Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 87
1.
Biochem Pharmacol ; 168: 26-37, 2019 10.
Article En | MEDLINE | ID: mdl-31202736

Constitutive androstane receptor (CAR), a member of the nuclear receptor superfamily, is retained as an inactive form phosphorylated at threonine in the cytoplasm of hepatocytes. Upon activation, CAR is dephosphorylated to move into the nucleus and induces the transcription of genes. Thus, nuclear translocation is a key step for CAR activation in hepatocytes. However, this nuclear translocation has not been demonstrated in conventional two-dimensionally-cultured immortalized cell lines such as HepG2, in which CAR spontaneously accumulates in the nucleus. In this study, we showed that treatment with the indirect CAR activator phenobarbital activated transcription of the CYP3A4 gene in three-dimensionally (3D)-cultured HepG2 cells. CAR was retained as its phosphorylated form in the cytoplasm and was translocated to the nucleus in 3D-cultured HepG2 cells in response to treatment with phenobarbital. Moreover, okadaic acid and epidermal growth factor, were found to repress phenobarbital-induced CAR nuclear translocation and subsequent activation of the CYP3A4 gene promoter. These results suggested that 3D-cultured HepG2 cells properly regulated CAR activation as has been observed in hepatocytes.


Phenobarbital/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Active Transport, Cell Nucleus/drug effects , Cell Nucleus/metabolism , Constitutive Androstane Receptor , Cytochrome P-450 CYP3A/genetics , Cytoplasm/metabolism , Epidermal Growth Factor/pharmacology , Hep G2 Cells , Humans , Okadaic Acid/pharmacology , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects
2.
J Pharmacol Sci ; 137(4): 350-358, 2018 Aug.
Article En | MEDLINE | ID: mdl-30150146

Astrocytes have shown longstanding promise as therapeutic targets for various central nervous system diseases. To facilitate drug development targeting astrocytes, we have recently developed a new conditionally immortalized human astrocyte cell line, termed HASTR/ci35 cells. In this study, in order to further increase their chances to contribute to various astrocyte studies, we report on the development of a culture method that improves HASTR/ci35 cell differentiation status and provide several proofs related to their astrocyte characteristics. The culture method is based on the simultaneous elimination of serum effects and immortalization signals. The results of qPCR showed that the culture method significantly enhanced several astrocyte marker gene expression levels. Using the differentiated HASTR/ci35, we examined their response profiles to nucleotide treatment and inflammatory stimuli, along with their membrane fatty acid composition. Consequently, we found that they responded to ADP or UTP treatment with a transient increase of intracellular Ca2+ concentration, and that they could show reactive response to interleukin-1ß treatments. Furthermore, the membrane phospholipids of the cells were enriched with polyunsaturated fatty acids. To summarize, as a unique human astrocyte model carrying the capability of a differentiation induction properties, HASTR/ci35 cells are expected to contribute substantially to astrocyte-oriented drug development studies.


Astrocytes , Cell Culture Techniques/methods , Central Nervous System Agents/pharmacology , Drug Discovery/methods , Adenosine Diphosphate/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Calcium/metabolism , Cell Differentiation , Cell Line , Fatty Acids/metabolism , Fatty Acids, Unsaturated/pharmacology , Gene Expression , Humans , Inflammation , Interleukin-1beta/metabolism , Phospholipids/metabolism , Uridine Triphosphate/pharmacology
3.
Cancer Lett ; 433: 107-116, 2018 10 01.
Article En | MEDLINE | ID: mdl-29960051

Cancer-type organic anion transporting polypeptide 1B3 (Ct-OATP1B3) has been identified as a cancer-specific transcript in various solid cancers, including colorectal cancer. Given its excellent cancer-specific expression profile, we hypothesized that Ct-OATP1B3 could represent a promising target for cancer-specific expression of the suicide gene, herpes simplex virus 1 thymidine kinase (HSV-tk), via a spliceosome-mediated RNA trans-splicing (SMaRT) approach. SMaRT technology is used to recombine two RNA molecules to generate a chimeric transcript. In this study, we engineered an RNA trans-splicing molecule carrying a translation-defective HSV-tk sequence (RTM44), which was capable of inducing its own trans-splicing to the desired Ct-OATP1B3 pre-mRNA target. RTM44 expression in LS180 cells resulted in generation of Ct-OATP1B3/HSV-tk fusion mRNA. A functional translation start site contributed by the target pre-mRNA restored HSV-tk protein expression, rendering LS180 cells sensitive to ganciclovir treatment in vitro and in xenografted mice. The observed effects are ascribed to accurate and efficient trans-splicing, as they were absent in cells carrying a splicing-deficient mutant of RTM44. Collectively, our data highlights Ct-OATP1B3 as an ideal target for the HSV-tk SMaRT suicide system, which opens up new translational avenues for Ct-OATP1B3-targeted cancer therapy.


Colorectal Neoplasms/therapy , Ganciclovir/administration & dosage , Genetic Therapy/methods , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Spliceosomes/genetics , Thymidine Kinase/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Combined Modality Therapy , Ganciclovir/pharmacology , Genetic Vectors/administration & dosage , HCT116 Cells , HT29 Cells , Humans , Mice , Recombinant Fusion Proteins/metabolism , Simplexvirus/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Thymidine Kinase/metabolism , Trans-Splicing , Xenograft Model Antitumor Assays
4.
Biol Pharm Bull ; 41(6): 972-977, 2018.
Article En | MEDLINE | ID: mdl-29863087

P-Glycoprotein (P-gp), encoded by the MDR1 (ABCB1) gene in humans and by Mdr1a and Mdr1b genes in rodents, is a member of the superfamily of ATP-binding cassette transporters. Since P-gp is constitutively expressed in numerous tissues and exhibits a broad specificity in substrate recognition, it can play a crucial role in limiting the absorption and distribution of xenobiotics by decreasing their intracellular accumulation. The expression of P-gp is regulated by various nuclear receptors such as pregnane X receptor (PXR). Although the characterization of P-gp induction by PXR ligands is a crucial goal for predicting pharmacokinetics of drugs, findings regarding the induction of P-gp by PXR ligands in vivo are still controversial. In this study, we examined the effect of pregnenolone 16α-carbonitrile (PCN), a murine PXR ligand, on the expression of Mdr1a/1b mRNA and P-gp protein in the intestine, brain and liver of mice. The results showed that PCN increased the expression of both Mdr1a/1b mRNA and P-gp protein in the intestine and the brain. The present study provided the first evidence that P-gp is inducible by PCN in the large intestine. The results also showed that P-gp protein was induced by PCN in the cortex but not in the whole brain. On the other hand, PCN increased the expression of Mdr1a/1b mRNA in the liver, although no increase was observed in the expression of P-gp protein. These results suggested different effect of PCN on the expression of P-gp protein in the intestine, brain and liver of mice.


ATP Binding Cassette Transporter, Subfamily B/metabolism , Brain/drug effects , Intestines/drug effects , Liver/drug effects , Pregnenolone Carbonitrile/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Brain/metabolism , Intestinal Mucosa/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , RNA, Messenger/metabolism
5.
AAPS J ; 20(3): 61, 2018 04 09.
Article En | MEDLINE | ID: mdl-29858698

Cytochrome P450 3A (CYP3A) enzymes metabolize approximately half of all drugs on the market. Since the endogenous compounds 4ß-hydroxycholesterol (4ß-HC) and 25-hydroxycholesterol (25-HC) are generated from cholesterol via CYP3A enzymes, we examined whether the plasma levels of 4ß-HC and 25-HC reflect hepatic CYP3A4 activity by using a CYP3A-humanized mouse model, in which the function of endogenous Cyp3a was genetically replaced by human CYP3A. CYP3A-humanized mice have great advantages for evaluation of the relationship between hepatic CYP3A protein levels and plasma and hepatic levels of 4ß-HC and 25-HC. Levels of CYP3A4 protein in the liver microsomes of CYP3A-humanized mice were increased by treatment with pregnenolone-16α-carbonitrile, a CYP3A inducer. Hepatic and plasma levels of 4ß-HC and 25-HC normalized by cholesterol were significantly correlated with hepatic CYP3A4 protein levels. In addition, in vitro studies using human liver microsomes showed that the formation of 4ß-HC was strongly inhibited by a CYP3A inhibitor, while the inhibitory effect of the CYP3A inhibition on the formation of 25-HC was weak. These results suggested that CYP3A mainly contributed to the formation of 4ß-HC in human liver microsomes, whereas other factors may be involved in the formation of 25-HC. In conclusion, the in vivo studies using CYP3A-humanized mice suggest that plasma 4ß-HC and 25-HC levels reflect hepatic CYP3A4 activity. Furthermore, taking the results of in vitro studies using human liver microsomes into consideration, 4ß-HC is a more reliable biomarker of hepatic CYP3A activity.


Biomarkers/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Hydroxycholesterols/metabolism , Animals , Chromatography, Liquid , Humans , Hydroxycholesterols/blood , Male , Mice , Microsomes, Liver/enzymology , Tandem Mass Spectrometry
6.
Drug Metab Dispos ; 46(11): 1756-1766, 2018 11.
Article En | MEDLINE | ID: mdl-29777024

P-glycoprotein (P-gp), encoded by the MDR1 gene in humans and by the Mdr1a/1b genes in rodents, is expressed in numerous tissues and performs as an efflux pump to limit the distribution and absorption of many drugs. Owing to species differences of P-gp between humans and rodents, it is difficult to predict the impact of P-gp on pharmacokinetics and the tissue distribution of P-gp substrates in humans from the results of animal experiments. Therefore, we generated a novel P-gp humanized mouse model by using a mouse artificial chromosome (MAC) vector [designated human MDR1-MAC (hMDR1-MAC) mice]. The results showed that hMDR1 mRNA was expressed in various tissues of hMDR1-MAC mice. Furthermore, the expression of human P-gp was detected in the brain capillary fraction and plasma membrane fraction of intestinal epithelial cells isolated from hMDR1-MAC mice, although the expression levels of intestinal P-gp were extremely low. Thus, we evaluated the function of human P-gp at the blood-brain barrier of hMDR1-MAC mice. The brain-to-plasma ratios of P-gp substrates in hMDR1-MAC mice were much lower than those in Mdr1a/1b-knockout mice, and the brain-to-plasma ratio of paclitaxel was significantly increased by pretreatment with a P-gp inhibitor in hMDR1-MAC mice. These results indicated that the hMDR1-MAC mice are the first P-gp humanized mice expressing functional human P-gp at the blood-brain barrier. This mouse is a promising model with which to evaluate species differences of P-gp between humans and mice in vivo and to estimate the brain distribution of drugs in humans while taking into account species differences of P-gp.


ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/metabolism , Chromosomes/metabolism , Pharmaceutical Preparations/metabolism , Animals , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Cell Line , Chickens/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Tissue Distribution/physiology
7.
Biol Pharm Bull ; 41(5): 697-706, 2018.
Article En | MEDLINE | ID: mdl-29709907

Carboxylesterase 2 (CES2), which is a member of the serine hydrolase superfamily, is primarily expressed in the human small intestine, where it plays an important role in the metabolism of ester-containing drugs. Therefore, to facilitate continued progress in ester-containing drug development, it is crucial to evaluate how CES2-mediated hydrolysis influences its intestinal permeability characteristics. Human colon carcinoma Caco-2 cells have long been widely used in drug permeability studies as an enterocyte model. However, they are not suitable for ester-containing drug permeability studies due to the fact that Caco-2 cells express CES1 (which is not expressed in human enterocytes) but do not express CES2. To resolve this problem, we created a new Caco-2 cell line carrying the human small intestine-type CES expression profile. We began by introducing short-hairpin RNA for CES1 mRNA knockdown into Caco-2 cells to generate CES1-decifient Caco-2 cells (Caco-2CES1KD cells). Then, we developed Caco-2CES1KD cells that stably express CES2 (CES2/Caco-2CES1KD cells) and their control Mock/Caco-2CES1KD cells. The results of a series of functional expression experiments confirmed that CES2-specific activity, along with CES2 mRNA and protein expression, were clearly detected in our CES2/Caco-2CES1KD cells. Furthermore, we also confirmed that CES2/Caco-2CES1KD cells retained their tight junction formation property as well as their drug efflux transporter functions. Collectively, based on our results clearly showing that CES2/Caco-2CES1KD cells carry the human intestinal-type CES expression profile, while concomitantly retaining their barrier properties, it can be expected that this cell line will provide a promising in vitro model for ester-containing drug permeability studies.


Caco-2 Cells , Carboxylesterase/genetics , Intestinal Mucosa/metabolism , Carboxylesterase/metabolism , Humans , Permeability , RNA, Messenger/genetics , Thiazepines/pharmacology
8.
Biol Pharm Bull ; 41(3): 440-444, 2018.
Article En | MEDLINE | ID: mdl-29491221

Liver sinusoidal endothelial cells (LSECs), which are specialized endothelial cells that line liver sinusoids, have been reported to participate in a variety of liver functions, such as blood macromolecule clearance and factor VIII production. In addition, LSECs play crucial roles in liver regeneration following acute liver injury, as well as the development and progression of liver diseases or drug-induced hepatotoxicity. However, the molecular mechanisms underlying their roles remain mostly unknown. Therefore, in order to contribute to the clarification of those mechanisms, herein we report on the development of a new immortalized human LSEC (HLSEC) line. To produce this cell line, two immortalized genes were introduced into the primary HLSECs, which eventually resulted in the establishment of the HLSEC/conditionally immortalized, clone-J (HLSEC/ciJ). Consistent with the two-immortalized gene expression, HLSEC/ciJ showed excellent proliferation activity. Additionally, the results of gene expression analyses showed that several LSEC (as well as pan-endothelial) marker mRNAs and proteins were clearly expressed in HLSEC/ciJ. Furthermore, we found that adherence junction proteins were localized at the cell border in the HLSEC/ciJ monolayer, and that the cells exhibited a tube-like structure formation property. Taken together, the results obtained thus far indicate that we have successfully immortalized HLSECs, resulting in creation of HLSEC/ciJ, a cell line that possesses infinite proliferation ability while retaining possession of at least some HLSEC features. We believe that the HLSEC/ciJ have the potential to provide a valuable and unlimited alternative source of HLSECs for use in liver/LSEC physiology/pathophysiology, pharmacology, and toxicology studies.


Endothelial Cells/drug effects , Liver/cytology , Cell Culture Techniques , Cell Line , Cell Proliferation , Cryopreservation , DNA, Complementary/biosynthesis , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gene Expression , Hepatocytes , Humans , Liver/metabolism , RNA/biosynthesis
9.
Biol Pharm Bull ; 41(3): 445-449, 2018.
Article En | MEDLINE | ID: mdl-29491222

Cancer-type organic anion transporting polypeptide 1B3 (Ct-OATP1B3) mRNA is a variant isoform of the liver-type OATP1B3. Because Ct-OATP1B3 mRNA shows an excellent cancer-specific expression profile in colorectal cancer (CRC), and that its expression levels are associated with CRC prognosis, it holds the potential to become a useful CRC detection and diagnosis biomarker. While the potential is currently justified only at the tissue level, if existence of Ct-OATP1B3 mRNA in CRC-derived extracellular vesicles (EVs) is validated, the findings could enhance its translational potential as a CRC detection and diagnosis biomarker. Therefore, this study aims at proving that Ct-OATP1B3 mRNA exists in CRC-derived EVs, and can be detected using serum specimens. To examine the possibility of Ct-OATP1B3 mRNA being existed in extracellular milieu, we isolated EVs from the human CRC (HCT116, HT-29, and SW480) cell lines, and prepared their cDNAs. The RT-PCR results showed that Ct-OATP1B3 mRNA was clearly present in EVs derived from the human CRC cell lines. Then, in order to further explore the possibility that Ct-OATP1B3 mRNA in CRC-derived EVs can be detected in serum, we isolated serum EVs derived from human CRC xenograft mice, and then performed RT-PCR. The results showed that Ct-OATP1B3 mRNA could be found in all serum EV and CRC tissue samples of the mice examined. Collectively, our findings, which show that Ct-OATP1B3 mRNA exists in EVs and can be detected in (at least) mouse serum, strengthen the potential use of Ct-OATP1B3 mRNA as a serum-based CRC biomarker.


Biomarkers, Tumor/genetics , Colorectal Neoplasms/blood , Extracellular Vesicles/metabolism , RNA, Messenger/blood , RNA, Messenger/genetics , RNA, Neoplasm/blood , RNA, Neoplasm/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Prognosis , Real-Time Polymerase Chain Reaction , Solute Carrier Organic Anion Transporter Family Member 1B3/biosynthesis
10.
Mol Neurobiol ; 55(7): 5993-6006, 2018 Jul.
Article En | MEDLINE | ID: mdl-29128907

While pericytes wrap around microvascular endothelial cells throughout the human body, their highest coverage rate is found in the brain. Brain pericytes actively contribute to various brain functions, including the development and stabilization of the blood-brain barrier (BBB), tissue regeneration, and brain inflammation. Accordingly, detailed characterization of the functional nature of brain pericytes is important for understanding the mechanistic basis of brain physiology and pathophysiology. Herein, we report on the development of a new human brain pericyte cell line, hereafter referred to as the human brain pericyte/conditionally immortalized clone 37 (HBPC/ci37). Developed via the cell conditionally immortalization method, these cells exhibited excellent proliferative ability at 33 °C. However, when cultured at 37 °C, HBPC/ci37 cells showed a differentiated phenotype that was marked by morphological alterations and increases in several pericyte-enriched marker mRNA levels, such as platelet-derived growth factor receptor ß. It was also found that HBPC/ci37 cells possessed the facilitative ability of in vitro BBB formation and differentiation into a neuronal lineage. Furthermore, HBPC/ci37 cells exhibited the typical "reactive" features of brain pericytes in response to pro-inflammatory cytokines. To summarize, our results clearly demonstrate that HBPC/ci37 cells possess the ability to perform several key brain pericyte functions while also showing the capacity for extensive and continuous proliferation. Based on these findings, it can be expected that, as a unique human brain pericyte model, HBPC/ci37 cells have the potential to contribute to significant advances in the understanding of human brain pericyte physiology and pathophysiology.


Brain/cytology , Fetus/cytology , Pericytes/cytology , Adipogenesis , Capillaries/metabolism , Cell Differentiation , Cell Line, Transformed , Cytokines/metabolism , Endothelial Cells/cytology , Gene Expression Profiling , Humans , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Neurogenesis , Pericytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
11.
Xenobiotica ; 48(11): 1098-1105, 2018 Nov.
Article En | MEDLINE | ID: mdl-29095659

Rifampicin (RIF), a typical ligand of human pregnane X receptor (PXR), powerfully induces the expression of cytochrome P450 3A4 (CYP3A4) in humans. Although it is thought that RIF is not a ligand of rodent PXR, treatment with high-dose RIF (e.g. more than 20 mg/kg) increases the expression of CYP3A in the mouse liver. In this study, we investigated whether the induction of CYP3A by high-dose RIF in the mouse liver is mediated via indirect activation of mouse PXR (mPXR). The results showed that high-dose RIF increased the expression of CYP3A11 and other PXR-target genes in the liver of wild-type mice but not PXR-knockout mice. However, the results of reporter gene and ligand-dependent assembly assays showed that RIF does not activate mPXR in a ligand-dependent manner. In addition, high-dose RIF stimulated nuclear accumulation of mPXR in the mouse liver, and geldanamycin and okadaic acid attenuated the induction of Cyp3a11 and other PXR-target genes in primary hepatocytes, suggesting that high-dose RIF triggers nuclear translocation of mPXR. In conclusion, the present study suggests that high-dose RIF stimulates nuclear translocation of mPXR in the liver of mice by indirect activation, resulting in the transactivation of Cyp3a11 and other PXR-target genes.


Cytochrome P-450 CYP3A/genetics , Membrane Proteins/genetics , Receptors, Steroid/metabolism , Rifampin/administration & dosage , Animals , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P450 Family 2/genetics , Gene Expression Regulation, Enzymologic/drug effects , Genes, Reporter , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Okadaic Acid/pharmacology , Pregnane X Receptor , Receptors, Steroid/genetics , Rifampin/adverse effects
12.
Sci Rep ; 7(1): 15189, 2017 11 09.
Article En | MEDLINE | ID: mdl-29123154

Cytochrome P450, family 3, subfamily A (CYP3A) enzymes metabolize approximately 50% of commercially available drugs. Recently, we developed fully humanized transchromosomic (Tc) CYP3A mice with the CYP3A cluster including CYP3A4, CYP3A5, CYP3A7, and CYP3A43. Our humanized CYP3A mice have the CYP3A5*3 (g.6986G) allele, resulting in the almost absence of CYP3A5 protein expression in the liver and intestine. To produce model mice for predicting CYP3A5's contribution to pharmacokinetics, we performed a single-nucleotide polymorphism (SNP) modification of CYP3A5 (g.6986G to A, *3 to *1) on the CYP3A cluster using genome editing in  both mouse ES cells and fertilized eggs, and produced humanized CYP3A5*1 mice recapitulating the CYP3A5*1 carrier phenotype in humans. The humanized CYP3A mouse with CYP3A5*1 is the first Tc mouse for predicting the SNP effect on pharmacokinetics in humans. The combination of Tc technology and genome editing enables the production of useful humanized models that reflect humans with different SNPs.


Cytochrome P-450 CYP3A/genetics , Gene Editing , Models, Animal , Pharmacogenetics/methods , Polymorphism, Single Nucleotide , Animals , Animals, Genetically Modified , Humans , Mice
13.
Drug Metab Pharmacokinet ; 32(5): 265-272, 2017 Oct.
Article En | MEDLINE | ID: mdl-28942083

Pregnane X receptor (PXR) is localized in the cytoplasm of liver cells, whereas it is localized in the nucleus of monolayer-cultured HepG2 cells. Since cultured cells are affected by the microenvironment in which they are grown, we studied the effect of three-dimensional (3D) culture on the localization of PXR in HepG2 cells using the hanging drop method. The results showed that PXR was retained in the cytoplasm of HepG2 cells and other human hepatocarcinoma cell lines (FLC5, FLC7 and Huh7) when they were cultured by the hanging drop method. Treatment with rifampicin, a ligand of PXR, translocated PXR from the cytoplasm to nucleus and increased expression levels of CYP3A4 mRNA in HepG2 cells cultured by the hanging drop method. These findings suggest that 3D culture is a key factor determining the intracellular localization of PXR in human hepatocarcinoma cells and that PXR that becomes retained in the cytoplasm of HepG2 cells with 3D culture has functions of nuclear translocation and regulation of target genes in response to human PXR ligands. Three-dimensionally cultured hepatocarcinoma cells would be a useful tool to evaluate induction potency of drug candidates and also to study mechanisms of nuclear translocation of PXR by human PXR ligands.


Cell Culture Techniques/methods , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Receptors, Steroid/analysis , Receptors, Steroid/metabolism , Active Transport, Cell Nucleus/drug effects , Cell Nucleus/drug effects , Cytochrome P-450 CYP3A/genetics , Cytoplasm/drug effects , Hep G2 Cells , Humans , Pregnane X Receptor , Receptors, Steroid/genetics , Rifampin/pharmacology
14.
Biomark Med ; 11(8): 629-639, 2017 Aug.
Article En | MEDLINE | ID: mdl-28592144

AIM: We aimed to clarify diagnostic and prognostic biomarker potentials of cancer-type organic anion transporting polypeptide 1B3 (Ct-OATP1B3) mRNA in colorectal cancer (CRC) patients. PATIENTS & METHODS: Ct-OATP1B3 mRNA levels in 97 CRC and adjacent normal colon tissues were measured by real-time PCR. The receiver operating characteristic curve analysis and the Kaplan-Meier curve analysis were performed to characterize its biomarker potentials. RESULTS: Ct-OATP1B3 mRNA showed noticeable diagnostic power (the area under the receiver operating characteristic = 0.91) in the CRC patients. Additionally, the higher/lower mRNA expression was clearly associated with better/poorer overall survival in the CRC patients (p < 0.05). CONCLUSION: Ct-OATP1B3 mRNA has the potential to be a tissue-based biomarker for definitive diagnosis and prognostic stratification in CRC.

15.
Drug Metab Lett ; 11(1): 60-67, 2017 11 17.
Article En | MEDLINE | ID: mdl-28393714

BACKGROUND: Cytochrome P450 3A4 (CYP3A4) is an important drug-metabolizing enzyme that is expressed in the liver and small intestine of humans. Various factors influence the expression of CYP3A4, but gender difference in CYP3A4 expression remains debatable. OBJECTIVE: To clarify gender difference of hepatic and intestinal CYP3A4 in CYP3A-humanized mice generated by a human artificial chromosome (HAC) vector system. The CYP3A-humanized (CYP3AHAC) mice have essential regulatory regions, including promoters and enhancers, and unknown elements affecting the expression of CYP3A4. METHODS: We examined the expression and activity of hepatic and intestinal CYP3A4 in male and female CYP3A-HAC mice. CYP3A activity was determined as α- and 4-hydroxylation activity of triazolam in liver and intestinal microsomes. Expression level of CYP3A protein was determined by Western blot analysis. Expression level of CYP3A4 mRNA was measured by quantitative real-time PCR. RESULTS: The results showed that triazolam hydroxylation activities and protein levels of CYP3A in the liver were significantly higher in female than in male CYP3A-HAC mice, whereas those in the intestine were not significantly different between the genders. In addition, the expression of CYP3A4 mRNA showed a tendency similar to that found for the activity and expression of CYP3A protein in the liver and intestine of CYP3A-HAC mice. CONCLUSION: These findings suggest that the expression and activity levels of CYP3A4 in the liver are higher in females than in males, whereas there is no gender difference in the levels in the intestine of CYP3A-HAC mice.


Cytochrome P-450 CYP3A/metabolism , Intestine, Small/enzymology , Liver/enzymology , Animals , Chromosomes, Artificial, Human , Cytochrome P-450 CYP3A/genetics , Female , Gene Expression , Humans , Male , Mice
16.
Eur J Drug Metab Pharmacokinet ; 42(6): 965-972, 2017 Dec.
Article En | MEDLINE | ID: mdl-28447323

BACKGROUND AND OBJECTIVE: Human in vitro and dog in vitro/in vivo researches indicate that the drug-drug interaction (DDI) of decreased plasma valproic acid (VPA) concentration by co-administration of carbapenem antibiotics is caused by inhibition of acylpeptide hydrolase (APEH)-mediated VPA acylglucuronide (VPA-G) hydrolysis by carbapenems. In this study, we investigated VPA disposition and APEH activities in TK-NOG chimeric mice, whose livers were highly replaced with human hepatocytes, to evaluate the utility of this animal model and the clinical relevance of the DDI mechanism. METHODS: VPA and VPA-G concentrations in plasma, urinary excretion of VPA-G and APEH activity in humanized livers were measured after co-administration of VPA with meropenem (MEPM) to chimeric mice. RESULTS: After co-administration with MEPM to the chimeric mice, plasma VPA concentration more rapidly decreased than without the co-administration. An increase in plasma AUC and urinary excretion of VPA-G was also observed. APEH activity in humanized livers was strongly inhibited even at 24 h after co-administration of MEPM to the chimeric mice. CONCLUSION: The DDI of VPA with carbapenems was successfully observed in chimeric mice with humanized livers. The DDI was caused by long-lasting inhibition of hepatic APEH-mediated VPA-G hydrolysis by carbapenems, which strongly supports the APEH-mediated mechanism of the clinical DDI. This is the first example showing the usefulness of chimeric mice with humanized livers for evaluation of a DDI via non-cytochrome P450 enzyme.


Carbapenems/pharmacology , Liver/drug effects , Liver/metabolism , Valproic Acid/pharmacokinetics , Animals , Drug Interactions , Humans , Meropenem , Mice , Peptide Hydrolases/metabolism , Thienamycins/pharmacology , Transplantation Chimera/blood , Transplantation Chimera/metabolism , Transplantation Chimera/urine , Valproic Acid/blood , Valproic Acid/urine
17.
Drug Metab Pharmacokinet ; 32(1): 46-52, 2017 Feb.
Article En | MEDLINE | ID: mdl-28131653

Benzbromarone (BBR) is a potent uricosuric drug that can cause serious liver injury. Our recent study suggested that 1'-hydroxy BBR, one of major metabolites of BBR, is metabolized to a cytotoxic metabolite that could be detoxified by glutathione (GSH). The aim of this study was to clarify whether GSH adducts are formed from 1'-hydroxy BBR in human liver microsomes (HLM). Incubation of 1'-hydroxy BBR with GSH in HLM did not result in the formation of GSH adducts, but 1',6-dihydroxy BBR was formed. In addition, incubation of 1',6-dihydroxy BBR with GSH in HLM resulted in the formation of three novel GSH adducts (M1, M2 and M3). The structures of M1 and M2 were estimated to be GSH adducts in which the 1-hydroxyethyl group at the C-2 position and the hydroxyl group at the C-1' position of 1',6-dihydroxy BBR were substituted by GSH, respectively. We also found that the 6-hydroxylation of 1'-hydroxy BBR is mainly catalyzed by CYP2C9 and that several CYPs and/or non-enzymatic reaction are involved in the formation of GSH adducts from 1',6-dihydroxy BBR. The results indicate that 1'-hydroxy BBR is metabolized to reactive metabolites via 1',6-dihydroxy BBR formation, suggesting that these reactive metabolites are responsible for BBR-induced liver injury.


Benzbromarone/analogs & derivatives , Benzbromarone/metabolism , Glutathione/metabolism , Microsomes, Liver/metabolism , Benzbromarone/adverse effects , Benzbromarone/chemistry , Glutathione/chemistry , Humans , Inactivation, Metabolic , Molecular Structure
18.
Drug Metab Pharmacokinet ; 32(1): 116-119, 2017 Feb.
Article En | MEDLINE | ID: mdl-27916488

Entecavir (ETV) and tenofovir (TFV) are essential nucleoside analogues in current hepatitis B virus (HBV) treatments. Since these drugs target the HBV polymerase that is localized within human hepatocytes, determining of their cellular uptake process is an important step in fully understanding their pharmacological actions. However, the human hepatic transporters responsible for their uptake have remained unidentified. Therefore, this study aimed at identifying the primary ETV and TFV uptake transporter(s) in human hepatocytes. In transport assays, temperature-sensitive ETV and TFV uptake by human hepatocytes were observed, and their uptake were strongly inhibited by bromosulfophthalein, which is an inhibitor of organic anion transporters/organic anion transporting polypeptides (OATs/OATPs). Given these results, ETV and TFV uptake activities in several human OAT/OATP expression systems were examined. The results showed that, among the transporters tested, only OAT2 possessed ETV transport activity. On the other hand, none of the transporters showed any TFV uptake activity. To summarize, our results identify that human OAT2 is an ETV transporter, thereby suggesting that it plays an important part in the mechanisms underlying ETV antiviral activity. Furthermore, although the hepatic TFV transporters remain unknown, our results have, at least, clarified that these two anti-HBV drugs have different hepatocyte entry routes.


Guanine/analogs & derivatives , Organic Anion Transporters, Sodium-Independent/metabolism , Biological Transport/drug effects , Guanine/antagonists & inhibitors , Guanine/metabolism , Guanine/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Sulfobromophthalein/pharmacology , Temperature , Tenofovir/antagonists & inhibitors , Tenofovir/metabolism , Tenofovir/pharmacokinetics
19.
Drug Metab Pharmacokinet ; 31(5): 340-348, 2016 Oct.
Article En | MEDLINE | ID: mdl-27567379

Pregnane X receptor (PXR) is involved in the transactivation of ABCB1 gene by rifampicin (RIF). However, we found that increase in ABCB1 mRNA by RIF was observed in LS180 cells but not in HepG2 cells. Since both cell lines expressed PXR equally, we hypothesized that a factor(s) other than PXR is responsible for PXR-mediated transactivation of the ABCB1 gene. Reporter activities of a distal enhancer module containing direct repeat 4 (DR4) motifs were increased by RIF in LS180 cells but not in HepG2 cells. Mutation of the DR4 motifs diminished the increase in reporter activities in LS180 cells. Gene subtraction showed that epithelial-specific ETS factor 3 (ESE-3) is a transcription factor enriched in LS180 cells compared to HepG2 cells. When ESE-3 and PXR were co-expressed in HepG2 cells, reporter activities were increased by RIF, which were completely abolished by mutation of DR4 motifs. Chromatin immunoprecipitation assays showed specific binding of ESE-3 to the region containing the DR4 motifs of the ABCB1 gene. Finally, knock-down of ESE-3 in LS180 cells resulted in a decrease in the induction of ABCB1 mRNA. These results suggest that ESE-3 is a factor responsible for PXR-mediated transactivation of the ABCB1 gene by RIF in LS180 cells.


Liver/metabolism , Receptors, Steroid/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Cell Line, Tumor , Genes, Reporter/genetics , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Humans , Liver/drug effects , Mutation/drug effects , Mutation/genetics , Pregnane X Receptor , RNA, Messenger/genetics , Rifampin/pharmacology , Transcriptional Activation/drug effects
20.
J Steroid Biochem Mol Biol ; 163: 121-8, 2016 10.
Article En | MEDLINE | ID: mdl-27137100

Testosterone regulates cellular functions in the prostate through activation of the androgen receptor (AR), which may enhance expression levels of cholesterogenic enzymes through activation of sterol regulatory element-binding protein2 (SREBP2). Because testosterone is inactivated to 6ß-hydroxytestosterone by cytochrome P450 3A (CYP3A), we examined the effects of Cyp3a deficiency on circulating testosterone levels and its effects on activation of the AR and expression levels of cholesterogenic enzymes in the prostate using Cyp3a-knockout (Cyp3a(-/-)) mice. The results showed that Cyp3a(-/-) mice had remarkably increased free testosterone levels in plasma along with suppressed testosterone 6ß-hydroxylation activities in liver microsomes, suggesting that Cyp3a is a major determinant of systemic levels of testosterone in mice. The results also showed that mRNA expression levels of the AR target genes were increased significantly, and that AR bindings to the promoter region of the AR target genes were more abundant in the prostates of Cyp3a(-/-) mice. These findings suggest that AR activation was stimulated in the prostate of Cyp3a(-/-) mice. In addition, the protein expression levels of SREBP cleavage-activating protein (SCAP), mRNA expression levels of SREBP2 target genes and total cholesterol contents were increased in the prostates of Cyp3a(-/-) mice. The findings suggest that Cyp3a deficiency stimulated the expression of Scap via activation of the AR, which elevated cholesterogenic gene expression levels through activation of SREBP2 and increased total cholesterol contents in the prostate.


Cytochrome P-450 Enzyme System/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Microsomes, Liver/enzymology , Receptors, Androgen/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Animals , Binding Sites , Cholesterol/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/deficiency , Gene Expression Regulation , Hydroxytestosterones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic , Prostate , Protein Binding , Receptors, Androgen/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 2/metabolism , Testosterone/metabolism
...