Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
World J Hepatol ; 4(5): 176-83, 2012 May 27.
Article in English | MEDLINE | ID: mdl-22662286

ABSTRACT

AIM: To optimize a xeno-free cryopreservation protocol for primary human hepatocytes. METHODS: The demand for cryopreserved hepatocytes is increasing for both clinical and research purposes. Despite several hepatocyte cryopreservation protocols being available, improvements are urgently needed. We first compared controlled rate freezing to polystyrene box freezing and did not find any significant change between the groups. Using the polystyrene box freezing, we compared two xeno-free freezing solutions for freezing of primary human hepatocytes: a new medium (STEM-CELLBANKER, CB), which contains dimethylsulphoxide (DMSO) and anhydrous dextrose, both permeating and non-permeating cryoprotectants, and the frequently used DMSO - University of Wisconsin (DMSO-UW) medium. The viability of the hepatocytes was assessed by the trypan blue exclusion method as well as a calcein-esterase based live-dead assay before and after cryopreservation. The function of the hepatocytes was evaluated before and after cryopreservation by assessing enzymatic activity of 6 major cytochrome P450 isoforms (CYPs): CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and CYP3A7. RESULTS: The new cryoprotectant combination preserved hepatocyte viability significantly better than the standard DMSO-UW protocol (P < 0.01). There was no significant difference in viability estimation between both the trypan blue (TB) and the Live-Dead Assay methods. There was a correlation between viability of fresh hepatocytes and the difference in cell viability between CB and DMSO protocols (r(2) = 0.69) using the TB method. However, due to high within-group variability in the activities of the major CYPs, any statistical between-group differences were precluded. Cryopreservation of human hepatocytes using the cryoprotectant combination was a simple and xeno-free procedure yielding better hepatocyte viability. Thus, it may be a better alternative to the standard DMSO-UW protocol. Estimating CYP activities did not seem to be a relevant way to compare hepatocyte function between different groups due to high normal variability between different liver samples. CONCLUSION: The cryoprotectant combination may be a better alternative to the standard DMSO-UW protocol in primary human hepatocyte cryopreservation.

2.
Hepatology ; 53(3): 996-1006, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21319191

ABSTRACT

UNLABELLED: We reported previously that mice overexpressing cytochrome P450 7a1 (Cyp7a1; Cyp7a1-tg mice) are protected against high fat diet-induced hypercholesterolemia, obesity, and insulin resistance. Here, we investigated the underlying mechanism of bile acid signaling in maintaining cholesterol homeostasis in Cyp7a1-tg mice. Cyp7a1-tg mice had two-fold higher Cyp7a1 activity and bile acid pool than did wild-type mice. Gallbladder bile acid composition changed from predominantly cholic acid (57%) in wild-type to chenodeoxycholic acid (54%) in Cyp7a1-tg mice. Cyp7a1-tg mice had higher biliary and fecal cholesterol and bile acid secretion rates than did wild-type mice. Surprisingly, hepatic de novo cholesterol synthesis was markedly induced in Cyp7a1-tg mice but intestine fractional cholesterol absorption in Cyp7a1-tg mice remained the same as wild-type mice despite the presence of increased intestine bile acids. Interestingly, hepatic but not intestinal expression of several cholesterol (adenosine triphosphate-binding cassette G5/G8 [ABCG5/G8], scavenger receptor class B, member 1) and bile acid (ABCB11) transporters were significantly induced in Cyp7a1-tg mice. Treatment of mouse or human hepatocytes with a farnesoid X receptor (FXR) agonist GW4064 or bile acids induced hepatic Abcg5/g8 expression. A functional FXR binding site was identified in the Abcg5 gene promoter. Study of tissue-specific Fxr knockout mice demonstrated that loss of the Fxr gene in the liver attenuated bile acid induction of hepatic Abcg5/g8 and gallbladder cholesterol content, suggesting a role of FXR in the regulation of cholesterol transport. CONCLUSION: This study revealed a new mechanism by which increased Cyp7a1 activity expands the hydrophobic bile acid pool, stimulating hepatic cholesterol synthesis and biliary cholesterol secretion without increasing intestinal cholesterol absorption. This study demonstrated that Cyp7a1 plays a critical role in maintaining cholesterol homeostasis and underscores the importance of bile acid signaling in regulating overall cholesterol homeostasis.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol 7-alpha-Hydroxylase/biosynthesis , Cholesterol/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/metabolism , Animals , Hepatocytes/drug effects , Homeostasis , Humans , Isoxazoles/pharmacology , Lipoproteins/metabolism , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/agonists
3.
Methods Mol Biol ; 640: 417-30, 2010.
Article in English | MEDLINE | ID: mdl-20645065

ABSTRACT

De novo synthesis of bile acids is a liver-specific function that is difficult to maintain in cultured cells. There are significant species differences in both types of bile acids formed and more importantly in the regulation of bile acid homeostasis. This highlights the need for a good human in vitro model. Isolated primary human hepatocytes have the capacity to synthesize normal conjugated bile acids at a rate similar to that in vivo. In this chapter we describe the importance of different culture conditions such as choice of substrate, media and supplements on the total bile acid production as wells as the bile acid composition.


Subject(s)
Bile Acids and Salts/metabolism , Hepatocytes/metabolism , Animals , Cell Culture Techniques/methods , Cells, Cultured , Collagen/isolation & purification , Collagen/metabolism , Drug Combinations , Hepatocytes/cytology , Humans , Laminin/isolation & purification , Laminin/metabolism , Liver/cytology , Liver/metabolism , Mice , Models, Biological , Proteoglycans/isolation & purification , Proteoglycans/metabolism , Rats
4.
Genes Dev ; 24(4): 381-95, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20159957

ABSTRACT

The orphan receptor LRH-1 and the oxysterol receptors LXRalpha and LXRbeta are established transcriptional regulators of lipid metabolism that appear to control inflammatory processes. Here, we investigate the anti-inflammatory actions of these nuclear receptors in the hepatic acute phase response (APR). We report that selective synthetic agonists induce SUMOylation-dependent recruitment of either LRH-1 or LXR to hepatic APR promoters and prevent the clearance of the N-CoR corepressor complex upon cytokine stimulation. Investigations of the APR in vivo, using LXR knockout mice, indicate that the anti-inflammatory actions of LXR agonists are triggered selectively by the LXRbeta subtype. We further find that hepatic APR responses in small ubiquitin-like modifier-1 (SUMO-1) knockout mice are increased, which is due in part to diminished LRH-1 action at APR promoters. Finally, we provide evidence that the metabolically important coregulator GPS2 functions as a hitherto unrecognized transrepression mediator of interactions between SUMOylated nuclear receptors and the N-CoR corepressor complex. Our study extends the knowledge of anti-inflammatory mechanisms and pathways directed by metabolic nuclear receptor-corepressor networks to the control of the hepatic APR, and implies alternative pharmacological strategies for the treatment of human metabolic diseases associated with inflammation.


Subject(s)
Acute-Phase Reaction/immunology , Intracellular Signaling Peptides and Proteins/immunology , Liver/immunology , Orphan Nuclear Receptors/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Small Ubiquitin-Related Modifier Proteins/immunology , Animals , Anti-Inflammatory Agents/immunology , COS Cells , Chlorocebus aethiops , Female , Gene Expression Regulation , HeLa Cells , Humans , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Biochem Biophys Res Commun ; 364(4): 743-7, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-17976534

ABSTRACT

The conversion of cholesterol to bile acids is a key pathway for elimination of cholesterol from the body, thereby reducing the risk of arteriosclerosis. Moderate consumption of ethanol has been shown to have preventive effects on cardiovascular disease and decrease the risk of gallstone formation. In the present study primary human hepatocytes were used to investigate if ethanol affected bile acid synthesis. Hepatocytes were prepared from donor liver (n=11) and treated with ethanol, 7.7 or 50 mM, for 24 h. mRNA levels for enzymes in bile acid synthesis pathways were studied and bile acid synthesis was analyzed. Treatment with 7.7 mM ethanol increased cholic acid synthesis by 20% and treatment with 50 mM ethanol up-regulated cholic acid formation by 60%. The synthesis of cholic acid increased more than that of chenodeoxycholic acid, indicating that the classical pathway for bile acid synthesis was up-regulated. Increased bile acid levels in the cells treated with ethanol were seen after approximately 20 h. mRNA expression of CYP7A1, CYP27A1, and CYP8B1 in the hepatocytes was not affected by alcohol exposure.


Subject(s)
Bile Acids and Salts/biosynthesis , Ethanol/administration & dosage , Hepatocytes/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Humans , Up-Regulation/drug effects
6.
Proc Natl Acad Sci U S A ; 104(40): 15665-70, 2007 Oct 02.
Article in English | MEDLINE | ID: mdl-17895379

ABSTRACT

Coordinated regulation of bile acid biosynthesis, the predominant pathway for hepatic cholesterol catabolism, is mediated by few key nuclear receptors including the orphan receptors liver receptor homolog 1 (LRH-1), hepatocyte nuclear factor 4alpha (HNF4alpha), small heterodimer partner (SHP), and the bile acid receptor FXR (farnesoid X receptor). Activation of FXR initiates a feedback regulatory loop via induction of SHP, which suppresses LRH-1- and HNF4alpha-dependent expression of cholesterol 7alpha hydroxylase (CYP7A1) and sterol 12alpha hydroxylase (CYP8B1), the two major pathway enzymes. Here we dissect the transcriptional network governing bile acid biosynthesis in human liver by identifying GPS2, a stoichiometric subunit of a conserved corepressor complex, as a differential coregulator of CYP7A1 and CYP8B1 expression. Direct interactions of GPS2 with SHP, LRH-1, HNF4alpha, and FXR indicate alternative coregulator recruitment strategies to cause differential transcriptional outcomes. In addition, species-specific differences in the regulation of bile acid biosynthesis were uncovered by identifying human CYP8B1 as a direct FXR target gene, which has implications for therapeutic approaches in bile acid-related human disorders.


Subject(s)
Bile Acids and Salts/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Transcription, Genetic , Carcinoma, Hepatocellular , Cell Line, Tumor , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Humans , Liver Neoplasms , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Steroid 12-alpha-Hydroxylase/genetics , Steroid 12-alpha-Hydroxylase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Biochem Biophys Res Commun ; 360(2): 437-40, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17601491

ABSTRACT

The metabolic regulator fibroblast growth factor 21 (FGF21) has antidiabetic properties in animal models of diabetes and obesity. Using quantitative RT-PCR, we here show that the hepatic gene expression of FGF21 is regulated by the peroxisome proliferator-activated receptor alpha (PPARalpha). Fasting or treatment of mice with the PPARalpha agonist Wy-14,643 induced FGF21 mRNA by 10-fold and 8-fold, respectively. In contrast, FGF21 mRNA was low in PPARalpha deficient mice, and fasting or treatment with Wy-14,643 did not induce FGF21. Obese ob/ob mice, known to have increased PPARalpha levels, displayed 12-fold increased hepatic FGF21 mRNA levels. The potential importance of PPARalpha for FGF21 expression also in human liver was shown by Wy-14,643 induction of FGF21 mRNA in human primary hepatocytes, and PPARalpha response elements were identified in both the human and mouse FGF21 promoters. Further studies on the mechanisms of regulation of FGF21 by PPARalpha in humans will be of great interest.


Subject(s)
Fibroblast Growth Factors/metabolism , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , PPAR alpha/metabolism , Animals , Humans , Male , Mice , Mice, Knockout , Tumor Cells, Cultured
8.
Mol Pharmacol ; 72(4): 947-55, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17628011

ABSTRACT

The liver is central to the maintenance of glucose and lipid homeostasis, and liver X receptors (LXRs) are key regulators of expression of the genes involved. So far, effects of activation of LXR in human hepatocytes have not been well characterized. Here we show that treatment of primary human hepatocytes with the synthetic LXR ligand 3-[3-[N-(2-chloro-3-trifluoromethylbenzyl)-(2,2-diphenylethyl)amino]propyloxy]phenylacetic acid hydrochloride (GW3965) results in reduced output of bile acids and very low density lipoprotein triglycerides and induced expression of adipose differentiation-related protein accompanied by increased lipid storage. Genome wide-expression profiling identified novel human LXR target genes in the glycolytic and lipogenic pathways and indicated that LXR activation reduced hepatic insulin sensitivity. Comparative experiments showed significant differences in the response to GW3965 between human and rat hepatocytes, raising the question as to how well rodent models reflect the human situation. In summary, the risk of hepatic steatosis upon pharmaceutical targeting of LXR may be a particularly serious consequence in humans.


Subject(s)
Benzoates/pharmacology , Benzylamines/pharmacology , DNA-Binding Proteins/agonists , Hepatocytes/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Female , Hepatocytes/metabolism , Humans , Insulin/pharmacology , Ligands , Liver X Receptors , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Orphan Nuclear Receptors , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Species Specificity
9.
J Clin Invest ; 117(7): 1968-78, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17571164

ABSTRACT

Niemann-Pick C1-like 1 (NPC1L1) is required for cholesterol absorption. Intestinal NPC1L1 appears to be a target of ezetimibe, a cholesterol absorption inhibitor that effectively lowers plasma LDL-cholesterol in humans. However, human liver also expresses NPC1L1. Hepatic function of NPC1L1 was previously unknown, but we recently discovered that NPC1L1 localizes to the canalicular membrane of primate hepatocytes and that NPC1L1 facilitates cholesterol uptake in hepatoma cells. Based upon these findings, we hypothesized that hepatic NPC1L1 allows the retention of biliary cholesterol by hepatocytes and that ezetimibe disrupts hepatic function of NPC1L1. To test this hypothesis, transgenic mice expressing human NPC1L1 in hepatocytes (L1-Tg mice) were created. Hepatic overexpression of NPC1L1 resulted in a 10- to 20-fold decrease in biliary cholesterol concentration, but not phospholipid and bile acid concentrations. This decrease was associated with a 30%-60% increase in plasma cholesterol, mainly because of the accumulation of apoE-rich HDL. Biliary and plasma cholesterol concentrations in these animals were virtually returned to normal with ezetimibe treatment. These findings suggest that in humans, ezetimibe may reduce plasma cholesterol by inhibiting NPC1L1 function in both intestine and liver, and hepatic NPC1L1 may have evolved to protect the body from excessive biliary loss of cholesterol.


Subject(s)
Azetidines/pharmacology , Bile/drug effects , Bile/metabolism , Cholesterol/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoproteins E/blood , Bile Acids and Salts/metabolism , Cell Membrane/metabolism , Cholesterol/blood , Ezetimibe , Humans , Liver/metabolism , Male , Membrane Proteins/genetics , Membrane Transport Proteins , Mice , Mice, Transgenic , Phospholipids/metabolism
10.
Biochem Biophys Res Commun ; 330(2): 395-9, 2005 May 06.
Article in English | MEDLINE | ID: mdl-15796896

ABSTRACT

A great number of nuclear factors are involved in the negative feedback mechanism regulating bile acid synthesis. There are two major ways for the negative feedback to effect the synthesis; the SHP-dependent, involving FXR, and the SHP-independent way, affecting HNF-4alpha. We studied 23 patients with gallstone disease. Eight patients were treated with chenodeoxycholic acid, 7 with cholestyramine prior to operation, and 8 served as controls. Liver biopsies were analyzed with Real-time-PCR. In the cholestyramine-treated group mRNA levels of CYP7A1 were increased about 10-fold. Treatment with CDCA decreased the mRNA levels of CYP7A1 by about 70%. The mRNA levels of CYP8B1, CYP27A1, and CYP7B1 were not significantly altered in the treated groups. The analysis of mRNA levels for HNF-4alpha showed 64% higher levels in the cholestyramine-treated group compared to the controls. These levels showed positive and highly significant correlation to the levels of mRNA of CYP7A1 when studied in all three groups together. FXR, SHP, and LRH-1/FTF were not significantly affected by the different treatments. Our results indicate that when bile acid synthesis is upregulated by cholestyramine treatment the SHP-independent pathway for controlling CYP7A1 transcription dominates over the SHP-dependent pathway.


Subject(s)
Bile Acids and Salts/biosynthesis , Cholesterol 7-alpha-Hydroxylase/metabolism , DNA-Binding Proteins/physiology , Feedback , Gene Expression Regulation, Enzymologic/physiology , Liver/metabolism , Phosphoproteins/physiology , Transcription Factors/physiology , Cholesterol 7-alpha-Hydroxylase/genetics , Hepatocyte Nuclear Factor 4 , Humans , Liver/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...