Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
ACS Med Chem Lett ; 15(4): 546-554, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38628802

ABSTRACT

Chronic hepatitis B (CHB) virus infection afflicts hundreds of millions of people and causes nearly one million deaths annually. The high levels of circulating viral surface antigen (HBsAg) that characterize CHB may lead to T-cell exhaustion, resulting in an impaired antiviral immune response in the host. Agents that suppress HBsAg could help invigorate immunity toward infected hepatocytes and facilitate a functional cure. A series of dihydropyridoisoquinolizinone (DHQ) inhibitors of human poly(A) polymerases PAPD5/7 were reported to suppress HBsAg in vitro. An example from this class, RG7834, briefly entered the clinic. We set out to identify a potent, orally bioavailable, and safe PAPD5/7 inhibitor as a potential component of a functional cure regimen. Our efforts led to the identification of a dihydropyridophthalazinone (DPP) core with improved pharmacokinetic properties. A conformational restriction strategy and optimization of core substitution led to GS-8873, which was projected to provide deep HBsAg suppression with once-daily dosing.

2.
Toxicol Sci ; 186(2): 298-308, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35134235

ABSTRACT

The nonclinical safety profile of GS-8873, a hepatitis B virus RNA transcript inhibitor was evaluated in rat and monkey 13-week toxicity studies with 8-week recovery phases. Vehicle or GS-8873 was dosed orally for 13 weeks at 2, 6, 20, and 60 mg/kg/day to Wistar Han rats and at 0.5, 1.5, 3, and 6 mg/kg/day to cynomolgus monkeys. In vitro and in vivo screening results from an analog discovered prior to GS-8873 informed the 13-week toxicology study designs. Neuroelectrophysiology and neurobehavioral evaluations were included at weeks 4 and 13 of the dosing and recovery phases for GS-8873. No adverse neurobehavioral effects were observed. Significant nerve conduction velocity (NCV) decreases and latency increases occurred at the high doses after 4 weeks of dosing. By week 13, dose-responsive NCV reductions and latency increases worsened across all dose groups compared with controls. Some reversal occurred 8 weeks after the last dose administered, but not to vehicle control levels. A minimal, axonal degeneration was observed in rat spinal and peripheral nerves across dose groups compared with controls. No monkey nervous system microscopic findings were observed. No-observed-adverse-effect-levels could not be determined for either species due to the neuroelectrophysiology findings and development was halted in the interest of safety. A retrospective risk assessment approach utilizing benchmark dose (BMD) modeling contributed 13-week NCV BMDL estimates (lower limits of the 95% confidence interval) in lieu of no-observed-adverse-effect-levels. The best-fitted models extrapolated NCV BMDLs for the rat caudal and monkey sural nerve at 0.3 and 0.1 mg/kg/day, respectively.


Subject(s)
Antiviral Agents , Hepatitis B virus , Administration, Oral , Animals , Antiviral Agents/pharmacology , Antiviral Agents/toxicity , Haplorhini , Hepatitis B virus/drug effects , Rats , Rats, Wistar , Retrospective Studies
3.
Toxicol Appl Pharmacol ; 368: 49-54, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30794826

ABSTRACT

INTRODUCTION: Nonalcoholic steatohepatitis (NASH) afflicts 20-36% of individuals with nonalcoholic fatty liver disease (NAFLD). A lipotoxic hepatic environment, altered innate immune signaling and inflammation are defining features of progression to NASH. Activated resident liver macrophages express folate receptor beta (FR-ß) which may be an indicator of progression from steatosis to NASH. The goals of this study were to characterize FR-ß protein expression in human NAFLD and rodent models of NASH, and demonstrate liver targeting of an FR-ß imaging agent to the liver of a rodent NASH model using FR-ß. METHODS: Rat liver lysates from methionine choline deficient (MCD) fed rats, high fat diet (HFD) and methionine choline sufficient (MC+) rat controls were analyzed for hepatic FR-ß protein. The FR-ß-targeted agent, Etarfolatide was injected into MCD and MC + -fed C57BL/6 mice for efficient FastSPECT hepatic imaging. Additionally, FR-ß expression across the stages of human NAFLD from normal to NASH was assessed. RESULTS: FastSPECT images show targeting of Etarfolatide to the liver of mice fed 8 weeks of MCD diet but not control-fed mice. The MCD rat model exhibited significantly increased protein expression of hepatic FR-ß in contrast to HFD or normal samples. Similarly human liver samples categorized as NASH Fatty or NASH Not Fatty showed elevated FR-ß protein when compared to normal liver. FR-ß transcript expression levels were elevated across both NASH Fatty and NASH Not Fatty samples. CONCLUSION: The findings in this study indicate that FR-ß expression in NASH may be harnessed to target agents directly to the liver.


Subject(s)
Folate Receptor 2/metabolism , Liver/diagnostic imaging , Liver/metabolism , Macrophages/metabolism , Molecular Imaging/methods , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Non-alcoholic Fatty Liver Disease/metabolism , Tomography, Emission-Computed, Single-Photon , Animals , Biomarkers/metabolism , Choline Deficiency/complications , Diet, High-Fat , Disease Models, Animal , Folate Receptor 2/genetics , Folic Acid/administration & dosage , Folic Acid/analogs & derivatives , Humans , Male , Methionine/deficiency , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Organotechnetium Compounds/administration & dosage , Predictive Value of Tests , Radiopharmaceuticals/administration & dosage , Rats, Sprague-Dawley
4.
Obesity (Silver Spring) ; 25(6): 1069-1076, 2017 06.
Article in English | MEDLINE | ID: mdl-28452429

ABSTRACT

OBJECTIVE: Characteristic pathological changes define the progression of steatosis to nonalcoholic steatohepatitis (NASH) and are correlated to metabolic pathways. A common rodent model of NASH is the methionine and choline deficient (MCD) diet. The objective of this study was to perform full metabolomic analyses on liver samples to determine which pathways are altered most pronouncedly in this condition in humans, and to compare these changes to rodent models of nonalcoholic fatty liver disease (NAFLD). METHODS: A principal component analysis for all 91 metabolites measured indicated that metabolome perturbation is greater and less varied for humans than for rodents. RESULTS: Metabolome changes in human and rat NAFLD were greatest for the amino acid and bile acid metabolite families (e.g., asparagine, citrulline, gamma-aminobutyric acid, lysine); although, in many cases, the trends were reversed when compared between species (cholic acid, betaine). CONCLUSIONS: Overall, these results indicate that metabolites of specific pathways may be useful biomarkers for NASH progression, although these markers may not correspond to rodent NASH models. The MCD model may be useful when studying certain end points of NASH; however, the metabolomics results indicate important differences between humans and rodents in the biochemical pathogenesis of the disease.


Subject(s)
Metabolomics/methods , Obesity/complications , Animals , Diet , Disease Progression , Humans , Liver/pathology , Male , Non-alcoholic Fatty Liver Disease/metabolism , Rats , Rats, Sprague-Dawley
5.
Liver Int ; 37(7): 1074-1081, 2017 07.
Article in English | MEDLINE | ID: mdl-28097795

ABSTRACT

BACKGROUND & AIMS: N-linked glycosylation of proteins is critical for proper protein folding and trafficking to the plasma membrane. Drug transporters are one class of proteins that have reduced function when glycosylation is impaired. N-linked glycosylation of plasma proteins has also been investigated as a biomarker for several liver diseases, including non-alcoholic fatty liver disease (NAFLD). The purpose of this study was to assess the transcriptomic expression of genes involved in protein processing and glycosylation, and to determine the glycosylation status of key drug transporters during human NAFLD progression. METHODS: Human liver samples diagnosed as healthy, steatosis, and non-alcoholic steatohepatitis (NASH) were analysed for gene expression of glycosylation-related genes and for protein glycosylation using immunoblot. RESULTS: Genes involved in protein processing in the ER and biosynthesis of N-glycans were significantly enriched for down-regulation in NAFLD progression. Included in the down regulated N-glycan biosynthesis category were genes involved in the oligosaccharyltransferase complex, N-glycan quality control, N-glycan precursor biosynthesis, N-glycan trimming to the core, and N-glycan extension from the core. N-glycan degradation genes were unaltered in the progression to NASH. Immunoblot analysis of the uptake transporters organic anion transporting polypeptide-1B1 (OATP1B1), OATP1B3, OATP2B1, and Sodium/Taurocholate Co-transporting Polypeptide (NTCP) and the efflux transporter multidrug resistance-associated protein 2 (MRP2) demonstrated a significant loss of glycosylation following the progression to NASH. CONCLUSIONS: These data suggest that the loss of glycosylation of key uptake and efflux transporters in humans NASH may influence transporter function and contribute to altered drug disposition observed in NASH.


Subject(s)
Liver/metabolism , Membrane Transport Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Protein Processing, Post-Translational , Biological Transport , Blotting, Western , Case-Control Studies , Endoplasmic Reticulum/metabolism , Gene Expression Profiling , Glycosylation , Humans , Membrane Transport Proteins/genetics , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/genetics , Transcriptome
6.
Biochem Pharmacol ; 122: 62-71, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27836672

ABSTRACT

The molecular mechanisms behind the transition from simple steatosis to nonalcoholic steatohepatitis (NASH) in nonalcoholic fatty liver disease (NAFLD) are not clearly understood. This hinders development of effective therapies for treatment and prevention of NASH. In this study expression profiling data from normal, steatosis, and NASH human livers were used to predict transcription factors that are misregulated as mechanistic features of NAFLD progression. Previously-published human NAFLD gene expression profiling data from normal, steatosis, and NASH livers were subjected to transcription factor binding site enrichment analysis. Selected transcription factors that bind enriched transcription factor binding sites were analyzed for changes in expression. Distinct transcription factor binding sites were enriched in genes significantly up- or down-regulated in NASH livers. Those enriched in up-regulated genes were bound by transcription factors such as FOXA, CEBP, and HNF1 family members, while those enriched in down-regulated genes were bound by nuclear receptors involved in xenobiotic sensing and lipid metabolism. Levels of mRNA and protein for selected transcription factors were significantly changed during disease progression. The study indicates that NAFLD progression involves changes in activity or expression of transcription factors that regulate genes involved in hepatic processes known to be altered in NASH. Transcription factors such as PPAR receptors, FoxA family members, and HNF4A might be targeted therapeutically to prevent NAFLD progression.


Subject(s)
Gene Expression Regulation/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Transcription Factors/metabolism , Binding Sites , Disease Progression , Down-Regulation , Gene Expression Profiling , Humans , Protein Binding , Up-Regulation
7.
Toxicol Sci ; 149(2): 312-25, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26519955

ABSTRACT

Current strategies for predicting adverse health outcomes of environmental chemicals are centered on early key events in toxicity pathways. However, quantitative relationships between early molecular changes in a given pathway and later health effects are often poorly defined. The goal of this study was to evaluate short-term key event indicators using qualitative and quantitative methods in an established pathway of mouse liver tumorigenesis mediated by peroxisome proliferator-activated receptor alpha (PPARα). Male B6C3F1 mice were exposed for 7 days to di (2-ethylhexyl) phthalate (DEHP), di-n-octyl phthalate (DNOP), and n-butyl benzyl phthalate (BBP), which vary in PPARα activity and liver tumorigenicity. Each phthalate increased expression of select PPARα target genes at 7 days, while only DEHP significantly increased liver cell proliferation labeling index (LI). Transcriptional benchmark dose (BMDT) estimates for dose-related genomic markers stratified phthalates according to hypothetical tumorigenic potencies, unlike BMDs for non-genomic endpoints (relative liver weights or proliferation). The 7-day BMDT values for Acot1 as a surrogate measure for PPARα activation were 29, 370, and 676 mg/kg/day for DEHP, DNOP, and BBP, respectively, distinguishing DEHP (liver tumor BMD of 35 mg/kg/day) from non-tumorigenic DNOP and BBP. Effect thresholds were generated using linear regression of DEHP effects at 7 days and 2-year tumor incidence values to anchor early response molecular indicators and a later phenotypic outcome. Thresholds varied widely by marker, from 2-fold (Pdk4 and proliferation LI) to 30-fold (Acot1) induction to reach hypothetical tumorigenic expression levels. These findings highlight key issues in defining thresholds for biological adversity based on molecular changes.


Subject(s)
Liver Neoplasms, Experimental/chemically induced , PPAR alpha/physiology , Animals , Benchmarking , Body Weight/drug effects , Cell Proliferation , Diethylhexyl Phthalate/toxicity , Dose-Response Relationship, Drug , Linear Models , Liver/metabolism , Liver/pathology , Male , Mice , Oxidative Stress , Phthalic Acids/toxicity , Polymerase Chain Reaction
8.
Carcinogenesis ; 36(7): 782-91, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25913432

ABSTRACT

Environmental exposures occurring early in life may have an important influence on cancer risk later in life. Here, we investigated carryover effects of dichloroacetic acid (DCA), a small molecule analog of pyruvate with metabolic programming properties, on age-related incidence of liver cancer. The study followed a stop-exposure/promotion design in which 4-week-old male and female B6C3F1 mice received the following treatments: deionized water alone (dH2O, control); dH2O with 0.06% phenobarbital (PB), a mouse liver tumor promoter; or DCA (1.0, 2.0 or 3.5g/l) for 10 weeks followed by dH2O or PB (n = 20-30/group/sex). Pathology and molecular assessments were performed at 98 weeks of age. In the absence of PB, early-life exposure to DCA increased the incidence and number of hepatocellular tumors in male and female mice compared with controls. Significant dose trends were observed in both sexes. At the high dose level, 10 weeks of prior DCA treatment induced comparable effects (≥85% tumor incidence and number) to those seen after continuous lifetime exposure. Prior DCA treatment did not enhance or inhibit the carcinogenic effects of PB, induce persistent liver cytotoxicity or preneoplastic changes on histopathology or alter DNA sequence variant profiles within liver tumors compared with controls. Distinct changes in liver messenger RNA and micro RNA profiles associated with prior DCA treatment were not apparent at 98 weeks. Our findings demonstrate that early-life exposure to DCA may be as carcinogenic as life-long exposures, potentially via epigenetic-mediated effects related to cellular metabolism.


Subject(s)
Dichloroacetic Acid/pharmacology , Liver Neoplasms/chemically induced , Animals , DNA Methylation/drug effects , Dichloroacetic Acid/administration & dosage , Dichloroacetic Acid/toxicity , Dose-Response Relationship, Drug , Eating , Environmental Pollutants/toxicity , Female , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Mice, Inbred Strains , MicroRNAs , Phenobarbital/toxicity , RNA, Messenger
9.
Drug Metab Dispos ; 43(2): 266-72, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25488932

ABSTRACT

Nonalcoholic fatty liver disease is the most common chronic liver disease, which can progress to nonalcoholic steatohepatitis (NASH). Previous investigations demonstrated alterations in the expression and activity of hepatic drug transporters in NASH. Moreover, studies using rodent models of cholestasis suggest that compensatory changes in kidney transporter expression occur to facilitate renal excretion during states of hepatic stress; however, little information is currently known regarding extrahepatic regulation of drug transporters in NASH. The purpose of the current study was to investigate the possibility of renal drug transporter regulation in NASH across multiple experimental rodent models. Both rat and mouse NASH models were used in this investigation and include: the methionine and choline-deficient (MCD) diet, atherogenic diet, fa/fa rat, ob/ob and db/db mice. Histologic and pathologic evaluations confirmed that the MCD and atherogenic rats as well as the ob/ob and db/db mice all developed NASH. In contrast, the fa/fa rats did not develop NASH but did develop extensive renal injury compared with the other models. Renal mRNA and protein analyses of xenobiotic transporters suggest that compensatory changes occur in NASH to favor increased xenobiotic secretion. Specifically, both apical efflux and basolateral uptake transporters are induced, whereas apical uptake transporter expression is repressed. These results suggest that NASH may alter the expression and potentially function of renal drug transporters, thereby impacting drug elimination mechanisms in the kidney.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Disease Models, Animal , Gene Expression Regulation , Kidney/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Gene Expression Profiling , Kidney/pathology , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Multidrug Resistance-Associated Proteins/genetics , Non-alcoholic Fatty Liver Disease/pathology , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats, Mutant Strains , Rats, Sprague-Dawley , Reproducibility of Results
10.
Amino Acids ; 47(3): 603-15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534430

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a globally widespread disease of increasing clinical significance. The pathological progression of the disease from simple steatosis to nonalcoholic steatohepatitis (NASH) has been well defined, however, the contribution of altered branched chain amino acid metabolomic profiles to the progression of NAFLD is not known. The three BCAAs: leucine, isoleucine and valine are known to mediate activation of several important hepatic metabolic signaling pathways ranging from insulin signaling to glucose regulation. The purpose of this study is to profile changes in hepatic BCAA metabolite levels with transcriptomic changes in the progression of human NAFLD to discover novel mechanisms of disease progression. Metabolomic and transcriptomic data sets representing the spectrum of human NAFLD (normal, steatosis, NASH fatty, and NASH not fatty livers) were utilized for this study. During the transition from steatosis to NASH, increases in the levels of leucine (127% of normal), isoleucine (139%), and valine (147%) were observed. Carnitine metabolites also exhibited significantly elevated profiles in NASH fatty and NASH not fatty samples and included propionyl, hexanoyl, lauryl, acetyl and butyryl carnitine. Amino acid and BCAA metabolism gene sets were significantly enriched among downregulated genes during NASH. These cumulative alterations in BCAA metabolite and amino acid metabolism gene profiles represent adaptive physiological responses to disease-induced hepatic stress in NASH patients.


Subject(s)
Isoleucine/metabolism , Leucine/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Valine/metabolism , Carnitine/genetics , Carnitine/metabolism , Female , Gene Expression Profiling , Humans , Isoleucine/genetics , Leucine/genetics , Male , Metabolomics , Non-alcoholic Fatty Liver Disease/genetics , Signal Transduction/genetics , Valine/genetics
11.
Toxicol Sci ; 142(1): 45-55, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25080921

ABSTRACT

Hepatic drug metabolizing enzymes and transporters play a crucial role in determining the fate of drugs, and alterations in liver function can place individuals at greater risk for adverse drug reactions (ADRs). We have shown that nonalcoholic steatohepatitis (NASH) leads to changes in the expression and localization of enzymes and transporters responsible for the disposition of numerous drugs. The purpose of this study was to determine the effect of NASH on methotrexate (MTX) disposition and the resulting toxicity profile. Sprague Dawley rats were fed either a control or methionine-choline-deficient diet for 8 weeks to induce NASH, then administered a single ip vehicle, 10, 40, or 100 mg/kg MTX injection followed by blood, urine, and feces collection over 96 h with terminal tissue collection. At the onset of dosing, Abcc1-4, Abcb1, and Abcg2 were elevated in NASH livers, whereas Abcc2 and Abcb1 were not properly localized to the membrane, similar to that previously observed in human NASH. NASH rodents receiving 40-100 mg/kg MTX exhibited hepatocellular damage followed by initiation of repair, whereas damage was absent in controls. NASH rodents receiving 100 mg/kg MTX exhibited slightly greater renal toxicity, indicating multiple organ toxicity, despite the majority of the dose being excreted by 6 h. Intestinal toxicity in NASH however, was strikingly less severe than controls, and coincided with reduced fecal MTX excretion. Because MTX-induced gastrointestinal toxicity limits the dose escalation necessary for cancer remission, these data suggest a greater risk for life-threatening MTX-induced hepatic and renal toxicity in NASH in the absence of overt gastrointestinal toxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Drug-Related Side Effects and Adverse Reactions/metabolism , Methotrexate/toxicity , Non-alcoholic Fatty Liver Disease/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Chemical and Drug Induced Liver Injury/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/etiology , Feces/chemistry , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Methotrexate/blood , Methotrexate/pharmacokinetics , Methotrexate/urine , Multidrug Resistance-Associated Protein 2 , Non-alcoholic Fatty Liver Disease/complications , Rats, Sprague-Dawley , Tissue Distribution
12.
J Hepatol ; 61(1): 139-47, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24613363

ABSTRACT

BACKGROUND & AIMS: A genome wide association study and multiple pharmacogenetic studies have implicated the hepatic uptake transporter organic anion transporting polypeptide-1B1 (OATP1B1) in the pharmacokinetics and musculoskeletal toxicity of statin drugs. Other OATP uptake transporters can participate in the transport of pravastatin, partially compensating for the loss of OATP1B1 in patients carrying the polymorphism. Non-alcoholic steatohepatitis (NASH) in humans and in a diet-induced rodent model alter the expression of multiple OATP transporters. METHODS: To determine how genetic alteration in one Oatp transporter can interact with NASH-associated changes in Oatp expression we measured the disposition of intravenously administered pravastatin in Slco1b2 knockout (Slco1b2(-/-)) and wild-type (WT) mice fed either a control or a methionine and choline deficient (MCD) diet to induce NASH. RESULTS: Genetic loss of Oatp1b2, the rodent ortholog of human OATP1B transporters, caused a modest increase in pravastatin plasma concentrations in mice with healthy livers. Although a diet-induced model of NASH decreased the expression of multiple hepatic Oatp transporters, it did not alter the disposition of pravastatin compared to WT control mice. In contrast, the combination of NASH-associated decrease in compensatory Oatp transporters and Oatp1b2 genetic loss caused a synergistic increase in plasma area under the curve (AUC) and tissue concentrations in kidney and muscle. CONCLUSIONS: Our data show that NASH alters the expression of multiple hepatic uptake transporters which, due to overlapping substrate specificity among the OATP transporters, may combine with the pharmacogenetic loss of OATP1B1 to increase the risk of statin-induced adverse drug reactions.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Organic Anion Transporters, Sodium-Independent/deficiency , Organic Anion Transporters, Sodium-Independent/genetics , Pravastatin/pharmacokinetics , Animals , Biological Transport, Active , Choline/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Liver/metabolism , Liver-Specific Organic Anion Transporter 1 , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscles/drug effects , Muscles/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Pravastatin/adverse effects , Pravastatin/blood
13.
Drug Metab Dispos ; 42(4): 586-95, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24384915

ABSTRACT

Nonalcoholic fatty liver disease is a prevalent form of chronic liver disease that can progress to the more advanced stage of nonalcoholic steatohepatitis (NASH). NASH has been shown to alter drug transporter regulation and may have implications in the development of adverse drug reactions. Several experimental rodent models have been proposed for the study of NASH, but no single model fully recapitulates all aspects of the human disease. The purpose of the current study was to determine which experimental NASH model best reflects the known alterations in human drug transporter expression to enable more accurate drug disposition predictions in NASH. Both rat and mouse NASH models were used in this investigation and include the methionine and choline deficient (MCD) diet model, atherogenic diet model, ob/ob and db/db mice, and fa/fa rats. Pathologic scoring evaluations demonstrated that MCD and atherogenic rats, as well as ob/ob and db/db mice, developed NASH. Liver mRNA and protein expression analyses of drug transporters showed that in general, efflux transporters were induced and uptake transporters were repressed in the rat MCD and the mouse ob/ob and db/db models. Lastly, concordance analyses suggest that both the mouse and rat MCD models as well as mouse ob/ob and db/db NASH models show the most similarity to human transporter mRNA and protein expression. These results suggest that the MCD rat and mouse model, as well as the ob/ob and db/db mouse models, may be useful for predicting altered disposition of drugs with similar kinetics across humans and rodents.


Subject(s)
Disease Models, Animal , Fatty Liver/etiology , Fatty Liver/metabolism , Organic Anion Transporters/metabolism , Pharmaceutical Preparations/metabolism , Animals , Choline Deficiency/complications , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diet, Atherogenic/adverse effects , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Obesity/complications , Obesity/genetics , Organic Anion Transporters/genetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
14.
J Pharmacol Exp Ther ; 348(3): 452-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24403518

ABSTRACT

Simvastatin (SIM)-induced myopathy is a dose-dependent adverse drug reaction (ADR) that has been reported to occur in 18.2% of patients receiving a 40- to 80-mg dose. The pharmacokinetics of SIM hydroxy acid (SIMA), the bioactive form of SIM, and the occurrence of SIM-induced myopathy are linked to the function of the organic anion transporting polypeptide (Oatp) hepatic uptake transporters. Genetic polymorphisms in SLCO1B1, the gene for human hepatic OATP1B1, cause decreased elimination of SIMA and increased risk of developing myopathy. Nonalcoholic steatohepatitis (NASH) is the most severe form of nonalcoholic fatty liver disease, and is known to alter drug transporter expression and drug disposition. The purpose of this study was to assess the metabolism and disposition of SIM in a diet-induced rodent model of NASH. Rats were fed a methionine- and choline-deficient diet for 8 weeks to induce NASH and SIM was administered intravenously. Diet-induced NASH caused increased plasma retention and decreased biliary excretion of SIMA due to decreased protein expression of multiple hepatic Oatps. SIM exhibited increased volume of distribution in NASH as evidenced by increased muscle, decreased plasma, and no change in biliary concentrations. Although Cyp3a and Cyp2c11 proteins were decreased in NASH, no alterations in SIM metabolism were observed. These data, in conjunction with our previous data showing that human NASH causes a coordinated downregulation of hepatic uptake transporters, suggest that NASH-mediated transporter regulation may play a role in altered SIMA disposition and the occurrence of myopathy.


Subject(s)
Fatty Liver/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Liver/metabolism , Organic Anion Transporters/metabolism , Simvastatin/analogs & derivatives , Animals , Down-Regulation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Male , Non-alcoholic Fatty Liver Disease , Rats , Rats, Sprague-Dawley , Simvastatin/adverse effects , Simvastatin/metabolism
15.
J Appl Toxicol ; 34(6): 726-32, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24217942

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a progressive form of non-alcoholic fatty liver disease (NAFLD) and is a major cause of liver cirrhosis and hepatic failure. The methionine choline-deficient diet (MCD) is a frequently used hepatotoxicity animal model of NASH that induces hepatic transaminase (ALT, AST) elevations and hepatobiliary histological changes similar to those observed in human NASH. Liver-specific microRNA-122 (miR-122) has been shown as a key regulator of cholesterol and fatty acid metabolism in adult liver, and has recently been proposed as a sensitive and specific circulating biomarker of hepatic injury. The purpose of this study was to assess miR-122 serum levels in mice receiving an MCD diet for 0, 3, 7, 14, 28 and 56 days and compare the performance vs. routine clinical chemistry when benchmarked against the histopathological liver findings. MiR-122 levels were quantified in serum using RT-qPCR. Both miR-122 and ALT/AST levels were significantly elevated in serum at all timepoints. MiR-122 levels increased on average by 40-fold after 3 days of initiating the MCD diet, whereas ALT and AST changes were 4.8- and 3.3-fold, respectively. In general, miR-122 levels remained elevated across all time points, whereas the ALT/AST increases were less robust but correlated with the progressive severity of NASH as assessed by histopathology. In conclusion, serum levels of miR-122 can potentially be used as a sensitive biomarker for the early detection of hepatotoxicity and can aid in monitoring the extent of NAFLD-associated liver injury in mouse efficacy models.


Subject(s)
Choline Deficiency/complications , Methionine , MicroRNAs/blood , Non-alcoholic Fatty Liver Disease/genetics , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Disease Models, Animal , Early Diagnosis , Genetic Markers , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/pathology , Predictive Value of Tests , Severity of Illness Index , Time Factors
16.
Dig Dis Sci ; 59(2): 365-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24048683

ABSTRACT

BACKGROUND: The worldwide prevalences of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are estimated to range from 30 to 40 % and 5-17 %, respectively. Hepatocellular carcinoma (HCC) is primarily caused by hepatitis B infection, but retrospective data suggest that 4-29 % of NASH cases will progress to HCC. Currently the connection between NASH and HCC is unclear. AIMS: The purpose of this study was to identify changes in expression of HCC-related genes and metabolite profiles in NAFLD progression. METHODS: Transcriptomic and metabolomic datasets from human liver tissue representing NAFLD progression (normal, steatosis, NASH) were utilized and compared to published data for HCC. RESULTS: Genes involved in Wnt signaling were downregulated in NASH but have been reported to be upregulated in HCC. Extracellular matrix/angiogenesis genes were upregulated in NASH, similar to reports in HCC. Iron homeostasis is known to be perturbed in HCC and we observed downregulation of genes in this pathway. In the metabolomics analysis of hepatic NAFLD samples, several changes were opposite to what has been reported in plasma of HCC patients (lysine, phenylalanine, citrulline, creatine, creatinine, glycodeoxycholic acid, inosine, and alpha-ketoglutarate). In contrast, multiple acyl-lyso-phosphatidylcholine metabolites were downregulated in NASH livers, consistent with observations in HCC patient plasma. CONCLUSIONS: These data indicate an overlap in the pathogenesis of NAFLD and HCC where several classes of HCC related genes and metabolites are altered in NAFLD. Importantly, Wnt signaling and several metabolites are different, thus implicating these genes and metabolites as mediators in the transition from NASH to HCC.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cluster Analysis , Databases, Genetic , Fatty Liver/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Metabolomics , Non-alcoholic Fatty Liver Disease , Signal Transduction/genetics
17.
Toxicol Sci ; 137(1): 26-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24097666

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) may progress from simple steatosis to severe, nonalcoholic steatohepatitis (NASH) in 7%-14% of the U.S. population through a second "hit" in the form of increased oxidative stress and inflammation. Endoplasmic reticulum (ER) stress signaling and the unfolded protein response (UPR) are triggered when high levels of lipids and misfolded proteins alter ER homeostasis creating a lipotoxic environment within NAFLD livers. The objective of this study was to determine the coordinate regulation of ER stress-associated genes in the progressive stages of human NAFLD. Human liver samples categorized as normal, steatosis, NASH (Fatty), and NASH (Not Fatty) were analyzed by individual Affymetrix GeneChip Human 1.0 ST microarrays, immunoblots, and immunohistochemistry. A gene set enrichment analysis was performed on autophagy, apoptosis, lipogenesis, and ER stress/UPR gene categories. An enrichment of downregulated genes in the ER stress-associated lipogenesis and ER stress/UPR gene categories was observed in NASH. Conversely, an enrichment of upregulated ER stress-associated genes for autophagy and apoptosis gene categories was observed in NASH. Protein expression of the adaptive liver response protein STC2 and the transcription factor X-box binding protein 1 spliced (XBP-1s) were significantly elevated among NASH samples, whereas other downstream ER stress proteins including CHOP, ATF4, and phosphorylated JNK and eIF2α were not significantly changed in disease progression. Increased nuclear accumulation of total XBP-1 protein was observed in steatosis and NASH livers. The findings reveal the presence of a coordinated, adaptive transcriptional response to hepatic ER stress in human NAFLD.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum/genetics , Fatty Liver/genetics , Lipogenesis/genetics , Liver/chemistry , Apoptosis/genetics , Autophagy/genetics , Blotting, Western , Cluster Analysis , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Profiling/methods , Gene Expression Regulation , Genetic Markers , Humans , Immunohistochemistry , Liver/pathology , Non-alcoholic Fatty Liver Disease , Oligonucleotide Array Sequence Analysis , Regulatory Factor X Transcription Factors , Signal Transduction/genetics , Transcription Factors/genetics , Transcription, Genetic , Unfolded Protein Response/genetics , X-Box Binding Protein 1
18.
J Pharmacol Exp Ther ; 347(1): 136-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23929842

ABSTRACT

Cholestasis results from interrupted bile flow and is associated with immune-mediated liver diseases. It is unclear how inflammation contributes to cholestasis. The aim of this study was to determine whether T and B cells contribute to hepatic transporter expression under basal and inflammatory conditions. C57BL/6J wild-type mice or strains lacking T, B, or both T and B cells were exposed to lipopolysaccharide (LPS) or saline, and livers were collected 16 hours later. Branched DNA signal amplification was used to assess mRNA levels of organic anion-transporting polypeptides (Oatp) 1a1, 1a4, and 1b2; organic cation transporter (Oct) 1; canalicular bile-salt export pump (Bsep); multidrug resistance-associated proteins (Mrp) 2 and 3; and sodium-taurocholate cotransporting polypeptide (Ntcp). Real-time polymerase chain reaction analysis was used to correlate changes of transporter expression with interleukin-1b (IL-1b), IL-6, IL-17A, IL-17F, tumor necrosis factor-α (TNF-α), and interferon-γ expression in the liver. LPS treatment inhibited Bsep and Oct1 mRNA expression, and this was abrogated with a loss of T cells, but not B cells. In addition, the absence of T cells increased Mrp2 mRNA expression, whereas B cell deficiency attenuated Oatp1a4 mRNA in LPS-treated mice. Oatp1a1, Oatp1b2, Ntcp, and Mrp3 were largely unaffected by T or B cell deficiency. Lymphocyte deficiency altered basal and inflammatory IL-6, but not TNF-α or IL-1b, mRNA expression. Taken together, these data implicate lymphocytes as regulators of basal and inflammatory hepatic transporter expression and suggest that IL-6 signaling may play a critical role.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Interleukin-6/biosynthesis , Lymphocytes/metabolism , Multidrug Resistance-Associated Proteins/biosynthesis , Octamer Transcription Factor-1/biosynthesis , Organic Cation Transport Proteins/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/physiology , Animals , Female , Gene Expression Regulation , Interleukin-6/physiology , Liver/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/physiology , Octamer Transcription Factor-1/physiology , Organic Cation Transport Proteins/physiology
19.
Toxicol Appl Pharmacol ; 268(2): 132-40, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23391614

ABSTRACT

Bile acids (BAs) have many physiological roles and exhibit both toxic and protective influences within the liver. Alterations in the BA profile may be the result of disease induced liver injury. Nonalcoholic fatty liver disease (NAFLD) is a prevalent form of chronic liver disease characterized by the pathophysiological progression from simple steatosis to nonalcoholic steatohepatitis (NASH). The hypothesis of this study is that the 'classical' (neutral) and 'alternative' (acidic) BA synthesis pathways are altered together with hepatic BA composition during progression of human NAFLD. This study employed the use of transcriptomic and metabolomic assays to study the hepatic toxicologic BA profile in progressive human NAFLD. Individual human liver samples diagnosed as normal, steatosis, and NASH were utilized in the assays. The transcriptomic analysis of 70 BA genes revealed an enrichment of downregulated BA metabolism and transcription factor/receptor genes in livers diagnosed as NASH. Increased mRNA expression of BAAT and CYP7B1 was observed in contrast to decreased CYP8B1 expression in NASH samples. The BA metabolomic profile of NASH livers exhibited an increase in taurine together with elevated levels of conjugated BA species, taurocholic acid (TCA) and taurodeoxycholic acid (TDCA). Conversely, cholic acid (CA) and glycodeoxycholic acid (GDCA) were decreased in NASH liver. These findings reveal a potential shift toward the alternative pathway of BA synthesis during NASH, mediated by increased mRNA and protein expression of CYP7B1. Overall, the transcriptomic changes of BA synthesis pathway enzymes together with altered hepatic BA composition signify an attempt by the liver to reduce hepatotoxicity during disease progression to NASH.


Subject(s)
Bile Acids and Salts/metabolism , Fatty Liver/metabolism , Liver/drug effects , Bile Acids and Salts/analysis , Bile Acids and Salts/genetics , Bile Acids and Salts/toxicity , Cluster Analysis , Disease Progression , Gene Expression Profiling , Humans , Metabolomics , Non-alcoholic Fatty Liver Disease
20.
Drug Metab Dispos ; 41(3): 554-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23223517

ABSTRACT

The UDP-glucuronosyltransferases (UGTs) and sulfotransferases (SULTs) represent major phase II drug-metabolizing enzymes that are also responsible for maintaining cellular homeostasis by metabolism of several endogenous molecules. Perturbations in the expression or function of these enzymes can lead to metabolic disorders and improper management of xenobiotics and endobiotics. Nonalcoholic fatty liver disease (NAFLD) represents a spectrum of liver damage ranging from steatosis to nonalcoholic steatohepatitis (NASH) and cirrhosis. Because the liver plays a central role in the metabolism of xenobiotics, the purpose of the current study was to determine the effect of human NAFLD progression on the expression and function of UGTs and SULTs in normal, steatosis, NASH (fatty), and NASH (not fatty/cirrhosis) samples. We identified upregulation of UGT1A9, 2B10, and 3A1 and SULT1C4 mRNA in both stages of NASH, whereas UGT2A3, 2B15, and 2B28 and SULT1A1, 2B1, and 4A1 as well as 3'-phosphoadenosine-5'-phosphosulfate synthase 1 were increased in NASH (not fatty/cirrhosis) only. UGT1A9 and 1A6 and SULT1A1 and 2A1 protein levels were decreased in NASH; however, SULT1C4 was increased. Measurement of the glucuronidation and sulfonation of acetaminophen (APAP) revealed no alterations in glucuronidation; however, SULT activity was increased in steatosis compared with normal samples, but then decreased in NASH compared with steatosis. In conclusion, the expression of specific UGT and SULT isoforms appears to be differentially regulated, whereas sulfonation of APAP is disrupted during progression of NAFLD.


Subject(s)
Fatty Liver/enzymology , Glucuronosyltransferase/metabolism , Liver/enzymology , Sulfotransferases/metabolism , Acetaminophen/metabolism , Biotransformation , Disease Progression , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression Regulation, Enzymologic , Glucuronides/metabolism , Humans , Isoenzymes , Liver/pathology , Non-alcoholic Fatty Liver Disease , RNA, Messenger/metabolism , Substrate Specificity , Sulfonic Acids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...