Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(7)2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38612422

ABSTRACT

As compounds of natural origin enter human body, it is necessary to investigate their possible interactions with the metabolism of drugs and xenobiotics in general, namely with the cytochrome P450 (CYP) system. Phytic acid (myo-inositol hexaphosphoric acid, IP6) is mainly present in plants but is also an endogenous compound present in mammalian cells and tissues. It has been shown to exhibit protective effect in many pathological conditions. For this paper, its interaction with CYPs was studied using human liver microsomes, primary human hepatocytes, the HepG2 cell line, and molecular docking. Docking experiments and absorption spectra demonstrated the weak ability of IP6 to interact in the heme active site of CYP1A. Molecular docking suggested that IP6 preferentially binds to the protein surface, whereas binding to the active site of CYP1A2 was found to be less probable. Subsequently, we investigated the ability of IP6 to modulate the metabolism of xenobiotics for both the mRNA expression and enzymatic activity of CYP1A enzymes. Our findings revealed that IP6 can slightly modulate the mRNA levels and enzyme activity of CYP1A. However, thanks to the relatively weak interactions of IP6 with CYPs, the chances of the mechanisms of clinically important drug-drug interactions involving IP6 are low.


Subject(s)
Phytic Acid , Xenobiotics , Humans , Animals , Molecular Docking Simulation , Cytochrome P-450 Enzyme System , RNA, Messenger , Mammals
2.
J Nutr Biochem ; 107: 109042, 2022 09.
Article in English | MEDLINE | ID: mdl-35533897

ABSTRACT

Modulation of gut microbiome composition seems to be a promising therapeutic strategy for a wide range of pathologic states. However, these microbiota-targeted interventions may affect production of microbial metabolites, circulating factors in the gut-liver axis influencing hepatic drug metabolism with possible clinical relevance. Butyrate, a short-chain fatty acid produced through microbial fermentation of dietary fibers in the colon, has well established anti-inflammatory role in the intestine, while the effect of butyrate on the liver is unknown. In this study, we have evaluated the effect of butyrate on hepatic AhR activity and AhR-regulated gene expression. We have showed that AhR and its target genes were upregulated by butyrate in dose-dependent manner in HepG2-C3 as well as in primary human hepatocytes. The involvement of AhR has been proved using specific AhR antagonists and siRNA-mediated AhR silencing. Experiments with AhR reporter cells have shown that butyrate regulates the expression of AhR target genes by modulating the AhR activity. Our results suggest also epigenetic action by butyrate on AhR and its repressor (AHRR) presumably through mechanisms based on HDAC inhibition in the liver. Our results demonstrate that butyrate may influence the drug-metabolizing ability of liver enzymes e.g., through the interaction with AhR-dependent pathways.


Subject(s)
Butyrates , Gastrointestinal Microbiome , Butyrates/metabolism , Butyrates/pharmacology , Colon/metabolism , Fatty Acids, Volatile/metabolism , Humans , Liver/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
3.
Curr Res Toxicol ; 2: 149-158, 2021.
Article in English | MEDLINE | ID: mdl-34345857

ABSTRACT

The hepatic cytochrome p450's (CYP) are of major importance for the metabolism of xenobiotics and knowledge about their regulation is crucial. This knowledge often originates from cell models; primary human hepatocytes (PHH) being the gold standard. However, due to limited availability of high-quality human donor organs, basic knowledge on alternative models are needed. Primary porcine hepatocytes (PPH) have been suggested as an alternative to PHH. Unfortunately, data comparing the response in gene-transcription to standard CYP inducers between PHH and PPH are missing. In the present study we, cultured PHH and PPH under the same conditions, treated them with standard inducers of the CYP1-3 and determined the response in gene and protein expression. The results demonstrated that in both species TCDD and omeprazole caused an increase in CYP1A/B expression. In PPH, CITCO increased the content of CYP1A/B. For the CYP2B/C/D's, phenobarbital and rifampicin caused increases in expression. For the CYP2D's, TCDD and omeprazole caused increased gene expression in PPH, which were not the case for PHH. Both phenobarbital, rifampicin and omeprazole increased CYP3A expression in PHH and PPH. Moreover, TCDD increased the gene expression of CYP3A in PPH; this was not the case for PHH. Multivariate data analysis found no difference in gene expression between PHH and PPH for phenobarbital, rifampicin and CITCO. However, differential clustering was observed for TCDD and omeprazole. In conclusion, despite model specificity, there are a high number of similar responses, and experiments investigating mRNA regulation made in PPH permits for a reliable translation into human setting.

4.
Biotechniques ; 69(6): 436-442, 2020 12.
Article in English | MEDLINE | ID: mdl-33103926

ABSTRACT

The aim of this study was to assess two protocols for their capacities to simultaneously isolate RNA, mtDNA and ncDNA from mammalian cells. We compared the Invitrogen TRIzol-based method and Qiagen DNeasy columns, using the HepG2 cell line and human primary glioblastoma stem cells. Both methods allowed the isolation of all three types of nucleic acids and provided similar yields in mtDNA. However, the yield in ncDNA was more than tenfold higher on columns, as observed for both cell types. Conversely, the TRIzol method proved more reproducible and was the method of choice for isolating RNA from glioblastoma cells, as demonstrated for the housekeeping genes RPLP0 and RPS9.


Subject(s)
Biochemistry/methods , Cell Nucleus/metabolism , DNA, Mitochondrial/isolation & purification , Mammals/metabolism , RNA/isolation & purification , Animals , Glioblastoma/metabolism , Glioblastoma/pathology , Hep G2 Cells , Humans , Neoplastic Stem Cells/metabolism , RNA, Messenger/isolation & purification , Reagent Kits, Diagnostic
5.
Food Chem Toxicol ; 141: 111403, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32387306

ABSTRACT

The prevalence of metabolic syndrome (MetS), elevating cardiovascular risks, is increasing worldwide, with no available global therapeutic options. The intake of plain, mineral or biocompatible modified waters was shown to prevent some MetS features. This study was designed to analyze, in mice fed a high fat and sucrose diet (HFSD), the effects on MetS features of the daily intake of a reverse osmosed, weakly remineralized, water (OW) and of an OW dynamized by a physical processing (ODW), compared to tap water (TW). The HFSD was effective at inducing major features of MetS such as obesity, hepatic steatosis and inflammation, blood dyslipidemia, systemic glucose intolerance and muscle insulin resistance. Compared to TW, OW intake decreased hepatic fibrosis and inflammation, and mitigated hepatic steatosis and dyslipidemia. ODW intake further improved skeletal muscle insulin sensitivity and systemic glucose tolerance. This study highlights the deleterious metabolic impacts of the daily intake of TW, in combination with a high energy diet, and its possible involvement in MetS prevalence increase. In addition, it demonstrates that biocompatible modified water may be promising non-pharmaceutical, cost-effective tools for nutritional approaches in the treatment of MetS.


Subject(s)
Biocompatible Materials , Diet, High-Fat , Drinking Water , Metabolic Syndrome/prevention & control , Obesity/etiology , Animals , Basal Metabolism , Biomarkers/metabolism , Insulin Resistance , Lipogenesis , Liver Glycogen/metabolism , Male , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Obesity/complications
6.
Drug Metab Dispos ; 46(3): 223-236, 2018 03.
Article in English | MEDLINE | ID: mdl-29269410

ABSTRACT

Growth factors have key roles in liver physiology and pathology, particularly by promoting cell proliferation and growth. Recently, it has been shown that in mouse hepatocytes, epidermal growth factor receptor (EGFR) plays a crucial role in the activation of the xenosensor constitutive androstane receptor (CAR) by the antiepileptic drug phenobarbital. Due to the species selectivity of CAR signaling, here we investigated epidermal growth factor (EGF) role in CAR signaling in primary human hepatocytes. Primary human hepatocytes were incubated with CITCO, a human CAR agonist, or with phenobarbital, an indirect CAR activator, in the presence or absence of EGF. CAR-dependent gene expression modulation and PXR involvement in these responses were assessed upon siRNA-based silencing of the genes that encode CAR and PXR. EGF significantly reduced CAR expression and prevented gene induction by CITCO and, to a lower extent, by phenobarbital. In the absence of EGF, phenobarbital and CITCO modulated the expression of 144 and 111 genes, respectively, in primary human hepatocytes. Among these genes, only 15 were regulated by CITCO and one by phenobarbital in a CAR-dependent manner. Conversely, in the presence of EGF, CITCO and phenobarbital modulated gene expression only in a CAR-independent and PXR-dependent manner. Overall, our findings suggest that in primary human hepatocytes, EGF suppresses specifically CAR signaling mainly through transcriptional regulation and drives the xenobiotic response toward a pregnane X receptor (PXR)-mediated mechanism.


Subject(s)
Epidermal Growth Factor/metabolism , Hepatocytes/metabolism , Peroxisome-Targeting Signal 1 Receptor/metabolism , Recoverin/metabolism , Adult , Aged , Cells, Cultured , ErbB Receptors/metabolism , Female , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Humans , Male , Middle Aged , Oximes/pharmacology , Phenobarbital/pharmacology , Signal Transduction/drug effects , Thiazoles/pharmacology , Transcription, Genetic/drug effects
7.
J Tissue Eng Regen Med ; 12(4): 1062-1073, 2018 04.
Article in English | MEDLINE | ID: mdl-29106037

ABSTRACT

Liver failure remains the leading cause of post-operative mortality after hepatectomy. This study investigated the effect of treatment with allogenic mesenchymal stem cells (MSCs) on survival and liver regeneration 48 hr and 7 days after 80% hepatectomy in C57Bl/6 mice. To optimize their biodistribution, MSCs were grown on acellular human amniotic membranes (HAM) and applied as a patch on the remnant liver. This approach was compared with MSC infusion and HAM patch alone. Hepatectomized mice without any treatment were used as control group. Survival rate was calculated and biological and histopathological parameters were analysed to monitor liver function and regeneration. MSCs grown on HAM retained their ability to proliferate, to differentiate into osteoblasts and adipocytes and to respond to pro-inflammatory stimuli. Extended hepatectomy (80%) led to liver failure that resulted in death within 72 hr in 76% of mice. MSC infusion showed an early but transitory positive effect on survival. MSC/HAM patches stimulated regeneration and significantly improved survival rate (54% vs. 24% in the control group at 7 days). They also decreased the severity of hepatectomy-induced steatosis, suggesting a modulation of lipid metabolism in hepatocytes. MSCs were still present on HAM at Days 2 and 7 posthepatectomy. In conclusion, engineered tissue constructs that combine MSCs and HAM improve survival and liver regeneration after 80% hepatectomy in mice. These encouraging results pave the way to potential clinical application.


Subject(s)
Amnion , Hepatectomy , Liver Regeneration , Liver , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Allografts , Animals , Humans , Liver/metabolism , Liver/surgery , Mice , Mice, Transgenic
8.
Nat Commun ; 7: 12203, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27417143

ABSTRACT

The accumulation of lipid droplets (LD) is frequently observed in hepatitis C virus (HCV) infection and represents an important risk factor for the development of liver steatosis and cirrhosis. The mechanisms of LD biogenesis and growth remain open questions. Here, transcriptome analysis reveals a significant upregulation of septin 9 in HCV-induced cirrhosis compared with the normal liver. HCV infection increases septin 9 expression and induces its assembly into filaments. Septin 9 regulates LD growth and perinuclear accumulation in a manner dependent on dynamic microtubules. The effects of septin 9 on LDs are also dependent on binding to PtdIns5P, which, in turn, controls the formation of septin 9 filaments and its interaction with microtubules. This previously undescribed cooperation between PtdIns5P and septin 9 regulates oleate-induced accumulation of LDs. Overall, our data offer a novel route for LD growth through the involvement of a septin 9/PtdIns5P signalling pathway.


Subject(s)
Hepacivirus/pathogenicity , Lipid Droplets/metabolism , Microtubules/metabolism , Phosphatidylinositol Phosphates/metabolism , Septins/metabolism , Cell Line, Tumor , Gene Expression Regulation , Hepacivirus/physiology , Hepatitis C/metabolism , Host-Pathogen Interactions/physiology , Humans , Lipid Droplets/drug effects , Lipid Metabolism/physiology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/virology , Microtubules/virology , Oleic Acid/pharmacology , Septins/genetics , Virus Replication
9.
Toxicol Sci ; 148(1): 261-75, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26259606

ABSTRACT

Cytochrome P450 (CYP) expression and activity are not homogeneous in the liver lobules. Indeed, CYPs are mainly expressed and induced in centrilobular hepatocytes. The wingless-type MMTV integration site family (WNT)/ß-catenin pathway was identified as a major regulator of this zonal organization. We have recently demonstrated that in primary human hepatocytes (PHHs), the expression of CYP2E1, CYP1A2, and aryl hydrocarbon receptor (AhR), but not of CYP3A4, is regulated by the WNT/ß-catenin pathway in response to WNT3a, its canonical activator. Here, we investigated whether glycogen synthase kinase 3ß (GSK3ß) inhibitors, which mimic the action of WNT molecules, could be used in PHHs to activate the ß-catenin pathway to study CYP expression. We assessed the activity of 6BIO (6-bromoindirubin-3'-oxime), CHIR99021 (6-((2-((4-(2,4-dichlorophenyl)-5-(4methyl-1H-imidazol-2-yl)pyrimidin-2-yl)amino)ethyl)amino) nicotinonitrile), and GSK3iXV (Pyridocarbazolo-cyclopentadienyl Ruthenium complex GSK3 inhibitor XV) that belong to structurally different families of GSK3ß inhibitors. Using small interfering RNAs, reporter gene assays, and molecular docking predictions, we demonstrated that GSK3ß inhibitors can activate the WNT/ß-catenin pathway in PHHs to regulate CYP2E1 expression. We also found that 6BIO and GSK3iXV are AhR full agonists that participate, through AhR signaling, to CYP1A2 induction. Conversely, CHIR99021 is an AhR partial agonist, and a pregnane X receptor ligand and partial agonist, thus regulating CYP1A2 and CYP3A4 gene expression in a ß-catenin-independent manner. In conclusion, GSK3ß inhibitors can activate the WNT/ß-catenin pathway in PHHs. Nevertheless, their role in CYP regulation should be analyzed with caution as these molecules can interact with xenosensors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/agonists , Cytochrome P-450 Enzyme Inducers/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hepatocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Steroid/agonists , beta Catenin/agonists , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cells, Cultured , Cytochrome P-450 Enzyme Inducers/chemistry , Cytochrome P-450 Enzyme Inducers/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Enzyme Induction/drug effects , Female , Genes, Reporter/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Indoles/pharmacology , Male , Molecular Docking Simulation , Organometallic Compounds/pharmacology , Oximes/pharmacology , Pregnane X Receptor , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA Interference , Receptors, Aryl Hydrocarbon/chemistry , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
10.
Mol Pharmacol ; 86(6): 624-34, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25228302

ABSTRACT

The wingless-type MMTV integration site family (WNT)/ß-catenin/adenomatous polyposis coli (CTNNB1/APC) pathway has been identified as a regulator of drug-metabolizing enzymes in the rodent liver. Conversely, little is known about the role of this pathway in drug metabolism regulation in human liver. Primary human hepatocytes (PHHs), which are the most physiologically relevant culture system to study drug metabolism in vitro, were used to investigate this issue. This study assessed the link between cytochrome P450 expression and WNT/ß-catenin pathway activity in PHHs by modulating its activity with recombinant mouse Wnt3a (the canonical activator), inhibitors of glycogen synthase kinase 3ß, and small-interfering RNA to invalidate CTNNB1 or its repressor APC, used separately or in combination. We found that the WNT/ß-catenin pathway can be activated in PHHs, as assessed by universal ß-catenin target gene expression, leucine-rich repeat containing G protein-coupled receptor 5. Moreover, WNT/ß-catenin pathway activation induces the expression of CYP2E1, CYP1A2, and aryl hydrocarbon receptor, but not of CYP3A4, hepatocyte nuclear factor-4α, or pregnane X receptor (PXR) in PHHs. Specifically, we show for the first time that CYP2E1 is transcriptionally regulated by the WNT/ß-catenin pathway. Moreover, CYP2E1 induction was accompanied by an increase in its metabolic activity, as indicated by the increased production of 6-OH-chlorzoxazone and by glutathione depletion after incubation with high doses of acetaminophen. In conclusion, the WNT/ß-catenin pathway is functional in PHHs, and its induction in PHHs represents a powerful tool to evaluate the hepatotoxicity of drugs that are metabolized by CYP2E1.


Subject(s)
Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP2E1/genetics , Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Receptors, Aryl Hydrocarbon/genetics , Wnt Signaling Pathway/physiology , beta Catenin/physiology , Adult , Aged , Cell Line , Cytochrome P-450 CYP3A/genetics , Female , Humans , Male , Middle Aged
11.
Gut ; 63(9): 1490-500, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24153249

ABSTRACT

OBJECTIVE: Adult primary human hepatocytes (PHHs) support the complete infection cycle of natural HCV from patients' sera. The molecular details underlying sera infectivity towards these cells remain largely unknown. Therefore, we sought to gain a deeper comprehension of these features in the most physiologically relevant culture system. DESIGN: Using kinetic experiments, we defined the optimal conditions to infect PHH and explored the link between cell organisation and permissivity. Based on their infectivity, about 120 sera were classified in three groups. Concentration of 52 analytes was measured in 79 selected sera using multiplexed immunobead-based analyte profiling. RESULTS: PHH permissivity towards HCV infection negatively correlated with cell polarisation and formation of functional bile canaliculi. PHH supported HCV replication for at least 2 weeks with de novo virus production. Depending on their reactivity, sera could be classified in three groups of high, intermediate or low infectivity toward PHH. Infectivity could not be predicted based on the donors' clinical characteristics, viral load or genotype. Interestingly, highly infectious sera displayed a specific cytokine profile with low levels of most of the 52 tested analytes. Among them, 24 cytokines/growth factors could impact hepatocyte biology and infection efficiency. CONCLUSIONS: We identified critical factors leading to efficient PHH infection by HCV sera in vitro. Overall, we showed that this cellular model provides a useful tool for studying the mechanism of HCV infection in its natural host cell, selecting highly infectious isolates, and determining the potency of drugs towards various HCV strains.


Subject(s)
Hepacivirus/pathogenicity , Hepatocytes/virology , Adult , Biomarkers/metabolism , Cell Culture Techniques/methods , Cell Line , Cells, Cultured , Cytokines/metabolism , Hepacivirus/metabolism , Hepatocytes/physiology , Humans , Kinetics , Models, Immunological , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serum/virology
12.
Methods Mol Biol ; 640: 447-62, 2010.
Article in English | MEDLINE | ID: mdl-20645067

ABSTRACT

Investigations on the biology of hepatitis C virus (HCV) have been hampered by the lack of small animal models. Efforts have therefore been directed to designing practical and robust cellular models of human origin able to support HCV replication and production in a reproducible and physiologically pertinent manner. Different systems have been constructed based on hepatoma or other cell lines, sub-genomic and genomic replicons, productive replicons, and immortalized hepatocytes. Although these models are practical for high-throughput screenings, they present several drawbacks related to the nature of the virions and the fact that the cells are not differentiated. Adult primary human hepatocytes infected with natural serum-derived HCV virions represent the model that most closely mimics the physiological situation. This chapter describes our experience with this culture model.


Subject(s)
Hepacivirus/physiology , Hepatitis C/drug therapy , Hepatitis C/virology , Hepatocytes/virology , Interferons/therapeutic use , Cells, Cultured , Hepacivirus/isolation & purification , Hepatocytes/drug effects , Host-Pathogen Interactions , Humans , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...