Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Environ Res ; 239(Pt 2): 117341, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37839534

ABSTRACT

BACKGROUND: Per- and polyfluoroalkyl substances (PFAS) constitute a large group of compounds that are water, stain, and oil repellent. Numerous sources contribute to the blood levels of PFAS in the European population. The main contributor for perfluorooctanoic acid (PFOA) is food, house dust, consumer products and personal care products (PCPs). OBJECTIVES: The purpose of the present work is to calculate the dietary and dermal external exposure to PFOA, estimate the aggregated internal exposure from diet and PCPs using a PBPK model, and compare estimates with measured concentrations. METHODS: Detailed information on diet and PCP use from the EuroMix study is combined with concentration data of PFOA in food and PCPs in a probabilistic exposure assessment. A physiologically based pharmacokinetic model (PBPK) was further refined by incorporating a dermal exposure pathway, and changes in the kidney and faecal excretion. RESULTS: The aggregated internal exposure using the PBPK model shows that the major contributor to the internal exposure is diet for both males and females. The estimated internal exposure of PFOA for the EuroMix population was in the same range but lower than the measured blood concentrations using the lower bound (LB) external exposure estimates, showing that the LB estimates are underestimations. For seven females the internal exposure of PFOA were higher from PCPs than from diet. CONCLUSION: PCPs and diet contributed in the same range to the internal PFOA exposure for several women participating in EuroMix. This calls for additional studies on exposure to PFOA and possibly other PFAS from PCPs, especially for women. Overall, PBPK modelling was shown as valuable tool in understanding the sources of PFOA exposure and in guiding risk assessments and regulatory decisions.


Subject(s)
Alkanesulfonic Acids , Cosmetics , Environmental Pollutants , Fluorocarbons , Male , Humans , Female , Biological Monitoring , Diet , Norway , Environmental Pollutants/metabolism
2.
Environ Res ; 195: 110795, 2021 04.
Article in English | MEDLINE | ID: mdl-33524335

ABSTRACT

BACKGROUND: Human exposure to chemicals through the oral, dermal, or inhalation routes is significant. To assess this exposure, a human biomonitoring study was conducted in Norway to examine the plausibility of source-to-dose calculations for chemical mixtures. Per- and polyfluoroalkyl substances (PFASs) are man-made compounds used for their surfactant properties, and several are persistent and bioaccumulative. Some PFASs are toxic and are regarded as endocrine disruptors and have been shown to suppress immune function and affect cholesterol homeostasis. Using the participants from the EuroMix BM study, we set out to describe PFAS concentrations and to evaluate associations with diet and use of personal care products (PCPs). METHODS: Participants (44 males and 100 females) kept detailed diaries on their food consumption and their PCP use for two non-consecutive days. All urine (24 h) and blood samples were collected at the end of each study day. Levels of 25 PFASs were analysed in serum from study day 1 using a high throughput online solid phase extraction ultra-high-performance liquid chromatography tandem mass spectrometry method. Multivariable linear regressions were performed between each food and PCP category and each chemical and were sex-stratified when the consumption of food or use of PCPs was significantly different between men and women. RESULTS: Eight PFASs were detected in all analysed samples (PFHxS, PFHpS, PFOS, PFOA, PFNA, PFDA, PFUnDA and PFDoDA), and four PFASs were below the limit of detection (PFOPA, PFDPA, PFHxA, and EtFOSA). Several PFASs were found to be positively associated with fish consumption (PFOS, PFNA, PFUnDA, PFDoDA, PFDA, PFDS and PFTrDA). Sunscreen, mouthwash, and lip gloss/lip balm were found to be positively associated with PFASs (PFOA, PFTrDA, and PFOSA). CONCLUSION: The participants in the EuroMix study were exposed to PFASs through their diet and PCP use. Several foods and PCPs were found to be potential sources of exposure to PFASs.


Subject(s)
Alkanesulfonic Acids , Cosmetics , Endocrine Disruptors , Environmental Pollutants , Fluorocarbons , Alkanesulfonic Acids/analysis , Animals , Biological Monitoring , Environmental Pollutants/analysis , Female , Fluorocarbons/analysis , Humans , Male , Norway
3.
Andrology ; 4(6): 1102-1114, 2016 11.
Article in English | MEDLINE | ID: mdl-27575329

ABSTRACT

Diet-induced obesity is known to impair male reproduction and may aggravate the male reproductive toxicity of the food contaminant acrylamide. Exposure of male mice to acrylamide induces paternally mediated pre- and post-implantation losses because of spermatozoal toxicity and these effects are potentiated in mice fed a high-fat diet. Glycidamide - an acrylamide metabolite - is the primary mediator of reproductive effects in males. The mechanisms causing the interaction between diet and acrylamide are not clear. However, diet-induced obesity is associated with oxidative stress in male reproductive tissues which might contribute to increased germ cell susceptibility. In this study, we investigated whether a moderate diet-induced obesity regimen could interfere with glycidamide-induced spermatozoal toxicity and increase oxidative stress. For this purpose, sperm chromatin integrity, oxidised DNA and protein levels, transcript levels of oxidative stress responsive genes and glycidamide-induced DNA and haemoglobin adducts were analysed in samples from male mice exposed to a high-fat diet for 6 weeks in combination with a single glycidamide exposure 7 days prior to sacrifice. We found that glycidamide-induced sperm DNA fragmentation was markedly higher in obese than in lean mice. However, the levels of oxidised DNA and/or protein in blood, liver and testicular tissue was lower in obese than in lean mice. Accompanying the reduced level of oxidised macromolecules, the transcript levels of several oxidative stress-related genes were altered in the liver and testis from obese mice suggesting induction of an antioxidant response in these animals. The haemoglobin-glycidamide adduct levels were higher in obese than in lean animals, whereas obesity did not seem to increase the level of glycidamide-induced DNA adducts. These findings show that a moderate diet-induced obesity regimen may potentiate glycidamide-induced sperm cells toxicity and suggest that the increase in glycidamide-induced sperm toxicity observed in obese mice does not depend on overt oxidative stress.


Subject(s)
Chromatin/metabolism , Epoxy Compounds/pharmacology , Obesity/metabolism , Oxidative Stress/physiology , Spermatozoa/metabolism , Animals , DNA Fragmentation/drug effects , Diet, High-Fat , Liver/drug effects , Liver/metabolism , Male , Mice , Oxidation-Reduction , Oxidative Stress/drug effects , Spermatozoa/drug effects , Testis/drug effects , Testis/metabolism
4.
Andrology ; 2(2): 234-43, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24459046

ABSTRACT

An increased global prevalence of obesity coincides with an apparent decline in male sperm quality and a possible association between these pathologies has been suggested. In this study, we examined the effects of obesity on sperm chromatin integrity using two mouse models of obesity. In one group of mice, obesity was induced by a high-fat diet (HFD) (diet-induced obesity; DIO model), whereas in the other group, leptin deficiency was used to study the effects of obesity independently of the influence of dietary factors. Sperm chromatin integrity is recognized as an important measure of male infertility, and was analysed by the sperm chromatin structure assay. We found increased sperm DNA fragmentation in both groups of obese mice compared to lean mice, whereas the percentage of immature spermatozoa was not increased by obesity. The DIO model reflects the human condition more closely than the leptin-deficient model and was therefore selected for examination of the transcriptional response of a selection of marker genes in the testis by quantitative real-time PCR. The analysis of transcript levels of the selected testicular marker genes showed moderate, but significant, up-regulation of the Cyp2e1, Cyp19a1, Tnf and Pparg genes in DIO mice compared to lean mice. In conclusion, a clear positive correlation between body mass index and sperm DNA fragmentation was found in two mouse models of obesity. However, the variability in sperm DNA fragmentation within the two groups of obese animals was high. The observed changes in the transcript level of the marker genes suggest that there may be a local response in testicular cells to the HFD regimen with a potential impact on intratesticular signalling and spermatogenesis.


Subject(s)
Chromatin/genetics , DNA Fragmentation , Obesity/genetics , Spermatozoa/cytology , Animals , Aromatase/biosynthesis , Aromatase/genetics , Body Mass Index , Cytochrome P-450 CYP2E1/biosynthesis , Cytochrome P-450 CYP2E1/genetics , Diet, High-Fat , Gene Expression , Infertility, Male/genetics , Leptin/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/biosynthesis , PPAR gamma/genetics , Semen Analysis , Tumor Necrosis Factors/biosynthesis , Tumor Necrosis Factors/genetics , Up-Regulation
5.
Oncogene ; 27(19): 2763-71, 2008 Apr 24.
Article in English | MEDLINE | ID: mdl-18026139

ABSTRACT

p53 plays a major role in the prevention of tumor development. It responds to a range of potentially oncogenic stresses by activating protective mechanisms, most notably cell-cycle arrest and apoptosis. The p53 gene is also induced during normal liver regeneration, and it has been hypothesized that p53 serve as a proliferative 'brake' to control excessive proliferation. However, it has lately been shown that p53 inhibition reduces hepatocyte growth factor-induced DNA synthesis of primary hepatocytes. Here we show that epidermal growth factor (EGF) activated p53 in a phosphatidylinositol-3 kinase-dependent way, and thus induced the cyclin-dependent kinase inhibitor p21(Cip1) in primary rat hepatocytes. p53 inactivation with a dominant-negative mutant (p53(V143A)) attenuated EGF-induced DNA synthesis and was associated with reduced CDK2 phosphorylation and retinoblastoma protein hyperphosphorylation. When p21(Cip1) was ectopically expressed in p53-inactivated cells, these effects were neutralized. In conclusion, our results demonstrate that in normal hepatocytes, EGF-induced expression of p53 is involved in regulating CDK2- and CDK4 activity, through p21(Cip1) expression.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , S Phase/physiology , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , Enzyme Activation/physiology , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/physiology , Male , Phosphatidylinositol 3-Kinases/physiology , Rats , Rats, Wistar , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology
6.
Cell Prolif ; 40(4): 475-87, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17635516

ABSTRACT

INTRODUCTION/OBJECTIVES: Cell cycle progression is driven by the coordinated regulation of cyclin-dependent kinases (CDKs). In response to mitogenic stimuli, CDK4 and CDK2 form complexes with cyclins D and E, respectively, and translocate to the nucleus in the late G(1) phase. It is an on-going discussion whether mammalian cells need both CDK4 and CDK2 kinase activities for induction of S phase. METHODS AND RESULTS: In this study, we have explored the role of CDK4 activity during G(1) progression of primary rat hepatocytes. We found that CDK4 activity was restricted by either inhibiting growth factor induced cyclin D1-induction with the PI3K inhibitor LY294002, or by transient transfection with a dominant negative CDK4 mutant. In both cases, we observed reduced CDK2 nuclear translocation and reduced CDK2-Thr160 phosphorylation. Furthermore, reduced pRb hyperphosphorylation and reduced cellular proliferation were observed. Ectopic expression of cyclin D1 alone was not sufficient to induce CDK4 nuclear translocation, CDK2 activity or cell proliferation. CONCLUSIONS: Thus, epidermal growth factor-induced CDK4 activity was necessary for CDK2 activation and for hepatocyte proliferation. These results also suggest that, in addition to regulating cyclin D1 expression, PI3K is involved in regulation of nuclear shuttling of cyclin-CDK complexes in G(1) phase.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Hepatocytes/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/enzymology , Cell Proliferation , Cells, Cultured , Cyclin D1/metabolism , Cyclin E/biosynthesis , Cyclin-Dependent Kinase 4/chemistry , Epidermal Growth Factor/pharmacology , Hepatocytes/drug effects , Male , Rats , Rats, Wistar , Threonine/metabolism
7.
Mol Carcinog ; 28(2): 84-96, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10900465

ABSTRACT

The hepatic carcinogen 2-acetylaminofluorene (AAF) exerts its effect as a tumor promoter by mitoinhibition of normal hepatocytes. Initiated cells proliferate selectively and develop into preneoplastic foci and subsequently into carcinomas. To study whether some of the mitoinhibitory effects of AAF could be attributed to an influence on intracellular signal transduction, growth factor signaling was studied in cultured hepatocytes from rats fed AAF for 7 d. Activation through the epidermal growth factor receptor (EGFR) was used to probe possible changes in downstream mitogenic signaling mechanisms. The proliferative response to epidermal growth factor (EGF), measured as proliferating cell nuclear antigen expression and thymidine incorporation, was almost completely inhibited in hepatocytes exposed to AAF. Neither EGFR protein levels nor EGF binding was notably altered in AAF-exposed hepatocytes as opposed to normal hepatocytes. The initial tyrosine phosphorylation of EGFR and downstream activation of Sos, Raf-1, and extracellular signal-regulated protein kinase (ERK) were similar in AAF-treated and control hepatocytes. Even though ERK phosphorylation was unaffected, a remarkable (80%) reduction of ERK nuclear accumulation was observed in AAF-exposed hepatocytes immediately after mitogen stimulation. EGFR tyrosine phosphorylation and downstream signaling lasted 6 h in control cells versus 2 h in AAF-exposed hepatocytes. We previously demonstrated that AAF inhibits the growth factor-dependent induction of cyclin D1 and arrests hepatocyte cell-cycle progression before the p21/CIP1-controlled DNA-damage check point. The present data indicate that the DNA-damaging carcinogen AAF induces growth inhibition by a distinct inhibition of ERK nuclear accumulation after mitogen stimulation. Inhibition of intracellular signal transduction may represent a novel mechanism of growth arrest. Mol. Carcinog. 28:84-96, 2000.


Subject(s)
2-Acetylaminofluorene/pharmacology , Carcinogens/pharmacology , Cell Nucleus/enzymology , Epidermal Growth Factor/pharmacology , Liver/drug effects , Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Division/drug effects , Cells, Cultured , ErbB Receptors/metabolism , Liver/enzymology , Phosphorylation , Protein Binding , Rats
8.
Mol Carcinog ; 27(3): 190-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10708481

ABSTRACT

Growth arrest in G(1) is a common cellular response to DNA damage. In the present study, liver regeneration was combined with continuous exposure for 2-acetylaminofluorene (AAF) to study mechanisms of carcinogen-induced growth arrest in vivo. Growth arrest of uninitiated hepatocytes is central for AAF-induced promotion of premalignant lesions in rat liver. To characterize this growth arrest, we examined the activity of cyclin-dependent kinase (Cdk) 2 in unexposed liver and in AAF-exposed liver after growth induction by partial hepatectomy (PH). Rats were fed either a control diet or an AAF-supplemented diet. After 7 d, a two-third PH was performed and the animals were killed after 0, 12, 18, 24, and 36 h. Kinase assays showed that cyclin E- and Cdk2-associated activities were lower in AAF-exposed liver than in unexposed liver after PH. Although the total cellular levels of cyclin E and Cdk2 were similar, cyclin E-Cdk2 assembly was markedly reduced. In unexposed hepatocytes, Cdk2 translocated to the nuclei after PH. Much of the nuclear Cdk2 was in a rapidly migrating form, presumably representing the Thr160-phosphorylated form of Cdk2. In contrast, in AAF-exposed liver both nuclear Cdk2 accumulation and Thr160-phosphorylation of Cdk2 were reduced. Although p53 and p21(waf1/cip1) were induced by AAF, the binding of p21 to cyclin E and Cdk2 was not increased in growth arrested liver. In conclusion, hepatocyte growth arrest caused by AAF exposure was characterized by a lowered Cdk2 activity that was accompanied by a reduced assembly of cyclin E-Cdk2 complexes but not by binding of p21.


Subject(s)
2-Acetylaminofluorene/pharmacology , CDC2-CDC28 Kinases , Carcinogens/pharmacology , Cell Nucleus/metabolism , Cyclin-Dependent Kinases/metabolism , Liver/drug effects , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Division , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Activation , Liver/enzymology , Liver/metabolism , Liver Regeneration , Male , Phosphorylation , Rats , Rats, Inbred F344 , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
9.
J Histochem Cytochem ; 48(1): 21-33, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10653583

ABSTRACT

After binding of epidermal growth factor (EGF), the EGF receptor (EGFR) becomes autophosphorylated via tyrosine. The ligand-activated receptor is internalized by endocytosis and subsequently degraded in the lysosomal pathway. To follow EGFR activation after EGF stimulation, we generated antisera to the EGFR phosphotyrosine sites pY992 and pY1173. The SH2 region of Shc binds to both these sites. Both antisera identified EGFR after EGF binding and did not crossreact with the unactivated receptor. The intracellular distribution of phosphorylated EGFR after ligand binding was traced by two-color immunofluorescence confocal microscopy and immunoelectron microscopy. Before EGF stimulation EGFR was primarily located along the cell surface. When internalization of activated EGFR was inhibited by incubation with EGF on ice, Y992- and Y1173-phosphorylated EGFR were located along the plasma membrane. Ten minutes after internalization at 37C, Y992- and Y1173-phosphorylated EGFR were almost exclusively located in early endosomes, as shown by co-localization with EEA1. Immunoelectron microscopy confirmed that phosphorylated EGFR was located in intracellular vesicles resembling early endosomes. After EGF stimulation, the adaptor protein Shc redistributed to EGFR-containing early endosomes. Our results indicate that EGFR activation of Shc via tyrosine-phosphorylated Y992 and Y1173 occurred in early endocytic compartments, and support a role for membrane trafficking in intracellular signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Endosomes/ultrastructure , Epidermal Growth Factor/pharmacology , ErbB Receptors/isolation & purification , Phosphoproteins/isolation & purification , Proteins/isolation & purification , Binding, Competitive , Biological Transport , Cell Compartmentation , Cell Membrane/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , HeLa Cells , Humans , Intracellular Membranes/metabolism , Microscopy, Immunoelectron , Phosphorylation , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
10.
Mol Carcinog ; 24(1): 36-46, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10029409

ABSTRACT

2-Acetylaminofluorene (AAF) is a potent tumor promoter in rat liver carcinogenesis models. In the resistant hepatocyte model, AAF is combined with a growth stimulus for efficient promotion of preneoplastic lesions. The promoting property of AAF in this model is closely associated with mito-inhibition of normal hepatocytes, an effect to which initiated cells are resistant. How AAF induces growth arrest is not known, but genotoxic as well as non-genotoxic effects have been implicated. To elucidate the mechanisms of AAF-induced mito-inhibition, we studied the expression of the tumor suppressor protein p53 and the cyclin-dependent kinase (cdk) complexes mediating G1 progression and S-phase entry. Hepatocytes were isolated from male Fisher 344 rats fed either a control diet or a diet supplemented with 0.02% AAF for 1 wk and cultured in a defined serum-free medium containing epidermal growth factor, insulin, and dexamethasone. Thymidine labeling revealed a profound inhibition of DNA synthesis in AAF-exposed cells compared with control cells. The retinoblastoma protein did not become hyperphosphorylated in AAF-exposed cells. Thus, inhibition of G1 cyclin-cdk activity was implied as a cause of growth arrest. Indeed, G1 cell-cycle arrest was accompanied by reduced induction and nuclear accumulation of the cyclin D1-cdk4 complex and inhibited nuclear translocation of cdk2. Furthermore, the growth arrest was not mediated through p21/waf1 upregulation, although nuclear levels of p53 were increased. Thus, carcinogen-induced mito-inhibition may be effected by altered levels and localization of G1 cyclin-cdk complexes, independent of the upregulation of cdk inhibitory proteins.


Subject(s)
2-Acetylaminofluorene/pharmacology , Carcinogens/pharmacology , Cell Cycle/genetics , Cyclins/genetics , Gene Expression Regulation/drug effects , Genes, p53 , Liver/drug effects , Animals , Cell Division/drug effects , Cells, Cultured , Culture Media, Serum-Free , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , DNA/biosynthesis , Enzyme Inhibitors , G1 Phase , Kinetics , Liver/cytology , Liver/metabolism , Male , Rats , Rats, Inbred F344 , Thymidine/metabolism , Time Factors , Tumor Suppressor Protein p53/biosynthesis
11.
Tidsskr Nor Laegeforen ; 119(29): 4323-8, 1999 Nov 30.
Article in Norwegian | MEDLINE | ID: mdl-10667131

ABSTRACT

The scandal in Belgium last spring has drawn attention to the environmental hazards of dioxins. Previous production of pesticides and widespread combustion of organic material in the presence of chloride have lead to environmental accumulation of these toxicants, which more precisely are termed polychlorinated dibenzo-p-dioxins and dibenzofurans. Their very long biological half-lives in combination with detectable biological effects at very low concentrations have caused health concerns. Chloracne is the only well documented health effect in man, but there are experimental evidence for carcinogenic, teratogenic, reproductive and immunosuppressive effects. In this presentation we review current knowledge about the cellular effects of dioxins. Dioxins bind to and exert their effects through the cytoplasmic aryl hydrocarbon receptor, which acts as a transcription factor and regulates a number of cytokines and microsomal enzymes. Furthermore, dioxins interfere with hormonal signalling, and anti-oestrogenic effects, vitamin A inhibition and thyroxin mimicry have been reported. Recently, effects on intracellular growth factor signalling have been demonstrated. Dioxins inhibit epidermal growth factor receptor, activate protein kinase C and other intracellular signal transducers, and activate transcription factors. As overall understanding of their cellular mechanisms of toxicity is lacking, we do not possess a complete basis for estimating the adverse health effects of this group of environmental toxicants.


Subject(s)
Dioxins/adverse effects , Environmental Pollutants/adverse effects , Polychlorinated Dibenzodioxins/adverse effects , Animals , Apoptosis/drug effects , Benzofurans/adverse effects , Benzofurans/chemistry , Benzofurans/poisoning , Benzofurans/toxicity , Carcinogens , Dioxins/chemistry , Dioxins/poisoning , Dioxins/toxicity , Environmental Pollutants/poisoning , Environmental Pollutants/toxicity , Hormones/metabolism , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/poisoning , Immunosuppressive Agents/toxicity , Polychlorinated Dibenzodioxins/chemistry , Polychlorinated Dibenzodioxins/poisoning , Polychlorinated Dibenzodioxins/toxicity , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/metabolism , Reproduction/drug effects , Risk Factors , Teratogens
12.
Carcinogenesis ; 19(9): 1559-63, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9771925

ABSTRACT

Exposure of cells to a variety of stresses such as heat, radiation and xenobiotics leads to increased expression of heat-shock proteins (HSPs). HSPs protect cells against irreversible protein damage and are involved in adaptive responses to stress stimuli. Some HSPs are overexpressed in neoplasias, possibly contributing to the increased drug tolerance often observed in such lesions. We have studied HSP expression in two experimental rat hepatocarcinogenesis models. Our aim was to clarify whether they are involved in stress adaptation in hepatocytes during carcinogen exposure, and whether HSPs may contribute to xenobiotic resistance in preneoplastic lesions. The complete carcinogen 2-acetylaminofluorene (AAF) was used in a continuous feeding protocol, and in the resistant hepatocyte model where the growth of diethylnitrosamine initiated lesions is efficiently promoted. Of the HSPs tested, only heat-shock protein 27 (hsp27) was induced during continuous AAF exposure. After 4 weeks of feeding AAF, increased hsp27 expression was noted in hepatocytes in perivenous areas of the liver lobule, possibly mediating an adaptive response to stress caused by reactive AAF metabolites. Enzyme altered preneoplastic foci were not found to overexpress HSPs. Thus, HSP induction does not seem to be a general mechanism underlying the increased stress tolerance observed in such lesions.


Subject(s)
2-Acetylaminofluorene/toxicity , Carcinogens/toxicity , Heat-Shock Proteins/biosynthesis , Liver Neoplasms, Experimental/metabolism , Liver/drug effects , Precancerous Conditions/metabolism , Animals , Liver/metabolism , Liver Neoplasms, Experimental/chemically induced , Male , Precancerous Conditions/chemically induced , Rats , Rats, Inbred F344
13.
Exp Cell Res ; 243(1): 161-72, 1998 Aug 25.
Article in English | MEDLINE | ID: mdl-9716460

ABSTRACT

We investigated the ability of endocytosed activated epidermal growth factor receptors (EGFR) to induce expression of the cyclin-interacting protein p21/CIP1 in A431 cells. Transforming growth factor alpha (TGFalpha) and EGF both induced tyrosine phosphorylation, induction of p21/CIP1, and thereby inhibition of DNA synthesis. TGFalpha is released from the EGFR when the TGFalpha-EGFR complex encounters low pH upon endocytosis. Consistently, we found more rapid dephosphorylation of the EGFR and less induction of p21/CIP1 by TGFalpha than by EGF. This difference was abolished upon neutralizing endosomal pH by the carboxylic ionophore monensin or the proton ATPase inhibitor bafilomycin A1. When surface-bound TGFalpha was removed by acid stripping and endosomal pH was neutralized with bafilomycin A1, TGFalpha stimulated EGFR tyrosine phosphorylation, induced p21/CIP1, and inhibited DNA synthesis. This strongly suggests that p21/CIP1 can be induced by endocytosed, activated EGFR and that endocytosed EGFR can affect cell growth.


Subject(s)
Cyclins/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Macrolides , Anti-Bacterial Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cyclin-Dependent Kinase Inhibitor p21 , DNA/biosynthesis , Endocytosis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Microscopy, Fluorescence , Monensin/pharmacology , Phosphorylation , Time Factors , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured
14.
J Histochem Cytochem ; 44(3): 227-33, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8648082

ABSTRACT

Transforming growth factor-alpha (TGF-alpha) and hepatocyte growth factor (HGF) are strong hepatocyte mitogens and important regulators of liver regeneration. The TGF-alpha receptor EGFr appears primarily to mediate a proliferative signal, whereas mitogenic, motogenic, and morphogenic effects have been attributed to activation of the HGF receptor Met. We have studied the localization of Met and EGFr in normal and carcinogen-treated rat livers. Oval cells and preneoplastic lesions were induced by diethylnitrosamine initiation, followed by promotion with 2-acetylaminofluorene combined with a partial hepatectomy. Different liver cell populations and their receptor expression were characterized by two-color immunofluorescence and confocal laser scanning microscopy. Hepatocytes were detected by keratin K8 staining, and oval cells and bile ducts were recognized by keratin K19 expression. Enzyme-altered preneoplastic lesions ere identified by expression of placental glutathione S-transferase (GST-pi). Staining for these cellular markers was combined with immunodetection of EGFr and Met. Normal liver exhibited strong staining for EGFr in hepatocytes, whereas blood vessels, bile ducts, and some sinusoidal cells were Met-positive. In carcinogen-treated livers, oval cells showed Met but not EGFr immunostaining. GST-pi-positive foci displayed EGFr immunostaining at a similar intensity as surrounding hepatocytes, whereas Met was not detected. Our data indicate that putative liver cells (oval cells) have a growth receptor phenotype similar to that of bile ducts, whereas preneoplastic live lesions appear hepatocyte-like. These results indicate that the preferential proliferation of preneoplastic liver lesions compared to surrounding hepatocytes is not associated with an altered EGFr or Met phenotype.


Subject(s)
Carcinogens/administration & dosage , Diethylnitrosamine/administration & dosage , ErbB Receptors/analysis , Liver/metabolism , Receptor Protein-Tyrosine Kinases/analysis , Animals , Immunohistochemistry , Liver/pathology , Male , Proto-Oncogene Proteins c-met , Rats , Rats, Inbred F344
15.
Histochem Cell Biol ; 104(4): 287-94, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8548562

ABSTRACT

To elucidate cell differentiation in liver carcinogenesis, we have studied the CCAAT/enhancer-binding protein (C/EBP). C/EBP is a positive-acting transcription factor important for the maintenance of liver-specific functions. It is associated with differentiation and regarded as an anti-proliferative agent. We have studied the expression and localization of C/EBP during sequential rat liver carcinogenesis. Two-color immunohistochemistry and confocal laser scan microscopy demonstrated C/EBP in hepatocyte nuclei and preneoplastic liver lesions, but not in bile ducts, non-parenchymal cells or oval cells. Both western blotting and immunohistochemistry revealed down-regulation of C/EBP during normal regeneration and when regeneration was inhibited by the carcinogen, 2-acetylaminofluorene. A similar down-regulation was shown by western blotting in hepatocytes grown in culture. Our data suggest that the altered metabolic phenotype of preneoplastic liver lesions was not caused by a change in the expression of C/EBP. Furthermore, the data favor a hepatocyte derivation of preneoplastic liver lesions.


Subject(s)
DNA-Binding Proteins/metabolism , Liver Neoplasms, Experimental/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , 2-Acetylaminofluorene/pharmacology , Animals , Blotting, Western , CCAAT-Enhancer-Binding Proteins , Carcinogens/pharmacology , Cell Separation , DNA-Binding Proteins/biosynthesis , Down-Regulation/physiology , Enhancer Elements, Genetic , Fluorescent Antibody Technique, Direct , Immunohistochemistry , Liver Neoplasms, Experimental/chemically induced , Liver Regeneration/drug effects , Liver Regeneration/physiology , Male , Microscopy, Confocal , Nuclear Proteins/biosynthesis , Precancerous Conditions/metabolism , Rats , Rats, Inbred F344 , Rats, Wistar , Tumor Cells, Cultured
17.
J Rural Health ; 10(4): 273-8, 1994.
Article in English | MEDLINE | ID: mdl-10139175

ABSTRACT

An important aspect of primary care physician availability is the retention of physicians once they have located. While retention has been under-researched compared to recruitment, it is especially important in rural areas where physician shortages already exist. This study reports the results of a retention survey completed by 132 primary care physicians in rural eastern Kentucky. The survey sets up an objective, hypothetical retention scenario and asks physicians to respond to structured questions and to an open-ended question about factors not appearing in the survey. In response to the structured portion of the survey, physicians indicate that relief coverage is the most important factor in rural physician retention. A content analysis of 75 open-ended responses reveals that besides the other factors in the survey, "sociocultural integration" is the pre-eminent retention issue for rural practitioners. This article concludes that the role of the local rural community may be more important in retention than in recruitment. Finally, it is suggested that additional in-depth qualitative research be conducted within the local contexts to enhance the understanding of rural physician retention processes.


Subject(s)
Personnel Loyalty , Physicians, Family/supply & distribution , Professional Practice Location/statistics & numerical data , Rural Health/statistics & numerical data , Attitude of Health Personnel , Data Collection , Evaluation Studies as Topic , Female , Health Services Research , Humans , Kentucky , Male , Physicians, Family/statistics & numerical data , Primary Health Care/statistics & numerical data , Workforce
SELECTION OF CITATIONS
SEARCH DETAIL