Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Biomed Pharmacother ; 165: 115098, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37437378

ABSTRACT

As a final common pathway of renal injuries, renal fibrosis leads to chronic kidney disease (CKD). Currently, there is no safe and effective therapy to prevent the progression of renal fibrosis to CKD. Inhibition of transforming growth factor-ß1 (TGF-ß1) pathway is proposed as one of the most promising approaches for anti-renal fibrosis therapies. This study aimed to identify novel anti-fibrotic agents using the TGF-ß1-induced fibrosis in renal proximal tubule epithelial cells (RPTEC) and characterize their mechanism of action as well as in vivo efficacy. By screening 362 natural product-based compounds for their ability to reduce collagen accumulation assessed by picro-sirius red (PSR) staining in RPTEC cells, a chalcone derivative AD-021 was identified as an anti-fibrotic agent with IC50 of 14.93 µM. AD-021 suppressed TGF-ß1-induced collagen production, expression of pro-fibrotic proteins (fibronectin and α-smooth muscle actin (αSMA)), and Smad-dependent and Smad-independent signaling pathways via suppression of TGF-ß receptor II (TGFßRII) phosphorylation in RPTEC cells. Furthermore, TGF-ß1-induced mitochondrial fission in RPTEC cells was ameliorated by AD-021 via mechanisms involving inhibition of Drp1 phosphorylation. In a mouse model of unilateral ureteral obstruction (UUO)-induced renal fibrosis, AD-021 reduced plasma TGF-ß1, ameliorated renal fibrosis and improved renal function. Collectively, AD-021 represents a novel class of natural product-based anti-fibrotic agent that has therapeutic potential in the prevention of fibrosis-associated renal disorders including CKD.


Subject(s)
Chalcone , Chalcones , Kidney Diseases , Renal Insufficiency, Chronic , Ureteral Obstruction , Mice , Animals , Transforming Growth Factor beta1/metabolism , Antifibrotic Agents , Chalcones/pharmacology , Chalcones/therapeutic use , Chalcones/metabolism , Chalcone/pharmacology , Chalcone/therapeutic use , Kidney Diseases/metabolism , Kidney , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy , Fibrosis
2.
Fundam Clin Pharmacol ; 37(4): 833-842, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36843181

ABSTRACT

Buspirone, a cationic drug, is an anxiolytic and antidepressant drug. However, whether buspirone and its metabolites are interacted with organic cationic transporter remains uncertain. In this study, we examined the interaction of buspirone and its major metabolites 1-(2-pyrimidinyl)piperazine (1-PP) and 6-hydroxybuspirone (6'-OH-Bu) with hOCTs using human hepatocellular carcinoma (HepG2), human colorectal adenocarcinoma (Caco-2) cells, and S2 cells expressing OCT1 (S2hOCT1), 2 (S2hOCT2), or 3 (S2hOCT3). Coadministration of buspirone and fluorescent 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP+ ) was examined using HepG2 cells, and [3 H]-1-methyl-4-phenylpyridinium (MPP+ ) transport was assessed in S2 cell overexpressing hOCTs. The results showed that ASP+ transport was suppressed by buspirone with an IC50 of 26.3 ± 2.9 µM without any cytotoxic effects in HepG2 expressing hOCTs cells. Consistently, buspirone strongly inhibited [3 H]-MPP+ uptake by S2hOCT1, S2hOCT2, and S2hOCT3 cells with an IC50s of 89.0 ± 1.3 µM, 43.7 ± 7.5 µM, and 20.4 ± 1.0 µM, respectively. Nonetheless, 6'-OH-Bu and 1-PP caused weak or no inhibition on ASP+ and [3 H]-MPP+ transport. These findings suggest the potential interaction of buspirone with organic cation drugs that are handled by hOCT3. However, further clinical relevance is needed to support these findings for preventing drug-drug interaction in patients who take prescribed drugs together with buspirone.


Subject(s)
Buspirone , Organic Cation Transport Proteins , Humans , Buspirone/pharmacology , Caco-2 Cells , Organic Cation Transporter 2 , Organic Cation Transporter 1/metabolism , Cations/metabolism
3.
Biomed Pharmacother ; 148: 112732, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35217281

ABSTRACT

Colistin is a last-resort polypeptide antibiotic widely used to treat against multidrug-resistant Gram-negative bacterial infections. However, this treatment is associated with nephrotoxicity. The aim of this study was to examine the potential protective effect of panduratin A, a bioactive compound of Boesenbergia rotunda, on colistin-induced nephrotoxicity in both in vivo and in vitro models. Intraperitoneal injection of 15 mg/kg colistin for 7 days markedly promoted renal tubular degeneration, increased blood urea nitrogen (BUN) levels, and upregulated the expression of renal injury biomarker and apoptosis proteins. In addition, treatment with colistin increased oxidative stress and apoptosis in mice kidney tissues. Interestingly, these defects were attenuated when co-administered of colistin with panduratin A (2.5 or 25 mg/kg). The underlying mechanisms of panduratin A attenuating colistin toxicity was investigated in human renal proximal tubular cells (RPTEC/TERT1). The mechanisms by which colistin-triggered cytotoxicity was determined by analysis of cell death, reactive oxygen species (ROS) levels, mitochondria function as well as the expression of proteins related to apoptosis pathway. Colistin treatment (200 µg/ml) significantly increased cell apoptosis, elevated ROS production, reduced mitochondrial membrane potential, and decreased anti-apoptotic protein (Bcl-2) expression. These effects were notably suppressed by co-treatment with panduratin A (5 µM). Collectively, panduratin A exerts as a novel nephroprotective agent to protect against colistin-induced renal injury by attenuating mitochondrial damage and renal cell apoptosis.


Subject(s)
Apoptosis/drug effects , Chalcones/pharmacology , Colistin/adverse effects , Kidney Diseases/drug therapy , Mitochondria/drug effects , Protective Agents/pharmacology , Animals , Anti-Bacterial Agents/adverse effects , Cell Line , Colistin/pharmacology , Epithelial Cells/drug effects , Humans , Kidney/drug effects , Kidney/injuries , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Zingiberaceae/chemistry
4.
Genetics ; 214(4): 1079-1090, 2020 04.
Article in English | MEDLINE | ID: mdl-32005656

ABSTRACT

Gout is a common arthritis caused by monosodium urate crystals. The heritability of serum urate levels is estimated to be 30-70%; however, common genetic variants account for only 7.9% of the variance in serum urate levels. This discrepancy is an example of "missing heritability." The "missing heritability" suggests that variants associated with uric acid levels are yet to be found. By using genomic sequences of the ToMMo cohort, we identified rare variants of the SLC22A12 gene that affect the urate transport activity of URAT1. URAT1 is a transporter protein encoded by the SLC22A12 gene. We grouped the participants with variants affecting urate uptake by URAT1 and analyzed the variance of serum urate levels. The results showed that the heritability explained by the SLC22A12 variants of men and women exceeds 10%, suggesting that rare variants underlie a substantial portion of the "missing heritability" of serum urate levels.


Subject(s)
Gout/genetics , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Polymorphism, Single Nucleotide , Uric Acid/blood , Animals , Female , Gene-Environment Interaction , Humans , Male , Middle Aged , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Uric Acid/metabolism , Xenopus
6.
Bioorg Med Chem Lett ; 28(10): 1915-1918, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29655983

ABSTRACT

p-Boronophenylalanine (l-BPA) is applied in clinical settings as a boron carrier for boron neutron capture therapy (BNCT) to cure malignant melanomas. Structural modification or derivatization of l-BPA, however, to improve its uptake efficiency into tumor cells has scarcely been investigated. We successfully synthesized (S)-2-amino-3-(4-boronophenyl)-2-methylpropanoic acid in enantioenriched form as a novel candidate molecule for BNCT. Key steps to enhance the efficiency of this synthesis were enantioselective alkylation of N-protected alanine tert-butyl ester with a Maruoka catalyst and Miyaura borylation reaction to install the boron functionality.


Subject(s)
Boron Compounds/chemistry , Phenylalanine/analogs & derivatives , Alkylation , Boron Compounds/chemical synthesis , Catalysis , Coordination Complexes/chemistry , Palladium/chemistry , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Stereoisomerism
7.
J Pharmacol Sci ; 136(4): 196-202, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29656005

ABSTRACT

The purpose of this study was to create novel urate under-excretion animal models using pyrazinamide and to evaluate whether dihydropyridine calcium channel blockers (CCBs) have uricosuric effects in vivo. Adult male ICR mice were treated with pyrazinamide, vehicle (dimethyl sulfoxide: DMSO), or tap water. Thirty minutes later, pyrazinamide-treated mice were given benzbromarone, losartan, nilvadipine, nitrendipine, nifedipine or azelnidipine. Six hours after the second administration, urine (by urinary bladder puncture) and plasma were collected to measure uric acid and creatinine levels, and fractional excretion of uric acid (FEUA) and creatinine clearance (Ccr) were calculated and evaluated. There was no significant difference in the levels of plasma uric acid, plasma creatinine, Ccr, urinary N-acetyl-ß-d-glucosaminidase (NAG) and urinary NAG-creatinine ratio between water, DMSO, and pyrazinamide-treated mice. But the FEUA of pyrazinamide-treated mice was significantly lower than water mice. The FEUA was significantly higher in mice taking the dihydropyridine CCBs (nilvadipine, nitrendipine, nifedipine, and high-dose azelnidipine) than in pyrazinamide-treated mice. There was no significant difference in Ccr. Thus, a novel animal model created with PZA administration was useful as a urate under-excretion animal model that was probably URAT1-mediated, and the uricosuric effects of dihydropyridine CCBs were confirmed in vivo.


Subject(s)
Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Models, Animal , Uricosuric Agents , Animals , Creatinine/blood , Creatinine/urine , DNA-Binding Proteins , Male , Mice, Inbred ICR , Organic Anion Transporters , Uric Acid/blood , Uric Acid/urine
8.
Diabetes Obes Metab ; 20(4): 1061-1065, 2018 04.
Article in English | MEDLINE | ID: mdl-29171930

ABSTRACT

An integrated analysis was performed with data from 4 phase 2 and phase 3 studies of tofogliflozin in which patients with type 2 diabetes mellitus received the sodium-glucose cotransporter 2 inhibitor tofogliflozin for up to 24 weeks. Sex differences, baseline haemoglobin A1c (HbA1c) and serum uric acid (UA) levels, and log10 -transformed urinary N-acetyl-ß-D-glucosaminidase ratio were significantly correlated with the reduction in serum UA levels at both 4 and 24 weeks in multivariate analysis (respectively, P < .0001). The decrease in HbA1c levels was greatest in the group with the highest baseline HbA1c level (quartile 4; HbA1c > 8.6%) and lowest in the group with the lowest baseline HbA1c level (quartile 1; HbA1c ≤ 7.4%). The decrease in serum UA levels was greatest in the quartile 1 group and lowest in the quartile 4 group. In most groups, the maximum decrease in serum UA levels was seen in the first 4 weeks, while the maximum decrease in HbA1c was seen at week 24. Thus, serum UA levels were significantly decreased in patients with moderate HbA1c levels.


Subject(s)
Benzhydryl Compounds/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Glycated Hemoglobin/drug effects , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Uric Acid/blood , Adult , Blood Glucose/drug effects , Blood Glucose/metabolism , Clinical Trials, Phase II as Topic/statistics & numerical data , Clinical Trials, Phase III as Topic/statistics & numerical data , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/epidemiology , Down-Regulation/drug effects , Female , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Treatment Outcome
9.
Tumour Biol ; 39(3): 1010428317694545, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28347255

ABSTRACT

Unlike normal cells, cancer cells undergo unlimited growth and multiplication, causing them to require massive amounts of amino acid to support their continuous metabolism. Among the amino acid transporters expressed on the plasma membrane, l-type amino acid transporter-1, a Na+-independent neutral amino acid transporter, is highly expressed in many types of human cancer including cholangiocarcinoma. Our previous study reported that l-type amino acid transporter-1 and its co-functional protein CD98 were highly expressed and implicated in cholangiocarcinoma progression and carcinogenesis. Therefore, this study determined the effect of JPH203, a selective inhibitor of l-type amino acid transporter-1 activity, on cholangiocarcinoma cell inhibition both in vitro and in vivo. JPH203 dramatically suppressed [14C]l-leucine uptake as well as cell growth in cholangiocarcinoma cell lines along with altering the expression of l-type amino acid transporter-1 and CD98 in response to amino acid depletion. We also demonstrated that JPH203 induced both G2/M and G0/G1 cell cycle arrest, as well as reduced the S phase accompanied by altered expression of the proteins in cell cycle progression: cyclin D1, CDK4, and CDK6. There was also cell cycle arrest of the related proteins, P21 and P27, in KKU-055 and KKU-213 cholangiocarcinoma cells. Apoptosis induction, detected by an increase in trypan blue-stained cells along with a cleaved caspase-3/caspase-3 ratio, occurred in JPH203-treated cholangiocarcinoma cells at the highest concentration tested (100 µM). As expected, daily intravenous administration of JPH203 (12.5 and 25 mg/kg) significantly inhibited tumor growth in KKU-213 cholangiocarcinoma cell xenografts in the nude mice model in a dose-dependent manner with no statistically significant change in the animal's body weight and with no differences in the histology and appearance of the internal organs compared with the control group. Our study demonstrates that suppression of l-type amino acid transporter-1 activity using JPH203 might be used as a new therapeutic strategy for cholangiocarcinoma treatment.


Subject(s)
Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Large Neutral Amino Acid-Transporter 1/biosynthesis , Animals , Apoptosis/drug effects , Benzoxazoles/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/pathology , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Leucine/metabolism , Mice , Molecular Targeted Therapy , Tyrosine/administration & dosage , Tyrosine/analogs & derivatives , Xenograft Model Antitumor Assays
10.
Parasitol Int ; 66(4): 471-478, 2017 Aug.
Article in English | MEDLINE | ID: mdl-26657242

ABSTRACT

L-type amino acid transporter 1 (LAT1) is highly expressed in various human cancers, including cholangiocarcinoma (CCA), the most common cancer in Northeast Thailand. Chronic inflammation and oxidative stress induced by liver fluke, Opisthorchis viverrini, infection has been recognized as the major cause of CCA in this area. We show here that an increased expression of LAT1 and its co-functional protein CD98 are found during carcinogenesis induced by Ov in hamster CCA tissues. We also demonstrate that oxidative stress induced by H2O2 is time-dependent and dramatically activates LAT1 and CD98 expression in immortal cholangiocytes (MMNK1). In addition, H2O2 treatment increased LAT1 and CD98 expression, as well as an activated form of AKT and mTOR in MMNK1 and CCA cell lines (KKU-M055 and KKU-M213). We also show that suppression of PI3K/AKT pathway activity with a dual PI3K/mTOR inhibitor, BEZ235, causes a reduction in LAT1 and CD98 expression in KKU-M055 and KKU-M213 in parallel with a reduction of activated AKT and mTOR. Interestingly, high expression of LAT1 in human CCA tissues is a significant prognostic factor for shorter survival. Taken together, our data show that LAT1 expression is significantly associated with CCA progression and cholangiocarcinogenesis induced by oxidative stress. Moreover, the expression of LAT1 and CD98 in CCA is possibly regulated by the PI3K/AKT signaling pathway.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Fusion Regulatory Protein-1/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Opisthorchiasis/metabolism , Opisthorchis/physiology , Adult , Aged , Animals , Bile Duct Neoplasms/diagnosis , Cell Line , Cholangiocarcinoma/diagnosis , Cricetinae , Female , Fusion Regulatory Protein-1/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Male , Mesocricetus , Middle Aged , Opisthorchiasis/complications , Opisthorchiasis/diagnosis , Oxidative Stress , Prognosis , Tissue Array Analysis , Up-Regulation
11.
Anat Sci Int ; 92(2): 200-206, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27614971

ABSTRACT

Organic anions (OAs) are secreted in renal proximal tubules in two steps. In the first step, OAs are transported from the blood through basolateral membranes into proximal tubular cells. The prototypical substrate for renal organic anion transport systems, para-aminohippurate (PAH), is transported across basolateral membranes of proximal tubular cells via OAT1 (SLC22A6) and OAT3 (SLC22A8) against an electrochemical gradient in exchange for intracellular dicarboxylates. In the second step, OAs exit into urine through apical membranes of proximal tubules. This step is thought to be performed by multidrug efflux transporters and a voltage-driven organic anion transporter. However, the molecular nature and precise functional properties of these efflux systems are largely unknown. Recently, we characterized an orphan transporter known as human type I sodium-phosphate transporter 4, hNPT4 (SLC17A3), using the Xenopus oocyte expression system. hNPT4 acts as a voltage-driven efflux transporter ("human OATv1") for several OAs such as PAH, estrone sulfate, diuretic drugs, and urate. Here, we describe a model for an OA secretory pathway in renal tubular cells in which OAs exit cells and enter the tubular lumen via hOATv1 (hNPT4). Additionally, hOATv1 functions as a common renal secretory pathway for both urate and drugs, indicating that hOATv1 may be a leak pathway for excess urate that is reabsorbed via apical URAT1 to control the intracellular urate levels. Therefore, we propose a molecular mechanism for the induction of hyperuricemia by diuretics: the diuretics enter proximal tubular cells via basolateral OAT1 and/or OAT3 and may then interfere with the NPT4-mediated apical urate efflux in the renal proximal tubule.


Subject(s)
Kidney Tubules, Proximal/metabolism , Organic Anion Transporters/metabolism , Biological Transport , Humans , Kidney/metabolism
12.
J Pharmacol Sci ; 132(3): 201-204, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27567475

ABSTRACT

L-type amino acid transporter 1 (LAT1, SLC7A5) incorporates essential amino acids into cells. Recent studies have shown that LAT1 is a predominant transporter in various human cancers. However, the function of LAT1 in thymic carcinoma remains unknown. Here we demonstrate that LAT1 is a critical transporter for human thymic carcinoma cells. LAT1 was strongly expressed in human thymic carcinoma tissues. LAT1-specific inhibitor significantly suppressed leucine uptake and growth of Ty82 human thymic carcinoma cell lines, suggesting that thymic carcinoma takes advantage of LAT1 as a quality transporter and that LAT1-specific inhibitor might be clinically beneficial in therapy for thymic carcinoma.


Subject(s)
Amino Acids/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Thymoma/metabolism , Thymus Neoplasms/metabolism , Benzoxazoles/pharmacology , Cell Line, Tumor , Humans , Large Neutral Amino Acid-Transporter 1/biosynthesis , Leucine/metabolism , Thymoma/drug therapy , Thymus Neoplasms/drug therapy , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
13.
Xenobiotica ; 46(7): 641-650, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26576923

ABSTRACT

1. Green tea extract (GTE) and EGCG have previously shown to increase the uptake of MPP+ into Caco-2 cells. However, whether GTE and its derivatives interact with renal basolateral organic cation transporter 2 (Oct2) which plays a crucial role for cationic clearance remains unknown. Thus, this study assessed the potential of drug-green tea (GT) catechins and its derivatives interactions with rat Oct2 using renal cortical slices and S2 stably expressing rat Oct2 (S2rOct2). 2. Both GTE and ECG inhibited MPP+ uptake in renal slices in a concentration-dependent manner (IC50 = 2.71 ± 0.360 mg/ml and 0.87 ± 0.151 mM), and this inhibitory effect was reversible. Inhibition of [3H]MPP+ transport in S2rOct2 by either GTE or ECG (IC50 = 1.90 ± 0.087 mg/ml and 1.67 ± 0.088 mM) was also observed. 3. The weak and reversible interactions of GTE and ECG with rOct2 indicate that consumption of GT beverages could not interfere with cationic drugs secreted via renal OCT2 in humans. However, the rise of therapeutic use of GTE and ECG might have to take into account the significant possibility of adverse drug-green tea catechins interactions which could alter renal organic cation drug clearance.

14.
J Endocrinol ; 224(3): 205-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25486965

ABSTRACT

Oral ingestion of carbohydrate triggers glucagon-like peptide 1 (GLP1) secretion, but the molecular mechanism remains elusive. By measuring GLP1 concentrations in murine portal vein, we found that the ATP-sensitive K(+) (KATP) channel is not essential for glucose-induced GLP1 secretion from enteroendocrine L cells, while the sodium-glucose co-transporter 1 (SGLT1) is required, at least in the early phase (5 min) of secretion. By contrast, co-administration of the α-glucosidase inhibitor (α-GI) miglitol plus maltose evoked late-phase secretion in a glucose transporter 2-dependent manner. We found that GLP1 secretion induced by miglitol plus maltose was significantly higher than that by another α-GI, acarbose, plus maltose, despite the fact that acarbose inhibits maltase more potently than miglitol. As miglitol activates SGLT3, we compared the effects of miglitol on GLP1 secretion with those of acarbose, which failed to depolarize the Xenopus laevis oocytes expressing human SGLT3. Oral administration of miglitol activated duodenal enterochromaffin (EC) cells as assessed by immunostaining of phosphorylated calcium-calmodulin kinase 2 (phospho-CaMK2). In contrast, acarbose activated much fewer enteroendocrine cells, having only modest phospho-CaMK2 immunoreactivity. Single administration of miglitol triggered no GLP1 secretion, and GLP1 secretion by miglitol plus maltose was significantly attenuated by atropine pretreatment, suggesting regulation via vagal nerve. Thus, while α-GIs generally delay carbohydrate absorption and potentiate GLP1 secretion, miglitol also activates duodenal EC cells, possibly via SGLT3, and potentiates GLP1 secretion through the parasympathetic nervous system.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Enteroendocrine Cells/drug effects , Glucagon-Like Peptide 1/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Sodium-Glucose Transport Proteins/metabolism , 1-Deoxynojirimycin/pharmacology , Acarbose/pharmacology , Animals , Enteroendocrine Cells/metabolism , Glucose Transporter Type 2/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transporter 1/physiology , Xenopus laevis
15.
J Pharmacol Sci ; 124(4): 511-3, 2014.
Article in English | MEDLINE | ID: mdl-24671056

ABSTRACT

Endothelial cell proliferation supporting angiogenesis requires sufficient nutrient supply because of facilitated intracellular metabolism. However, little is known about the mechanism for the promotion of nutrient incorporation in proliferating endothelial cells. Here we show that L-type amino acid transporter 1 (LAT1) is a major transporter of essential amino acids in human umbilical vein endothelial cells (HUVECs). Growing HUVECs express a certain level of LAT1. A LAT1-specific inhibitor suppressed leucine uptake, cell proliferation, and tube formation of HUVECs. Therefore, LAT1 acts to support effective uptake of amino acids, which is critical for the optimal function of HUVECs for angiogenesis.


Subject(s)
Amino Acids, Essential/metabolism , Endothelial Cells/metabolism , Large Neutral Amino Acid-Transporter 1/physiology , Umbilical Veins/cytology , Umbilical Veins/metabolism , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Neovascularization, Physiologic
16.
J Immunol ; 191(8): 4080-5, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24038088

ABSTRACT

Activation of T cells accompanies remarkable enhancement of metabolism. Sufficient and continuous nutrient supply is therefore important to support immune reaction in T cells. However, the mechanism of the promotion of nutrient incorporation in activated T cells has not been elucidated. In this study, we show that L-type amino acid transporter 1 (LAT1) is a major transporter for essential amino acids into activated human T cells. CD3/CD28 stimulation in primary human T cells triggered dramatic induction of LAT1 expression mediated by NF-κB and AP-1. Functional disturbance of LAT1 by a specific inhibitor and by small interfering RNA in human T cells suppressed essential amino acid uptake and induced a stress response mediated by DNA damage-inducible transcript 3 to attenuate cytokine production via inhibition of NF-κB and NFAT activities. These results uncover the previously unknown mechanism by which T cells accelerate essential amino acid uptake upon activation and adapt to essential amino acid starvation. Our results also raise the possibility for application of an LAT1 inhibitor as a new drug for therapy of disease caused by exaggerated immune response.


Subject(s)
Amino Acids, Essential/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Amino Acid Transport Systems , Biological Transport , CD28 Antigens/metabolism , CD3 Complex/metabolism , Cells, Cultured , Humans , Large Neutral Amino Acid-Transporter 1/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering , Transcription Factor AP-1/metabolism , Transcription Factor CHOP/metabolism
17.
Toxicol Pathol ; 41(3): 480-6, 2013.
Article in English | MEDLINE | ID: mdl-22968287

ABSTRACT

The present study describes atypical Leydig cell (LC) hyperplasia in 20-week-old Sprague-Dawley rats with low testosterone and high luteinizing hormone levels after prenatal administration of 100 mg/kg/day di(n-butyl) phthalate on days 12 to 21 postconception. Light microscopy revealed LC hyperplasia surrounded by severely degenerated seminiferous tubules. Aggregated LCs had large ovoid nuclei with nucleoli and abundant eosinophilic cytoplasm. Immunohistochemical analysis showed expression of proliferating cell nuclear antigen and vimentin in many hyperplastic LCs. Electron microscopy revealed atypical nuclei, abundant free ribosomes, stripped rough endoplasmic reticulum, intermediate-size filaments, elongated cytoplasmic filopodia, atypical tight junctions, and cilia formations, but smooth endoplasmic reticulum was scarcely observed.


Subject(s)
Dibutyl Phthalate/toxicity , Leydig Cells/drug effects , Leydig Cells/pathology , Luteinizing Hormone/blood , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Testosterone/metabolism , Animals , Female , Histocytochemistry , Hyperplasia/chemically induced , Hyperplasia/pathology , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Testis/chemistry , Testis/drug effects , Testis/pathology
18.
Drug Des Devel Ther ; 6: 323-39, 2012.
Article in English | MEDLINE | ID: mdl-23152669

ABSTRACT

Human uric acid transporter 1 (hURAT1; SLC22A12) is a very important urate anion exchanger. Elevated urate levels are known to play a pivotal role in cardiovascular diseases, chronic renal disease, diabetes, and hypertension. Therefore, the development of potent uric acid transport inhibitors may lead to novel therapeutic agents to combat these human diseases. The current study investigates small molecular weight compounds and their ability to inhibit 14C-urate uptake in oocytes expressing hURAT1. Using the most promising drug candidates generated from our structure-activity relationship findings, we subsequently conducted in vitro hepatic metabolism and pharmacokinetic (PK) studies in male Sprague-Dawley rats. Compounds were incubated with rat liver microsomes containing cofactors nicotinamide adenine dinucleotide phosphate and uridine 5'-diphosphoglucuronic acid. In vitro metabolism and PK samples were analyzed using liquid chromatography/mass spectrometry-mass spectrometry methods. Independently, six different inhibitors were orally (capsule dosing) or intravenously (orbital sinus) administered to fasting male Sprague-Dawley rats. Blood samples were collected and analyzed; these data were used to compare in vitro and in vivo metabolism and to compute noncompartmental model PK values. Mono-oxidation (Phase I) and glucuronidation (Phase II) pathways were observed in vitro and in vivo. The in vitro data were used to compute hepatic intrinsic clearance, and the in vivo data were used to compute peak blood concentration, time after administration to achieve peak blood concentration, area under the curve, and orally absorbed fraction. The experimental data provide additional insight into the hURAT1 inhibitor structure-activity relationship and in vitro-in vivo correlation. Furthermore, the results illustrate that one may successfully prepare potent inhibitors that exhibit moderate to good oral bioavailability.


Subject(s)
Benzbromarone/analogs & derivatives , Benzbromarone/metabolism , Organic Anion Transporters/antagonists & inhibitors , Organic Cation Transport Proteins/antagonists & inhibitors , Animals , Area Under Curve , Biological Availability , Humans , Male , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
19.
Oncol Rep ; 28(3): 862-6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22736142

ABSTRACT

Tumor cells take up a massive amount of nutrition compared to normal cells for increased metabolism. Therefore, special transporters for organic materials are required to satisfy the powerful consumption of nutrition in tumor cells. L-type amino acid transporter 1 (LAT1) incorporates large neutral amino acids, most of which are also categorized as essential amino acids, into cells in a Na+-independent manner. Because of its high expression levels in a variety of cancer cells, it is speculated that LAT1 functions as a key transporter for highly effective delivery of essential amino acids into cancer cells. In this regard, LAT1 inhibitor is expected to have clinical benefit for cancer therapy. However, the molecular mechanism of enrichment of LAT1 in cancer cells remains poorly understood. Here, we show that a proto-oncogene, c-Myc, is a critical positive regulator of LAT1 expression in MIA Paca-2 human pancreatic cancer cells. The uptake of leucine, a representative neutral amino acid, was strictly dependent on LAT1 in MIA Paca-2 cells, and siRNA-mediated knockdown of LAT1 inhibited cell proliferation. Diminished c-Myc expression with siRNA resulted in severe reduction of LAT1 protein levels as well as mRNA levels, which, in turn, led to a significant defect of leucine incorporation. The LAT1 promoter has a canonical c-Myc binding sequence and overexpression of c-Myc increased LAT1 promoter activity, whereas mutation of c-Myc binding site diminished this effect. Our results suggest biological significance of LAT1 in tumor growth and molecular machinery that could explain why LAT1 is preferentially expressed in cancer cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Expression , Large Neutral Amino Acid-Transporter 1/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation , Consensus Sequence , Down-Regulation , Gene Knockdown Techniques , Genes, Reporter , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Leucine/metabolism , Luciferases/biosynthesis , Luciferases/genetics , Pancreatic Neoplasms , Promoter Regions, Genetic , Proto-Oncogene Mas , RNA, Small Interfering
20.
Drug Metab Pharmacokinet ; 27(1): 155-61, 2012.
Article in English | MEDLINE | ID: mdl-21914964

ABSTRACT

Many primary human tumors and tumor cell lines highly express human L-type amino acid transporter 1 (hLAT1); cancerous cells in vivo are strongly linked to LAT1 expression. Synthetic chemistry and in vitro screening efforts have afforded a variety of novel and highly hLAT1 selective compounds, such as JPH203 1. In a recent report, we demonstrated that 1 has potent in vitro and in vivo activity. JPH203 was intravenously administered to produce significant growth inhibition against HT-29 tumors transplanted in nude mice. The current work develops a robust LC/MS-MS method to monitor 1 and its major Phase II metabolite N-acetyl-JPH203 2 from biological samples. We have conducted in vitro and in vivo experiments and the major scientific findings are: i) the major route of biotransformation of 1 is Phase II metabolism to produce 2; ii) metabolite 2 is formed in various organs/tissues (i.e. blood, liver, kidney); and iii) as dogs, which are deficient in NAT genes, do not produce 2, the dog will not be an appropriate toxicological model to evaluate 1.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Benzoxazoles/pharmacokinetics , Large Neutral Amino Acid-Transporter 1/chemistry , Membrane Transport Modulators/metabolism , Membrane Transport Modulators/pharmacokinetics , Microsomes/metabolism , Tyrosine/analogs & derivatives , Acetylation , Animals , Antineoplastic Agents/analysis , Antineoplastic Agents/blood , Benzoxazoles/analysis , Benzoxazoles/blood , Benzoxazoles/metabolism , Biotransformation , Dogs , Humans , Intestine, Small/metabolism , Kidney/chemistry , Kidney/metabolism , Liver/chemistry , Liver/metabolism , Macaca fascicularis , Male , Membrane Transport Modulators/analysis , Membrane Transport Modulators/blood , Mice , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution , Tyrosine/analysis , Tyrosine/blood , Tyrosine/metabolism , Tyrosine/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...