesis in preneoplastic rat liver. Administration of a mixture of 4 ribo- or deoxyribonucleosides of adenine, guanine, cytosine and uracil/thymine, prevents growth inhibition but not inhibition of CH synthesis. The purpose of this paper was to identify the site of inhibition of CH synthesis by DHEA. Persistent nodules (PNs) were induced, in diethylnitrosamine-initiated male F344 rats, by 'resistant hepatocyte' protocol. Fifteen weeks after initiation, nodule bearing rats and normal controls received a diet containing 0.6% DHEA for 3 weeks. They were then killed. 3-Hydroxy-3-methylglutaryl-CoA reductase (HMGR) activity and mRNA levels were 18- and 14-fold higher, respectively in nodules than in normal liver. DHEA strongly inhibited HMGR activity in both tissues in vivo, but had a slight effect on HMGR activity, when added in vitro to the reaction mixture for determination of this activity. In vivo DHEA treatment caused a 65% decrease in the level of HMGR mRNA in PNs, which, however, does not seem to completely account for the decrease in HMGR activity (83%). Low density lipoprotein receptor (LDL-R) mRNA level underwent a slight decrease in PNs, with respect to control liver, which did not lead to a significant decrease in 125I-LDL binding to LDL-R. DHEA treatment caused 30% and 24% increases in LDL-R expression and 125I-LDL binding, respectively, in nodules. These observations indicate that in addition to HMGR gene expression, increased influx of LDL into preneoplastic cells may contribute to the deregulation of mevalonate synthesis by DHEA. The observation that HMGR activity and gene expression were still 3- to 5-fold higher in PNs of DHEA-treated rats than in control liver, and previous findings of preneoplastic liver cell growth in the presence of relatively low CH synthesis, suggest that even relatively low levels of mevalonate are sufficient for the growth of preneoplastic liver cells.


Asunto(s)
Deshidroepiandrosterona/farmacología , Hidroximetilglutaril-CoA Reductasas/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/metabolismo , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/metabolismo , 17-Cetosteroides/orina , Animales , Peso Corporal/efectos de los fármacos , Colesterol/biosíntesis , Expresión Génica/efectos de los fármacos , Lipoproteínas LDL/metabolismo , Hígado/anatomía & histología , Hígado/efectos de los fármacos , Neoplasias Hepáticas Experimentales/genética , Masculino , Ácido Mevalónico/metabolismo , Lesiones Precancerosas/genética , Ratas , Ratas Endogámicas F344 , Receptores de LDL/metabolismo
15.
Carcinogenesis ; 14(5): 1077-80, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-7684954

RESUMEN

Liver nodules and carcinomas, developing in F344 rats initiated with diethylnitrosamine, exhibit high ornithine decarboxylase (ODC) activity and DNA synthesis. ODC-related RNAs of 1.8, 2.1 and 2.6 kb are produced by normal rat liver. Early preneoplastic nodules, developing 10 weeks after initiation, showed overproduction of 1.8 and 2.1 kb RNAs, while the 2.6 kb RNA was barely detectable. Rises in the 1.8, 2.1 and 2.6 kb RNAs occur in late nodules (30 weeks after initiation) and in carcinomas. The comparison of different tissues for relative increase in ODC activity, RNA levels and DNA synthesis showed that these parameters behaved in the same way: highest increases occurred in early nodules and carcinomas. These observations suggest that overexpression of ODC gene and alterations in regulatory mechanisms of ODC gene expression may be implicated in the progression of preneoplastic lesions to malignancy. Southern blot analysis of PstI DNA digests revealed the presence of ODC gene rearrangement in two carcinomas and in one late nodule. However, the role of this phenomenon in the progression of preneoplastic lesions is unclear, due to the possibility that ODC pseudogenes are involved instead of or in addition to ODC gene.


Asunto(s)
2-Acetilaminofluoreno/toxicidad , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/inducido químicamente , Hígado/patología , Ornitina Descarboxilasa/genética , Lesiones Precancerosas/inducido químicamente , Animales , Northern Blotting , Hígado/efectos de los fármacos , Hígado/enzimología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Índice Mitótico/efectos de los fármacos , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , ARN/genética , ARN/aislamiento & purificación , Ratas , Ratas Endogámicas F344 , Valores de Referencia , gamma-Glutamiltransferasa/análisis , gamma-Glutamiltransferasa/metabolismo
16.
Carcinogenesis ; 17(8): 1765-8, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8761440

RESUMEN

The phylogenetically distant F344 and BN rat strains and their (BN x F344) F1 hybrids were compared for susceptibility to hepatocarcinogenesis using the 'resistant hepatocyte' model. Quantitative stereological analysis of frequency (number/liver) and size (mean volume and volume fraction) of placental form glutathione S-transferase (GST-P)-positive lesions was carried out at 8, 15 and 32 weeks after diethylnitrosamine initiation. The number/liver of GST-P-positive lesions at any time point was slightly higher in BN and (BN x F344) F1 rats than in F344 rats, but not statistically different. However, mean volume and volume fraction of GST-P positive lesions were much higher in F344 than in both BN and (BN x F344) F1 rats at any time point, with a difference of up to > 10-fold. GST-P-positive lesions exhibited a significantly higher labeling index and much lower remodeling in male F344 than in BN and (BN x F344) F1 rats. HCCs were present at 54-57 weeks after initiation in 77% of male F344 and in no (BN x F344) F1 rats and at 70 weeks HCCs were observed in 100% of male F344 and in 23% of (BN x F344) F1 rats. These results suggest that the BN rat strain is resistant to hepatocarcinogenesis and that its resistance is genetically transmitted as a dominant character to F1 hybrids of the BN strain with the F344 susceptible strain.


Asunto(s)
2-Acetilaminofluoreno/toxicidad , Carcinógenos/toxicidad , Genes Dominantes , Neoplasias Hepáticas Experimentales/inducido químicamente , Animales , Peso Corporal , Predisposición Genética a la Enfermedad , Glutatión Transferasa/metabolismo , Isoenzimas/metabolismo , Hígado/enzimología , Neoplasias Hepáticas Experimentales/genética , Masculino , Tamaño de los Órganos , Filogenia , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344 , Especificidad de la Especie
17.
Carcinogenesis ; 17(7): 1533-7, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8706260

RESUMEN

S-Adenosyl-L-methionine (SAM) is a strong chemopreventive agent of rat liver carcinogenesis. Examination was made to determine whether inhibition by SAM of the development of preneoplastic liver lesions persists to SAM withdrawal in diethylnitrosamine-initiated F344 rats promoted with thiobenzamide (TB). The rats were subjected, 2 weeks after initiation, to 5 weeks feeding with a 0.1% TB diet followed by a TB-free diet for 6 weeks and then a second TB treatment for 3 weeks. SAM (384 micromol/kg/day) was injected i.m. during the first TB cycle (treatment A) or for 6 weeks after the first TB cycle (treatment B). Many gamma-glutamyltranspeptidase (GGT)-positive lesions developed in initiated rats after the first TB cycle. They decreased in number after TB withdrawal, while partial recovery of lesion number and a great increase in volume occurred after the second TB cycle. Liver ornithine decarboxylase (ODC) activity and c-myc and c-Ha-ras mRNAs increased during the TB cycles and returned to normal liver values after TB withdrawal. Number and size of GGT-positive lesions, DNA synthesis of GGT-positive cells, liver ODC activity and c-myc and c-Ha-ras mRNA levels decreased as a consequence of SAM treatment A. The recovery of these parameters, induced by a second TB cycle in rats not treated with SAM, was prevented by SAM treatment B. These results suggest that SAM causes a persistent decrease in growth capacity of preneoplastic liver lesions in rats subjected to a diethylnitrosamine/TB protocol.


Asunto(s)
Anticarcinógenos/farmacología , Carcinógenos , Neoplasias Hepáticas/prevención & control , Hígado/efectos de los fármacos , Lesiones Precancerosas/prevención & control , S-Adenosilmetionina/farmacología , Animales , ADN/biosíntesis , Dietilnitrosamina , Expresión Génica/efectos de los fármacos , Genes myc , Genes ras , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Masculino , Ornitina Descarboxilasa/metabolismo , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/patología , ARN Mensajero/análisis , Ratas , Ratas Endogámicas F344 , Tioamidas , Factores de Tiempo , gamma-Glutamiltransferasa/metabolismo
18.
Carcinogenesis ; 16(2): 427-30, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7859377

RESUMEN

Chemoprevention of liver carcinogenesis by S-adenosyl-L-methionine (SAM) was studied in F344 male rats. The rats were given 1,2-dimethylhydrazine (1,2-DMH) 2 HCl (100 mg/kg, i.p.) 18 h after two-thirds hepatectomy. One week later they were fed a semisynthetic basal diet containing 1% orotic acid (OA) for 29 weeks. At this time the rats were transferred to the basal semisynthetic diet and were killed 3 weeks later. SAM treatment (384 mumol/kg/day, i.m.), was started 1 week after 1,2-DMH and was continued up to the end of the experiment. Controls received solvent alone. SAM exerted an inhibitory effect on the induction of preneoplastic and neoplastic lesions. For example, nodules with diameters of 1-2 and 2-6 mm exhibited a decrease in both incidence and number per liver, while no such inhibitory effect was seen in the category of larger nodules. Furthermore, hepatocellular carcinoma (HCC) also exhibited a decrease in the SAM-treated group. The number/liver and incidence were 0.04 and 4.8% respectively in the SAM-treated group, compared to 0.38 and 37.8% in the control group. Microscopic examination showed the presence of well-differentiated carcinomas and atypical nodules in control rats, while only one small, well-differentiated tumor and one nodule with patterns of initial transformation were seen in SAM-treated rats. No patchy staining of glutathione-S-transferase, indicative of remodeling, was observed in nodules of both SAM-treated and control rats. Nodules and HCCs developing in SAM-treated rats exhibited a relatively high number of apoptotic bodies. Apoptotic bodies count showed 2.8- and 1.8-fold increases in nodules and HCCs of SAM-treated rats with respect to controls. These results indicate that SAM exerts a chemopreventive effect on hepatocarcinogenesis induced by the OA model. SAM seems to be more effective in inhibiting nodule to HCC progression than on the growth of nodule per se. The inhibitory effect is associated with an increase in cell loss by apoptosis in nodules and HCC.


Asunto(s)
Anticarcinógenos/uso terapéutico , Cocarcinogénesis , Dimetilhidrazinas/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/prevención & control , Ácido Orótico/toxicidad , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/prevención & control , S-Adenosilmetionina/uso terapéutico , 1,2-Dimetilhidrazina , Animales , Hígado/efectos de los fármacos , Neoplasias Hepáticas Experimentales/patología , Masculino , Lesiones Precancerosas/patología , Ratas , Ratas Endogámicas F344
19.
Int J Cancer ; 68(1): 136-42, 1996 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-8895554

RESUMEN

We have investigated by restriction fragment analysis genomic abnormalities involving the c-myc gene in DNA isolated from adenomas and hepatocellular carcinomas (HCCs). Adenomas and HCCs were induced by the "resistant hepatocyte" protocol in diethylnitrosamine-initiated male F344 rats. Southern-blot analysis of EcoRI-restricted DNA from normal liver, early and late adenomas, 12 weeks (EAs) and 30 weeks (LAs) after initiation, and HCCs, showed 2 bands of 18 and 3.2 kb hybridizing with c-myc, in all tissues. c-myc amplification occurred in almost all HCCs, and in the majority of EAs and LAs. These results were confirmed by dilution analysis. c-myc amplification was also seen in adenomas and HCCs by Southern analysis with HindIII-restricted DNA, and in HCCs by differential PCR. c-myc mRNA increase occurred in all adenomas and HCCs, but it was higher in the lesions showing gene amplification. Moreover, a 13-kb DNA extraband, hybridizing with c-myc, was found in the HindIII-restricted DNA from HCCs, but not in normal liver and adenomas, and a 7.1-kb extra band was present in EcoRI-digested DNA from one LA. EcoRI-restricted DNA from some adenomas exhibited a decrease in intensity of the 18-kb fragment, and an increase in intensity of the 3.2-kb fragment. No alteration in banding pattern occurred in the beta-actin gene in adenomas. These results provide evidence of amplification and some other rearrangements involving the c-myc gene, in pre-malignant and malignant liver lesions, induced by the RH protocol, and suggest a role of c-myc rearrangement in the progression of adenomas to malignancy.


Asunto(s)
Genes myc/genética , Neoplasias Hepáticas/genética , Adenoma/inducido químicamente , Adenoma/genética , Animales , Northern Blotting , Southern Blotting , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Desoxirribonucleasa EcoRI , Desoxirribonucleasa HindIII , Dietilnitrosamina , Neoplasias Hepáticas/inducido químicamente , Masculino , Reacción en Cadena de la Polimerasa , Lesiones Precancerosas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344
20.
Hepatology ; 31(4): 956-65, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10733553

RESUMEN

Molecular mechanisms of basal and D-amphetamine (AMPH)-induced apoptosis were studied in rat liver nodules, 12 (N12) and 30 (N30) weeks after initiation, and in hepatocellular carcinoma (HCC) induced by diethylnitrosamine in rats subjected to resistant hepatocyte model. Basal apoptosis in hematoxylin/eosin- and propidium iodide-stained sections was higher in nodules and HCC than in normal livers. It sharply increased in all tissues 4 hours after AMPH treatment (10 mg/kg), and declined to basal levels at 8 to 12 hours in liver and N12, but remained high up to 18 hours in N30 and HCC. c-myc, Tgf-alpha, p53, and Bcl-X(S) messenger RNA (mRNA) levels were higher, and Bcl-2 mRNA was lower in N12 and/or N30 and HCC than in normal liver. Four hours after AMPH injection, increase in c-myc and decreases in Bcl-2 and Bcl-X(L) mRNAs occurred in all tissues, whereas p53, Bax, and Bcl-X(S) mRNAs increased in N30 and HCC. These changes disappeared in liver and N12 at 18 hours, but persisted in N30 and HCC. c-Myc, P53, Bcl-2, and Bax proteins in normal liver and HCC +/- AMPH showed similar patterns. Tgf-beta1, Tgf-beta-RIII, CD95, and CD95L mRNA levels underwent slight or no changes in any tissue +/- AMPH. Basal Hsp27 expression was high in nodules and HCC, and was stimulated by AMPH in liver and N12, but not in N30 and HCC. These data suggest a role of dysregulation of Bcl-2 family genes and, at least in atypical lesions, of p53 overexpression, in basal and AMPH-induced apoptosis in nodules and HCCs. Hsp27 does not appear to sufficiently protect atypical lesions against apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Dextroanfetamina/farmacología , Neoplasias Hepáticas Experimentales/patología , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Animales , Apoptosis/genética , Colorantes , Fragmentación del ADN , Dietilnitrosamina , Genes myc , Genes p53 , Cinética , Neoplasias Hepáticas Experimentales/inducido químicamente , Masculino , Propidio , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Proteína X Asociada a bcl-2 , Receptor fas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA