Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Immun Inflamm Dis ; 12(4): e1248, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38607233

ABSTRACT

INTRODUCTION: Regulatory CD4+ T cells (Tregs) are pivotal for inhibition of autoimmunity. Primary sclerosing cholangitis (PSC) is an autoimmune cholestatic liver disease of unknown etiology where contribution of Tregs is still unclear. Activation of the JAK-STAT pathway critically modifies functions of Tregs. In PSC, we studied activation of STAT proteins and Treg functions in response to cytokines. METHODS: In 51 patients with PSC, 10 disease controls (chronic replicative hepatitis C), and 36 healthy controls we analyzed frequencies of Foxp3+CD25+CD127lowCD4+ Tregs, their expression of ectonucleotidase CD39, and cytokine-induced phosphorylation of STAT1, 3, 5, and 6 using phospho-flow cytometry. In parallel, we measured cytokines IFN-gamma, interleukin (IL)-6, IL-2, and IL-4 in serum via bead-based immunoassays. RESULTS: In patients with PSC, ex vivo frequencies of peripheral Tregs and their expression of CD39 were significantly reduced (p < .05 each). Furthermore, serum levels of IFN-gamma, IL-6, IL-2, and IL-4 were markedly higher in PSC (p < .05 each). Unlike activation of STAT1, STAT5, and STAT6, IL-6 induced increased phosphorylation of STAT3 in Tregs of PSC-patients (p = .0434). Finally, STAT3 activation in Tregs correlated with leukocyte counts. CONCLUSIONS: In PSC, we observed enhanced STAT3 responsiveness of CD4+ Tregs together with reduced CD39 expression probably reflecting inflammatory activity of the disease.


Subject(s)
Cholangitis, Sclerosing , T-Lymphocytes , Humans , Interleukin-6 , Interleukin-2 , Interleukin-4 , Janus Kinases , STAT Transcription Factors , Signal Transduction , Cytokines , CD4-Positive T-Lymphocytes
2.
Hepatol Res ; 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38509789

ABSTRACT

AIM: Primary sclerosing cholangitis (PSC) is a rare cholestatic liver disease characterized by inflammation of the intra- and extrahepatic bile ducts. Pathogenesis of PSC is still enigmatic but is likely to be multifactorial. Recently, we identified an interleukin-6 (IL-6)-dependent signal transducer and activator of transcription 3 (STAT3) activation in CD4+ TH1 and TH17 cells in PSC. The IL-6/STAT3 pathway was shown to be regulated by protease-activated receptor 1 (PAR1) contributing to inflammation. The role of the PAR1 -506 deletion/insertion (Del/Ins) polymorphism in PSC has not yet been investigated. METHODS: Two hundred eighty four PSC patients (200 patients with inflammatory bowel diseases [IBD] and 84 without IBD) and 309 healthy controls were genotyped for PAR1 rs11267092 (-506 Del/Ins -13 bp). Results were correlated with clinical characteristics and transplant-free survival. RESULTS: The frequency of PAR1 -506 Ins allele carriers (Del/Ins and Ins/Ins) was significantly higher in PSC patients (57.0%) compared to healthy controls (39.8%). Furthermore, carriers of PAR1 -506 Ins allele were more likely to have PSC than noncarriers (odds ratio 2.01; 95% confidence interval, 1.45-2.79). Patients with PSC carrying the PAR1 -506 Ins allele showed significantly higher alanine aminotransferase serum levels (p = 0.0357) and a trend toward shorter transplant-free survival time compared to noncarriers (8.9 ± 6.6 years vs. 10.5 ± 7.1 years; p = 0.076). CONCLUSIONS: Our study shows that PAR1 -506 Ins is significantly more frequent in people with PSC. As PAR1 -506 Ins allele carriers tended to have a shorter transplant-free survival, PAR1 might play a role in the development and course of PSC.

3.
Epigenomics ; 13(4): 271-283, 2021 02.
Article in English | MEDLINE | ID: mdl-33432840

ABSTRACT

Aim: This study aimed to identify novel miRNAs (miRs) as regulators of UGT1A gene expression and to evaluate them as potential risk factors for the development of liver fibrosis/cirrhosis. Materials & methods: miRNA target sites in UDP-glucuronosyltransferase 1A (UGT1A) 3'-UTR were predicted and confirmed by luciferase assays, quantitative real-time PCR and western blot using HEK293, HepG2 and Huh7 cells. UGT1A and miRNA expression were analyzed in cirrhotic patients and a mouse model of alcoholic liver fibrosis. Results: miR-214-5p and miR-486-3p overexpression reduced UGT1A mRNA, protein levels and enzyme activity in HepG2 and Huh7 cells. miR-486-3p was upregulated in cirrhotic patients and fibrotic mice livers, whereas UGT1A mRNA levels were reduced. Conclusion: In conclusion, we identified two novel miRNAs capable to repress UGT1A expression in vitro and in vivo. Furthermore, miR-486-3p may represent a potential risk factor for the development or progression of liver fibrosis/cirrhosis by means of a reduced UGT1A-mediated detoxification activity.


Subject(s)
Glucuronosyltransferase/genetics , MicroRNAs/genetics , Animals , Cell Line , Cell Line, Tumor , HEK293 Cells , Hep G2 Cells , Humans , Liver/metabolism , Liver Cirrhosis/genetics , Male , Mice , Up-Regulation/genetics
4.
Hepatobiliary Surg Nutr ; 10(6): 766-781, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35004944

ABSTRACT

BACKGROUND: Coffee consumption has been shown to reduce the risk of liver fibrosis and is capable of inducing human UDP-glucuronosyltransferase (UGT) 1A genes. UGT1A enzymes act as indirect antioxidants catalyzing the elimination of reactive metabolites, which in turn are potent initiators of profibrotic mechanisms. The aim of this study was to analyze the role of UGT1A genes as effectors of the protective properties of coffee in bile duct ligation (BDL) induced liver fibrosis. METHODS: Fourteen days BDL with and without coffee pre- and co-treatment was performed in htgUGT1A-WT and htgUGT1A-SNP mice. Hepatic UGT1A mRNA expression levels, serum bilirubin and aminotransferase activities were determined. Liver fibrosis was assessed by collagen deposition, computational analysis of Sirius red tissue staining and expression of profibrotic marker genes. Oxidative stress was measured by hepatic peroxidase concentrations and immunofluorescence staining. RESULTS: UGT1A transcription was differentially activated in the livers of htgUGT1A-WT mice after BDL, in contrast to a reduced or absent induction in the presence of SNPs. Co-treated (coffee + BDL) htgUGT1A-WT-mice showed significantly increased UGT1A expression and protein levels and a considerably higher induction compared to water drinking WT mice (BDL), whereas in co-treated htgUGT1A-SNP mice absolute expression levels remained below those observed in htgUGT1A-WT mice. Collagen deposition, oxidative stress and the expression of profibrotic markers inversely correlated with UGT1A expression levels in htgUGT1A-WT and SNP mice after BDL and coffee + BDL co-treatment. CONCLUSIONS: Coffee exerts hepatoprotective and antioxidative effects via activation of UGT1A enzymes. Attenuated hepatic fibrosis as a result of coffee-mediated UGT1A induction during cholestasis was detected, while the protective action of coffee was lower in a common low-function UGT1A SNP haplotype present in 10% of the Caucasian population. This study suggests that coffee consumption might constitute a potential strategy to support the conventional treatment of cholestasis-related liver diseases.

5.
Cancer Sci ; 111(11): 4266-4275, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32860300

ABSTRACT

UDP-glucuronosyltransferase (UGT) 1A enzymes detoxify a broad array of exogenous compounds including environmental toxins and carcinogens. Case-control studies identified genetic variations in UGT1A genes leading to reduced glucuronidation activity, which were associated with hepatocellular carcinoma (HCC) formation and progression. The aim of the study was therefore to examine the direct effect of common UGT1A polymorphisms (SNPs) on HCC development and outcome in a diethylnitrosamine (DEN)-induced mouse model. Therefore, a single intraperitoneal DEN injection (20 mg/kg) was administered to 15-day-old htgUGT1A-WT and htgUGT1A-SNP mice (containing a human haplotype of 10 common UGT1A SNPs) either receiving water or coffee cotreatment for the following 39 weeks. After this time, tumor incidence, size (>1 mm), histology, liver-body ratio, serum aminotransferase activities, and UGT1A regulation and activity levels were determined. In DEN-treated htgUGT1A-SNP mice, a markedly higher number of tumors with a bigger cumulative diameter were detected. The relative liver weight and aminotransferase activity levels were also significantly higher in mice carrying UGT1A SNPs. After coffee + DEN cotreatment, susceptibility for tumor development and growth considerably decreased in both mouse lines, but was still higher in htgUGT1A-SNP mice. In conclusion, our study provides experimental evidence for the protective role of UGT1A enzymes in neoplastic transformation. These data confirm case-control studies implicating impaired UGT1A-mediated carcinogen detoxification as a risk factor for individual cancer disposition. Coffee treatment, which is able to activate UGT1A expression and activity, reduced HCC development and provides an explanation for the protective properties of coffee on liver diseases including liver cancer.


Subject(s)
Carcinogenesis/chemically induced , Carcinogenesis/genetics , Diethylnitrosamine/adverse effects , Genetic Predisposition to Disease , Glucuronosyltransferase/genetics , Polymorphism, Single Nucleotide , Animals , Biopsy , Disease Models, Animal , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Glucuronosyltransferase/blood , Humans , Liver Function Tests , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Transgenic , Transcriptome
6.
Br J Pharmacol ; 177(18): 4193-4208, 2020 09.
Article in English | MEDLINE | ID: mdl-32548889

ABSTRACT

BACKGROUND AND PURPOSE: Irinotecan, used in colorectal cancer therapy, is metabolized by glucuronidation involving different UDP-glucuronosyltransferase (UGT)1A isoforms leading to facilitated elimination from the body. Individuals homozygous for the genetic variants UGT1A1*28 (Gilbert syndrome) and UGT1A7*3 are more susceptible to irinotecan side effects, severe diarrhoea and leukopenia. The aim of this study was to investigate the protective effects and active constituents of coffee during irinotecan therapy using humanized transgenic (htg)UGT1A-WT and htgUGT1A-SNP (carry UGT1A1*28 and UGT1A7*3 polymorphisms) mice. EXPERIMENTAL APPROACH: HtgUGT1A mice were pretreated with coffee or caffeic acid (CA) + caffeic acid phenylethyl ester (CAPE) and injected with irinotecan. The effects of coffee and CA + CAPE were investigated using reporter gene assays, immunoblot, TaqMan-PCR, siRNA analyses and blood counts. KEY RESULTS: Only the combination of the two coffee ingredients, CA and CAPE, mediates protective effects of coffee in a model of irinotecan toxicity by activation of UGT1A genes. Coffee and CA + CAPE significantly increased UGT1A expression and activity along with SN-38 glucuronide excretion in irinotecan-injected htgUGT1A mice, resulting in significant improvement of leukopenia, intestinal oxidative stress and inflammation. CONCLUSION AND IMPLICATIONS: In this study, we identify the compounds responsible for mediating the previously reported coffee-induced activation of UGT1A gene expression. CA and CAPE represent key factors for the protective properties of coffee which are capable of reducing irinotecan toxicity, exerting antioxidant and protective effects. Provided that CA + CAPE do not affect irinotecan efficacy, they might represent a novel strategy for the treatment of irinotecan toxicity.


Subject(s)
Caffeic Acids , Coffee , Irinotecan , Leukopenia , Oxidative Stress , Animals , Caffeic Acids/pharmacology , Camptothecin/toxicity , Esters , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Irinotecan/toxicity , Leukopenia/chemically induced , Leukopenia/prevention & control , Mice
7.
Liver Int ; 40(7): 1645-1654, 2020 07.
Article in English | MEDLINE | ID: mdl-32378294

ABSTRACT

BACKGROUND AND AIMS: Primary sclerosing cholangitis (PSC) is a progressive cholestatic liver disease without a curative medical therapy. The human UDP-glucuronosyltransferases 1A play a major role in the detoxification and elimination of bilirubin, bile acids and xenobiotics. Whether genetic UGT1A variants determine course and outcome of PSC has not yet been described. METHODS: A large cohort of German PSC patients with a long-term-follow-up was genotyped for UGT1A variants including UGT1A1*28, UGT1A3-66 T>C and UGT1A7 p.N129K/p.R131K using TaqMan 5'-nuclease assays. Results were correlated with clinical characteristics and transplant-free survival. RESULTS: About 331 patients with PSC were included in the study (69.9% male, mean age at diagnosis 32.6 years). Median transplant-free survival was 14.9 years. Patients with wild-type alleles of all three UGT1A genes had a longer transplant-free survival (17.2 vs. 14.4 years, P = .048) than patients carrying a homozygous or heterozygous SNP variant in at least one of the UGT1A1, UGT1A3 or UGT1A7 genes. Additionally, we found that patients carrying wild-type alleles of all three UGT1A genes had lower serum bilirubin (25 vs. 38 µmol/L, P = .02) and serum cholesterol (195 vs. 223 mg/dL), P = .035) at first presentation. Furthermore, inflammatory bowel disease was found to be associated with wild-type UGT1A alleles (82.2% vs. 68.4%, P = .046). CONCLUSIONS: This large cohort shows an association with single nucleotide polymorphisms of the UGT1A1, UGT1A3 and UGT1A7 genes and outcome in PSC. Thus, UGT1A variants may represent a tool for the prognostic stratification of PSC patients and establish a link between disease progression and the regulation of detoxification by glucuronidation in PSC.


Subject(s)
Cholangitis, Sclerosing , Adult , Alleles , Cholangitis, Sclerosing/genetics , Female , Genotype , Glucuronosyltransferase/genetics , Humans , Male , Uridine Diphosphate
8.
Sci Rep ; 10(1): 8689, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457304

ABSTRACT

UDP-glucuronosyltransferases 1 A (UGT1A) enzymes are capable of detoxifying a broad range of endo- and xenobiotic compounds, which contributes to antioxidative effects, modulation of inflammation and cytoprotection. In the presence of low-function genetic UGT1A variants fibrosis development is increased in various diseases. This study aimed to examine the role of common UGT1A polymorphisms in NASH. Therefore, htgUGT1A-WT mice and htgUGT1A-SNP mice (carrying a common human haplotype present in 10% of the white population) were fed a high-fat Paigen diet for 24 weeks. Serum aminotransferase activities, hepatic triglycerides, fibrosis development and UGT1A expression were assessed. Microscopic examination revealed higher hepatic fat deposition and a significant induction of UGT1A gene expression in htgUGT1A-WT mice. In agreement with these observations, lower serum aminotransferase activities and lower expression levels of fibrosis-related genes were measured in htgUGT1A-SNP mice. This was accompanied by reduced PPARα protein levels in htgUGT1A-WT but not in SNP mice. Our data demonstrate a protective effect of a UGT1A SNP haplotype, leading to milder hepatic steatosis and NASH. Higher PPARα protein levels in animals with impaired UGT1A activity are the likely result of reduced glucuronidation of ligands involved in PPARα-mediated fatty acid oxidation and may lead to the observed protection in htgUGT1A-SNP mice.


Subject(s)
Gilbert Disease/genetics , Glucuronosyltransferase/genetics , Non-alcoholic Fatty Liver Disease/prevention & control , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Diet, High-Fat , Fibrosis , Gilbert Disease/metabolism , Gilbert Disease/pathology , Glucuronosyltransferase/metabolism , Haplotypes , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/pathology , PPAR alpha/genetics , PPAR alpha/metabolism , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Protein Isoforms/metabolism , Triglycerides/analysis
9.
Phytomedicine ; 56: 35-39, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30668351

ABSTRACT

BACKGROUND: The popularity of herbal medicines is rapidly increasing in many countries including the Western world where many individuals turn to natural products, because they promise a safe and natural remedy for a broad variety of health disorders or the prevention of disease development. Although therapy with a number of herbal products has demonstrated a promising potential and efficacy, insufficient information exists concerning their pharmacodynamics, pharmacokinetics and mode of action. HYPOTHESIS/PURPOSE: Aim of this study was to examine the role of human detoxifying UDP-glucuronosyltransferases (UGTs) in the mechanism underlying the protective antioxidant effects reported for propolis, artichoke and silymarin. METHODS: UGT1A induction was analyzed by reporter gene assays, siRNA mediated knockdown and enzyme activity assays. Antioxidant activity was measured using a hydrogen peroxide colorimetric assay. RESULTS: We identified propolis, artichoke and silymarin as potent activators of UGT1A transcription and enzyme activity in KYSE70 cells mediated by aryl hydrocarbon receptor AhR and nuclear factor E2-related factor 2 (Nrf2) signaling. Propolis, artichoke and silymarin significantly decreased tertiary butylhydroquinone (tBHQ)-induced hydrogen peroxide levels. This protective effect was significantly reduced by siRNA mediated knockdown of UGT1A expression. CONCLUSION: In conclusion, this study provides a possible molecular mechanism for protective antioxidant effects associated with the herbal drugs propolis, artichoke and silymarin. The herbal drug-mediated transcriptional upregulation of human detoxifying UGT1A enzymes via activation of AhR and Nrf2 leads to reduced hydrogen peroxide and oxidative stress. Because of UGT1A activation, the intake of these drugs could affect the therapeutic efficacy of other drugs when these also undergo metabolism by glucuronidation.


Subject(s)
Antioxidants/pharmacology , Cynara scolymus/chemistry , Glucuronosyltransferase/metabolism , Propolis/pharmacology , Silymarin/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Gene Knockdown Techniques , Glucuronosyltransferase/genetics , Humans , Hydrogen Peroxide/metabolism , Inactivation, Metabolic/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Plant Extracts/pharmacology , RNA, Small Interfering , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
10.
Free Radic Biol Med ; 108: 66-76, 2017 07.
Article in English | MEDLINE | ID: mdl-28300668

ABSTRACT

AIMS: Coffee consumption has been epidemiologically associated with a lower risk for liver cirrhosis and cancer. UDP-glucuronosyltransferases (UGT1A) catalyze the detoxification of reactive metabolites thereby acting as indirect antioxidants. Aim of the study was to examine UGT1A regulation in response to Benzo[α]pyrene (BaP) to elucidate the potentially protective effects of coffee on BaP-induced oxidative stress and toxicity. RESULTS: In cell culture (HepG2, KYSE70 cells) and in htgUGT1A-WT mice, UGT1A transcription was activated by BaP, while it was reduced or absent htgUGT1A-SNP (containing 10 commonly occurring UGT1A-SNPs) mice. siRNA-mediated knockdown identified aryl hydrocarbon receptor (AhR) and nuclear factor erythroid2-related factor-2 (Nrf2) as mediators of BaP-induced UGT1A upregulation. Exposure to coffee led to a reduction of BaP-induced production of reactive oxygen species in vitro and in htgUGT1A-WT and -SNP mice. After UGT1A silencing by UGT1A-specific siRNA in cell culture, the coffee-mediated reduction of ROS production was significantly impaired compared to UGT1A expressing cells. CONCLUSION: A common UGT1A haplotype, prevalent in 9% (homozygous) of the White population, significantly impairs the expression of UGT1A enzymes in response to the putative tobacco carcinogen BaP and is likely to represent a significant risk factor for reduced detoxification and increased genotoxicity. Coffee was demonstrated to inhibit BaP-induced production of oxidative stress by UGT1A activation, and is therefore an attractive candidate for chemoprotection in risk groups for HCC or other tumors.


Subject(s)
Carcinoma, Hepatocellular/epidemiology , Coffea , Coffee/metabolism , Glucuronosyltransferase/metabolism , Liver Cirrhosis/epidemiology , Liver Neoplasms/epidemiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Benzopyrenes/toxicity , Carcinoma, Hepatocellular/diet therapy , Carcinoma, Hepatocellular/genetics , Eating , Gene Expression Regulation, Neoplastic , Genotype , Glucuronosyltransferase/genetics , Hep G2 Cells , Humans , Liver Cirrhosis/diet therapy , Liver Cirrhosis/genetics , Liver Neoplasms/diet therapy , Liver Neoplasms/genetics , Male , Mice , Mice, Transgenic , Oxidative Stress , Polymorphism, Single Nucleotide , Prevalence , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Risk , Tobacco Smoking/adverse effects , White People
11.
Hepatology ; 64(5): 1667-1682, 2016 11.
Article in English | MEDLINE | ID: mdl-27302828

ABSTRACT

Acetaminophen (APAP, paracetamol) poisoning is a leading cause of acute liver failure (ALF) in humans and induces hepatocyte necrosis, followed by activation of the innate immune system, further aggravating liver injury. The role of infiltrating monocytes during the early phase of ALF is still ambiguous. Upon experimental APAP overdose in mice, monocyte-derived macrophages (MoMFs) massively accumulated in injured liver within 12-24 hours, whereas the number of tissue-resident macrophages (Kupffer cells) decreased. Influx of MoMFs is dependent on the chemokine receptor, chemokine (C-C motif) receptor 2 (CCR2), given that Ccr2-/- mice display reduced infiltration of monocytes and attenuated liver injury post-APAP overdose at early time points. As evidenced by intravital multiphoton microscopy of Ccr2 reporter mice, CCR2+ monocytes infiltrate liver as early as 8-12 hours post-APAP overdose and form dense cellular clusters around necrotic areas. CCR2+ MoMFs express a distinct pattern of inflammatory, but also repair-associated, genes in injured livers. Adoptive transfer experiments revealed that MoMFs primarily exert proinflammatory functions early post-APAP, thereby aggravating liver injury. Consequently, early pharmacological inhibition of either chemokine (C-C motif) ligand (CCL2; by the inhibitor, mNOX-E36) or CCR2 (by the orally available dual CCR2/CCR5 inhibitor, cenicriviroc) reduces monocyte infiltration and APAP-induced liver injury (AILI) in mice. Importantly, neither the early nor continuous inhibition of CCR2 hinder repair processes during resolution from injury. In line with this, human livers of ALF patients requiring liver transplantation reveal increased CD68+ hepatic macrophage numbers with massive infiltrates of periportal CCR2+ macrophages that display a proinflammatory polarization. CONCLUSION: Infiltrating monocyte-derived macrophages aggravate APAP hepatotoxicity, and the pharmacological inhibition of either CCL2 or CCR2 might bear therapeutic potential by reducing the inflammatory reaction during the early phase of AILI. (Hepatology 2016;64:1667-1682).


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Antipyretics/adverse effects , Liver Failure, Acute/chemically induced , Receptors, CCR2/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Monocytes/chemistry , Receptors, CCR2/analysis , Severity of Illness Index
12.
J Hepatol ; 59(4): 797-804, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23714156

ABSTRACT

BACKGROUND & AIMS: Gender influences incidence, progression, and therapy of hepatogastrointestinal diseases. The aim of this study was to elucidate the molecular mechanism of gender-specific UDP-glucuronosyltransferases (UGT1A) regulation, representing important hepatogastrointestinal detoxification enzymes for xenobiotics, drugs, and endobiotics. METHODS: UGT1A-gene activation was studied by reporter gene experiments and estrogen receptor alpha (ESR1/ERα) co-transfection using KYSE70- and HepG2 cells (male origin), and SW403 cells (female origin). Cell lines, and humanized transgenic UGT1A (htgUGT1A) mice (female/male) were treated with the ERα inhibitor tamoxifen. UGT1A mRNA expression was analyzed by TaqMan PCR, the recruitment of ERα, histone deacetylases (HDAC), and the aryl hydrocarbon receptor (AhR) by chromatin immunoprecipitation (ChIP), and ERα expression in gastrointestinal mouse tissues by Western blot and immunofluorescence. RESULTS: In KYSE70 cells (male), UGT1A gene expression was induced 5-10 fold, and inhibited in the presence of ERα by 55-77%. In SW403 (female) cells, absent inducibility was restored after tamoxifen treatment. In the jejunum and colon of tgUGT1A mice, UGT1A induction that was exclusively detected in male mice could be restored in female mice after tamoxifen pre-treatment. ChIP assays demonstrated the recruitment of ERα and HDACs to the xenobiotic response elements of UGT1A promoters during gene repression. Western blot showed higher ERα expression in the female jejunum and colon. CONCLUSIONS: We show gender-specific transcriptional control of UGT1A genes in jejunum and colon, which is repressed by ERα and the recruitment of HDCAs to the UGT1A promoter sequence in females. A molecular mechanism controlling gender-specific drug metabolism and its therapeutic reversal is demonstrated.


Subject(s)
Estrogen Receptor alpha/metabolism , Glucuronosyltransferase/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Animals , Cell Line , Coffee , Colon/metabolism , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Enzymologic/drug effects , Hep G2 Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , Jejunum/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Sex Characteristics , Tamoxifen/pharmacology
13.
Hepatology ; 55(6): 1912-21, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22213127

ABSTRACT

UNLABELLED: Gilbert syndrome (GS) is characterized by intermittent unconjugated hyperbilirubinemia without structural liver damage, affecting about 10% of the white population. In GS the UGT1A1*28 variant reduces bilirubin conjugation by 70% and is associated with irinotecan and protease inhibitor side effects. The aim of this study was to characterize potential in vivo consequences of UGT1A gene variability in GS. Three hundred GS patients (UGT1A1*28 homozygous) and 249 healthy blood donors (HBD) were genotyped for UGT1A (UGT1A1*28, UGT1A3-66 T>C, UGT1A6*3a, UGT1A7*3) and transporter single nucleotide polymorphisms (SNPs) (SCLO1B1 p.V174A, SCLO1B1 p.N130D, ABCC2 p.I1324I, ABCC2-24 UTR) using TaqMan-5'-nuclease-assays. A humanized transgenic UGT1A-SNP and corresponding wildtype mouse model were established carrying the GS-associated UGT1A variant haplotype. UGT1A transcript and protein expression, and transcriptional activation were studied in vivo. Homozygous UGT1A1*28 GS individuals were simultaneously homozygous for UGT1A3-66 T>C (91%), UGT1A6*2a (77%), and UGT1A7*3 (77%). Seventy-six percent of GS and only 9% of HBD were homozygous for the variant haplotype spanning four UGT1A genes. SCLO1B1 and ABCC2 SNPs showed no differences. In transgenic humanized UGT1A SNP and wildtype mice this UGT1A haplotype led to lower UGT1A messenger RNA (mRNA) expression and UGT1A protein synthesis. UGT1A transcriptional activation by dioxin, phenobarbital, and endotoxin was significantly reduced in SNP mice. CONCLUSION: Our data redefine the genetic basis behind GS. In vivo data studying the genotype present in 76% of GS individuals suggest that transcription and transcriptional activation of glucuronidation genes responsible for conjugation and detoxification is directly affected, leading to lower responsiveness. This study suggests that GS should be considered a potential risk factor for drug toxicity.


Subject(s)
Gilbert Disease/genetics , Glucuronides/metabolism , Glucuronosyltransferase/genetics , Haplotypes , Polymorphism, Single Nucleotide , Animals , Blood Donors , Gilbert Disease/metabolism , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multidrug Resistance-Associated Protein 2 , Polychlorinated Dibenzodioxins/pharmacology , Transcriptional Activation/drug effects
14.
Gastroenterology ; 139(5): 1699-710, 1710.e1-2, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20600030

ABSTRACT

BACKGROUND & AIMS: Coffee is one of the most widely consumed beverages worldwide. Epidemiologic data indicate that coffee consumption protects against the progression of chronic liver disease and development of hepatocellular carcinoma and diabetes, but the mechanisms are not clear. UDP glucuronosyltransferases (UGT1A) are proteins with indirect antioxidant, cytoprotective, and genoprotective capabilities; we examined UGT1A regulation in response to coffee in cultured cells and mice. METHODS: HepG2 and CaCo2 cells were incubated with regular, metal- or paper-filtered, decaffeinated, or instant coffee; green or black tea; cocoa; or metabolic products of caffeine. The effects of UGT1A regulation were investigated with reporter gene assays, immunoblot, TaqMan polymerase chain reaction, mutagenesis, and short interfering (si)RNA analyses. We also studied the effects of coffee in humanized transgenic mice that express human UGT1A. RESULTS: Incubation of cells with coffee induced transcription of UGT1A1 (5.4-fold), UGT1A3 (5.2-fold), UGT1A4 (4.8-fold), UGT1A7 (6.2-fold), UGT1A8 (5.2-fold), UGT1A9 (3.5-fold), and UGT1A10 (6.1-fold). Induction was independent of caffeine, methylxanthines, or the diterpenes cafestol and kahweol. Mutagenesis and short interfering RNA knockdown studies showed that UGT1A is regulated by the aryl hydrocarbon receptor (AhR) and the nuclear factor erythroid-related factor 2 (Nrf2) by cis-acting antioxidant and xenobiotic response elements (ARE/XRE). In transgenic UGT1A mice, administration of coffee resulted in a 10- and 14-fold induction of UGT1A transcription in liver and stomach, respectively. CONCLUSIONS: UGT1A genes are induced in vitro and in vivo by coffee, independent of caffeine content, cafestol, or kahweol. Coffee up-regulates glucuronidation by AhR signaling and Nrf2 binding to the ARE/XRE. Glucuronidation could mediate the protective and antioxidant effects of coffee.


Subject(s)
Coffee , Gastric Mucosa/metabolism , Gene Expression Regulation, Neoplastic , Glucuronosyltransferase/genetics , Liver/metabolism , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Blotting, Western , Chronic Disease , Disease Models, Animal , Female , Glucuronosyltransferase/biosynthesis , Humans , Liver/pathology , Male , Mice , Mice, Transgenic , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach/pathology , Tumor Cells, Cultured
15.
Drug Metab Dispos ; 38(7): 1246-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20406851

ABSTRACT

Substrates for glucuronidation include endogenous and xenobiotic compounds such as environmental carcinogens and drugs, as well as the chemotherapeutic agent irinotecan. The UDP-glucuronosyltransferase (UGT) 1A7 gene is expressed in the upper gastrointestinal tract and the lung but is not expressed in the liver. The transcriptional regulation of UGT1A7 and the putative influence of single nucleotide polymorphisms (SNPs) are incompletely characterized. UGT1A8, UGT1A9, and UGT1A10, which are highly homologous to UGT1A7, have been reported to be transcriptionally regulated by hepatocyte nuclear factors (HNFs). In this study, we show the activation of UGT1A7 by the aforementioned transcription factors. Sequence analyses, mutagenesis, reporter gene experiments, small interfering RNA silencing, chromatin immunoprecipitation, and electromobility shift assays identified five HNF binding sites in the proximal promoter region of UGT1A7 that were regulated by HNF1alpha and HNF4alpha. Activation by HNF1alpha was lower in the presence of the UGT1A7 -57G SNP. In contrast to liver-expressed UGT1A9, transcriptional activation of UGT1A7 by HNF4alpha was lower and dependent on higher HNF4alpha concentrations, which may contribute to the observed differences in tissue expression patterns. Therefore, a specific role of HNF in the transcriptional control of UGT1A7 is shown and characterized, which may contribute to its tissue specificity and function.


Subject(s)
Gene Expression Regulation, Enzymologic/genetics , Glucuronosyltransferase/biosynthesis , Glucuronosyltransferase/genetics , Hepatocyte Nuclear Factor 1-alpha/physiology , Hepatocyte Nuclear Factor 4/physiology , Base Sequence , Binding Sites/genetics , Cells, Cultured , Humans , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Transcriptional Activation
16.
J Hepatol ; 52(4): 570-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20189675

ABSTRACT

BACKGROUND & AIMS: Cholestasis is a serious complication of many liver diseases leading to increased serum bile acids (BA) and their conjugates. Chenodeoxycholic (CDCA) acid is a substrate of the human hepatic UDP-glucuronosyltransferase (UGT) 1A3. UGT1A3 may, therefore, be a BA-inducible gene relevant to BA regulation. METHODS: BA and human bile were used to induce UGT1A3 in HepG2 cells. Genomic DNA was analyzed by PCR amplification and sequencing. Transcriptional regulation was studied by DNA mutagenesis, RT-PCR, luciferase reporter gene constructs and electrophoretic mobility shift assays (EMSA). RESULTS: CDCA differentially induced UGT1A3 but not UGT1A4 expression. Bile from ursodeoxycholic acid (UDCA)-treated and untreated patients differentially induced UGT1A3. A farnesoid X receptor (FXR) half-site DNA motif was identified in the UGT1A3 5' upstream region. The FXR inducer GW4064 activated UGT1A3 transcription, and electrophoretic mobility shift assays identified UGT1A3 as a FXR target gene. CONCLUSIONS: Transcriptional regulation of the human bile acid and xenobiotic UGT1A3 by its substrate CDCA and FXR is shown. CDCA glucuronidation can be controlled by feed back inhibition proceeding via the glucuronidation of CDCA. UDCA does not induce UGT1A3 transcription. Since UGT1A3 is significantly induced by xenobiotics this physiologically links xenobiotic and bile acid metabolism to cholestasis.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Bile Acids and Salts/pharmacology , Chenodeoxycholic Acid/metabolism , Chenodeoxycholic Acid/pharmacology , Cholestasis/genetics , Cholestasis/metabolism , Cholestasis/physiopathology , Gene Expression/physiology , Genes, Reporter/physiology , Genetic Variation , Hep G2 Cells , Humans , Lithocholic Acid/metabolism , Lithocholic Acid/pharmacology , Luciferases/genetics , Mutagenesis, Site-Directed , Promoter Regions, Genetic/genetics , Transcriptional Activation/physiology , Ursodeoxycholic Acid/metabolism , Ursodeoxycholic Acid/pharmacology , Xenobiotics/metabolism
17.
J Biol Chem ; 285(9): 5993-6002, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20053997

ABSTRACT

The defense against oxidative stress is a critical feature that prevents cellular and DNA damage. UDP-glucuronosyltransferases (UGTs) catalyze the glucuronidation of xenobiotics, mutagens, and reactive metabolites and thus act as indirect antioxidants. Aim of this study was to elucidate the regulation of UGTs expressed in the mucosa of the gastrointestinal tract by xenobiotics and the main mediator of antioxidant defense, Nrf2 (nuclear factor erythroid 2-related factor 2). Xenobiotic (XRE) and antioxidant (ARE) response elements were detected in the promoters of UGT1A8, UGT1A9, and UGT1A10. Reporter gene experiments demonstrated XRE-mediated induction by dioxin in addition to tert-butylhydroquinone (ARE)-mediated induction of UGT1A8 and UGT1A10, which are expressed in extrahepatic tissue in humans in vivo. The responsible XRE and ARE motifs were identified by mutagenesis. Small interfering RNA knockdown, electrophoretic mobility shifts, and supershifts identified a functional interaction of Nrf2 and the aryl hydrocarbon receptor (AhR). Induction of UGT1A8 and UGT1A10 requires Nrf2 and AhR. It proceeds by utilizing XRE- as well as ARE-binding motifs. In summary, we demonstrate the coordinated AhR- and Nrf2-dependent transcriptional regulation of human UGT1As. Cellular protection by glucuronidation is thus inducible by xenobiotics via AhR and by oxidative metabolites via Nrf2 linking glucuronidation to cellular protection and defense against oxidative stress.


Subject(s)
Glucuronosyltransferase/metabolism , Metabolic Detoxication, Phase II , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Receptors, Aryl Hydrocarbon/metabolism , Xenobiotics/pharmacology , Antioxidants , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Glucuronides , Glucuronosyltransferase/genetics , Humans , NF-E2-Related Factor 2/physiology , Response Elements , Transcription, Genetic
18.
J Hepatol ; 50(5): 1010-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19303655

ABSTRACT

BACKGROUND/AIMS: Gilbert's syndrome is a frequent genetic conjugation abnormality associated with adverse drug effects. Genetic UDP glucuronosyltransferase (UGT)1A gene variants can influence gene transcription, inducibility and glucuronidation activity. Protease inhibitors used in human immunodeficiency virus (HIV) infection and chronic viral hepatitis can inhibit UGTs. Indinavir (IDV) can lead to hyperbilirubinemia in Gilbert's syndrome (UGT1A1*28), which does not explain interindividual severity differences and may thus involve additional UGT1A variants. METHODS: One hundred and twenty-five HIV patients receiving IDV and 427 healthy blood donors were genotyped for the presence of UGT1A1*28, UGT1A3 -66T/C, UGT1A7 -57T/G, UGT1A7(N129K/R131K) using Taqman 5' nuclease assays. RESULTS: Hyperbilirubinemia was observed in 42%. UGT1A1*28 frequencies did not differ between HIV patients and controls but were significantly higher in hyperbilirubinemic patients. The frequency of homozygous carriers of the 4 UGT1A marker haplotype increased with hyperbilirubinemia affecting all patients with bilirubin levels >85 micromol/l. CONCLUSIONS: In IDV treatment the risk of severe hyperbilirubinemia is associated with genetic variants of the UGT1A3 and UGT1A7 genes in addition to Gilbert's syndrome (UGT1A1*28). This haplotype is a useful predictor of protease inhibitor-induced side effects.


Subject(s)
Gilbert Disease/drug therapy , Gilbert Disease/genetics , Glucuronosyltransferase/genetics , Hyperbilirubinemia/chemically induced , Hyperbilirubinemia/epidemiology , Indinavir/adverse effects , Protease Inhibitors/adverse effects , Adult , Aged , Case-Control Studies , Female , Genotype , Haplotypes/genetics , Humans , Indinavir/therapeutic use , Male , Middle Aged , Predictive Value of Tests , Protease Inhibitors/therapeutic use , Risk Factors
19.
Crit Rev Clin Lab Sci ; 45(6): 485-530, 2008.
Article in English | MEDLINE | ID: mdl-19003600

ABSTRACT

The substrate spectrum of human UDP-glucuronosyltransferase 1A (UGT1A) proteins includes the glucuronidation of non-steroidal anti-inflammatory drugs, anticonvulsants, chemotherapeutics, steroid hormones, bile acids, and bilirubin. The unique genetic organization of the human UGT1A gene locus, and an increasing number of functionally relevant genetic variants define tissue specificity as well as a broad range of interindividual variabilities of glucuronidation. Genetic UGT1A variability has been conserved throughout the protein's evolution and shows ethnic diversity. It is the biochemical and genetic basis for clinical phenotypes such as Gilbert's syndrome and Crigler-Najjar's disease as well as for the potential for severe, unwanted drug side effects such as in irinotecan treatment. UGT1A variants influence the metabolic effects of xenobiotic exposure and therefore have been linked to cancer risk. Detailed knowledge of the organization, function, and pharmacogenetics of the human UGT1A gene locus is likely to significantly contribute to the improvement of drug safety and efficacy as well as to the provision of steps toward the goal of individualized drug therapy and disease risk prediction.


Subject(s)
Evolution, Molecular , Genetic Variation , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hyperbilirubinemia/metabolism , Neoplasms/genetics , Pharmaceutical Preparations/metabolism , Gene Components , Humans , Hyperbilirubinemia/etiology , Molecular Structure , Pharmacogenetics , Substrate Specificity
20.
J Mol Diagn ; 10(6): 549-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18832463

ABSTRACT

Gilbert's syndrome causes mild, unconjugated hyperbilirubinemia and is present in approximately 10% of the Caucasian population. The basis of the disorder is a 70% reduction in bilirubin glucuronidation catalyzed by the UDP-glucuronosyltransferase 1A1 (UGT1A1), which, in Caucasians, is the result of a homozygous TA insertion into the promoter region of the UGT1A1 gene (UGT1A1*28). Homozygous carriers of UGT1A1*28 as well as those with additional UGT1A variants can suffer from severe irinotecan toxicity or jaundice during treatment with the protease inhibitor atazanavir. UGT1A1*28 genotyping identifies patients at risk for drug toxicity and can increase drug safety by dose individualization. Rapid and facile UGT1A1*28 genotyping is therefore of great clinical importance. Two hundred ninety-one patients with suspected Gilbert's syndrome were genotyped using the TaqMan 5'nuclease assay with minor groove binder-non fluorescent quench probes; results were confirmed by direct sequencing. Ninety-six patients (33%) were homozygous for UGT1A1*28, which was verified by direct sequencing of a different PCR product showing 100% concordance with the TaqMan PCR results. We describe a novel UGT1A1*28 genotyping method that employs allelic discrimination by TaqMan PCR. This assay provides a rapid, high-throughput, and cost-effective method for Gilbert's syndrome genotyping, which is of value for pretreatment screening of potential irinotecan toxicity. The method utilizes a technological platform that is widely used in clinical practice and could therefore be easily adapted for routine clinical applications.


Subject(s)
Alleles , DNA Mutational Analysis/methods , Genetic Markers , Gilbert Disease/genetics , Glucuronosyltransferase/genetics , Polymerase Chain Reaction/methods , Aged , Female , Genotype , Humans , Male , Middle Aged , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...