Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34299017

ABSTRACT

Oxygen deficiency in cells, tissues, and organs can not only prevent the proper development of biological functions but it can also lead to several diseases and disorders. In this sense, the kidney deserves special attention since hypoxia can be considered an important factor in the pathophysiology of both acute kidney injury and chronic kidney disease. To provide better knowledge to unveil the molecular mechanisms involved, new studies are necessary. In this sense, this work aims to study, for the first time, an in vitro model of hypoxia-induced metabolic alterations in human proximal tubular HK-2 cells because renal proximal tubules are particularly susceptible to hypoxia. Different groups of cells, cultivated under control and hypoxia conditions at 0.5, 5, 24, and 48 h, were investigated using untargeted metabolomic approaches based on reversed-phase liquid chromatography-mass spectrometry. Both intracellular and extracellular fluids were studied to obtain a large metabolite coverage. On the other hand, multivariate and univariate analyses were carried out to find the differences among the cell groups and to select the most relevant variables. The molecular features identified as affected metabolites were mainly amino acids and Amadori compounds. Insights about their biological relevance are also provided.


Subject(s)
Cell Hypoxia , Chromatography, Reverse-Phase/methods , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Metabolomics/methods , Tandem Mass Spectrometry/methods , Activation, Metabolic/genetics , Activation, Metabolic/physiology , Cell Hypoxia/genetics , Cell Line , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Metabolome/genetics , Multivariate Analysis , Principal Component Analysis
2.
FEBS Open Bio ; 10(10): 1985-2002, 2020 10.
Article in English | MEDLINE | ID: mdl-32794647

ABSTRACT

Tubular 3D liver tissue with enhanced capillary-like structures branching from a large main channel is potentially useful for drug discovery because the perfusable main channel and capillary-like structures enable mass transfer into and out from the tissue. Tubular liver tissue is comprised of the hepatocellular carcinoma cell line HepG2, human umbilical vein endothelial cells (HUVECs), and mesenchymal stem cells (MSCs), using a perfusion device functioning as the interface for an external pump. This study aimed to compare the expression of genes involved in drug metabolism between 2D-cultured hepatocellular carcinoma cells and 3D-cultured tubular liver tissue. Gene expression profiles of 2D-cultured cells and tubular liver tissue were compared using RNA sequencing. Multidimensional scaling analysis revealed that culture dimensionality had a more prominent effect on gene expression profiles than perfusion conditions. More specifically, genes involved in drug metabolism such as CYP2D6, CYP2E1, NNMT, and SLC28A1 were slightly upregulated in the 3D cultures, while certain genes such as ALDH1B1, ALDH1A2, and SULT1E1 were downregulated. These results indicate that gene expression profiles are largely influenced by culture dimensionality and are potentially useful to researchers intending to switch from 2D culture to 3D culture of hepatocellular carcinoma or other tissue types.


Subject(s)
Activation, Metabolic/genetics , Cell Culture Techniques/methods , Organoids/metabolism , Carcinoma, Hepatocellular/metabolism , Coculture Techniques , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Hep G2 Cells , Hepatocytes/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Mesenchymal Stem Cells/metabolism , Organoids/drug effects , Perfusion , Pharmaceutical Preparations/metabolism
3.
PLoS Biol ; 18(1): e3000612, 2020 01.
Article in English | MEDLINE | ID: mdl-31986134

ABSTRACT

Antibiotic resistance increasingly limits the success of antibiotic treatments, and physicians require new ways to achieve efficient treatment despite resistance. Resistance mechanisms against a specific antibiotic class frequently confer increased susceptibility to other antibiotic classes, a phenomenon designated collateral sensitivity (CS). An informed switch of antibiotic may thus enable the efficient treatment of resistant strains. CS occurs in many pathogens, but the mechanisms that generate hypersusceptibility are largely unknown. We identified several molecular mechanisms of CS against the antibiotic nitrofurantoin (NIT). Mutants that are resistant against tigecycline (tetracycline), mecillinam (ß-lactam), and protamine (antimicrobial peptide) all show CS against NIT. Their hypersusceptibility is explained by the overexpression of nitroreductase enzymes combined with increased drug uptake rates, or increased drug toxicity. Increased toxicity occurs through interference of the native drug-response system for NIT, the SOS response, with growth. A mechanistic understanding of CS will help to develop drug switches that combat resistance.


Subject(s)
Drug Collateral Sensitivity/genetics , Nitrofurantoin/pharmacology , Activation, Metabolic/drug effects , Activation, Metabolic/genetics , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/drug effects , Microbial Sensitivity Tests , Mutation/drug effects , Nitrofurantoin/pharmacokinetics , Organisms, Genetically Modified , Prodrugs/pharmacokinetics , Salmonella enterica/drug effects , Salmonella enterica/genetics , Salmonella enterica/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Cancer Res ; 79(22): 5849-5859, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31506334

ABSTRACT

Genetic and epigenetic changes (e.g., histone methylation) contribute to cancer development and progression, but our understanding of whether and how specific mutations affect a cancer's sensitivity to histone demethylase (KDM) inhibitors is limited. Here, we evaluated the effects of a panel of KDM inhibitors on lung adenocarcinomas (LuAC) with various mutations. Notably, LuAC lines harboring KRAS mutations showed hypersensitivity to the histone H3K27 demethylase inhibitor GSK-J4. Specifically, GSK-J4 treatment of KRAS mutant-containing LuAC downregulated cell-cycle progression genes with increased H3K27me3. In addition, GSK-J4 upregulated expression of genes involved in glutamine/glutamate transport and metabolism. In line with this, GSK-J4 reduced cellular levels of glutamate, a key source of the TCA cycle intermediate α-ketoglutarate (αKG) and of the antioxidant glutathione, leading to reduced cell viability. Supplementation with an αKG analogue or glutathione protected KRAS-mutant LuAC cells from GSK-J4-mediated reductions in viability, suggesting GSK-J4 exerts its anticancer effects by inducing metabolic and oxidative stress. Importantly, KRAS knockdown in mutant LuAC lines prevented GSK-J4-induced decrease in glutamate levels and reduced their susceptibility to GSK-J4, whereas overexpression of oncogenic KRAS in wild-type LuAC lines sensitized them to GSK-J4. Collectively, our study uncovers a novel association between a genetic mutation and KDM inhibitor sensitivity and identifies the underlying mechanisms. This suggests GSK-J4 as a potential treatment option for cancer patients with KRAS mutations. SIGNIFICANCE: This study not only provides a novel association between KRAS mutation and GSK-J4 sensitivity but also demonstrates the underlying mechanisms, suggesting a potential use of GSK-J4 in cancer patients with KRAS mutations.


Subject(s)
Activation, Metabolic/genetics , Adenocarcinoma of Lung/genetics , Benzazepines/pharmacology , Lung Neoplasms/genetics , Oncogenes/genetics , Oxidative Stress/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/pharmacology , A549 Cells , Activation, Metabolic/drug effects , Adenocarcinoma of Lung/pathology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Histones/genetics , Humans , Lung Neoplasms/pathology , Methylation/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Oxidative Stress/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
5.
Toxicol Sci ; 172(2): 279-291, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31501904

ABSTRACT

Context-specific GEnome-scale metabolic Network REconstructions (GENREs) provide a means to understand cellular metabolism at a deeper level of physiological detail. Here, we use transcriptomics data from chemically-exposed rat hepatocytes to constrain a GENRE of rat hepatocyte metabolism and predict biomarkers of liver toxicity using the Transcriptionally Inferred Metabolic Biomarker Response algorithm. We profiled alterations in cellular hepatocyte metabolism following in vitro exposure to four toxicants (acetaminophen, carbon tetrachloride, 2,3,7,8-tetrachlorodibenzodioxin, and trichloroethylene) for six hour. TIMBR predictions were compared with paired fresh and spent media metabolomics data from the same exposure conditions. Agreement between computational model predictions and experimental data led to the identification of specific metabolites and thus metabolic pathways associated with toxicant exposure. Here, we identified changes in the TCA metabolites citrate and alpha-ketoglutarate along with changes in carbohydrate metabolism and interruptions in ATP production and the TCA Cycle. Where predictions and experimental data disagreed, we identified testable hypotheses to reconcile differences between the model predictions and experimental data. The presented pipeline for using paired transcriptomics and metabolomics data provides a framework for interrogating multiple omics datasets to generate mechanistic insight of metabolic changes associated with toxicological responses.


Subject(s)
Activation, Metabolic/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Metabolic Networks and Pathways/drug effects , Transcriptome/drug effects , Acetaminophen/toxicity , Activation, Metabolic/genetics , Animals , Biomarkers/metabolism , Carbon Tetrachloride/toxicity , Cells, Cultured , Computational Biology , Gene Expression Profiling , Male , Metabolic Networks and Pathways/genetics , Metabolomics , Polychlorinated Dibenzodioxins/toxicity , Primary Cell Culture , Rats, Sprague-Dawley , Trichloroethylene/toxicity
6.
Semin Nephrol ; 39(2): 132-140, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30827336

ABSTRACT

The kidneys are a frequent target organ for toxicity from exposures to various environmental chemicals and agents. To understand the risk to human health from such exposures, it is important to consider both the underlying chemical and pathologic mechanisms and factors that may modify susceptibility to injury. Choices of exemplary environmental agents to review are based on those with selective effects on the kidneys and for which significant amounts of mechanistic and human data are available. These include the heavy metals cadmium and arsenic, fluoride, the organic solvents trichloroethylene and perchloroethylene, drinking water disinfection by-products haloacids, food and herbal drug contaminants aristolochic acid and melamine, and heat stress. Some common mechanistic features of all these diverse exposures are highlighted, and include oxidative stress and mitochondrial damage. Two major genetic factors that are discussed include genetic polymorphisms in plasma membrane transporters that catalyze uptake and accumulation or efflux and elimination of environmental chemicals, and genetic polymorphisms in bioactivation enzymes that generate toxic and reactive metabolites. Identification of methods to prevent environmental toxicant-associated kidney damage and understanding the genetic factors that influence kidney function and the kidney's response to exposures can be applied to refine risk assessments.


Subject(s)
Acute Kidney Injury/chemically induced , Metals, Heavy/adverse effects , Renal Insufficiency, Chronic/chemically induced , Solvents/adverse effects , Activation, Metabolic/genetics , Acute Kidney Injury/genetics , Aristolochic Acids/adverse effects , Arsenic/adverse effects , Cadmium/adverse effects , Drug Contamination , Fluorides/adverse effects , Food Contamination , Humans , Kidney Cortex Necrosis , Membrane Transport Proteins/genetics , Oxidative Stress , Plant Preparations , Renal Insufficiency, Chronic/genetics , Tetrachloroethylene/adverse effects , Triazines/adverse effects , Trichloroethylene/adverse effects
7.
Clin Pharmacol Ther ; 105(2): 448-457, 2019 02.
Article in English | MEDLINE | ID: mdl-29998574

ABSTRACT

CYP3A enzymes participate in the elimination of ticagrelor and the bioactivation of clopidogrel and prasugrel. We studied the effects of functional CYP3A genetic variants (CYP3A4*22; rs35599367 and CYP3A5*3; rs776746) on the pharmacokinetics and pharmacodynamics of ticagrelor, clopidogrel, and prasugrel. Six healthy volunteers with the CYP3A4*1/*22 and CYP3A5*3/*3 genotype (CYP3A4*22 carriers), eight with the CYP3A4*1/*1 and CYP3A5*1/*3 genotype (CYP3A5 expressors), and 11-13 with the CYP3A4*1/*1 and CYP3A5*3/*3 genotypes (controls) ingested single doses of ticagrelor, clopidogrel, and prasugrel on separate occasions. Ticagrelor area under the plasma concentration-time curve (AUC) was 89% (P = 0.004) higher in CYP3A4*22 carriers than in controls. CYP3A4*22 carriers also showed more pronounced platelet inhibition at 24 hours after ticagrelor ingestion than the controls (43% vs. 21%; P = 0.029). The CYP3A5 genotype did not affect ticagrelor pharmacokinetics. Neither CYP3A5 nor CYP3A4 genotypes significantly affected prasugrel or clopidogrel. In conclusion, the CYP3A4*22 allele markedly impairs ticagrelor elimination enhancing its antiplatelet effect.


Subject(s)
Clopidogrel/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Platelet Aggregation Inhibitors/pharmacokinetics , Prasugrel Hydrochloride/pharmacokinetics , Ticagrelor/pharmacokinetics , Activation, Metabolic/genetics , Adult , Area Under Curve , Clopidogrel/pharmacology , Female , Genetic Variation , Genotype , Healthy Volunteers , Humans , Male , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Function Tests , Prasugrel Hydrochloride/pharmacology , Ticagrelor/pharmacology , Young Adult
8.
PLoS One ; 13(6): e0199122, 2018.
Article in English | MEDLINE | ID: mdl-29920533

ABSTRACT

INTRODUCTION: The prostate-specific antigen (PSA) based prostate cancer (PC) screening is currently being debated. The current assessment is to understand the variability of detecting high-risk PC in a NZ cohort in comparison to a US cohort with better PSA screening facilities. Aldo-keto reductase 1C3 (AKR1C3) is known for multiple functions with a potential to regulate subsequent PSA levels. Therefore, we wish to understand the influence of tobacco smoking and the AKR1C3 rs12529 gene polymorphism in this variability. METHOD: NZ cohort (n = 376) consisted of 94% Caucasians while the US cohort consisted of African Americans (AA), n = 202, and European Americans (EA), n = 232. PSA level, PC grade and stage at diagnosis were collected from hospital databases for assigning high-risk PC status. Tobacco smoking status and the AKR1C3 rs12529 SNP genotype were considered as confounding variables. Variation of the cumulative % high-risk PC (outcome variable) with increasing PSA intervals (exposure factor) was compared between the cohorts using the Kolmogorov-Smirnov test. Comparisons were carried out with and without stratifications made using confounding variables. RESULTS: NZ cohort has been diagnosed at a significantly higher mean age (66.67± (8.08) y) compared to both AA (62.65±8.17y) and EA (64.83+8.56y); median PSA (NZ 8.90ng/ml compared to AA 6.86ng/ml and EA 5.80ng/ml); and Gleason sum (NZ (7) compared EA (6)) (p<0.05). The cumulative % high-risk PC detection shows NZ cohort with a significantly lower diagnosis rates at PSA levels between >6 - <10ng/ml compared to both US groups (p<0.05). These were further compounded significantly by smoking status and genetics. CONCLUSIONS: High-risk PCs recorded at higher PSA levels in NZ could be due to factors including lower levels of PSA screening and subsequent specialist referrals for biopsies. These consequences could be pronounced among NZ ever smokers carrying the AKR1C3 rs12529 G alleles making them a group that requires increased PSA screening attention.


Subject(s)
Adenocarcinoma/genetics , Aldo-Keto Reductase Family 1 Member C3/genetics , Early Detection of Cancer , Genetic Variation , Neoplasm Proteins/genetics , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Smoking/metabolism , Activation, Metabolic/genetics , Adenocarcinoma/blood , Adenocarcinoma/enzymology , Adenocarcinoma/ethnology , Black or African American , Aged , Delayed Diagnosis , Europe/ethnology , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , New Zealand , Prostate-Specific Antigen/biosynthesis , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/blood , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/ethnology , Risk , Smoking/epidemiology , Smoking/genetics , Social Determinants of Health , United States , White People
9.
J Neurosci ; 38(20): 4811-4828, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29695414

ABSTRACT

Schwann cells (SCs) are endowed with a remarkable plasticity. When peripheral nerves are injured, SCs dedifferentiate and acquire new functions to coordinate nerve repair as so-called repair SCs. Subsequently, SCs redifferentiate to remyelinate regenerated axons. Given the similarities between SC dedifferentiation/redifferentiation in injured nerves and in demyelinating neuropathies, elucidating the signals involved in SC plasticity after nerve injury has potentially wider implications. c-Jun has emerged as a key transcription factor regulating SC dedifferentiation and the acquisition of repair SC features. However, the upstream pathways that control c-Jun activity after nerve injury are largely unknown. We report that the mTORC1 pathway is transiently but robustly reactivated in dedifferentiating SCs. By inducible genetic deletion of the functionally crucial mTORC1-subunit Raptor in mouse SCs (including male and female animals), we found that mTORC1 reactivation is necessary for proper myelin clearance, SC dedifferentiation, and consequently remyelination, without major alterations in the inflammatory response. In the absence of mTORC1 signaling, c-Jun failed to be upregulated correctly. Accordingly, a c-Jun binding motif was found to be enriched in promoters of genes with reduced expression in injured mutants. Furthermore, using cultured SCs, we found that mTORC1 is involved in c-Jun regulation by promoting its translation, possibly via the eIF4F-subunit eIF4A. These results provide evidence that proper c-Jun elevation after nerve injury involves also mTORC1-dependent post-transcriptional regulation to ensure timely dedifferentiation of SCs.SIGNIFICANCE STATEMENT A crucial evolutionary acquisition of vertebrates is the envelopment of axons in myelin sheaths produced by oligodendrocytes in the CNS and Schwann cells (SCs) in the PNS. When myelin is damaged, conduction of action potentials along axons slows down or is blocked, leading to debilitating diseases. Unlike oligodendrocytes, SCs have a high regenerative potential, granted by their remarkable plasticity. Thus, understanding the mechanisms underlying SC plasticity may uncover new therapeutic targets in nerve regeneration and demyelinating diseases. Our work reveals that reactivation of the mTORC1 pathway in SCs is essential for efficient SC dedifferentiation after nerve injury. Accordingly, modulating this signaling pathway might be of therapeutic relevance in peripheral nerve injury and other diseases.


Subject(s)
Cell Dedifferentiation , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Proto-Oncogene Proteins c-jun/biosynthesis , Schwann Cells , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Activation, Metabolic/genetics , Activation, Metabolic/physiology , Animals , Eukaryotic Initiation Factor-4F/genetics , Female , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Knockout , Mutation/genetics , Myelin Sheath/metabolism , Proto-Oncogene Proteins c-jun/genetics , Rats , Rats, Sprague-Dawley , Regulatory-Associated Protein of mTOR/genetics , Signal Transduction/physiology
10.
Adv Exp Med Biol ; 1043: 141-151, 2017.
Article in English | MEDLINE | ID: mdl-29224094

ABSTRACT

Recent studies have demonstrated that in mice, the estrogen receptor alpha (ERα) is expressed in the liver and has a direct effect on the regulation of the hepatic genes relevant for energy metabolism and drug metabolism. The sex-related differential expression of the hepatic ERα raises the questions as to whether this receptor is responsible for the sexual differences observed in the physiopathology of the liver.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Liver/metabolism , Sex Characteristics , Activation, Metabolic/genetics , Animals , Energy Metabolism/genetics , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation , Inactivation, Metabolic/genetics , Male , Mice , Reproduction/genetics , Sex Factors , Signal Transduction , Transcription, Genetic
11.
Br J Clin Pharmacol ; 83(3): 572-583, 2017 03.
Article in English | MEDLINE | ID: mdl-27718269

ABSTRACT

AIMS: Oxidative bioactivation of amodiaquine (AQ) by cytochrome P450s to a reactive quinoneimine is considered as an important mechanism underlying its idiosyncratic hepatotoxicity. However, because internal exposure to its major metabolite N-desethylamodiaquine (DEAQ) is up to 240-fold higher than AQ, bioactivation of DEAQ might significantly contribute to covalent binding. The aim of the present study was to compare the kinetics of bioactivation of AQ and DEAQ by human liver microsomes (HLM) and to characterize the CYPs involved in bioactivation of AQ and DEAQ. METHODS: Glutathione was used to trap reactive metabolites formed in incubations of AQ and DEAQ with HLM and recombinant human cytochrome P450s (hCYPs). Kinetics of bioactivation of AQ and DEAQ in HLM and involvement of hCYPs were characterized by measuring corresponding glutathione conjugates (AQ-SG and DEAQ-SG) using a high-performance liquid chromatography method. RESULTS: Bioactivation of AQ and DEAQ in HLM both exhibited Michaelis-Menten kinetics. For AQ bioactivation, enzyme kinetical parameters were Km , 11.5 ± 2.0 µmol l-1 , Vmax , 59.2 ± 3.2 pmol min-1  mg-1 and CLint , 5.15 µl min-1  mg-1 . For DEAQ, parameters for bioactivation were Km , 6.1 ± 1.3 µmol l-1 , Vmax , 5.5 ± 0.4 pmol min-1  mg-1 and CLint 0.90 µl min-1  mg-1 . Recombinant hCYPs and inhibition studies with HLM showed involvement of CYP3A4, CYP2C8, CYP2C9 and CYP2D6 in bioactivation. CONCLUSIONS: The major metabolite DEAQ is likely to be quantitatively more important than AQ with respect to hepatic exposure to reactive metabolites in vivo. High expression of CYP3A4, CYP2C8, CYP2C9, and CYP2D6 may be risk factors for hepatotoxicity caused by AQ-therapy.


Subject(s)
Activation, Metabolic/genetics , Amodiaquine/analogs & derivatives , Amodiaquine/pharmacokinetics , Microsomes, Liver/enzymology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Glutathione/metabolism , Humans , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism
12.
Behav Brain Res ; 316: 261-270, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27618763

ABSTRACT

When food is restricted to a brief fixed period every day, animals show an increase in temperature, corticosterone concentration and locomotor activity for 2-3h before feeding time, termed food anticipatory activity. Mechanisms and neuroanatomical circuits responsible for food anticipatory activity remain unclear, and may involve both oscillators and networks related to temporal conditioning. Rabbit pups are nursed once-a-day so they represent a natural model of circadian food anticipatory activity. Food anticipatory behavior in pups may be associated with neural circuits that temporally anticipate feeding, while the nursing event may produce consummatory effects. Therefore, we used New Zealand white rabbit pups entrained to circadian feeding to investigate the hypothesis that structures related to reward expectation and conditioned emotional responses would show a metabolic rhythm anticipatory of the nursing event, different from that shown by structures related to reward delivery. Quantitative cytochrome oxidase histochemistry was used to measure regional brain metabolic activity at eight different times during the day. We found that neural metabolism peaked before nursing, during food anticipatory behavior, in nuclei of the extended amygdala (basolateral, medial and central nuclei, bed nucleus of the stria terminalis), lateral septum and accumbens core. After pups were fed, however, maximal metabolic activity was expressed in the accumbens shell, caudate, putamen and cortical amygdala. Neural and behavioral activation persisted when animals were fasted by two cycles, at the time of expected nursing. These findings suggest that metabolic activation of amygdala-septal-accumbens circuits involved in temporal conditioning may contribute to food anticipatory activity.


Subject(s)
Activation, Metabolic/physiology , Amygdala/metabolism , Food , Motivation/physiology , Nucleus Accumbens/metabolism , Septum of Brain/metabolism , Activation, Metabolic/genetics , Age Factors , Animals , Animals, Newborn , Circadian Rhythm/physiology , Conditioning, Operant/physiology , Electric Stimulation , Electron Transport Complex IV/metabolism , Fasting , Locomotion/physiology , Motivation/genetics , Rabbits , Reward
13.
Pharmacogenomics J ; 16(3): 220-30, 2016 06.
Article in English | MEDLINE | ID: mdl-26076923

ABSTRACT

The aim of the study was to determine the effect of carboxylesterase 1 (CES1) genetic variation on the activation of angiotensin-converting enzyme inhibitor (ACEI) prodrugs. In vitro incubation study of human liver, intestine and kidney s9 fractions demonstrated that the ACEI prodrugs enalapril, ramipril, perindopril, moexipril and fosinopril are selectively activated by CES1 in the liver. The impact of CES1/CES1VAR and CES1P1/CES1P1VAR genotypes and diplotypes on CES1 expression and activity on enalapril activation was investigated in 102 normal human liver samples. Neither the genotypes nor the diplotypes affected hepatic CES1 expression and activity. Moreover, among several CES1 nonsynonymous variants studied in transfected cell lines, the G143E (rs71647871) was a loss-of-function variant for the activation of all ACEIs tested. The CES1 activity on enalapril activation in human livers with the 143G/E genotype was approximately one-third of that carrying the 143G/G. Thus, some functional CES1 genetic variants (for example, G143E) may impair ACEI activation, and consequently affect therapeutic outcomes of ACEI prodrugs.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/metabolism , Carboxylic Ester Hydrolases/genetics , Liver/enzymology , Pharmacogenomic Variants/genetics , Polymorphism, Single Nucleotide , Prodrugs/metabolism , Activation, Metabolic/genetics , Adult , Aged , Aged, 80 and over , Carboxylic Ester Hydrolases/metabolism , Cell Line , Female , Genotype , Humans , Hydrolysis , Intestines/enzymology , Kidney/enzymology , Kinetics , Male , Middle Aged , Phenotype , Transfection , Young Adult
14.
Cancer Chemother Pharmacol ; 76(5): 1081-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26242222

ABSTRACT

PURPOSE: To describe concentration versus time profiles of capecitabine and its metabolites 5'-DFUR, 5'-DFCR and 5-FU, depending on tablet formulation and on frequent and/or relevant genetic polymorphisms of cytidine deaminase, dihydropyrimidine dehydrogenase, thymidylate synthase and methylenetetrahydrofolate reductase (MTHFR). METHODS: In 46 cancer patients on chronic capecitabine treatment, who voluntarily participated in the study, individual therapeutic doses were replaced on four consecutive mornings by the study medication. The appropriate number of 500 mg test (T) or reference (R) capecitabine tablets was given in randomly allocated sequences TRTR or RTRT (replicate design). Average bioavailability was assessed by ANOVA. RESULTS: Thirty female and 16 male patients suffering from gastrointestinal or breast cancer (mean age 53.4 years; mean dose 1739 mg) were included. The T/R ratios for AUC0-t(last) and C max were 96.7 % (98 % CI 90.7-103.2 %) and 87.2 % (98 % CI 74.9-101.5 %), respectively. Within-subject variability for AUC0-t(last) and C max (coefficient of variation for R) was 16.5 and 30.2 %, respectively. Similar results were seen for all metabolites. No serious adverse events occurred. For the MTHFR C677T (rs1801133) genotype, an increasing number of 677C alleles showed borderline correlation with an increasing elimination half-life of capecitabine (p = 0.043). CONCLUSIONS: The extent of absorption was similar for T and R, but the rate of absorption was slightly lower for T. While such differences are not considered as clinically relevant, formal bioequivalence criteria were missed. A possible, probably indirect role of the MTHFR genotype in pharmacokinetics of capecitabine and/or 5-FU should be investigated in further studies.


Subject(s)
Activation, Metabolic/genetics , Antimetabolites, Antineoplastic/pharmacokinetics , Capecitabine/pharmacokinetics , Cytidine Deaminase/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Prodrugs/pharmacokinetics , Thymidylate Synthase/genetics , Administration, Oral , Adult , Aged , Alleles , Antimetabolites, Antineoplastic/administration & dosage , Area Under Curve , Capecitabine/administration & dosage , Carboxylesterase/metabolism , Cytidine Deaminase/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/metabolism , Dihydrouracil Dehydrogenase (NADP)/genetics , Dihydrouracil Dehydrogenase (NADP)/metabolism , Female , Floxuridine/metabolism , Fluorouracil/metabolism , Genotype , Humans , Liver/enzymology , Male , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Middle Aged , Neoplasm Proteins/metabolism , Polymorphism, Single Nucleotide , Prodrugs/administration & dosage , Tablets , Therapeutic Equivalency , Thymidine Phosphorylase/metabolism , Thymidylate Synthase/metabolism
15.
PLoS One ; 10(7): e0132561, 2015.
Article in English | MEDLINE | ID: mdl-26147597

ABSTRACT

BACKGROUND: The cytochrome P450 (CYP450) 2C19 681 genotypes affect the antiplatelet activity of clopidogrel. We investigated the correlation of CYP 2C19 681G > A mutation with clopidogrel resistance (CR). Additionally, we studied the effect of CR on clinical prognosis of patients with acute coronary syndrome (ACS). METHODS: One hundred ten ACS patients undergoing percutaneous coronary intervention, who were followed-up for 1 year, were included in the study. The patients were co-administered aspirin 100 mg/d and clopidogrel 75mg/d following a loading dose of 300 mg. CR was assessed on the basis of polymorphism observed in the CYP2C19 subgroup. RESULTS: Patients in GG genotype group exhibited greater inhibition of platelet aggregation than patients in GA and AA genotype groups (16.2 ± 10.1%; 10.2 ± 9.9%; 8.0 ± 5.9%, respectively, p < 0.01). CYP2C19 681GG genotype group was associated with lower CR than CYP2C19 681A allele (GA + AA) group (9/59 vs. (12+5)/51; p = 0.009). Over a follow-up of 12 months, the incidence of recurrent angina, acute myocardial infarction, and intra-stent thrombosis in CYP2C19 681 GG carriers was significantly lower than that in CYP2C19 681A allele (GA + AA) group (2/59 vs. 8/51, 1/59 vs. 6/51, 0 vs. 4/51, respectively, p < 0.05). CONCLUSION: CYP 2C19*2 is associated with reduced clopidogrel antiplatelet activity and might be an important marker for poor prognosis of ACS.


Subject(s)
Acute Coronary Syndrome/genetics , Cytochrome P-450 CYP2C19/genetics , Drug Resistance/genetics , Platelet Aggregation Inhibitors/pharmacokinetics , Polymorphism, Single Nucleotide , Prodrugs/pharmacokinetics , Ticlopidine/analogs & derivatives , Activation, Metabolic/genetics , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/surgery , Aged , Alleles , Angina Pectoris/epidemiology , Angina Pectoris/genetics , Aspirin/therapeutic use , Clopidogrel , Coronary Restenosis/epidemiology , Coronary Restenosis/genetics , Coronary Thrombosis/epidemiology , Coronary Thrombosis/genetics , Cytochrome P-450 CYP2C19/physiology , Drug Therapy, Combination , Female , Follow-Up Studies , Genetic Predisposition to Disease , Genotype , Humans , Incidence , Male , Middle Aged , Myocardial Infarction/epidemiology , Myocardial Infarction/genetics , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/therapeutic use , Prodrugs/therapeutic use , Prognosis , Prospective Studies , Recurrence , Stents , Ticlopidine/pharmacokinetics , Ticlopidine/therapeutic use , Treatment Outcome
16.
Atherosclerosis ; 242(1): 102-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26188531

ABSTRACT

OBJECTIVES: Dual antiplatelet therapy with aspirin and a platelet P2Y12 ADP receptor antagonist is the cornerstone of treatment following percutaneous coronary intervention (PCI). Several clinical and genetic factors can cause suboptimal clopidogrel response. We examined the impact of endothelial dysfunction on clopidogrel response variability in subjects with stable coronary artery disease (CAD) after PCI. METHODS: We consecutively enrolled 198 patients with stable CAD one month after successful PCI. All patients were receiving dual antiplatelet therapy (clopidogrel 75 mg and aspirin 100 mg/day). Platelet reactivity was measured by VerifyNow P2Y12 assay (Accumetrics, San Diego, CA). VerifyNow reports its results in P2Y12 reaction units (PRU) and the diagnostic cut-off value is 230. Endothelial function was evaluated by flow mediated dilation (FMD). RESULTS: Patients with high on treatment platelet reactivity (32% of the study population), compared to subjects with low on treatment platelet reactivity, presented decreased FMD values (4.35 ± 2.22% vs. 5.74 ± 3.29%, p = 0.01). Moreover, an inverse association between endothelial function measurement and platelet reactivity (r = -0.24, p = 0.001) was found. Importantly, multivariate analysis after adjustment for age, gender and confounders revealed by the univariate analysis (left ventricle ejection fraction, body mass index, diabetes, dyslipidemia, coronary lesion number) showed that for every decrease in FMD by 1% there is an anticipated increased in the odds of patients to have HPR by 1.66 (95% CI 1.03-2.57, p = 0.037). CONCLUSIONS: Endothelial dysfunction is associated with clopidogrel response variability in patients after PCI receiving dual antiplatelet therapy. These findings shed some light on the mechanisms affecting individual platelet response to antiplatelet therapy and may explain the non-straight forward association between clopidogrel dose, platelet inhibition and cardiovascular outcome.


Subject(s)
Aspirin/therapeutic use , Coronary Disease/drug therapy , Endothelium, Vascular/physiopathology , Platelet Aggregation Inhibitors/therapeutic use , Purinergic P2Y Receptor Antagonists/therapeutic use , Thrombophilia/drug therapy , Ticlopidine/analogs & derivatives , Activation, Metabolic/genetics , Aged , Brachial Artery/physiopathology , Clopidogrel , Coronary Disease/blood , Coronary Disease/physiopathology , Coronary Disease/surgery , Cytochrome P-450 CYP2C19/genetics , Diabetes Mellitus/epidemiology , Drug Synergism , Drug Therapy, Combination , Female , Genotype , Humans , Hyperlipidemias/epidemiology , Male , Middle Aged , Obesity/epidemiology , Percutaneous Coronary Intervention , Platelet Function Tests , Postoperative Complications/prevention & control , Prospective Studies , Risk Factors , Smoking/epidemiology , Stents , Thrombophilia/prevention & control , Ticlopidine/therapeutic use , Vasodilation
17.
Drug Metab Dispos ; 43(7): 916-21, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25922528

ABSTRACT

4-Aminobiphenyl (ABP), a prototypical aromatic amine carcinogen in rodents and humans, requires bioactivation to manifest its toxic effects. A traditional model of ABP bioactivation, based on in vitro enzyme kinetic evidence, had postulated initial N-hydroxylation by the cytochrome P450 isoform CYP1A2. This is followed by phase 2 O-conjugation and hydrolysis to form a reactive nitrenium ion that covalently binds to DNA and produces tumor-initiating mutations. However, Cyp1a2(-/-) mice still possess significant liver ABP N-hydroxylation activity, DNA damage, and incidence of ABP-induced liver tumors, and in vivo induction of CYP1A2 paradoxically reduces levels of ABP-induced DNA damage. Competing ABP detoxification pathways can include N-acetylation by arylamine N-acetyltransferase 1 (NAT1) and/or NAT2; however, wild-type and Nat1/2(-/-) mice have similar in vivo ABP clearance rates. Together, these studies suggest the existence of novel ABP bioactivating and clearance/detoxification enzymes. In the present study, we detected similar reductions in Vmax for ABP N-hydroxylation by liver microsomes from Cyp1a2(-/-) and Cyp2e1(-/-) mice when compared with wild-type mice. In addition, recombinant mouse CYP1A2 and CYP2E1 were both able to N-hydroxylate ABP in mouse hepatoma cells. However, the in vivo clearance of ABP was significantly reduced in Cyp1a2(-/-) but not in Cyp2e1(-/-) mice. Our results support a significant role for CYP2E1 as a novel ABP N-oxidizing enzyme in adult mice, and suggest a more important contribution of CYP1A2 to the in vivo plasma clearance and thus detoxification of ABP.


Subject(s)
Aminobiphenyl Compounds/metabolism , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Acetylation , Activation, Metabolic/genetics , Aminobiphenyl Compounds/pharmacokinetics , Animals , Arylamine N-Acetyltransferase/genetics , Arylamine N-Acetyltransferase/metabolism , Cell Line, Tumor , DNA Damage , Hydroxylation , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microsomes, Liver/enzymology
18.
Circ J ; 79(1): 85-90, 2015.
Article in English | MEDLINE | ID: mdl-25744629

ABSTRACT

BACKGROUND: The loss-of-function genotype of cytochrome P450 2C19 (CYP2C19) has been proposed as a risk factor for stent thrombosis in patients with drug-eluting stent implantation. The aim of this study was to clarify the clinical features of patients with angioscopically-detected in-stent mural thrombi (ISMT). METHODS AND RESULTS: Enrolled were 100 stented segments in 55 patients with stable angina (20 bare-metal stents; 39 Cypher sirolimus-eluting stents [SES]; 26 Endeavor zotarolimus-eluting stents [ZES]; 13 Xience V everolimus-eluting stents; and 2 Nobori biolimus-eluting stents). Dual antiplatelet therapy (100 mg aspirin+75 mg clopidogrel once daily) had been continued since stenting. A poor metabolizer (PM) of clopidogrel was defined as a homozygote of CYP2C19 loss-of-function alleles. Coronary angioscopy revealed ISMT in 6 patients (5 SES, 1 ZES). Between the ISMT group and control group (n=49), there were no significant differences with regards to the VerifyNow P2Y12platelet function assay or in-stent endothelial coverage grade. Exact logistic regression analyses with stepwise forward selection at a significance level of 0.10 were performed to reveal predictive variables for ISMT (respectively: odds ratio, 95% confidence interval, P value: CYP2C19 PM genotype (3.28, 0.88-24.80, 0.09), SES implantation (3.37, 0.90-28.09, 0.08), and presence of yellow plaque (3.69, 1.14-25.70, 0.02). CONCLUSIONS: Patients with ISMT were characterized by SES implantation, poor clopidogrel metabolism, and in-stent yellow plaque.


Subject(s)
Angioscopy , Coronary Thrombosis/etiology , Cytochrome P-450 CYP2C19/genetics , Drug-Eluting Stents , Platelet Aggregation Inhibitors/pharmacokinetics , Polymorphism, Single Nucleotide , Ticlopidine/analogs & derivatives , Activation, Metabolic/genetics , Aged , Alleles , Angina Pectoris/etiology , Aspirin/therapeutic use , Clopidogrel , Coronary Angiography , Coronary Thrombosis/diagnostic imaging , Coronary Thrombosis/genetics , Coronary Thrombosis/prevention & control , Cytochrome P-450 CYP2C19/deficiency , Cytochrome P-450 CYP2C19/metabolism , Drug Resistance/genetics , Drug Therapy, Combination , Drug-Eluting Stents/classification , Female , Genotype , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/therapeutic use , Proton Pump Inhibitors/pharmacology , Receptors, Purinergic P2Y12/analysis , Ticlopidine/pharmacokinetics , Ticlopidine/therapeutic use
19.
Br J Clin Pharmacol ; 79(3): 501-13, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25264565

ABSTRACT

AIMS: The rare association of flupirtine with liver injury is most likely caused by reactive quinone diimines and their oxidative formation may be influenced by the activities of N-acetyltransferases (NAT) that conjugate the less toxic metabolite D13223, and by glucuronosyltransferases (UGT) and glutathione S-transferases (GST) that generate stable terminal glucuronides and mercapturic acid derivatives, respectively. The influence of genetic polymorphisms of NAT2, UGT1A1 and GSTP1 on generation of the terminal mercapturic acid derivatives and analgesic effects was evaluated to identify potential genetic risk factors for hepatotoxicity of flupirtine. METHODS: Metabolic disposition of flupirtine was measured after intravenous administration (100 mg), after swallowing an immediate-release (IR) tablet (100 mg) and after repeated administration of modified release (MR) tablets (400 mg once daily 8 days) in 36 selected healthy subjects. Analgesic effects were measured using pain models (delayed onset of muscle soreness, electric pain). RESULTS: Flupirtine IR was rapidly but incompletely absorbed (∼ 72%). Repeated administration of flupirtine MR showed lower bioavailability (∼ 60%). Approximately 12% of bioavailable flupirtine IR and 8% of bioavailable flupiritine MR was eliminated as mercapturic acid derivatives into the urine independent of the UGT1A1, NAT2 and GSTP1 genotype. Carriers of variant GSTP1 alleles showed lower bioavailability but increased intestinal secretion of flupirtine and increased efficiency in experimental pain. Flupirtine was not a substrate for ABCB1 and ABCC2. CONCLUSIONS: Formation of mercapturic acid derivatives is a major elimination route for flupirtine in man. However, the theoretically toxic pathway is not influenced by the frequent polymorphisms of UGT1A1, NAT2 and GSTP1.


Subject(s)
Acetylcysteine , Aminopyridines , Analgesics , Arylamine N-Acetyltransferase/genetics , Glucuronosyltransferase/genetics , Glutathione S-Transferase pi/genetics , Polymorphism, Genetic , Acetylcysteine/analogs & derivatives , Acetylcysteine/metabolism , Activation, Metabolic/drug effects , Activation, Metabolic/genetics , Administration, Oral , Adult , Aminopyridines/administration & dosage , Aminopyridines/adverse effects , Aminopyridines/pharmacokinetics , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/pharmacokinetics , Animals , Arylamine N-Acetyltransferase/metabolism , Biological Availability , Cross-Over Studies , Delayed-Action Preparations , Dogs , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Glucuronosyltransferase/metabolism , Glutathione S-Transferase pi/metabolism , Healthy Volunteers , Humans , Injections, Intravenous , Madin Darby Canine Kidney Cells , Male , Multidrug Resistance-Associated Protein 2 , Pain Threshold/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...