Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Int J Mol Sci ; 23(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35955741

ABSTRACT

Dofetilide is a rapid delayed rectifier potassium current inhibitor widely used to prevent the recurrence of atrial fibrillation and flutter. The clinical use of this drug is associated with increases in QTc interval, which predispose patients to ventricular cardiac arrhythmias. The mechanisms involved in the disposition of dofetilide, including its movement in and out of cardiomyocytes, remain unknown. Using a xenobiotic transporter screen, we identified MATE1 (SLC47A1) as a transporter of dofetilide and found that genetic knockout or pharmacological inhibition of MATE1 in mice was associated with enhanced retention of dofetilide in cardiomyocytes and increased QTc prolongation. The urinary excretion of dofetilide was also dependent on the MATE1 genotype, and we found that this transport mechanism provides a mechanistic basis for previously recorded drug-drug interactions of dofetilide with various contraindicated drugs, including bictegravir, cimetidine, ketoconazole, and verapamil. The translational significance of these observations was examined with a physiologically-based pharmacokinetic model that adequately predicted the drug-drug interaction liabilities in humans. These findings support the thesis that MATE1 serves a conserved cardioprotective role by restricting excessive cellular accumulation and warrant caution against the concurrent administration of potent MATE1 inhibitors and cardiotoxic substrates with a narrow therapeutic window.


Subject(s)
Anti-Arrhythmia Agents , Atrial Fibrillation , Animals , Anti-Arrhythmia Agents/pharmacology , Humans , Mice , Phenethylamines/pharmacology , Sulfonamides/therapeutic use
2.
Mol Cancer Ther ; 20(11): 2207-2217, 2021 11.
Article in English | MEDLINE | ID: mdl-34518298

ABSTRACT

Acute myeloid leukemia (AML) with an FLT3 internal tandem duplication (FLT3-ITD) mutation is an aggressive hematologic malignancy associated with frequent relapse and poor overall survival. The tyrosine kinase inhibitor gilteritinib is approved for the treatment of relapse/refractory AML with FLT3 mutations, yet its mechanism of action is not completely understood. Here, we sought to identify additional therapeutic targets that can be exploited to enhance gilteritinib's antileukemic effect. Based on unbiased transcriptomic analyses, we identified the glutamine transporter SNAT1 (SLC38A1) as a novel target of gilteritinib that leads to impaired glutamine uptake and utilization within leukemic cells. Using metabolomics and metabolic flux analyses, we found that gilteritinib decreased glutamine metabolism through the TCA cycle and cellular levels of the oncometabolite 2-hydroxyglutarate. In addition, gilteritinib treatment was associated with decreased ATP production and glutathione synthesis and increased reactive oxygen species, resulting in cellular senescence. Finally, we found that the glutaminase inhibitor CB-839 enhanced antileukemic effect of gilteritinib in ex vivo studies using human primary FLT3-ITD-positive AML cells harboring mutations in the enzyme isocitrate dehydrogenase, which catalyzes the oxidative decarboxylation of isocitrate, producing α-ketoglutarate. Collectively, this work has identified a previously unrecognized, gilteritinib-sensitive metabolic pathway downstream of SLC38A1 that causes decreased glutaminolysis and disruption of redox homeostasis. These findings provide a rationale for the development and therapeutic exploration of targeted combinatorial treatment strategies for this subset of relapse/refractory AML.


Subject(s)
Aniline Compounds/therapeutic use , Glutamine/drug effects , Leukemia, Myeloid, Acute/drug therapy , Pyrazines/therapeutic use , fms-Like Tyrosine Kinase 3/metabolism , Aniline Compounds/pharmacology , Animals , Female , Humans , Mice , Pyrazines/pharmacology
3.
Handb Exp Pharmacol ; 266: 41-80, 2021.
Article in English | MEDLINE | ID: mdl-33963461

ABSTRACT

The blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB) represent major control checkpoints protecting the CNS, by exerting selective control over the movement of organic cations and anions into and out of the CNS compartment. In addition, multiple CNS cell types, e.g., astrocytes, ependymal cells, microglia, contribute to processes that maintain the status quo of the CNS milieu. To fulfill their roles, these barriers and cell types express a multitude of transporter proteins from dozens of different transporter families. Fundamental advances over the past few decades in our knowledge of transporter substrates, expression profiles, and consequences of loss of function are beginning to change basic theories regarding the contribution of various cell types and clearance networks to coordinated neuronal signaling, complex organismal behaviors, and overall CNS homeostasis. In particular, transporters belonging to the Solute Carrier (SLC) superfamily are emerging as major contributors, including the SLC22 organic cation/anion/zwitterion family of transporters (includes OCT1-3 and OCTN1-3), the SLC29 facilitative nucleoside family of transporters (includes PMAT), and the SLC47 multidrug and toxin extrusion family of transporters (includes MATE1-2). These transporters are known to interact with neurotransmitters, antidepressant and anxiolytic agents, and drugs of abuse. Clarifying their contributions to the underlying mechanisms regulating CNS permeation and clearance, as well as the health status of astrocyte, microglial and neuronal cell populations, will drive new levels of understanding as to maintenance of the CNS milieu and approaches to new therapeutics and therapeutic strategies in the treatment of CNS disorders. This chapter highlights organic cation transporters belonging to the SLC superfamily known to be expressed in the CNS, providing an overview of their identification, mechanism of action, CNS expression profile, interaction with neurotransmitters and antidepressant/antipsychotic drugs, and results from behavioral studies conducted in loss of function models (knockout/knockdown).


Subject(s)
Antipsychotic Agents , Blood-Brain Barrier , Biological Transport , Blood-Brain Barrier/metabolism , Membrane Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism
4.
Xenobiotica ; 50(5): 588-592, 2020 May.
Article in English | MEDLINE | ID: mdl-31448977

ABSTRACT

Indoxyl sulfate (IS), a highly protein-bound nephro-cardiovascular toxin, was poorly removed by hemodialysis. IS exists as anions in the body and the renal excretion is mediated by organic anion transporter 1 (OAT1) and OAT3. Acidic antibiotics such as cephalosporins and fluoroquinolones were putative substrates/inhibitors of OATs. We hypothesized that cephalosporins and fluoroquinolones might compete with IS for OAT1- and/or OAT3-mediated renal excretions.This study investigated the effects of ciprofloxacin, cefuroxime, cefotaxime, cefazolin and ofloxacin on the intravenous pharmacokinetics of IS in rats. IS was intravenously injected with and without each individual antibiotics, and the concentrations of IS in serum and lysate were determined by HPLC.The results showed that ciprofloxacin significantly increased AUC0-t and T1/2 of IS by 272% and 491%, respectively, and decreased the clearance by 71%. However, ofloxacin, cefuroxime, cefotaxime and cefazolin did not alter the pharmacokinetics of IS. Furthermore, cell line study showed that ciprofloxacin inhibited the OAT3-mediated transport of IS.This study indicates 30 mg/kg of ciprofloxacin decreased the clearance of IS through inhibition on the OAT3-mediated transport, whereas 50 mg/kg of ofloxacin, cefuroxime, cefotaxime and cefazolin did not show significant influence.


Subject(s)
Anti-Bacterial Agents/pharmacology , Indican/metabolism , Animals , Cardiovascular System , Humans , Indican/toxicity , Kidney , Rats , Renal Elimination
5.
Psychopharmacology (Berl) ; 236(7): 2093-2104, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30805668

ABSTRACT

RATIONALE: 2-Amino-6-chloro-3,4-dihydroquinazoline (e.g., A6CDQ) represents a novel putative antidepressant originally thought to act through a 5-HT3 serotonin receptor antagonist mechanism. Here, we investigated this further by examining a positional isomer of A6CDQ (i.e., A7CDQ). MATERIALS AND METHODS: 5-HT3 receptor and transporter activity (uptake-1 and uptake-2) were investigated using a variety of in vitro assays and the in vivo mouse tail suspension test (TST). RESULTS: Although A7CDQ binds at 5-HT3 receptors with low affinity (Ki = 1975 nM) compared to A6CDQ (Ki = 80 nM), it retained 5-HT3 receptor antagonist action (IC50 = 5.77 and 0.26 µM, respectively). In the mouse TST A7CDQ produced antidepressant-like actions (ED50 = 0.09 mg/kg) comparable to that of A6CDQ. In addition, A6CDQ was found to be a 5-HT releasing agent (Km = 2.8 µM) at hSERT and a reuptake inhibitor (IC50 = 1.8 µM) at hNET, whereas A7CDQ was a weak reuptake inhibitor (Km = 43.6 µM) at SERT but a releasing agent (EC50 = 3.3 µM) at hNET. Moreover, A6CDQ and A7CDQ were potent inhibitors of uptake-2 (e.g.; OCT3 IC50 = 3.9 and 5.9 µM, respectively). CONCLUSIONS: A simple shift of a substituent in a common quinazoline scaffold from one position to another (i.e., a chloro group from the 6- to the 7-position) resulted in a common action in the TST but via a somewhat different mechanism. A6CDQ and A7CDQ might represent the first members of a new class of potential antidepressants with a unique multi-modal mechanism of action.


Subject(s)
Antidepressive Agents/chemistry , Antidepressive Agents/therapeutic use , Hindlimb Suspension/psychology , Quinazolines/chemistry , Quinazolines/therapeutic use , Animals , Antidepressive Agents/pharmacology , Depression/drug therapy , Depression/psychology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , HEK293 Cells , Hindlimb Suspension/adverse effects , Humans , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred ICR , Quinazolines/pharmacology , Serotonin Antagonists/pharmacology , Xenopus laevis
6.
Hum Mol Genet ; 27(18): 3246-3256, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29905862

ABSTRACT

The transcription factor 4 (TCF4) locus is a robust association finding with schizophrenia (SCZ), but little is known about the genes regulated by the encoded transcription factor. Therefore, we conducted chromatin immunoprecipitation sequencing (ChIP-seq) of TCF4 in neural-derived (SH-SY5Y) cells to identify genome-wide TCF4 binding sites, followed by data integration with SCZ association findings. We identified 11 322 TCF4 binding sites overlapping in two ChIP-seq experiments. These sites are significantly enriched for the TCF4 Ebox binding motif (>85% having ≥1 Ebox) and implicate a gene set enriched for genes downregulated in TCF4 small-interfering RNA (siRNA) knockdown experiments, indicating the validity of our findings. The TCF4 gene set was also enriched among (1) gene ontology categories such as axon/neuronal development, (2) genes preferentially expressed in brain, in particular pyramidal neurons of the somatosensory cortex and (3) genes downregulated in postmortem brain tissue from SCZ patients (odds ratio, OR = 2.8, permutation P < 4x10-5). Considering genomic alignments, TCF4 binding sites significantly overlapped those for neural DNA-binding proteins such as FOXP2 and the SCZ-associated EP300. TCF4 binding sites were modestly enriched among SCZ risk loci from the Psychiatric Genomic Consortium (OR = 1.56, P = 0.03). In total, 130 TCF4 binding sites occurred in 39 of the 108 regions published in 2014. Thirteen genes within the 108 loci had both a TCF4 binding site ±10kb and were differentially expressed in siRNA knockdown experiments of TCF4, suggesting direct TCF4 regulation. These findings confirm TCF4 as an important regulator of neural genes and point toward functional interactions with potential relevance for SCZ.


Subject(s)
Gene Regulatory Networks/genetics , Genome, Human/genetics , Schizophrenia/genetics , Transcription Factor 4/genetics , Binding Sites/genetics , Brain/metabolism , Brain/pathology , Chromatin Immunoprecipitation , Gene Ontology , Genetic Predisposition to Disease , Humans , Neurogenesis/genetics , Postmortem Changes , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Schizophrenia/physiopathology , Somatosensory Cortex/metabolism , Somatosensory Cortex/pathology
7.
J Food Drug Anal ; 26(2S): S45-S60, 2018 04.
Article in English | MEDLINE | ID: mdl-29703386

ABSTRACT

Many drugs, hormones, components of herbal medicines, environmental pesticides and toxins are Solute Carrier family 22 (SLC22) substrates. The last twenty years has seen great progress in determining SLC22 tissue expression profiles, membrane localization, energetics, substrate profiles and biopharmaceutical significance. However, much still remains to be answered in terms of SLC22 family member's roles in 'normal' physiology as compared to pathophysiological states, as well as in drug interactions that impact pharmacokinetics, efficacy and toxicity. This review begins with a brief synopsis of SLC22 family discovery, function and tissue expression. Subsequent sections provide examples establishing a role for SLC22 transporters in food-drug, herbal supplement-drug, endogenous substrate-drug and drug-drug interactions.


Subject(s)
Drug Interactions , Food-Drug Interactions , Organic Cation Transport Proteins/metabolism , Animals , Dietary Supplements/adverse effects , Dietary Supplements/analysis , Humans , Multigene Family , Organic Cation Transport Proteins/genetics
8.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1068-1069: 64-70, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29031110

ABSTRACT

A rapid and selective method for the quantitation of neurotransmitters, l-Glutamic acid (GA) and γ-Aminobutyric acid (GABA), was developed and validated using gas chromatography-tandem mass spectrometry (GC-MS/MS). The novel method utilized a rapid online hot GC inlet gas phase sample derivatization and fast GC low thermal mass technology. The method calibration was linear from 0.5 to 100µg/mL, with limits of detections of 100ng/mL and 250ng/mL for GA and GABA, respectively. The method was used to investigate the effects of deletion of organic anion transporter 1 (Oat1) or Oat3 on murine CNS levels of GA and GABA at 3 and 18 mo of age, as compared to age matched wild-type (WT) animals. Whole brain concentrations of GA were comparable between WT, Oat1-/-, and Oat3-/- 18 mo at both 3 and 18 mo of age. Similarly, whole brain concentrations of GABA were not significantly altered in either knockout mouse strain at 3 or 18 mo of age, as compared to WT. These results indicate that the developed GC-MS/MS method provides sufficient sensitivity and selectivity for the quantitation of these neurotransmitters in mouse brain tissue. Furthermore, these results suggest that loss of Oat1 or Oat3 function in isolation does not result in significant alterations in brain tissue levels of GA or GABA.


Subject(s)
Brain Chemistry/physiology , Gas Chromatography-Mass Spectrometry/methods , Glutamic Acid/analysis , Tandem Mass Spectrometry/methods , gamma-Aminobutyric Acid/analysis , Animals , Limit of Detection , Linear Models , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results
9.
Bioorg Med Chem Lett ; 27(18): 4440-4445, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28811134

ABSTRACT

Human organic cation transporters (OCTs) represent an understudied neurotransmitter uptake mechanism for which no selective agents have yet been identified. Several neurotransmitters (e.g. serotonin, norepinephrine) are low-affinity substrates for these transporters, but possess higher affinity for other transporters (e.g. the serotonin or norepinephrine transporters; SERT and NET, respectively). We have identified a new class of OCT inhibitors with a phenylguanidine structural scaffold. Here, we examine the actions of a series of such compounds and report preliminary structure-activity relationships (SARs) - the first dedicated SAR study of OCT3 action. Initial results showed that the presence of a substituent on the phenyl ring, as well as its position, contributes to the phenylguanidines' inhibitory potency (IC50 values ranging from 2.2 to >450µM) at hOCT3. There is a trend towards enhanced inhibitory potency of phenylguanidines with increased lipophilic character and the size of the substituent at the phenyl 4-position, with the latter reaching a ceiling effect. The first PiPT-based hOCT3 homology models were generated and are in agreement with our biological data.


Subject(s)
Guanidines/pharmacology , Organic Cation Transport Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , Guanidines/chemical synthesis , Guanidines/chemistry , Humans , Molecular Structure , Organic Cation Transport Proteins/metabolism , Structure-Activity Relationship
10.
Neuropharmacology ; 117: 182-194, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28192112

ABSTRACT

There is accumulating evidence that bumetanide, which has been used over decades as a potent loop diuretic, also exerts effects on brain disorders, including autism, neonatal seizures, and epilepsy, which are not related to its effects on the kidney but rather mediated by inhibition of the neuronal Na-K-Cl cotransporter isoform NKCC1. However, following systemic administration, brain levels of bumetanide are typically below those needed to inhibit NKCC1, which critically limits its clinical use for treating brain disorders. Recently, active efflux transport at the blood-brain barrier (BBB) has been suggested as a process involved in the low brain:plasma ratio of bumetanide, but it is presently not clear which transporters are involved. Understanding the processes explaining the poor brain penetration of bumetanide is needed for developing strategies to improve the brain delivery of this drug. In the present study, we administered probenecid and more selective inhibitors of active transport carriers at the BBB directly into the brain of mice to minimize the contribution of peripheral effects on the brain penetration of bumetanide. Furthermore, in vitro experiments with mouse organic anion transporter 3 (Oat3)-overexpressing Chinese hamster ovary cells were performed to study the interaction of bumetanide, bumetanide derivatives, and several known inhibitors of Oats on Oat3-mediated transport. The in vivo experiments demonstrated that the uptake and efflux of bumetanide at the BBB is much more complex than previously thought. It seems that both restricted passive diffusion and active efflux transport, mediated by Oat3 but also organic anion-transporting polypeptide (Oatp) Oatp1a4 and multidrug resistance protein 4 explain the extremely low brain concentrations that are achieved after systemic administration of bumetanide, limiting the use of this drug for targeting abnormal expression of neuronal NKCC1 in brain diseases.


Subject(s)
Blood-Brain Barrier/physiology , Brain/metabolism , Bumetanide/pharmacokinetics , Multidrug Resistance-Associated Proteins/physiology , Organic Anion Transporters, Sodium-Independent/physiology , Organic Cation Transport Proteins/physiology , Animals , Biological Transport/drug effects , Blood-Brain Barrier/drug effects , Brain/drug effects , Bumetanide/analogs & derivatives , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Diffusion , Female , Membrane Transport Modulators/pharmacology , Mice , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/metabolism , Probenecid/pharmacology
11.
Eur J Pharm Sci ; 101: 66-70, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28185988

ABSTRACT

Chronic kidney disease (CKD) is a health problem worldwide. Indoxyl sulfate (IS) is a nephro-cardiovascular toxin accumulated in CKD patients and cannot be removed through hemodialysis. The renal excretion of IS was mediated by organic anion transporters (OATs) OAT 1 and OAT 3. Because a number of nonsteroidal anti-inflammatory drugs (NSAIDs) have been reported to inhibit OATs, we hypothesize that NSAIDs might inhibit the renal excretion of IS. Rats were intravenously injected IS with and without diclofenac, ketoprofen or salicylic acid, individually. Blood samples were collected at predetermined time points and the concentrations of IS were determined by HPLC method. The results showed that diclofenac and ketoprofen at 10.0mg/kg significantly decreased the systemic clearance of IS by 71% and 82%, and increased the MRT of IS by 106% and 105%, respectively, whereas salicylic acid did not exhibit significant effects. Cell studies indicated that diclofenac and ketoprofen inhibited the uptake of IS mediated by OAT 1 and OAT 3. In conclusion, diclofenac and ketoprofen inhibited the excretion of IS through inhibition on OAT 1 and OAT 3.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cardiovascular System/drug effects , Indican/urine , Kidney/drug effects , Renal Elimination/drug effects , Toxins, Biological/urine , Animals , CHO Cells , Cell Line , Cricetulus , Diclofenac/pharmacology , Dogs , HEK293 Cells , Humans , Ketoprofen/pharmacology , Kidney/metabolism , Madin Darby Canine Kidney Cells , Male , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Rats , Rats, Sprague-Dawley , Salicylic Acid/pharmacology
12.
Sci Rep ; 5: 16226, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26552961

ABSTRACT

Chronic kidney disease (CKD) is a major health problem worldwide. Indoxyl sulfate (IS) and p-cresyl sulfate (PCS) are highly protein-bound nephro-cardiovascular toxins, which are not efficiently removed through hemodialysis. The renal excretions of IS and PCS were mediated by organic anion transporters (OATs) such as OAT1 and OAT3. Green tea (GT) is a popular beverage containing plenty of catechins. Previous pharmacokinetic studies of teas have shown that the major molecules present in the bloodstream are the glucuronides/sulfates of tea catechins, which are putative substrates of OATs. Here we demonstrated that GT ingestion significantly elevated the systemic exposures of endogenous IS and PCS in rats with chronic renal failure (CRF). More importantly, GT also significantly increased the levels of serum creatinine (Cr) and blood urea nitrogen (BUN) in CRF rats. Mechanism studies indicated that the serum metabolites of GT (GTM) inhibited the uptake transporting functions of OAT1 and OAT3. In conclusion, GT inhibited the elimination of nephro-cardiovascular toxins such as IS and PCS, and deteriorated the renal function in CRF rats.


Subject(s)
Tea/chemistry , Toxins, Biological/metabolism , Adenine/pharmacology , Animals , CHO Cells , Catechin/analysis , Catechin/pharmacology , Creatinine/blood , Cresols/blood , Cresols/pharmacokinetics , Cricetinae , Cricetulus , Disease Models, Animal , Glucuronides/chemistry , HEK293 Cells , Humans , Indican/blood , Indican/pharmacokinetics , Kidney/drug effects , Kidney/metabolism , Male , Organic Anion Transport Protein 1/genetics , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Rats , Rats, Sprague-Dawley , Renal Insufficiency/metabolism , Renal Insufficiency/pathology , Sulfates/chemistry , Sulfuric Acid Esters/blood , Sulfuric Acid Esters/pharmacokinetics , Tea/metabolism , Toxins, Biological/chemistry
13.
Drug Metab Dispos ; 43(8): 1156-68, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986849

ABSTRACT

The recent symposium on "Target-Site" Drug Metabolism and Transport that was sponsored by the American Society for Pharmacology and Experimental Therapeutics at the 2014 Experimental Biology meeting in San Diego is summarized in this report. Emerging evidence has demonstrated that drug-metabolizing enzyme and transporter activity at the site of therapeutic action can affect the efficacy, safety, and metabolic properties of a given drug, with potential outcomes including altered dosing regimens, stricter exclusion criteria, or even the failure of a new chemical entity in clinical trials. Drug metabolism within the brain, for example, can contribute to metabolic activation of therapeutic drugs such as codeine as well as the elimination of potential neurotoxins in the brain. Similarly, the activity of oxidative and conjugative drug-metabolizing enzymes in the lung can have an effect on the efficacy of compounds such as resveratrol. In addition to metabolism, the active transport of compounds into or away from the site of action can also influence the outcome of a given therapeutic regimen or disease progression. For example, organic anion transporter 3 is involved in the initiation of pancreatic ß-cell dysfunction and may have a role in how uremic toxins enter pancreatic ß-cells and ultimately contribute to the pathogenesis of gestational diabetes. Finally, it is likely that a combination of target-specific metabolism and cellular internalization may have a significant role in determining the pharmacokinetics and efficacy of antibody-drug conjugates, a finding which has resulted in the development of a host of new analytical methods that are now used for characterizing the metabolism and disposition of antibody-drug conjugates. Taken together, the research summarized herein can provide for an increased understanding of potential barriers to drug efficacy and allow for a more rational approach for developing safe and effective therapeutics.


Subject(s)
Pharmaceutical Preparations/metabolism , Animals , Biological Transport , Biological Transport, Active , Drug Delivery Systems , Humans , Inactivation, Metabolic
14.
Pharm Res ; 31(12): 3503-14, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24962510

ABSTRACT

PURPOSE: To evaluate organic anion transporter-mediated drug-drug interaction (DDI) potential for individual active components of Danshen (Salvia miltiorrhiza) vs. combinations using in vitro and in silico approaches. METHODS: Inhibition profiles for single Danshen components and combinations were generated in stably-expressing human (h)OAT1 and hOAT3 cells. Plasma concentration-time profiles for compounds were estimated from in vivo human data using an i.v. two-compartment model (with first-order elimination). The cumulative DDI index was proposed as an indicator of DDI potential for combination products. This index was used to evaluate the DDI potential for Danshen injectables from 16 different manufacturers and 14 different lots from a single manufacturer. RESULTS: The cumulative DDI index predicted in vivo inhibition potentials, 82% (hOAT1) and 74% (hOAT3), comparable with those observed in vitro, 72 ± 7% (hOAT1) and 81 ± 10% (hOAT3), for Danshen component combinations. Using simulated unbound Cmax values, a wide range in cumulative DDI index between manufacturers, and between lots, was predicted. Many products exhibited a cumulative DDI index > 1 (50% inhibition). CONCLUSIONS: Danshen injectables will likely exhibit strong potential to inhibit hOAT1 and hOAT3 function in vivo. The proposed cumulative DDI index might improve prediction of DDI potential of herbal medicines or pharmaceutical preparations containing multiple components.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Organic Anion Transporters/metabolism , Salvia miltiorrhiza/chemistry , Animals , CHO Cells , Cell Line , Computer Simulation , Cricetinae , Cricetulus , Drug Carriers , Drug Interactions , Drugs, Chinese Herbal/pharmacokinetics , Humans , Injections , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism
15.
Semin Nephrol ; 34(2): 191-208, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24780473

ABSTRACT

Chronic kidney disease (CKD) is a condition that affects approximately 10% of the adult population in developed countries. In patients with CKD adequate renal clearance is compromised, resulting in the accumulation of a plethora of uremic solutes. These uremic retention solutes, also known as uremic toxins, are a heterogeneous group of organic compounds, many are too large to be filtered (middle molecules) or are protein-bound. Tubular secretion shifts the binding and allows for active secretion of such solutes. To mediate urinary solute excretion, renal proximal tubules are equipped with a range of transporters that cooperate in basolateral uptake and luminal excretion. These putative uremic toxins are poorly filtered across dialysis membranes because they are protein bound and current dialysis therapy does not correct the full spectrum of uremic toxicity. Residual renal function, which may represent an important contribution of solutes secreted by the proximal tubule rather than unreabsorbed filtrate, is an important predictor of survival of CKD patients. Many of the transporters that mediate the renal excretion of uremic retention solutes were first recognized as mediators of drug trafficking and drug-drug interactions, and a considerable amount of literature concerning the actions of these transporters antedates the recognition of their importance in the proximal renal tubular transport of uremic retention solutes. These transporters include members belonging to the organic cation/anion/zwitterion solute carrier family, such as the organic anion transporters (OAT)1, OAT3, and OATP4C1, and to the adenosine triphosphate binding cassette superfamily of transmembrane transporters, including the multidrug resistance proteins and breast cancer resistance protein. This article draws on this body of information to describe the renal tubular clearance mechanisms for uremic toxins, as well as the intracellular events associated with their accumulation, involving activation of the aryl hydrocarbon receptor, disturbance of mitochondrial functioning, and competition with metabolizing enzymes.


Subject(s)
Kidney Glomerulus/metabolism , Kidney Tubules/metabolism , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Uremia/metabolism , Animals , Humans , Organic Anion Transporters/physiology , Organic Cation Transport Proteins/physiology
16.
Cell Metab ; 19(4): 653-66, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24703697

ABSTRACT

Gestational diabetes (GDM) results from failure of the ß cells to adapt to increased metabolic demands; however, the cause of GDM and the extremely high rate of progression to type 2 diabetes (T2D) remains unknown. Using metabolomics, we show that the furan fatty acid metabolite 3-carboxy-4-methyl-5-propyl-2-furanpropanoic acid (CMPF) is elevated in the plasma of humans with GDM, as well as impaired glucose-tolerant and T2D patients. In mice, diabetic levels of plasma CMPF induced glucose intolerance, impaired glucose-stimulated insulin secretion, and decreased glucose utilization. Mechanistically, we show that CMPF acts directly on the ß cell, causing impaired mitochondrial function, decreasing glucose-induced ATP accumulation, and inducing oxidative stress, resulting in dysregulation of key transcription factors and ultimately reduced insulin biosynthesis. Importantly, specifically blocking its transport through OAT3 or antioxidant treatment could prevent CMPF-induced ß cell dysfunction. Thus, CMPF provides a link between ß cell dysfunction and GDM/T2D that could be targeted therapeutically.


Subject(s)
Furans/blood , Insulin-Secreting Cells/pathology , Mitochondria/pathology , Models, Biological , Organic Anion Transporters, Sodium-Independent/metabolism , Propionates/blood , Adenosine Triphosphate/metabolism , Animals , Furans/adverse effects , Humans , Insulin/biosynthesis , Insulin-Secreting Cells/drug effects , Metabolomics , Mice , Mitochondria/drug effects , Oxidative Stress/physiology , Propionates/adverse effects , Transcription Factors/metabolism
17.
Curr Pharm Des ; 20(10): 1472-86, 2014.
Article in English | MEDLINE | ID: mdl-23789957

ABSTRACT

A major function of the blood brain barrier (BBB) and blood cerebrospinal fluid barrier (BCSFB) is to exert selective control over the flux of organic cations and anions into and out of the CNS compartment. These barriers are dynamic tissues that accomplish this task by expressing dozens of transporter proteins representing numerous transporter families. One such family, belonging to the Solute Carrier (SLC) superfamily, is the organic cation/anion/zwitterion (SLC22) family of transporters, which includes the organic cation transporters (OCTs/OCTNs) and organic anion transporters (OATs). SLC22 transporters interact with a broad range of compounds that include drugs of abuse, environmental toxins/toxicants, opioid analgesics, antidepressant and anxiolytic agents and neurotransmitters and their metabolites. Defining the transport mechanisms controlling the CNS penetration, disposition and clearance of such compounds is fundamental to advancing our understanding of the underlying mechanisms that regulate CNS homeostasis and impact neuronal health. Such information might help direct efforts to improve the efficacy and clinical outcomes of current and future therapeutic agents used in the treatment of CNS disorders. This review focuses on highlighting the identification of the SLC22 transporter family, current knowledge of OCT and OAT expression within the CNS (including brain capillaries, choroid plexus and brain regions relevant to monoaminergic neuronal signaling), and recent data regarding behavioral changes related to mood and anxiety disorders and altered responses to stimulants and antidepressants in SLC22 loss of functions models (knockout/knockdown). In vitro and in vivo evidence of SLC22 localization and transport characteristics within the CNS compartment are summarized.


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Organic Anion Transporters/physiology , Organic Cation Transport Proteins/physiology , Animals , Humans , Ion Transport
18.
Fitoterapia ; 92: 206-10, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24291757

ABSTRACT

Human organic cation transporters (hOCTs; SLC22) are expressed in many organs, including intestine, liver, kidney, heart and brain, where they contribute to the absorption, distribution, and elimination of endogenous and exogenous substances. The alkaloids matrine and oxymatrine are widely used in herbal medicine for the treatment of cancer, as well as viral, and cardiac diseases. Their physicochemical properties indicated that they are potential inhibitors for hOCTs, leading to drug-drug interactions in vivo. Therefore, we assessed the inhibitory effects of matrine and oxymatrine on the function of hOCT1 (SLC22A1), hOCT2 (SLC22A2) and hOCT3 (SLC22A3) using stably transfected transporter-expressing cells. At 100-fold excess, oxymatrine exhibited marked inhibition of hOCT1-mediated substrate uptake (p<0.05), while matrine failed to produce significant inhibition on hOCT1. The IC50 value for oxymatrine on hOCT1 was estimated as 513±132 µM. While there was no significant inhibition of hOCT2 or hOCT3 at 100-fold excess, oxymatrine and matrine showed 42% and 88% inhibition of hOCT3-mediated substrate uptake at 3 and 6mM, respectively. Considering the potential intestinal lumen and reported plasma concentrations of matrine and oxymatrine, these data suggest that drug-drug interactions may occur during hOCT1-mediated hepatic and renal uptake and during hOCT3-mediated intestinal absorption.


Subject(s)
Alkaloids/pharmacology , Cations/metabolism , Kidney/drug effects , Organic Cation Transporter 1/antagonists & inhibitors , Plant Extracts/pharmacology , Quinolizines/pharmacology , Sophora/chemistry , Biological Transport , Drug Interactions , HEK293 Cells , Humans , Intestinal Absorption , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Transfection , Matrines
19.
Article in English | MEDLINE | ID: mdl-24022056

ABSTRACT

In order to elucidate the role of organic anion transporters (OATs) in the renal elimination of gallic acid and gentisic acid, a new, rapid, and sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method has been developed and validated for the simultaneous determination of gallic acid and gentisic acid in cell lysate, using Danshensu as the internal standard (IS). After a simple liquid-liquid extraction, the analytes were detected in negative ESI mode using selected reaction monitoring. The precursor-to-product ion transitions (m/z) were 169.0→125.0, 153.1→108.0, and 196.8→135.2 for gallic acid, gentisic acid, and the IS, respectively. Chromatographic separation was achieved on a C18 column using mobile phases consisting of water with 0.1% acetic acid (A) and acetonitrile with 0.05% formic acid. (B) The total run time was 3min and calibration curves were linear over the concentrations of 0.33-2400ng/mL for both compounds (r(2)>0.995). Good precision (between 3.11% and 14.1% RSD) and accuracy (between -12.7% and 11% bias) was observed for quality controls at concentrations of 0.33 (lower limit of quantification), 1, 50, and 2000ng/mL. The mean extraction recovery of gallic acid and gentisic acid was 80.7% and 83.5%, respectively. Results from post-column infusion and post-extraction methods indicated that the analytical method exhibited negligible matrix effects. Finally, this validated assay was successfully applied in a cellular uptake study to determine the intracellular concentrations of gallic acid and gentisic acid in OAT expressing cells.


Subject(s)
Chromatography, Liquid/methods , Gallic Acid/analysis , Gentisates/analysis , Organic Anion Transporters, Sodium-Independent/metabolism , Tandem Mass Spectrometry/methods , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Stability , Gallic Acid/chemistry , Gallic Acid/metabolism , Gentisates/chemistry , Gentisates/metabolism , Intracellular Space/chemistry , Intracellular Space/metabolism , Linear Models , Mice , Reproducibility of Results
20.
Biochem Pharmacol ; 86(7): 991-6, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23973525

ABSTRACT

Rhein, a major metabolite of the prodrug diacerein and a major component of the medicinal herb Rheum sp., is used for its beneficial effects in a variety of clinical applications including the treatment of osteoarthritis and diabetic nephropathy. The physicochemical properties of rhein are consistent with those of known organic anion transporter (OAT) substrates and inhibitors. Therefore, the inhibitory effect of rhein on human (h) OAT1, hOAT3, hOAT4, and murine (m) Oat1 and mOat3 was examined in heterologous cell lines stably expressing each transporter in isolation. Rhein was shown to potently inhibit hOAT1 and hOAT3, with IC50 estimates in the low nanomolar range (IC50=77.1±5.5 nM and 8.4±2.5 nM, respectively), while poor affinity was observed for hOAT4 (IC50>100 µM). Marked species differences were observed with hOAT1 and hOAT3 exhibiting 3- and 28-fold higher affinity for rhein as compared to their murine orthologs. The estimated drug-drug interaction (DDI) indices (>>0.1) indicated a very strong potential for clinically relevant, rhein perpetrated DDIs mediated by inhibition of hOAT1 (DDI index=5.0; 83% inhibition) and/or hOAT3 (DDI index=46; 98% inhibition) transport activity. These results suggested that rhein, from herbal medicines and/or prodrug conversion, may significantly impact the dosing, efficacy and toxicity (i.e., pharmacokinetics and pharmacodynamics) of co-administered hOAT1 and/or hOAT3 drug substrates.


Subject(s)
Anthraquinones/pharmacokinetics , Organic Anion Transport Protein 1/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Drug Interactions , Humans , Inhibitory Concentration 50 , Mice , Organic Anion Transport Protein 1/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/metabolism , Species Specificity , p-Aminohippuric Acid/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...