Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
J Intern Med ; 272(3): 201-23, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22789058

ABSTRACT

Inherited metabolic diseases of the liver are characterized by deficiency of a hepatic enzyme or protein often resulting in life-threatening disease. The remaining liver function is usually normal. For most patients, treatment consists of supportive therapy, and the only curative option is liver transplantation. Hepatocyte transplantation is a promising therapy for patients with inherited metabolic liver diseases, which offers a less invasive and fully reversible approach. Procedure-related complications are rare. Here, we review the experience of hepatocyte transplantation for metabolic liver diseases and discuss the major obstacles that need to be overcome to establish hepatocyte transplantation as a reliable treatment option in the clinic.


Subject(s)
Cell Transplantation/methods , Hepatocytes/cytology , Liver Diseases/therapy , Metabolism, Inborn Errors/therapy , Adaptive Immunity , Animals , Cell Culture Techniques , Cellular Senescence , Cryopreservation , Humans , Immunity, Innate , Immunosuppression Therapy , Tissue Donors , Transplantation Conditioning
2.
Transplant Proc ; 38(4): 1179-80, 2006 May.
Article in English | MEDLINE | ID: mdl-16757299

ABSTRACT

Hepatocyte transplantation has been used for temporary metabolic support of patients in end-stage liver failure awaiting whole organ transplantation as a method to support liver function and facilitate regeneration of the native liver in cases of fulminant hepatic failure and as a "cellular therapy" for patients with genetic defects in vital liver functions. The aim of this paper was to discuss the basic research that led to clinical hepatocyte transplantation, the published clinical experience with this experimental technique, and some possible future uses of hepatocyte transplantation.


Subject(s)
Hepatocytes/transplantation , Liver Diseases/surgery , Animals , Humans , Models, Animal
3.
Toxicol In Vitro ; 20(2): 135-53, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16359840

ABSTRACT

Anticancer drugs have a complex pharmacological and toxicological profile with a narrow therapeutic index. It is therefore critical to understand the factors that contribute to the marked intersubject variability in the pharmacokinetics and pharmacodynamics often observed with anticancer compounds. Since hepatic and extra-hepatic drug metabolism represents a major drug disposition pathway, extensive efforts are made to thoroughly investigate metabolism of anticancer compounds during the pre-clinical and clinical development phases as well as to address issues encountered during the clinical use of an approved drug. In recent years there has been a significant paradigm shift in pre-clinical/non-clinical drug metabolism studies. Most importantly, this has included a reduced reliance on animal models and increased use of human tissues (i.e. human liver microsomes and other cellular fractions, primary culture of human hepatocytes, cDNA expressed human-specific enzymes and cell-based reporter assays). Typically, experiments are performed using these tools to identify the phase I and/or phase II enzymes involved in metabolism of the drug/investigational agent and for metabolic fingerprinting. Additionally, issues pertaining to the rate, extent and mechanism(s) of the inhibition or induction of the metabolic pathways are also investigated. These studies provide important clues about various aspects of the disposition of a therapeutic agent including first-pass metabolism, elimination half-life, overall bioavailability and the potential for drug-drug interactions. The methodologies used for in vitro assessment of drug metabolism and their applications to drug development and clinical therapeutics with special emphasis on anticancer drugs are reviewed in this manuscript.


Subject(s)
Antineoplastic Agents/metabolism , Liver/metabolism , Biotransformation , Cells, Cultured , Drug Evaluation, Preclinical , Drug Interactions , Hepatocytes/metabolism , Humans , In Vitro Techniques , Liver/enzymology , Microsomes, Liver/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism
4.
Xenobiotica ; 35(5): 419-38, 2005 May.
Article in English | MEDLINE | ID: mdl-16012075

ABSTRACT

1. Previous studies reported that rat hepatocytes overlaid with extracellular matrix components (Matrigel) maintain the expression and responsiveness of drug-metabolizing enzymes. However, whether Matrigel provides similar advantages in human hepatocytes remains largely uncertain.2. The influence of Matrigel-overlay on the constitutive and phenobarbital- and oltipraz-inducible expression of nine biotransformation enzymes, cytochrome P450s 1A1, 1A2, 2B6, 3A4, and glutathione S-transferases A1, A2, M1, T1, P1, in primary human hepatocytes was evaluated.3. Hepatocytes from five livers were maintained on a rigid collagen substratum with or without Matrigel overlay and treated for 48?h with two doses of each inducer. Quantitative RT-PCR, and for selected genes, immunoblot and enzyme activity analyses, demonstrated that human hepatocytes overlaid with Matrigel showed consistently higher constitutive and inducible expression of biotransformation genes. 4. Phenobarbital-mediated responsiveness of cytochrome P450 2B6, a potential indicator of hepatocyte differentiation status, was markedly higher in overlaid relative to non-overlaid hepatocytes. 5. It is concluded that an Matrigel overlay facilitates the maintenance and induction of xenobiotic metabolizing enzymes in primary cultures of human hepatocytes.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic , Hepatocytes/enzymology , Pharmaceutical Preparations/metabolism , Adult , Animals , Biotransformation , Collagen , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/metabolism , Drug Combinations , Enzyme Induction , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Laminin , Middle Aged , Proteoglycans , Rodentia
5.
Scand J Gastroenterol ; 39(6): 584-93, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15223685

ABSTRACT

BACKGROUND: For hepatocyte transplantation as well as experimental purposes, it would be advantageous to be able to expand human hepatocytes in vitro. However, under serum-free conditions, even with supplements of HGF (hepatic growth factor) and EGF (epidermal growth factor), proliferation of human hepatocytes is hampered. The aim of this study was to identify differences in the proliferative capacity of cultured primary human hepatocytes related to the age of the liver donors. METHODS: Proliferation was determined by BrdU-uptake, ploidy was measured using propidium iodide staining and flow cytometry, and the expression of cell cycle related proteins was determined by Western blotting. RESULTS: During the initial culture, juvenile hepatocytes proliferated better than adult hepatocytes. The proliferation rate declined to barely detectable levels after 8 days in culture in both juvenile and adult hepatocytes. The higher proliferative capacity of juvenile hepatocytes was associated with a larger fraction of diploid cells and a higher viability. The expression of regulatory cell cycle related proteins was higher in juvenile than in adult hepatocytes. CONCLUSIONS: The proliferation of human hepatocytes in vitro is critically related to a large fraction of diploid hepatocytes. The expression of regulatory cell cycle proteins reflects the proliferative capacity of cultured human hepatocytes. Juvenile as compared to adult human hepatocytes may be better suited for expansion in culture and could have a stronger repopulation capacity in vivo.


Subject(s)
Cell Division/physiology , Hepatocytes/physiology , Age Factors , Aged , Cell Culture Techniques , Child , Child, Preschool , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Diploidy , Female , Hepatocytes/transplantation , Humans , Male , Middle Aged , Polyploidy , Proliferating Cell Nuclear Antigen/metabolism
6.
Xenobiotica ; 34(7): 619-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15672752

ABSTRACT

1. The naturally occurring compounds curcumin (CUR), 3,3'-diindolylmethane (DIM), isoxanthohumol (IXN), 8-prenylnaringenin (8PN), phenethyl isothiocyanate (PEITC) and sulforaphane (SFN) protect animals against chemically induced tumours. Putative chemoprotective mechanisms include modulated expression of hepatic biotransformation enzymes. However, few, if any, studies have used human primary cells as test models. 2. The present study investigated the effects of these phytochemicals on the expression of four carcinogenesis-relevant enzymes--cytochrome P450 (CYP)1A1 and 1A2, NAD(P)H:quinone oxidoreductase (NQO1) and glutathione S-transferase A1 (GSTA1)--in primary cultures of freshly isolated human hepatocytes. 3. Quantitative RT-PCR analyses demonstrated that CYP1A1 was up-regulated by PEITC and DIM in a dose-dependent manner. CYP1A2 transcription was significantly activated following DIM, IXN, 8PN and PEITC treatments. DIM exhibited a remarkably effective induction response of CYP1A1 (474-, 239- and 87-fold at 50, 25 and 10 microM, respectively) and CYP1A2 (113-, 70- and 31-fold at 50, 25 and 10 microM, respectively), that was semiquantitatively reflected in protein levels. NQO1 expression responded to PEITC (11 x at 25 microM), DIM (4.5 x at 50 microM) and SFN (5 x at 10 microM) treatments. No significant effects on GSTA1 transcription were seen. 4. The findings show novel and unexpected effects of these phytochemicals on the expression of human hepatic biotransformation enzymes that play key roles in chemical-induced carcinogenesis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinogens/metabolism , Enzymes/genetics , Enzymes/metabolism , Hepatocytes/drug effects , Anticarcinogenic Agents/metabolism , Carrier Proteins/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Curcumin/metabolism , Curcumin/pharmacology , Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/drug effects , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Enzymes/drug effects , Flavanones/metabolism , Flavanones/pharmacology , Gene Expression Regulation/drug effects , Glutathione Transferase , Hepatocytes/physiology , Humans , Inactivation, Metabolic , Indoles/metabolism , Indoles/pharmacology , Isothiocyanates/metabolism , Isothiocyanates/pharmacology , NAD(P)H Dehydrogenase (Quinone)/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Plants/chemistry , Sulfoxides , Thiocyanates/metabolism , Thiocyanates/pharmacology
7.
Drug Metab Dispos ; 28(11): 1270-3, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11038151

ABSTRACT

Milk thistle extract is one of the most commonly used nontraditional therapies, particularly in Germany. Milk thistle is known to contain a number of flavonolignans. We evaluated the effect of silymarin, on the activity of various hepatic drug-metabolizing enzymes in human hepatocyte cultures. Treatment with silymarin (0.1 and 0.25 mM) significantly reduced the activity of CYP3A4 enzyme (by 50 and 100%, respectively) as determined by the formation of 6-beta-hydroxy testosterone and the activity of uridine diphosphoglucuronosyl transferase (UGT1A6/9) (by 65 and 100%, respectively) as measured by the formation of 4-methylumbelliferone glucuronide. Silymarin (0.5 mM) also significantly decreased mitochondrial respiration as determined by MTT reduction in human hepatocytes. These observations point to the potential of silymarin to impair hepatic metabolism of certain coadministered drugs in humans. Indiscriminate use of herbal products may lead to altered pharmacokinetics of certain drugs and may result in increased toxicity of certain drugs.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Glucuronosyltransferase/antagonists & inhibitors , Hepatocytes/drug effects , Mixed Function Oxygenases/antagonists & inhibitors , Plants, Medicinal , Silybum marianum/chemistry , Silymarin/pharmacology , Cells, Cultured , Cytochrome P-450 CYP3A , Humans
8.
Drug Metab Dispos ; 28(10): 1192-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10997939

ABSTRACT

In primary human and porcine hepatocyte cultures, we investigated the relationship between metabolism and cytotoxicity of troglitazone. Treatment of human hepatocytes for 2 h with 10, 20, 25, 35, and 50 microM troglitazone in protein-free medium resulted in concentration-dependent decreases in total protein synthesis. Decreases at 10 and 20 microM were reversible by 24 h, however protein synthesis did not recover at concentrations >/=25 microM. Troglitazone at 50 microM caused cellular death. In porcine hepatocytes, 100 microM troglitazone was lethal, whereas at 50 microM, protein synthesis completely recovered by 24 h. Recovery in protein synthesis was associated with metabolism of parent drug, whereas toxicity correlated (r(2) = 0.82) with accumulation of unmetabolized troglitazone. By 1 h, in human hepatocytes, troglitazone was metabolized to similar amounts of sulfate and quinone metabolites with little glucuronide detected. In contrast, porcine hepatocytes metabolized troglitazone to the similar amounts of glucuronide and the quinone metabolites with little sulfate detected. Exposure of human hepatocytes to a combination of 10 microM troglitazone and 10 microM 2,4-dichloro-4-nitrophenol resulted in a 70% decrease in protein synthesis, associated with 90% inhibition in the formation of troglitazone sulfate, a 4-fold increase in unmetabolized troglitazone, and no effect on formation of the quinone metabolite. Treatment with a combination of acetaminophen or phenobarbital with 20 microM troglitazone resulted in sustained decrease in protein synthesis associated with inhibition of sulfation and accumulation of troglitazone. These results suggest that inhibition of troglitazone sulfation may result in increased hepatotoxicity due to exposure to parent drug, or increased metabolism by alternate pathways.


Subject(s)
Chromans/pharmacology , Hepatocytes/drug effects , Thiazoles/pharmacology , Thiazolidinediones , Acetaminophen/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Nitrophenols/pharmacology , Pentachlorophenol/pharmacology , Phenobarbital/pharmacology , Protein Biosynthesis , Proteins/drug effects , Swine , Troglitazone
10.
Clin Nucl Med ; 25(6): 447-50, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10836694

ABSTRACT

PURPOSE: Hepatocyte transplantation is useful for ex vivo gene therapy and liver repopulation. Methods for hepatic reconstitution were recently developed, but hepatocyte transplantation systems must be optimized. The authors report their experience with In-111 oxyquinolone labeling of a test dose of hepatocytes (108 cells) for noninvasive assessment of the biodistribution of transplanted hepatocytes in a 5-year-old child with omithine transcarbamoylase deficiency. MATERIALS AND METHODS: Donor hepatocytes (approximately 108) were radiolabeled using a commercially available In-111 oxyquinolone solution (specific activity of 1 mCi/ml). RESULTS: The overall labeling efficiency was 36.4%. A final dose of approximately 290 ,uCi of the In-111-labeled hepatocytes in 10 ml serum-free phosphate-buffered saline was infused percutaneously into the portal vein approximately 2.5 hours after their preparation. The study was performed 3 hours before cell transplantation (109 cells). Quantitative analysis of the biodistribution of In-111-labeled hepatocytes indicated that cells were predominantly localized in the liver immediately after portal vein-infused transplantation. The predominant hepatic distribution was persistent for as long as 7 days after the procedure, with an average liver-to-spleen ratio of 9.5 to 1. No significant pulmonary radiotracer uptake was present. CONCLUSION: These results indicate that In-111 labeling of hepatocytes is useful for the short-term noninvasive analysis of the biodistribution of transplanted hepatocytes.


Subject(s)
Cell Transplantation/methods , Indium Radioisotopes , Liver/cytology , Portal Vein , Quinolones , Radiopharmaceuticals , Cell Movement , Child, Preschool , Follow-Up Studies , Humans , Infusions, Intravenous , Liver/diagnostic imaging , Lung/diagnostic imaging , Male , Ornithine Carbamoyltransferase Deficiency Disease/therapy , Radionuclide Imaging , Spleen/diagnostic imaging
11.
J Biol Chem ; 275(36): 28083-92, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10846185

ABSTRACT

By using specific anti-ACAT-1 antibodies in immunodepletion studies, we previously found that ACAT-1, a 50-kDa protein, plays a major catalytic role in the adult human liver, adrenal glands, macrophages, and kidneys but not in the intestine. Acyl-coenzyme A:cholesterol acyltransferase (ACAT) activity in the intestine may be largely derived from a different ACAT protein. To test this hypothesis, we produced specific polyclonal anti-ACAT-2 antibodies that quantitatively immunodepleted human ACAT-2, a 46-kDa protein expressed in Chinese hamster ovary cells. In hepatocyte-like HepG2 cells, ACAT-1 comprises 85-90% of the total ACAT activity, with the remainder attributed to ACAT-2. In adult intestines, most of the ACAT activity can be immunodepleted by anti-ACAT-2. ACAT-1 and ACAT-2 do not form hetero-oligomeric complexes. In differentiating intestinal enterocyte-like Caco-2 cells, ACAT-2 protein content increases by 5-10-fold in 6 days, whereas ACAT-1 protein content remains relatively constant. In the small intestine, ACAT-2 is concentrated at the apices of the villi, whereas ACAT-1 is uniformly distributed along the villus-crypt axis. In the human liver, ACAT-1 is present in both fetal and adult hepatocytes. In contrast, ACAT-2 is evident in fetal but not adult hepatocytes. Our results collectively suggest that in humans, ACAT-2 performs significant catalytic roles in the fetal liver and in intestinal enterocytes.


Subject(s)
Intestinal Mucosa/enzymology , Intestine, Small/enzymology , Liver/enzymology , Sterol O-Acyltransferase/analysis , Sterol O-Acyltransferase/metabolism , Adolescent , Adult , Aged , Animals , CHO Cells , Carcinoma, Hepatocellular , Child , Cloning, Molecular , Cricetinae , Humans , Intestinal Mucosa/embryology , Intestine, Small/embryology , Isoenzymes/analysis , Isoenzymes/metabolism , Kinetics , Liver/embryology , Liver Neoplasms , Middle Aged , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Tumor Cells, Cultured
12.
Biochem Biophys Res Commun ; 273(1): 333-41, 2000 Jun 24.
Article in English | MEDLINE | ID: mdl-10873607

ABSTRACT

Human hepatocytes cultured serum-free for up to 6 weeks were used to study expression and induction of enzymes and membrane transport proteins involved in drug metabolism. Phase I drug metabolizing enzymes cytochrome P450 (CYP)1A1, CYP1A2, CYP2C9, CYP2C19, CYP2E1, and CYP3A4 were detected by Western blot analyses and, when appropriate, by enzymatic assays for ethoxyresorufin-O-deethylase(EROD)-activity and testosterone-6beta-hydroxylase(T6H)-activity. Expression of the membrane transporter multi-drug resistance protein (P-glycoprotein, MDR-1), multidrug resistance-associated protein (MRP-1), and lung-resistance protein (LRP) was maintained during the culture as detected by RT-PCR and Western blot analyses. Model inducers like rifampicin, phenobarbital, or 3-methylcholanthrene and beta-naphtoflavone were able to induce CYP1A or CYP3A4 as well as EROD or T6H activities for up to 30 days. CYP2C9, CYP2C19 and CYP2E1 expression was maintained but not inducible for 48 days. Also, rifampicin and phenobarbital were unable to increase MDR-1 and MRP-1 protein levels significantly.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/biosynthesis , Liver/drug effects , Liver/enzymology , Steroid 16-alpha-Hydroxylase , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/genetics , Blotting, Western , Cell Size/drug effects , Cells, Cultured , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Enzyme Induction/drug effects , Epidermal Growth Factor/pharmacology , Hepatocyte Growth Factor/pharmacology , Humans , Isoenzymes/analysis , Isoenzymes/biosynthesis , Liver/cytology , Liver/ultrastructure , Methylcholanthrene/pharmacology , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/metabolism , Multidrug Resistance-Associated Proteins , Neoplasm Proteins/genetics , Phenobarbital/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rifampin/pharmacology , Steroid Hydroxylases/metabolism , Vault Ribonucleoprotein Particles/genetics , beta-Naphthoflavone/pharmacology
13.
Nat Med ; 6(5): 564-7, 2000 May.
Article in English | MEDLINE | ID: mdl-10802713

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been reported to induce apoptosis in various tumor cells but not in nontransformed, normal cells. Preclinical studies in mice and nonhuman primates have shown that administration of TRAIL can induce apoptosis in human tumors, but that no cytotoxicity to normal organs or tissues is found. The susceptibility of tumor cells to TRAIL and an apparent lack of activity in normal cells has lead to a proposal to use TRAIL in cancer therapy. Here, we assessed the sensitivity of hepatocytes from rat, mouse, rhesus monkey and human livers to TRAIL-induced apoptosis. TRAIL induced apoptosis in normal human hepatocytes in culture but not in hepatocytes isolated from the other species. Human hepatocytes showed characteristic features of apoptosis, including cytoplasmic shrinkage, the activation of caspases and DNA fragmentation. Apoptosis and cell death in human hepatocytes was massive and rapid, occurring in more than 60% of the cells exposed to TRAIL within 10 hours. These results indicate that there are species differences in sensitivity to TRAIL, and that substantial liver toxicity might result if TRAIL were used in human cancer therapy.


Subject(s)
Apoptosis , Liver/drug effects , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis Regulatory Proteins , Humans , Liver/cytology , Macaca mulatta , Mice , RNA, Messenger/analysis , Rats , Receptors, Tumor Necrosis Factor/isolation & purification , Species Specificity , TNF-Related Apoptosis-Inducing Ligand
15.
Biochem Biophys Res Commun ; 269(1): 46-53, 2000 Mar 05.
Article in English | MEDLINE | ID: mdl-10694475

ABSTRACT

Since human hepatocytes are available only in limited number, the development of a serum-free culture system for long-term cultivation of differentiated and functional hepatocytes is of great importance. Here we describe the culture of human hepatocytes in a chemically defined serum-free medium for up to 5 weeks. Cell morphology was assayed by light and electron microscopy and revealed a well-preserved cellular morphology. Marker proteins for epithelial and bile duct cells, cytokeratin (CK) 18 and 19, and liver-specific proteins, like phosphoenolpyruvate carboxykinase-2 (PCK2) and serum proteins, were expressed. Liver-enriched transcription factors CCAAT/enhancer binding protein alpha (C/EBPalpha) and hepatocyte nuclear factor-4 (HNF-4), cytokine and mitogen activated factors (nuclear factor kappa B) NFkappaB, and activator protein-1 (AP-1) were maintained and active for several weeks in our cultures. In summary, our serum-free culture system allows the culture of differentiated human hepatocytes for several weeks. It may serve as a model system for metabolic, pharmacologic-toxicologic studies, and studies on human pathogens under defined chemical conditions.


Subject(s)
Liver/cytology , Liver/physiology , Transcription Factors/metabolism , Blood Proteins/genetics , Blood Proteins/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free , Gene Expression , Humans , Liver/metabolism , Microscopy, Electron , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transcription Factors/genetics
16.
J Biol Chem ; 275(12): 8806-11, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10722725

ABSTRACT

In rat liver epithelial cells constitutively expressing transforming growth factor alpha (TGFalpha), c-Met is constitutively phosphorylated in the absence of its ligand, hepatocyte growth factor. We proposed that TGFalpha and the autocrine activation of its receptor, epidermal growth factor receptor (EGFR), leads to phosphorylation and activation of c-Met. We found that there is constitutive c-Met phosphorylation in human hepatoma cell lines and the human epidermoid carcinoma cell line, A431 which express TGFalpha, but not in normal human hepatocytes. Constitutive c-Met phosphorylation in A431, HepG2, AKN-1, and HuH6 cells was inhibited by neutralizing antibodies against TGFalpha and/or EGFR. Exposure to exogenous TGFalpha or EGF increased the phosphorylation of c-Met in the human epidermoid carcinoma cell line, A431. The increase of c-Met phosphorylation by TGFalpha in A431 cells was inhibited by neutralizing antibodies against TGFalpha and/or EGFR and by the EGFR-specific inhibitor tyrphostin AG1478. These results indicate that constitutive c-Met phosphorylation, and the increase of c-Met phosphorylation by TGFalpha or EGF, in tumor cell lines is the result of the activation via EGFR. We found that c-Met in tumor cells co-immunoprecipitates with EGFR regardless of the existence of their ligands in tumor cells, but not in normal human hepatocytes. We conclude that c-Met associates with EGFR in tumor cells, and this association facilitates the phosphorylation of c-Met in the absence of hepatocyte growth factor. This cross-talk between c-Met and EGFR may have significant implications for altered growth control in tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor Cross-Talk , Animals , Carcinoma, Hepatocellular , Carcinoma, Squamous Cell , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Hepatocyte Growth Factor/pharmacology , Humans , Liver Neoplasms , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinazolines , Rats , Transforming Growth Factor alpha/pharmacology , Tumor Cells, Cultured , Tyrphostins/pharmacology
17.
Biochem Biophys Res Commun ; 265(2): 376-81, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10558875

ABSTRACT

Long-term cultures of human hepatocytes were maintained serum-free in a chemically defined medium in the presence of HGF and EGF for up to 30 days. STAT 1alpha/1beta, STAT 3, and STAT 5 were present and tyrosine phosphorylated throughout the culture period in the cytosol as well as the nucleus. We show by co-immunoprecipitation that a portion of the cellular pools of STAT 1alpha/1beta and STAT 5 is physically associated with c-MET and EGF-receptor. Co-immunoprecipitation of STAT 3 with STAT 5 did occur in the cytosol but not in the nucleus, suggesting dimerization of the two STAT family members. The observed differences of protein amounts and tyrosine phosphorylation between cytosol and nucleus, the association of STAT proteins with EGF-receptor and c-MET and with each other may all be involved in regulating the activity of the STAT transcription factors. It is intriguing to speculate that STAT 5 may have a modulating role in the regulation of STAT 3 activity.


Subject(s)
DNA-Binding Proteins/metabolism , ErbB Receptors/metabolism , Liver/metabolism , Milk Proteins , Proto-Oncogene Proteins c-met/metabolism , Trans-Activators/metabolism , Animals , Cattle , Cell Nucleus/metabolism , Cells, Cultured , Culture Media, Serum-Free , Cytosol/metabolism , DNA-Binding Proteins/chemistry , Humans , Mice , Phosphorylation , Rabbits , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , Trans-Activators/chemistry , Tyrosine/chemistry
18.
J Hepatol ; 31(3): 435-42, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10488701

ABSTRACT

BACKGROUND/AIMS: CD14 has been identified as a receptor for LPS and is present both in a membrane-bound and a soluble form. Membrane CD14 (mCD14) is predominantly expressed on monocytes, macrophages and granulocytes. The source of soluble CD14 (sCD14) is as yet unclear. Previous investigation using monocytes has shown that sCD14 can be derived either from the shedding of mCD14 or from direct secretion by monocytes. Whether the monocyte is the sole or even the major source of sCD14 is as yet uncertain. Clearance of LPS from the bloodstream is thought to be primarily mediated by the liver. Production of CD14 by hepatocytes would potentially provide a powerful mechanism by which the liver could clear LPS, and therefore we examined the ability of human hepatocytes to produce CD14. METHODS: Human hepatocytes were isolated using collagenase perfusion. RESULTS: Human hepatocytes were found to have CD14 mRNA by Northern blot analysis. Western blot analysis and immunohistochemical staining confirmed CD14 protein in primary hepatocyte cultures. Further studies showed that a liver epithelial-like cell line AKN-1 is capable of producing CD14. Comparisons of the size of hepatocyte-derived CD14 protein with the sCD14 protein from the human monocytic leukemia cell line HL60 suggested that a slightly larger form of CD14 is expressed by human liver cells. CONCLUSION: This is the first study to demonstrate production of CD14 by human hepatocytes.


Subject(s)
Lipopolysaccharide Receptors/biosynthesis , Liver/immunology , Cell Line , Epithelial Cells/immunology , Humans , Lipopolysaccharides/blood , Liver/cytology , Metabolic Clearance Rate , Solubility
19.
Drug Metab Dispos ; 27(10): 1194-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10497147

ABSTRACT

Troglitazone (TRO) is an insulin sensitizer used in the treatment of type II diabetes. TRO is known to increase the activity of cytochrome P-450 (CYP) 3A in vivo. We have investigated the effect of TRO on CYP3A protein content and the activity of CYP3A (as measured by the formation of 6beta-hydroxytestosterone formation) in primary cultures of human hepatocytes in comparison with rifampicin (RIF). Hepatocytes were isolated from four human livers by perfusion with collagenase, plated on collagen-coated plates, and maintained in William's E medium. After 48 h in culture, cells were exposed to RIF (10 microM) or TRO (0-50 microM) twice, each over a period of 24 h, and the activity of CYP3A was measured. TRO increased the activity of CYP3A in a concentration-dependent manner, reaching a maximal response at 5 microM. Pretreatment of the hepatocytes with 10 microM TRO or 10 microM RIF resulted in a 4- to 15-fold increase in the activity of CYP3A. Maximum increase in CYP3A protein was observed at 5 microM TRO. There was a significant correlation (R(2) = 0.89) between the content of immunoreactive CYP3A protein in the hepatocytes and the rate of formation of 6beta-hydroxytestosterone. These results indicate that TRO is a potent inducer of CYP3A and is similar to RIF in inducing CYP3A in human hepatocytes. At concentrations of 25 microM and above, TRO was toxic to the cells, as determined by a decrease in the activity of CYP3A, a reduction in the amount of immunoreactive protein, and changes in the morphology of the cells.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Chromans/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Hypoglycemic Agents/pharmacology , Liver/enzymology , Oxidoreductases, N-Demethylating/biosynthesis , Thiazoles/pharmacology , Thiazolidinediones , Cells, Cultured , Cytochrome P-450 CYP3A , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Humans , Rifampin/pharmacology , Troglitazone
20.
In Vitro Cell Dev Biol Anim ; 35(4): 190-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10478798

ABSTRACT

The isolation and characterization of human liver cell lines are rather difficult due to limited material and poor growth in cell culture. In this report, we present the isolation, culture and characterization of a new epithelial-like liver cell line (AKN-1) with a heterogeneous cell population and many characteristics of the biliary epithelium. The AKN-1 cell line stained positively with antibodies to epithelial cytokeratin polypetides CK 8, 18, and 19. In addition, the cell line expressed the anti-human epithelial-related antigen (MOC-31), the human epithelial antigen (HEA), and the gamma-glutamyl transpeptidase, the hematopoietic growth factor, stem cell factor, and also its receptor, c-kit. The cell line failed to express albumin and factor 8 by immunohistochemistry. It did show, however, a twofold increase in 7-ethoxyresorufin-O-deethylase activity. Cytogenetic characterization revealed rare breakpoints in chromosome 2, which to our knowledge, have not yet been reported in liver cells.


Subject(s)
Cell Line , Epithelial Cells/cytology , Liver/cytology , Biomarkers , Cell Differentiation , Epithelial Cells/physiology , Humans , Karyotyping , Liver/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...