Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Hum Mol Genet ; 14(19): 2839-49, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16115815

ABSTRACT

Diseases caused by mutations in lamins A and C (laminopathies) suggest a crucial role for A-type lamins in different cellular processes. Laminopathies mostly affect tissues of mesenchymal origin. As transforming growth factor-beta1 (TGF-beta1) signalling impinges on the retinoblastoma protein (pRB) and SMADs, we tested the hypothesis that lamins modulate cellular responses to TGF-beta1 signalling, via the regulation of these transcription factors in mesenchymal cells. Here, we report that A-type lamins are essential for the inhibition of fibroblast proliferation by TGF-beta1. TGF-beta1 dephosphorylated pRB through PP2A, both of which, we show, are associated with lamin A/C. In addition, lamin A/C modulates the effect of TGF-beta1 on collagen production, a marker of mesenchymal differentiation. Our findings implicate lamin A/C in control of gene activity downstream of TGF-beta1, via nuclear phosphatases such as PP2A. This biological function provides a novel explanation for the observed mesenchymal dysfunction in laminopathies.


Subject(s)
Lamin Type A/physiology , Phosphoprotein Phosphatases/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Cell Proliferation , Cells, Cultured , Fibroblasts/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Mice , Mice, Mutant Strains , Phosphorylation , Signal Transduction , Transforming Growth Factor beta1
2.
Biochem Pharmacol ; 61(8): 991-8, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11286990

ABSTRACT

The underlying mechanism of the antiproliferative effect of S (simvastatin), a HMG-CoA reductase inhibitor, in vascular smooth muscle cells (SMC) is still poorly understood. In the present study, we used synchronized human SMC, isolated from left interior mammary artery, as an in vitro model to test the effects of S on platelet-derived growth factor (PDGF)-induced DNA synthesis, extracellular-regulated kinase 1/2 (ERK1/2), p38/stress-activated protein kinase 2 (SAPK2), RhoA and Rac1 activation. ERK1/2 phosphorylation was triggered within 2 min of PDGF stimulation (early G1 phase) and was blocked by PD98059, a specific inhibitor of the ERK1/2 pathway, which also strongly inhibited PDGF-induced DNA synthesis (IC(50) = 10 micromol/L). PDGF quickly induced p38 phosphorylation (early G1 phase) and SB203580, a specific inhibitor of the p38/SAPK2 pathway, also blocked PDGF-induced DNA synthesis (IC(50) = 0.3 micromol/L). Translocation to the plasma membrane of small GTPases, such as RhoA and Rac1, could not be detected within 15 min of stimulation with PDGF or lysophosphatidic acid (LPA) (early G1 phase), but occurred after 24 hr of PDGF stimulation (late G1/S phase). S inhibited PDGF-induced DNA synthesis (IC(50) = 3.5 micromol/L), and this effect was dependent on intracellular mevalonate, farnesyl pyrophosphate, and geranylgeranyl pyrophosphate availability. The critical time period for the reversal of the S effect by mevalonate comprised both the early and late G1 phase of the SMC cycle. PDGF-induced ERK1/2 phosphorylation and PDGF-induced p38 phosphorylation were not markedly affected by S during the whole G1 phase. However, S treatment blocked the PDGF- and LPA-induced membrane translocation of RhoA that occurred during the late G1/S phase. In the case of Rac1, the same process was also inhibited by S treatment. We concluded from these results that, in SMC, the early events associated with ERK1/2 and p38 signal transduction pathways, recruited for PDGF-mediated DNA synthesis, were insensitive to S action, whereas the mevalonate-dependent, posttranslational modification of RhoA and Rac1 molecules, required for PDGF-induced membrane translocation, was blocked by this drug. These results suggest that the antiproliferative effect of S can be explained not only by the blockage of RhoA-mediated signaling events but also by Rac1-mediated signaling events.


Subject(s)
Muscle, Smooth/drug effects , Platelet-Derived Growth Factor/pharmacology , Signal Transduction/drug effects , Simvastatin/pharmacology , Anticholesteremic Agents/pharmacology , Cells, Cultured , DNA/drug effects , DNA/metabolism , Drug Interactions , Enzyme Activation , Guanosine Triphosphate/metabolism , Humans , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth/enzymology , Muscle, Smooth/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
3.
Biochem Pharmacol ; 60(8): 1061-8, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11007942

ABSTRACT

Anchoring of small G-proteins to cellular membranes via a covalently bound lipophylic prenyl group is essential for the functioning of these proteins. For example, the farnesylation of Ras by the action of the enzyme protein:farnesyl transferase (PFT) is pivotal for its signalling function in cell growth and differentiation. The development of inhibitors of PFT was triggered by the role of mutated Ras in certain types of cancer and by the observation that non-farnesylated Ras is inactive. Besides the screening of existing compounds for PFT inhibition, rational drug design has also led to new inhibitors. Our research is in the field of atherosclerosis and concerns the development of inhibitors of the growth of vascular smooth muscle cells. The latter process gives rise to reocclusion of the coronary artery (restenosis) after balloon angioplasty. We and others have developed several analogues of the two substrates of PFT, i.e. farnesyl pyrophosphate (FPP) and the so-called CAAX peptide consensus sequence, which were tested in vitro for the inhibition of PFT and of other enzymes involved in protein prenylation, such as protein:geranylgeranyl transferase-1 (PGGT-1). The FPP analogue TR006, a strong inhibitor of PFT (IC(50) of 67 nM), blocked the proliferation of cultured human smooth muscle cells and inhibited platelet-derived growth factor- and basic fibroblast growth factor-induced DNA synthesis. Similar but more highly charged compounds failed in this respect, probably because of an impaired uptake in the cells. Less charged derivatives were designed to circumvent this problem. The effect on the GF-induced activation of intermediates in signal transduction pathways was investigated in order to gain insight into the mechanism of action within the cells. TR006 decreased the bFGF activation of extracellular signal-regulated kinase 1 (ERK1), suggesting its involvement in inhibiting Ras activity. Although other analogues inhibited DNA synthesis, they affected neither ERK1 activation nor p38/stress-activated protein kinase 2 or Jun N-terminal kinase 1 activation. Since some of these compounds were also shown to be inhibitors of in vitro PGGT-1 activity, the geranylgeranylation of other G-proteins may be decreased by these compounds. Rho seems to be a good candidate as a target for inhibitors of PGGT-1. This uncertainty as to the mechanism of action within non-transformed as well as transformed cells applies to all prenylation inhibitors, but is not holding back their further development as drugs. Their current and possible future application as therapeutics in cancer, restenosis, angiogenesis, and osteoporosis is briefly discussed.


Subject(s)
Arteriosclerosis/drug therapy , Enzyme Inhibitors/therapeutic use , GTP-Binding Proteins/metabolism , Neovascularization, Pathologic/drug therapy , ras Proteins/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Animals , Arteriosclerosis/enzymology , Arteriosclerosis/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/enzymology , Bone Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Humans , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Osteoporosis/drug therapy , Protein Prenylation/drug effects , ras Proteins/antagonists & inhibitors
4.
Biopharm Drug Dispos ; 21(9): 353-64, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11523064

ABSTRACT

Six 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (the present cholesterol-lowering drugs known as statins), lovastatin (L), simvastatin (S), pravastatin (P), fluvastatin (F), atorvastatin (A) and cerivastatin (C) are shown to be potent inhibitors of cholesterol synthesis in human hepatocytes, the target tissue for these drugs in man. All six inhibited in the nM range (IC(50) values: 0.2-8.0 nM). As daily used cholesterol-lowering drugs they are likely coadministered with other drugs. While several cytochrome P450 (CYP) enzymes are involved in drug metabolism in the liver and thus play an important role in drug-drug interaction it was investigated which of these enzymes are influenced by the active forms of the six statins. These enzyme activities were studied in human liver microsomal preparations, and in simian and human hepatocytes in primary culture. The following CYP reactions were used: nifedipine aromatization (CYP3A4), testosterone 6beta-hydroxylation (CYP3A4), tolbutamide methylhydroxylation (CYP2C9), S-mephenytoin 4-hydroxylation (CYP2C19), bufuralol 1'-hydroxylation (CYP2D6), aniline 4-hydroxylation (CYP2E1), coumarin 7-hydroxylation (CYP2A6) and 7-ethoxyresorufin O-dealkylation (CYP1A1/2). In the human liver microsomes the statins (concentrations up to 400 microM) did not influence the CYP1A1/2 activity and hardly the CYP2A6 and CYP2E1 activities. Except P, the other five statins were stronger inhibitors of the CYP2C19 activity with IC(50) values around 200 microM and the same holds for the effect of A, C and F on the CYP2D6 activity. L and S were weaker inhibitors of the latter enzyme activity, whereas P did not influence both activities. About the same was observed for the statin effect on CYP2C9 activity, except that F was a strong inhibitor of this activity (IC(50) value: 4 microM). Using the assay of testosterone 6beta-hydroxylation the CYP3A4 activity was decreased by L, S and F with IC(50) values of about 200 microM and a little more by C and A (IC(50) around 100 microM). P had hardly an effect on this activity. To a somewhat less extent the same trend was seen when CYP3A4 activity was measured using nifedipine as substrate. The inhibitory effects observed in microsomes were verified in suspension culture of freshly isolated hepatocytes from Cynomolgus monkey (as a readily available model) and of human hepatocytes. In general the same trends were seen as in the human microsomes, except that in some cases the inhibition of the CYP activity was less, possibly by the induction of the particular CYP enzyme by incubation of the cells with a particular statin. F remained a strong inhibitor of CYP2C9 activity in human and monkey hepatocytes. A induced the CYP2C9 in monkey hepatocytes but was an inhibitor of the CYP2C9 in human hepatocytes. A, S, L and C were moderate inhibitors in both cellular systems of CYP3A4. P was not affecting any of the CYP activities in the three systems studied. It is concluded that different CYP enzymes interact with different statins and therefore differences in between these drugs are to be expected when drug-drug interaction is considered.


Subject(s)
Cholesterol/biosynthesis , Cytochrome P-450 Enzyme System/drug effects , Hepatocytes/enzymology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Animals , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Hepatocytes/drug effects , Humans , Macaca fascicularis
5.
Biochem Pharmacol ; 57(4): 365-73, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9933024

ABSTRACT

In this study, it was investigated whether and how inhibitors of protein:farnesyl transferase (PFT) can inhibit the proliferation of human smooth muscle cells (HSMC) in culture. Several farnesyl pyrophosphate (FPP) analogues were synthesized and tested in vitro for their specificity in inhibiting squalene synthase (SS), PFT, or protein:geranylgeranyl transferase-1 (PGGT-1) activities (the latter was determined using a newly designed assay). One of these compounds appeared to be a strong PFT inhibitor (IC50 value: 340 nM) and a weak inhibitor in the other two enzyme assays. This compound (designated as TR006) inhibited the farnesylation of Ras in a Ha-ras transfected cell line (Cohen et al., Biochem. Phamacol. 49: 839-845, 1995) and concomitantly slowed down the growth of these cells. Twenty-five microM of TR006 inhibited the proliferation of HSMC isolated from left internal mammary artery, as measured by counting the cells over a period of three cell cycles (10 days). A structurally related compound (TR007), a specific SS inhibitor, did not influence HSMC proliferation under the same conditions. The inhibition by TR006 was concentration-dependent. In HSMC, synchronized by serum depletion, platelet-derived growth factor (PDGF) or basic fibroblast growth factor (bFGF)-induced DNA synthesis was decreased by a 29-hr pretreatment with 100 microM of TR006, indicating that this inhibitor acted in an early phase of the cell cycle, probably by preventing protein isoprenylation. Some other FPP analogues with comparable IC50 values in the in vitro PFT assay were also able to decrease bFGF-induced DNA synthesis without affecting cell viability. A more negatively charged member of this group, TR018, did not influence the growth factor-induced DNA synthesis, probably due to an impaired uptake into the cells. However, the pivaloyloxomethyl derivative of this compound, which is uncharged, and is thought to be converted into TR018 within the cells, showed a strong decrease in bFGF-induced DNA synthesis in HSMC. These data suggest that the compounds investigated may be developed further for treatment of conditions in which undesirable proliferation of smooth muscle cells plays an important role.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Muscle, Smooth/drug effects , Polyisoprenyl Phosphates/pharmacology , Animals , Cell Division/drug effects , Cell Line , Cells, Cultured , Enzyme Inhibitors/pharmacology , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Fibroblast Growth Factor 2/pharmacology , Humans , Platelet-Derived Growth Factor/pharmacology , Rats , Sesquiterpenes
6.
Biochim Biophys Acta ; 1345(3): 259-68, 1997 Apr 21.
Article in English | MEDLINE | ID: mdl-9150246

ABSTRACT

The effects of 6 HMG-CoA reductase inhibitors: pravastatin, lovastatin, simvastatin, atorvastatin, fluvastatin and cerivastatin were analyzed in cultured human smooth muscle cells, fibroblasts, endothelial cells and myoblasts. In vascular smooth muscle cells, pravastatin was a much weaker inhibitor of cholesterol synthesis than the 5 other drugs which displayed equally strong inhibitory potency. The anti-proliferative effects of these 6 drugs were analyzed by measuring cell number and mitochondrial dehydrogenase activity (MTT assay) after 3 days of incubation. IC25 values for inhibition of proliferation were very similar among the 4 cell types and were in the following order of magnitude: pravastatin << lovastatin = simvastatin = atorvastatin = fluvastatin << cerivastatin. Only in the case of pravastatin was proliferation inhibited at lower concentration in smooth muscle cells than in the other cell types. Proliferation was also assessed by measuring DNA synthesis in these cells. A 3 day-incubation with 1 microM of pravastatin had no effect on this parameter in all 4 cell types. However, 1 microM of simvastatin or lovastatin caused either an inhibition (in smooth muscle cells and endothelial cells) or stimulation (in fibroblasts) of this process. The effects of simvastatin on cell number, mitochondrial dehydrogenase activity and DNA synthesis were counteracted by simultaneous mevalonate addition. Simvastatin treatment was also associated with a change in the post-translational modification of the ras protein in smooth muscle cells, probably by inhibition of its farnesylation. Moreover, simvastatin treatment blocked the PDGF and bFGF-induced DNA synthesis in synchronized smooth muscle cells, whereas it does not affect the fetal calf serum-induced DNA synthesis in synchronized fibroblasts, suggesting that simvastatin blocks various steps of the cell cycle and that this effect depends on the cell type and the growth signalling pathway activated.


Subject(s)
Anticholesteremic Agents/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Lovastatin/analogs & derivatives , Muscle, Smooth, Vascular/drug effects , Pravastatin/pharmacology , Cell Division/drug effects , Cells, Cultured , Cholesterol/biosynthesis , DNA/biosynthesis , Enzyme Inhibitors/pharmacology , Humans , Lovastatin/antagonists & inhibitors , Lovastatin/pharmacology , Simvastatin , ras Proteins/metabolism
7.
J Nutr ; 125(10): 2631-9, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7562100

ABSTRACT

Restriction of energy intake (ER), without malnutrition of essential nutrients, has repeatedly been demonstrated to increase longevity in rodents. In the antioxidant theory of aging the lack of balance between the generation of free radicals and free radical scavenging was thought to be a main causal agent, in the aging process. From this point of view the antiaging effect induced by ER might be due to the lower rate of free-radical production and related damage induced by a lower metabolic rate. The antiaging effects of ER might also occur in humans. This study explored the effects of a 10-week moderately energy-restricted diet (80% of habitual) in 24 non-obese middle-aged men (16 ER subjects, 8 controls) on resting metabolic rate (RMR) and indicators of the primary antioxidant defense system, oxidative stress and genotoxicity. RMR decreased significantly in both groups, even when adjustments were made for the change in body composition. The increase in blood vitamin C concentration correlated with the increase in urinary 8-hydroxydeoxyguanosine (80HdG) excretion. The change in urinary 80HdG excretion also correlated with the change in RMR per kg fat-free mass. No differences between groups were found for changes in indicators of genotoxicity, erythrocyte catalase, glutathione peroxidase and superoxide dismutase activity and in plasma vitamin E, A or beta-carotene concentrations. We conclude that 10 weeks of moderate ER did not affect indicators of antioxidative capacity, oxidative stress and genotoxicity of humans. Since subjects were not in energy balance at the end of the study, no conclusions can be made with respect to long-term effects.


Subject(s)
Energy Intake/physiology , Energy Metabolism/physiology , Oxidative Stress/physiology , 8-Hydroxy-2'-Deoxyguanosine , Adult , Ascorbic Acid/blood , Carotenoids/blood , Catalase/analysis , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/urine , Erythrocytes/enzymology , Free Radical Scavengers/metabolism , Glutathione Peroxidase/analysis , Humans , Male , Middle Aged , Superoxide Dismutase/analysis , Time Factors , Vitamin E/blood , beta Carotene
8.
Biochem Pharmacol ; 49(6): 839-45, 1995 Mar 15.
Article in English | MEDLINE | ID: mdl-7702642

ABSTRACT

The inhibitory potency of farnesyl pyrophosphate analogues was investigated on two farnesyl pyrophosphate-consuming enzymes: squalene synthase, a secondary regulation site in the cholesterol synthesis pathway, and protein:farnesyl transferase, which plays a role in the function of Ras-proteins. For the transferase determination a rapid in vitro assay, using Sepharose-bound Ras-peptides, was developed. The distinct farnesyl pyrophosphate analogues showed a different order of potency in the inhibition of these two enzymes. Using the farnesyl transferase assay with pre-p21Ha-ras as substrate the same result was obtained. The difference observed in the in vitro assays was also reflected in the inhibition of cholesterol synthesis, protein prenylation in general and Ha-ras farnesylation in Rat-1.H-ras13 cells, a rat fibroblast cell line that overproduces human p21Ha-ras. This work shows that farnesyl pyrophosphate analogues can be developed for specific inhibition of different processes such as cholesterol synthesis and protein prenylation.


Subject(s)
Alkyl and Aryl Transferases , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Polyisoprenyl Phosphates/pharmacology , Transferases/antagonists & inhibitors , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Microsomes, Liver/enzymology , Molecular Sequence Data , Oncogene Protein p21(ras)/biosynthesis , Oncogene Protein p21(ras)/metabolism , Polyisoprenyl Phosphates/chemistry , Rats , Sesquiterpenes
9.
Liver ; 15(1): 1-8, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7776851

ABSTRACT

Earlier data from experiments in rats have shown that administration of retinyl esters (vitamin A) strongly influences the effects of CCl4 on the liver. The accumulation of collagen was inhibited, but an increase in CCl4-toxicity with high mortality was observed. The present study was conducted to examine the effects of beta-carotene (provitamin A) on CCl4-related general and hepatic toxicity in rats. Oral administration of beta-carotene during CCl4-treatment resulted, biochemically, in a significantly lower increase in the hydroxyproline liver content and, histopathologically, in less severe liver fibrosis as compared with the liver of rats not treated with beta-carotene. The study also showed that beta-carotene administration could prevent the long-term loss of retinoids from the CCl4-injured liver. No significant toxic effects of beta-carotene, as previously found with retinyl esters (vitamin A), were observed. This experimental study suggests that beta-carotene has the therapeutic potential to decrease the severity of liver fibrosis without marked toxicity.


Subject(s)
Carbon Tetrachloride Poisoning/pathology , Carotenoids/pharmacology , Chemical and Drug Induced Liver Injury/pathology , Liver Cirrhosis, Experimental/pathology , Animals , Collagen/metabolism , Female , Hydroxyproline/metabolism , Liver/drug effects , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Function Tests , Rats , Rats, Inbred BN , beta Carotene
10.
J Hepatol ; 21(3): 394-402, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7836710

ABSTRACT

Rats were malnourished for 12 months with a highly inadequate fat-rich, calorie-sufficient but otherwise poly-deficient liquid diet composed of mashed potatoes with mayonnaise, comparable with the nutritional intake of many chronic alcoholics. When alcohol was incorporated into this diet, administered as whisky in drinking water available ad libitum, the livers of all eight rats showed increased fibrosis and cirrhosis as compared to the livers of the eight non-alcohol-treated, isocalorically fed, paired control rats. Alcohol-treated rats developed fibrosis and cirrhosis on a dietary fat content of 38% of total caloric intake and low blood alcohol levels, ranging from 50 to 126 mg/dl, due to gradual intake over the day and to low absolute intake (mean 11.9 +/- 0.6 g/kg per day). None of the rats died spontaneously. Malnutrition is likely to be an important factor in the development of the fibrosis of alcoholic liver disease, and this rat model may be used to study aspects of the pathogenesis.


Subject(s)
Alcoholism/complications , Liver Cirrhosis, Alcoholic/etiology , Liver Cirrhosis, Experimental/etiology , Nutrition Disorders/complications , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cholesterol/blood , Dietary Fats/administration & dosage , Disease Models, Animal , Ethanol/adverse effects , Ethanol/blood , Food, Formulated , Liver/metabolism , Liver/pathology , Liver/ultrastructure , Liver Cirrhosis, Alcoholic/blood , Liver Cirrhosis, Alcoholic/pathology , Liver Cirrhosis, Experimental/blood , Liver Cirrhosis, Experimental/pathology , Male , Microscopy, Electron , Rats , Triglycerides/blood , gamma-Glutamyltransferase/blood
11.
Hepatology ; 19(1): 193-201, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8276355

ABSTRACT

Earlier studies have shown that retinoid administration suppresses the generation of hepatic fibrosis and stimulates its regression in normal (i.e., vitamin A-sufficient) carbon tetrachloride-treated rats. This study focuses on the possible role of a marginal or deficient vitamin A status on carbon tetrachloride-induced fibrosis. This experimental study in rats shows that vitamin A status, reflected by hepatic retinoid content (retinol and retinyl esters), modulates the development of hepatic fibrosis induced by carbon tetrachloride. In rats with low hepatic retinoid levels (12 +/- 0.9 micrograms/gm liver), carbon tetrachloride-induced liver fibrosis was more pronounced than in rats with sufficient hepatic retinoid levels (1,065 +/- 327 micrograms/gm liver). Enhanced liver fibrogenesis was confirmed both morphologically and by a higher hydroxyproline content of the liver. It was associated with a reduced liver weight and the development of parenchymal regeneration nodules. Furthermore, carbon tetrachloride treatment itself reduced the hepatic retinoid content in rats independently of the liver vitamin A status before treatment and increased serum retinol levels in vitamin A-sufficient rats. The results show that the vitamin A status of the liver plays an important role in hepatic fibrogenesis. Low hepatic vitamin A levels, which can be the result not only of low dietary intake but also of interference with vitamin A metabolism by agents such as ethanol and carbon tetrachloride, may be a risk factor for the development of liver fibrosis. We suggest that retinoids modulate collagen synthesis and deposition irrespective of the degree of hepatocellular necrosis induced by carbon tetrachloride.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Liver Cirrhosis, Experimental/metabolism , Vitamin A Deficiency/complications , Animals , Carbon Tetrachloride , Collagen/metabolism , Female , Hydroxyproline/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/pathology , Rats , Rats, Inbred Strains , Retinoids/metabolism , Specific Pathogen-Free Organisms , Vitamin A/blood , Vitamin A Deficiency/metabolism
12.
J Hepatol ; 13(2): 249-55, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1744428

ABSTRACT

The inability of the 'ethanol/high vitamin A Lieber-DeCarli diet' to induce liver fibrosis in two different rat strains was further evaluated by determining changes in parameters of liver cell damage and of retinoid and lipid metabolism. In the ethanol/vitamin A-treated group, slight but constant hepatic cell damage, as indicated by elevated alanine aminotransferase, aspartate aminotransferase and glutamate dehydrogenase activities in blood, was already observed at 6 months and maintained until the time of death at 16 months. Serum gamma-glutamyl transaminase activities were not raised. Moderate parenchymal liver cell damage was not accompanied by fibrosis. Hypertriglyceridemia or hypercholesterolemia were observed at 6-16 months of chronic alcohol administration. This response was strain dependent. In ethanol-treated rats of both strains, total liver retinoids and serum retinol concentrations were not altered. Therefore, the hypothesis that interaction between alcohol and retinoids is a major factor in the pathogenesis of alcoholic liver disease, needs to be reconsidered.


Subject(s)
Diet , Disease Models, Animal , Ethanol/administration & dosage , Liver Cirrhosis, Alcoholic/etiology , Vitamin A/administration & dosage , Animals , Ethanol/toxicity , Lipid Metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/pathology , Male , Rats , Rats, Inbred BN , Rats, Inbred Strains , Vitamin A/blood , Vitamin A/toxicity
13.
Clin Chim Acta ; 177(2): 141-6, 1988 Oct 14.
Article in English | MEDLINE | ID: mdl-3233763

ABSTRACT

Procollagen type III N-peptide (PIII NP) and laminin P1 fragment (LP1) have both been proposed as markers of liver fibrosis. In this study we evaluated the diagnostic application of both peptides in alcoholic liver disease and primary biliary cirrhosis. Serum concentrations of the peptides were measured by radioimmunoassay. PIII NP and LP1 levels appeared to be significantly raised in patients with alcoholic and primary biliary cirrhosis. Patients with alcohol abuse without cirrhosis had normal or slightly elevated PIII NP levels, but significantly raised LP1 levels. There was a strong correlation between PIII NP and LP1 concentrations.


Subject(s)
Laminin/blood , Liver Cirrhosis, Alcoholic/diagnosis , Liver Cirrhosis, Biliary/diagnosis , Peptide Fragments/blood , Procollagen/blood , Humans , Radioimmunoassay
15.
Mech Ageing Dev ; 33(3): 305-12, 1986 Feb.
Article in English | MEDLINE | ID: mdl-3713267

ABSTRACT

Rates of fluid-phase endocytosis were determined in several organs and tissues of female WAG/Rij rats of several ages by using 125I-labelled polyvinylpyrrolidone ([125I]-PVP) as a marker. Liver, muscle and skin accounted for a high level of [125I]PVP uptake 28 h after injection. When PVP uptake was expressed per gram of organ/tissue, the liver and spleen were found to be the most active. An age-related increase in [125I]PVP uptake was seen at between 12 and 36 months of age in liver, kidneys and heart. Except for the kidneys this increase is caused by an increase in wet weight of these organs and not by an increase in the specific endocytic rate. These data, together with reported findings on the major sites of albumin catabolism, in liver, kidney, spleen and intestine, indicate that fluid-phase endocytosis is a main mechanism for the observed age-related increase in albumin elimination in these rats.


Subject(s)
Aging , Endocytosis , Povidone/metabolism , Serum Albumin/metabolism , Animals , Female , Metabolic Clearance Rate , Povidone/urine , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...