Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Med Chem ; 67(7): 5758-5782, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38511649

ABSTRACT

Eukaryotic translation initiation factor 2B (eIF2B) is a key component of the integrated stress response (ISR), which regulates protein synthesis and stress granule formation in response to cellular insult. Modulation of the ISR has been proposed as a therapeutic strategy for treatment of neurodegenerative diseases such as vanishing white matter (VWM) disease and amyotrophic lateral sclerosis (ALS) based on its ability to improve cellular homeostasis and prevent neuronal degeneration. Herein, we report the small-molecule discovery campaign that identified potent, selective, and CNS-penetrant eIF2B activators using both structure- and ligand-based drug design. These discovery efforts culminated in the identification of DNL343, which demonstrated a desirable preclinical drug profile, including a long half-life and high oral bioavailability across preclinical species. DNL343 was progressed into clinical studies and is currently undergoing evaluation in late-stage clinical trials for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Leukoencephalopathies , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Mutation , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Brain/metabolism , Leukoencephalopathies/metabolism
2.
J Med Chem ; 65(24): 16290-16312, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36469401

ABSTRACT

Dual leucine zipper kinase (DLK) and leucine zipper-bearing kinase (LZK) are regulators of neuronal degeneration and axon growth. Therefore, there is a considerable interest in developing DLK/LZK inhibitors for neurodegenerative diseases. Herein, we use ligand- and structure-based drug design approaches for identifying novel amino-pyrazine inhibitors of DLK/LZK. DN-1289 (14), a potent and selective dual DLK/LZK inhibitor, demonstrated excellent in vivo plasma half-life across species and is anticipated to freely penetrate the central nervous system with no brain impairment based on in vivo rodent pharmacokinetic studies and human in vitro transporter data. Proximal target engagement and disease relevant pathway biomarkers were also favorably regulated in an in vivo model of amyotrophic lateral sclerosis.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Humans , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Leucine Zippers , MAP Kinase Kinase Kinases , Central Nervous System/metabolism , Brain/metabolism , Neurodegenerative Diseases/metabolism
3.
Elife ; 72018 08 02.
Article in English | MEDLINE | ID: mdl-30070632

ABSTRACT

Reparative hepatocyte replication is impaired in chronic liver disease, contributing to disease progression; however, the underlying mechanism remains elusive. Here, we identify Map3k14 (also known as NIK) and its substrate Chuk (also called IKKα) as unrecognized suppressors of hepatocyte replication. Chronic liver disease is associated with aberrant activation of hepatic NIK pathways. We found that hepatocyte-specific deletion of Map3k14 or Chuk substantially accelerated mouse hepatocyte proliferation and liver regeneration following partial-hepatectomy. Hepatotoxin treatment or high fat diet feeding inhibited the ability of partial-hepatectomy to stimulate hepatocyte replication; remarkably, inactivation of hepatic NIK markedly increased reparative hepatocyte proliferation under these liver disease conditions. Mechanistically, NIK and IKKα suppressed the mitogenic JAK2/STAT3 pathway, thereby inhibiting cell cycle progression. Our data suggest that hepatic NIK and IKKα act as rheostats for liver regeneration by restraining overgrowth. Pathological activation of hepatic NIK or IKKα likely blocks hepatocyte replication, contributing to liver disease progression.


Subject(s)
End Stage Liver Disease/pathology , I-kappa B Kinase/physiology , Liver Failure, Acute/pathology , Liver Regeneration , Protein Serine-Threonine Kinases/physiology , Animals , Cells, Cultured , End Stage Liver Disease/etiology , End Stage Liver Disease/metabolism , Hepatectomy/adverse effects , Hepatocytes , Janus Kinase 2/metabolism , Liver Failure, Acute/etiology , Liver Failure, Acute/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , NF-kappaB-Inducing Kinase
4.
Drug Metab Dispos ; 45(12): 1317-1325, 2017 12.
Article in English | MEDLINE | ID: mdl-28986475

ABSTRACT

Variable drug responses depend on individual variation in the activity of drug-metabolizing enzymes, including cytochrome P450 enzymes (CYP). As the most common chronic liver disease in children and adults, nonalcoholic steatohepatitis (NASH) has been identified as a source of significant interindividual variation in hepatic drug metabolism. Compared with adults, children present age-related differences in pharmacokinetics and pharmacodynamics. The purpose of this study was to determine the impact of fatty liver disease severity on the activity of a variety of CYP enzymes in children and adolescents. Healthy and nonalcoholic fatty liver disease pediatric subjects aged 12-21 years inclusive received an oral cocktail of four probe drugs: caffeine (CYP1A2, 100 mg), omeprazole (CYP2C19, 20 mg), losartan (CYP2C9, 25 mg), and midazolam (CYP3A4, 2 mg). Venous blood and urine were collected before administration and 1, 2, 4, and 6 hours after administration. Concentrations of the parent drugs and CYP-specific metabolites were quantified in plasma and urine using liquid chromatography with tandem mass spectrometry. In plasma, the decreased metabolic area under the curve (AUC) ratio, defined as the metabolite AUC to parent AUC, of omeprazole indicated significant decreases of CYP2C19 (P = 0.002) enzymatic activities in NASH adolescents, while the urine analyses did not show significant differences and were highly variable. A comparison between the present in vivo pediatric studies and a previous ex vivo study in adults indicates distinct differences in the activities of CYP1A2 and CYP2C9. These data demonstrate that pediatric NASH presents an altered pattern of CYP activity and NASH should be considered as a confounder of drug metabolism for certain CYP enzymes. These differences could lead to future investigations that may reveal unexpected variable drug responses that should be considered in pediatric dosage recommendations.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Non-alcoholic Fatty Liver Disease/enzymology , Adolescent , Adult , Aging/metabolism , Area Under Curve , Child , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 Enzyme System/genetics , Female , Genotype , Humans , Male , Pharmaceutical Preparations/metabolism , Young Adult
5.
Drug Metab Dispos ; 43(6): 829-35, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25788542

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, representing a spectrum of liver pathologies that include simple hepatic steatosis and the more advanced nonalcoholic steatohepatitis (NASH). The current study was conducted to determine whether pediatric NASH also results in altered disposition of acetaminophen (APAP) and its two primary metabolites, APAP-sulfate and APAP-glucuronide. Pediatric patients with hepatic steatosis (n = 9) or NASH (n = 3) and healthy patients (n = 12) were recruited in a small pilot study design. All patients received a single 1000-mg dose of APAP. Blood and urine samples were collected at 1, 2, and 4 hours postdose, and APAP and APAP metabolites were determined by high-performance liquid chromatography. Moreover, human liver tissues from patients diagnosed with various stages of NAFLD were acquired from the Liver Tissue Cell Distribution System to investigate the regulation of the membrane transporters, multidrug resistance-associated protein 2 and 3 (MRP2 and MRP3, respectively). Patients with the more severe disease (i.e., NASH) had increased serum and urinary levels of APAP-glucuronide along with decreased serum levels of APAP-sulfate. Moreover, an induction of hepatic MRP3 and altered canalicular localization of the biliary efflux transporter, MRP2, describes the likely mechanism for the observed increase in plasma retention of APAP-glucuronide, whereas altered regulation of sulfur activation genes may explain decreased sulfonation activity in NASH. APAP-glucuronide and APAP-sulfate disposition is altered in NASH and is likely due to hepatic membrane transporter dysregulation as well as altered intracellular sulfur activation.


Subject(s)
Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Liver/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Up-Regulation , Acetaminophen/analogs & derivatives , Acetaminophen/blood , Acetaminophen/urine , Adolescent , Analgesics, Non-Narcotic/blood , Analgesics, Non-Narcotic/urine , Bile Canaliculi/metabolism , Bile Canaliculi/pathology , Biotransformation , Child , Cohort Studies , Fatty Liver/blood , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/urine , Female , Humans , Liver/pathology , Male , Multidrug Resistance-Associated Protein 2 , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/urine , Pilot Projects , Protein Transport
6.
Mol Metab ; 4(12): 951-60, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26909311

ABSTRACT

OBJECTIVE: Metabolic inflammation is believed to promote insulin resistance and type 2 diabetes progression in obesity. TRAF3, a cytoplasmic signaling protein, has been known to mediate/modulate cytokine signaling in immune cells. The goal is to define the metabolic function of hepatic TRAF3 in the setting of obesity. METHODS: Hepatocyte-specific TRAF3 knockout mice were generated using the loxp/albumin-cre system. Liver TRAF3 was deleted in adult obese mice via Cre adenoviral infection. Both high fat diet-induced and genetic obesity were examined. TRAF3 levels and insulin signaling were measured by immunoblotting. Insulin sensitivity, hepatic glucose production, and glucose metabolism were examined by glucose, insulin, and pyruvate tolerance tests. Hepatic steatosis was examined by Oil red O staining of liver sections and measuring liver triacylglycerol levels. RESULTS: Liver TRAF3 levels were lower in the fasted states in normal mice, and were aberrantly higher in obese mice and in mice with streptozotocin-induced hyperglycemia. Glucose directly increased TRAF3 levels in primary hepatocytes. Hepatocyte-specific deletion of TRAF3 decreased hyperinsulinemia, insulin resistance, glucose intolerance, and hepatic steatosis in mice with either high fat diet-induced obesity or genetic obesity (ob/ob); conversely, in lean mice, adenovirus-mediated overexpression of TRAF3 in the liver induced hyperinsulinemia, insulin resistance, and glucose intolerance. Deletion of TRAF3 enhanced the ability of insulin to stimulate phosphorylation of Akt in hepatocytes, whereas overexpression of TRAF3 suppressed insulin signaling. CONCLUSIONS: Glucose increases the levels of hepatic TRAF3. TRAF3 in turn promotes hyperglycemia through increasing hepatic glucose production, thus forming a glucose-TRAF3 reinforcement loop in the liver. This positive feedback loop may drive the progression of type 2 diabetes and nonalcoholic fatty liver disease in obesity.

7.
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
8.
Drug Metab Dispos ; 43(1): 93-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25349122

ABSTRACT

The ATP-binding cassette (ABC) family of transporters, including ABCC3, is a large family of efflux pumps that plays a pivotal role in the elimination of xenobiotics from the body. ABCC3 has been reported to be induced during hepatic stress conditions and through the progression of some forms of cancer. Several lines of evidence have implicated the transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in this induction. However, although rodent models have been investigated, a functional antioxidant response element (ARE) in the human ABCC3 gene has not been identified. The purpose of this study was to identify and characterize the ARE(s) responsible for mediating the Nrf2-dependent induction of the human ABCC3 gene. A high-throughput chromatin immunoprecipitation-sequencing analysis performed in A549 cells revealed a specific interaction between Nrf2 and the eighth intron of the human ABCC3 gene rather than the more prototypical flanking region of the gene. Subsequent in silico analysis of the intron identified two putative ARE elements that contained the core consensus ARE sequence commonly found in several Nrf2-responsive genes. Functional characterization of these two AREs using luciferase-reporter constructs with ARE mutant constructs revealed that one of these putative AREs is functionally active. Finally, DNA pull-down assays confirmed specific binding of these intronic AREs by Nrf2 in vitro. Our findings identify a functional Nrf2 response element within the eighth intron of the ABCC3 gene, which may provide mechanistic insight into the induction of ABCC3 during antioxidant response stimuli.


Subject(s)
Antioxidant Response Elements/genetics , Introns/genetics , Multidrug Resistance-Associated Proteins/genetics , Antioxidants/metabolism , Cell Line, Tumor , Humans , NF-E2-Related Factor 2/genetics , Transcription Factors/genetics
9.
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
10.
Expert Opin Drug Metab Toxicol ; 10(9): 1209-19, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24989624

ABSTRACT

INTRODUCTION: The pharmacokinetics (PK) of drugs and xenobiotics, namely pharmaceuticals, is influenced by a host of factors that include genetics, physiological factors and environmental stressors. The importance of disease on the disposition of xenobiotics has been increasingly recognized among medical professionals for alterations in key enzymes and membrane transporters that influence drug disposition and contribute to the development of adverse drug reactions. AREAS COVERED: This review will survey pertinent literature of how liver disease alters the PKs of drugs and other xenobiotics. The focus will be on nonalcoholic steatohepatitis as well as cholestatic liver diseases. A review of basic pharmacokinetic principles, with a special emphasis on xenobiotic metabolizing enzymes and membrane transporters, will be provided. Specifically, examples of how genetic alterations affect metabolism and excretion, respectively, will be highlighted. Lastly, the idea of 'extrahepatic' regulation will be explored, citing examples of how disease manifestation in the liver may affect drug disposition in distal sites, such as the kidney. EXPERT OPINION: An expert opinion will be provided highlighting the definite need for data in understanding extrahepatic regulation of membrane transporters in the presence of liver disease and its potential to dramatically alter the PK and toxicokinetic profile of numerous drugs and xenobiotics.


Subject(s)
Cholestasis/physiopathology , Non-alcoholic Fatty Liver Disease/physiopathology , Xenobiotics/pharmacokinetics , Animals , Humans , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Xenobiotics/adverse effects
11.
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
12.
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
13.
Am J Physiol Gastrointest Liver Physiol ; 304(1): G48-56, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23125159

ABSTRACT

Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of premature infants and is characterized by an extensive hemorrhagic inflammatory necrosis of the distal ileum and proximal colon. We have previously shown that, during the development of experimental NEC, the liver plays an important role in regulating inflammation in the ileum, and accumulation of ileal bile acids (BA) along with dysregulation of ileal BA transporters contributes to ileal damage. Given these findings, we speculated that hepatic BA transporters would also be altered in experimental NEC. Using both rat and mouse models of NEC, levels of Cyp7a1, Cyp27a1, and the hepatic BA transporters Bsep, Ntcp, Oatp2, Oatp4, Mrp2, and Mrp3 were investigated. In addition, levels of hepatic BA transporters were also determined when the proinflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-18, which are both elevated in NEC, are neutralized during disease development. Ntcp and Mrp2 were decreased in NEC, but elevated ileal BA levels were not responsible for these reductions. However, neutralization of TNF-α normalized Ntcp, whereas removal of IL-18 normalized Mrp2 levels. These data show that the hepatic transporters Ntcp and Mrp2 are downregulated, whereas Cyp27a1 is increased in rodent models of NEC. Furthermore, increased levels of TNF-α and IL-18 in experimental NEC may play a role in the regulation of Ntcp and Mrp2, respectively. These data suggest the gut-liver axis should be considered when therapeutic modalities for NEC are developed.


Subject(s)
Enterocolitis, Necrotizing/metabolism , Liver/metabolism , Multidrug Resistance-Associated Proteins/biosynthesis , Organic Anion Transporters, Sodium-Dependent/biosynthesis , Symporters/biosynthesis , Animals , Bile Acids and Salts/metabolism , Blotting, Western , Carrier Proteins/metabolism , Cholestanetriol 26-Monooxygenase/metabolism , DNA/biosynthesis , DNA/genetics , Down-Regulation , Enterocolitis, Necrotizing/pathology , Enterocytes/metabolism , Enterocytes/pathology , Interleukin-18/genetics , Interleukin-18/metabolism , Liver/pathology , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , RNA/biosynthesis , RNA/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology
14.
Drug Metab Dispos ; 40(9): 1817-24, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22699396

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is represented by a spectrum of liver pathologies ranging from simple steatosis to nonalcoholic steatohepatitis (NASH). Liver damage sustained in the progressive stages of NAFLD may alter the ability of the liver to properly metabolize and eliminate xenobiotics. The purpose of the current study was to determine whether NAFLD alters the disposition of the environmental toxicant arsenic. C57BL/6 mice were fed either a high-fat or a methionine-choline-deficient diet to model simple steatosis and NASH, respectively. At the conclusion of the dietary regimen, all mice were given a single oral dose of either sodium arsenate or arsenic trioxide. Mice with NASH excreted significantly higher levels of total arsenic in urine (24 h) compared with controls. Total arsenic in the liver and kidneys of NASH mice was not altered; however, NASH liver retained significantly higher levels of the monomethyl arsenic metabolite, whereas dimethyl arsenic was retained significantly less in the kidneys of NASH mice. NASH mice had significantly higher levels of the more toxic trivalent form in their urine, whereas the pentavalent form was preferentially retained in the liver of NASH mice. Moreover, hepatic protein expression of the arsenic biotransformation enzyme arsenic (3+ oxidation state) methyltransferase was not altered in NASH animals, whereas protein expression of the membrane transporter multidrug resistance-associated protein 1 was increased, implicating cellular transport rather than biotransformation as a possible mechanism. These results suggest that NASH alters the disposition of arsenical species, which may have significant implications on the overall toxicity associated with arsenic in NASH.


Subject(s)
Arsenates/pharmacokinetics , Arsenicals/pharmacokinetics , Environmental Pollutants/pharmacokinetics , Fatty Liver/metabolism , Liver/metabolism , Oxides/pharmacokinetics , Animals , Arsenates/toxicity , Arsenates/urine , Arsenic Trioxide , Arsenicals/urine , Biotransformation , Choline Deficiency/complications , Diet, High-Fat , Disease Models, Animal , Environmental Pollutants/toxicity , Environmental Pollutants/urine , Fatty Liver/etiology , Fatty Liver/urine , Kidney/metabolism , Liver/drug effects , Liver/pathology , Male , Methionine/deficiency , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Multidrug Resistance-Associated Proteins/metabolism , Non-alcoholic Fatty Liver Disease , Oxides/toxicity , Oxides/urine
15.
Drug Metab Dispos ; 40(3): 450-60, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22112382

ABSTRACT

Ezetimibe (EZE) lowers serum lipid levels by blocking cholesterol uptake in the intestine. Disposition of EZE and its pharmacologically active glucuronide metabolite (EZE-GLUC) to the intestine is dependent on hepatobiliary efflux. Previous studies suggested that hepatic transporter expression and function may be altered during nonalcoholic steatohepatitis (NASH). The purpose of the current study was to determine whether NASH-induced changes in the expression and function of hepatic transporters result in altered disposition of EZE and EZE-GLUC. Rats fed a methionine- and choline-deficient (MCD) diet for 8 weeks were administered 10 mg/kg EZE either by intravenous bolus or oral gavage. Plasma and bile samples were collected over 2 h followed by terminal urine and tissue collection. EZE and EZE-GLUC concentrations were determined by liquid chromatography-tandem mass spectrometry. The sinusoidal transporter Abcc3 was induced in MCD rats, which correlated with increased plasma concentrations of EZE-GLUC, regardless of dosing method. Hepatic expression of the biliary transporters Abcc2 and Abcb1 was also increased in MCD animals, but the biliary efflux of EZE-GLUC was slightly diminished, whereas biliary bile acid concentrations were unaltered. The cellular localization of Abcc2 and Abcb1 appeared to be internalized away from the canalicular membrane in MCD livers, providing a mechanism for the shift to plasma drug efflux. The combination of induced expression and altered localization of efflux transporters in NASH shifts the disposition profile of EZE-GLUC toward plasma retention away from the site of action. This increased plasma retention of drugs in NASH may have implications for the pharmacological effect and safety of numerous drugs.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Azetidines/pharmacokinetics , Fatty Liver/metabolism , Animals , Anticholesteremic Agents/metabolism , Azetidines/metabolism , Bile Acids and Salts/metabolism , Biliary Tract/metabolism , Biological Transport , Choline Deficiency/metabolism , Diet , Ezetimibe , Fatty Liver/genetics , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Methionine/deficiency , Methionine/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Non-alcoholic Fatty Liver Disease , Rats , Rats, Sprague-Dawley
16.
Drug Metab Dispos ; 39(12): 2395-402, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21878559

ABSTRACT

Transporters located on the sinusoidal and canalicular membranes of hepatocytes regulate the efflux of drugs and metabolites into blood and bile, respectively. Changes in the expression or function of these transporters during liver disease may lead to a greater risk of adverse drug reactions. Nonalcoholic fatty liver disease (NAFLD) is a progressive condition encompassing the relatively benign steatosis and the more severe, inflammatory state of nonalcoholic steatohepatitis (NASH). Here, we present an analysis of the effect of NAFLD progression on the major ATP-binding cassette (ABC) efflux transport proteins ABCC1-6, ABCB1, and ABCG2. Human liver samples diagnosed as normal, steatotic, NASH (fatty), and NASH (not fatty) were analyzed. Increasing trends in mRNA expression of ABCC1, ABCC4-5, ABCB1, and ABCG2 were found with NAFLD progression, whereas protein levels of all transporters exhibited increasing trends with disease progression. Immunohistochemical staining of ABCC3, ABCB1, and ABCG2 revealed no alterations in cellular localization during NAFLD progression. ABCC2 staining revealed an alternative mechanism of regulation in NASH in which the transporter appears to be internalized away from the canalicular membrane. This correlated with a preferential shift in the molecular mass of ABCC2 from 200 to 180 kDa in NASH, which has been shown to be associated with a loss of glycosylation and internalization of the protein. These data demonstrate increased expression of multiple efflux transporters as well as altered cellular localization of ABCC2 in NASH, which may have profound effects on the ability of patients with NASH to eliminate drugs in an appropriate manner.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Fatty Liver/metabolism , Disease Progression , Humans , Multidrug Resistance-Associated Protein 2 , Non-alcoholic Fatty Liver Disease
17.
Drug Metab Dispos ; 38(12): 2293-301, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20805291

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), which occurs in approximately 17 to 40% of Americans, encompasses progressive stages of liver damage ranging from steatosis to nonalcoholic steatohepatitis (NASH). Inflammation and oxidative stress are known characteristics of NAFLD; however, the precise mechanisms occurring during disease progression remain unclear. The purpose of the current study was to determine whether the expression or function of enzymes involved in the antioxidant response, NAD(P)H:quinone oxidoreductase 1 (NQO1), glutathione transferase (GST), and glutamate cysteine ligase, are altered in the progression of human NAFLD. Human livers staged as normal, steatotic, NASH (fatty), and NASH (not fatty) were obtained from the Liver Tissue Cell Distribution System. NQO1 mRNA, protein, and activity tended to increase with disease progression. mRNA levels of the GST isoforms A1, A2, A4, M3, and P1 increased with NAFLD progression. Likewise, GST A and P protein increased with progression; however, GST M protein levels tended to decrease. Of interest, total GST activity toward the substrate 1-chloro-2,4-dinitrobenzene decreased with NAFLD progression. GSH synthesis does not seem to be significantly dysregulated in NAFLD progression; however, the GSH/oxidized glutathione redox ratio seemed to be reduced with disease severity, indicating the presence of oxidative stress and depletion of GSH throughout progression of NAFLD. Malondialdehyde concentrations were significantly increased with disease progression, further indicating the presence of oxidative stress. Nuclear immunohistochemical staining of nuclear factor E2-related factor 2 (Nrf2), an indicator of activation of the transcription factor, was evident in all stages of NAFLD. The current data suggest that Nrf2 activation occurs in response to disease progression followed by induction of specific Nrf2 targets, whereas functionality of specific antioxidant defense enzymes seems to be impaired as NAFLD progresses.


Subject(s)
Antioxidants/metabolism , Fatty Liver/enzymology , Adult , Disease Progression , Fatty Liver/pathology , Glutamate-Cysteine Ligase/analysis , Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Glutathione Transferase/analysis , Glutathione Transferase/genetics , Humans , NAD(P)H Dehydrogenase (Quinone)/analysis , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...