Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Drug Metab Rev ; : 1-28, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967415

ABSTRACT

This review, part of a special issue on drug-drug interactions (DDIs) spearheaded by the International Society for the Study of Xenobiotics (ISSX) New Investigators, explores the critical role of drug transporters in absorption, disposition, and clearance in the context of DDIs. Over the past two decades, significant advances have been made in understanding the clinical relevance of these transporters. Current knowledge on key uptake and efflux transporters that affect drug disposition and development is summarized. Regulatory guidelines from the FDA, EMA, and PMDA that inform the evaluation of potential transporter-mediated DDIs are discussed in detail. Methodologies for preclinical and clinical testing to assess potential DDIs are reviewed, with an emphasis on the utility of physiologically based pharmacokinetic (PBPK) modeling. This includes the application of relative abundance and expression factors to predict human pharmacokinetics (PK) using preclinical data, integrating the latest regulatory guidelines. Considerations for assessing transporter-mediated DDIs in special populations, including pediatric, hepatic, and renal impairment groups, are provided. Additionally, the impact of transporters at the blood-brain barrier (BBB) on the disposition of CNS-related drugs is explored. Enhancing the understanding of drug transporters and their role in drug disposition and toxicity can improve efficacy and reduce adverse effects. Continued research is essential to bridge remaining gaps in knowledge, particularly in comparison with cytochrome P450 (CYP) enzymes.

2.
Nat Commun ; 15(1): 4380, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782905

ABSTRACT

SLC22A10 is an orphan transporter with unknown substrates and function. The goal of this study is to elucidate its substrate specificity and functional characteristics. In contrast to orthologs from great apes, human SLC22A10, tagged with green fluorescent protein, is not expressed on the plasma membrane. Cells expressing great ape SLC22A10 orthologs exhibit significant accumulation of estradiol-17ß-glucuronide, unlike those expressing human SLC22A10. Sequence alignments reveal a proline at position 220 in humans, which is a leucine in great apes. Replacing proline with leucine in SLC22A10-P220L restores plasma membrane localization and uptake function. Neanderthal and Denisovan genomes show proline at position 220, akin to modern humans, indicating functional loss during hominin evolution. Human SLC22A10 is a unitary pseudogene due to a fixed missense mutation, P220, while in great apes, its orthologs transport sex steroid conjugates. Characterizing SLC22A10 across species sheds light on its biological role, influencing organism development and steroid homeostasis.


Subject(s)
Primates , Animals , Humans , Amino Acid Sequence , Estradiol/metabolism , HEK293 Cells , Hominidae/genetics , Hominidae/metabolism , Mutation, Missense , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Primates/genetics , Pseudogenes , Substrate Specificity
3.
Nat Chem Biol ; 20(1): 62-73, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37474759

ABSTRACT

Cells interpret a variety of signals through G-protein-coupled receptors (GPCRs) and stimulate the generation of second messengers such as cyclic adenosine monophosphate (cAMP). A long-standing puzzle is deciphering how GPCRs elicit different physiological responses despite generating similar levels of cAMP. We previously showed that some GPCRs generate cAMP from both the plasma membrane and the Golgi apparatus. Here we demonstrate that cardiomyocytes distinguish between subcellular cAMP inputs to elicit different physiological outputs. We show that generating cAMP from the Golgi leads to the regulation of a specific protein kinase A (PKA) target that increases the rate of cardiomyocyte relaxation. In contrast, cAMP generation from the plasma membrane activates a different PKA target that increases contractile force. We further validated the physiological consequences of these observations in intact zebrafish and mice. Thus, we demonstrate that the same GPCR acting through the same second messenger regulates cardiac contraction and relaxation dependent on its subcellular location.


Subject(s)
Signal Transduction , Zebrafish , Mice , Animals , Cyclic AMP/metabolism , Second Messenger Systems , Myocytes, Cardiac , Receptors, G-Protein-Coupled/metabolism
4.
Res Sq ; 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37790518

ABSTRACT

SLC22A10 is classified as an orphan transporter with unknown substrates and function. Here we describe the discovery of the substrate specificity and functional characteristics of SLC22A10. The human SLC22A10 tagged with green fluorescent protein was found to be absent from the plasma membrane, in contrast to the SLC22A10 orthologs found in great apes. Estradiol-17ß-glucuronide accumulated in cells expressing great ape SLC22A10 orthologs (over 4-fold, p<0.001). In contrast, human SLC22A10 displayed no uptake function. Sequence alignments revealed two amino acid differences including a proline at position 220 of the human SLC22A10 and a leucine at the same position of great ape orthologs. Site-directed mutagenesis yielding the human SLC22A10-P220L produced a protein with excellent plasma membrane localization and associated uptake function. Neanderthal and Denisovan genomes show human-like sequences at proline 220 position, corroborating that SLC22A10 were rendered nonfunctional during hominin evolution after the divergence from the pan lineage (chimpanzees and bonobos). These findings demonstrate that human SLC22A10 is a unitary pseudogene and was inactivated by a missense mutation that is fixed in humans, whereas orthologs in great apes transport sex steroid conjugates.

5.
bioRxiv ; 2023 Aug 12.
Article in English | MEDLINE | ID: mdl-37609337

ABSTRACT

SLC22A10 is classified as an orphan transporter with unknown substrates and function. Here we describe the discovery of the substrate specificity and functional characteristics of SLC22A10. The human SLC22A10 tagged with green fluorescent protein was found to be absent from the plasma membrane, in contrast to the SLC22A10 orthologs found in great apes. Estradiol-17ß-glucuronide accumulated in cells expressing great ape SLC22A10 orthologs (over 4-fold, p<0.001). In contrast, human SLC22A10 displayed no uptake function. Sequence alignments revealed two amino acid differences including a proline at position 220 of the human SLC22A10 and a leucine at the same position of great ape orthologs. Site-directed mutagenesis yielding the human SLC22A10-P220L produced a protein with excellent plasma membrane localization and associated uptake function. Neanderthal and Denisovan genomes show human-like sequences at proline 220 position, corroborating that SLC22A10 were rendered nonfunctional during hominin evolution after the divergence from the pan lineage (chimpanzees and bonobos). These findings demonstrate that human SLC22A10 is a unitary pseudogene and was inactivated by a missense mutation that is fixed in humans, whereas orthologs in great apes transport sex steroid conjugates.

6.
Clin Pharmacol Ther ; 112(3): 527-539, 2022 09.
Article in English | MEDLINE | ID: mdl-35546260

ABSTRACT

Over the last decade, significant progress been made in elucidating the role of membrane transporters in altering drug disposition, with important toxicological consequences due to changes in localized concentrations of compounds. The topic of "Transporters and Toxicity" was recently highlighted as a scientific session at the International Transporter Consortium (ITC) Workshop 4 in 2021. The current white paper is not intended to be an extensive review on the topic of transporters and toxicity but an opportunity to highlight aspects of the role of transporters in various toxicities with clinically relevant implications as covered during the session. This includes a review of the role of solute carrier transporters in anticancer drug-induced organ injury, transporters as key players in organ barrier function, and the role of transporters in metal/metalloid toxicity.


Subject(s)
Membrane Transport Proteins , Humans
7.
ChemMedChem ; 16(5): 869-880, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33230949

ABSTRACT

The l-type amino acid transporter 1 (LAT1, SLC7A5) imports dietary amino acids and amino acid drugs (e. g., l-DOPA) into the brain, and plays a role in cancer metabolism. Though there have been numerous reports of LAT1-targeted amino acid-drug conjugates (prodrugs), identifying the structural determinants to enhance substrate activity has been challenging. In this work, we investigated the position and orientation of a carbonyl group in linking hydrophobic moieties including the anti-inflammatory drug ketoprofen to l-tyrosine and l-phenylalanine. We found that esters of meta-carboxyl l-phenylalanine had better LAT1 transport rates than the corresponding acylated l-tyrosine analogues. However, as the size of the hydrophobic moiety increased, we observed a decrease in LAT1 transport rate with a concomitant increase in potency of inhibition. Our results have important implications for designing amino acid prodrugs that target LAT1 at the blood-brain barrier or on cancer cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain/drug effects , Ketoprofen/pharmacology , Large Neutral Amino Acid-Transporter 1/metabolism , Prodrugs/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Brain/metabolism , Dose-Response Relationship, Drug , Humans , Ketoprofen/chemistry , Molecular Structure , Particle Size , Prodrugs/chemistry , Structure-Activity Relationship
8.
FASEB J ; 34(12): 15734-15752, 2020 12.
Article in English | MEDLINE | ID: mdl-33124720

ABSTRACT

The human solute carrier 22A (SLC22A) family consists of 23 members, representing one of the largest families in the human SLC superfamily. Despite their pharmacological and physiological importance in the absorption and disposition of a range of solutes, eight SLC22A family members remain classified as orphans. In this study, we used a multifaceted approach to identify ligands of orphan SLC22A15. Ligands of SLC22A15 were proposed based on phylogenetic analysis and comparative modeling. The putative ligands were then confirmed by metabolomic screening and uptake assays in SLC22A15 transfected HEK293 cells. Metabolomic studies and transporter assays revealed that SLC22A15 prefers zwitterionic compounds over cations and anions. We identified eight zwitterions, including ergothioneine, carnitine, carnosine, gabapentin, as well as four cations, including MPP+ , thiamine, and cimetidine, as substrates of SLC22A15. Carnosine was a specific substrate of SLC22A15 among the transporters in the SLC22A family. SLC22A15 transport of several substrates was sodium-dependent and exhibited a higher Km for ergothioneine, carnitine, and carnosine compared to previously identified transporters for these ligands. This is the first study to characterize the function of SLC22A15. Our studies demonstrate that SLC22A15 may play an important role in determining the systemic and tissue levels of ergothioneine, carnosine, and other zwitterions.


Subject(s)
Organic Cation Transport Proteins/genetics , Biological Transport/drug effects , Biological Transport/genetics , Carnitine/pharmacology , Carnosine/pharmacology , Cell Line , Ergothioneine/pharmacology , Gabapentin/pharmacology , Genomics/methods , HEK293 Cells , Humans , Ligands , Phylogeny , Transfection/methods
9.
Proc Natl Acad Sci U S A ; 117(27): 16009-16018, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571913

ABSTRACT

Food and drug products contain diverse and abundant small-molecule additives (excipients) with unclear impacts on human physiology, drug safety, and response. Here, we evaluate their potential impact on intestinal drug absorption. By screening 136 unique compounds for inhibition of the key intestinal transporter OATP2B1 we identified and validated 24 potent OATP2B1 inhibitors, characterized by higher molecular weight and hydrophobicity compared to poor or noninhibitors. OATP2B1 inhibitors were also enriched for dyes, including 8 azo (R-N=N-R') dyes. Pharmacokinetic studies in mice confirmed that FD&C Red No. 40, a common azo dye excipient and a potent inhibitor of OATP2B1, decreased the plasma level of the OATP2B1 substrate fexofenadine, suggesting that FD&C Red No. 40 has the potential to block drug absorption through OATP2B1 inhibition in vivo. However, the gut microbiomes of multiple unrelated healthy individuals as well as diverse human gut bacterial isolates were capable of inactivating the identified azo dye excipients, producing metabolites that no longer inhibit OATP2B1 transport. These results support a beneficial role for the microbiome in limiting the unintended effects of food and drug additives in the intestine and provide a framework for the data-driven selection of excipients. Furthermore, the ubiquity and genetic diversity of gut bacterial azoreductases coupled to experiments in conventionally raised and gnotobiotic mice suggest that variations in gut microbial community structure may be less important to consider relative to the high concentrations of azo dyes in food products, which have the potential to saturate gut bacterial enzymatic activity.


Subject(s)
Bacteria/metabolism , Excipients/metabolism , Food Additives/metabolism , Food , Gastrointestinal Microbiome/physiology , Intestinal Absorption/physiology , Organic Anion Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Anti-Allergic Agents/metabolism , Anti-Allergic Agents/pharmacokinetics , Azo Compounds , Bacteria/isolation & purification , Excipients/pharmacokinetics , Female , Food Additives/pharmacokinetics , Histamine H1 Antagonists, Non-Sedating/metabolism , Histamine H1 Antagonists, Non-Sedating/pharmacokinetics , Humans , Intestinal Absorption/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Terfenadine/analogs & derivatives , ATP-Binding Cassette Sub-Family B Member 4
10.
PLoS Genet ; 15(9): e1008208, 2019 09.
Article in English | MEDLINE | ID: mdl-31553721

ABSTRACT

Variation in steroid hormone levels has wide implications for health and disease. The genes encoding the proteins involved in steroid disposition represent key determinants of interindividual variation in steroid levels and ultimately, their effects. Beginning with metabolomic data from genome-wide association studies (GWAS), we observed that genetic variants in the orphan transporter, SLC22A24 were significantly associated with levels of androsterone glucuronide and etiocholanolone glucuronide (sentinel SNPs p-value <1x10-30). In cells over-expressing human or various mammalian orthologs of SLC22A24, we showed that steroid conjugates and bile acids were substrates of the transporter. Phylogenetic, genomic, and transcriptomic analyses suggested that SLC22A24 has a specialized role in the kidney and appears to function in the reabsorption of organic anions, and in particular, anionic steroids. Phenome-wide analysis showed that functional variants of SLC22A24 are associated with human disease such as cardiovascular diseases and acne, which have been linked to dysregulated steroid metabolism. Collectively, these functional genomic studies reveal a previously uncharacterized protein involved in steroid homeostasis, opening up new possibilities for SLC22A24 as a pharmacological target for regulating steroid levels.


Subject(s)
Organic Cation Transport Proteins/metabolism , Steroids/metabolism , Symporters/metabolism , Androsterone/analogs & derivatives , Androsterone/genetics , Androsterone/metabolism , Animals , Biological Transport , Genome-Wide Association Study/methods , HEK293 Cells , Humans , Metabolomics/methods , Models, Molecular , Organic Cation Transport Proteins/chemistry , Organic Cation Transport Proteins/genetics , Phylogeny , Polymorphism, Single Nucleotide , Symporters/chemistry , Symporters/genetics
11.
Bioorg Med Chem Lett ; 29(16): 2254-2258, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31248771

ABSTRACT

A series of 1,2,3-triazole analogs of the amino acids l-histidine and l-tryptophan were modeled, synthesized and tested for l-type amino acid transporter 1 (LAT1; SLC7A5) activity to guide the design of amino acid-drug conjugates (prodrugs). These triazoles were conveniently prepared by the highly convergent Huisgen 1,3-dipolar cycloaddition (Click Chemistry). Despite comparable predicted binding modes, triazoles generally demonstrated reduced cell uptake and LAT1 binding potency relative to their natural amino acid counterparts. The structure-activity relationship (SAR) data for these triazoles has important ramifications for treating cancer and brain disorders using amino acid prodrugs or LAT1 inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Histidine/pharmacology , Large Neutral Amino Acid-Transporter 1/metabolism , Neoplasms/drug therapy , Prodrugs/pharmacology , Triazoles/pharmacology , Tryptophan/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Brain Diseases/drug therapy , Brain Diseases/metabolism , Click Chemistry , Dose-Response Relationship, Drug , Histidine/chemistry , Humans , Molecular Structure , Neoplasms/metabolism , Prodrugs/chemical synthesis , Prodrugs/chemistry , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Tryptophan/chemistry
12.
J Med Chem ; 61(16): 7358-7373, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30048132

ABSTRACT

The L-type amino acid transporter 1 (LAT1, SLC7A5) transports essential amino acids across the blood-brain barrier (BBB) and into cancer cells. To utilize LAT1 for drug delivery, potent amino acid promoieties are desired, as prodrugs must compete with millimolar concentrations of endogenous amino acids. To better understand ligand-transporter interactions that could improve potency, we developed structural LAT1 models to guide the design of substituted analogues of phenylalanine and histidine. Furthermore, we evaluated the structure-activity relationship (SAR) for both enantiomers of naturally occurring LAT1 substrates. Analogues were tested in cis-inhibition and trans-stimulation cell assays to determine potency and uptake rate. Surprisingly, LAT1 can transport amino acid-like substrates with wide-ranging polarities including those containing ionizable substituents. Additionally, the rate of LAT1 transport was generally nonstereoselective even though enantiomers likely exhibit different binding modes. Our findings have broad implications to the development of new treatments for brain disorders and cancer.


Subject(s)
Large Neutral Amino Acid-Transporter 1/chemistry , Large Neutral Amino Acid-Transporter 1/metabolism , Structure-Activity Relationship , Amino Acid Transport Systems/chemistry , Amino Acid Transport Systems/metabolism , Antiporters/chemistry , Antiporters/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , HEK293 Cells , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Ligands , Molecular Docking Simulation , Phenylalanine/chemistry , Phenylalanine/metabolism , Stereoisomerism , Structural Homology, Protein , Substrate Specificity
13.
Mol Pharmacol ; 94(1): 689-699, 2018 07.
Article in English | MEDLINE | ID: mdl-29720497

ABSTRACT

Species differences in renal drug transporters continue to plague drug development with animal models failing to adequately predict renal drug toxicity. For example, adefovir, a renally excreted antiviral drug, failed clinical studies for human immunodeficiency virus due to pronounced nephrotoxicity in humans. In this study, we demonstrated that there are large species differences in the kinetics of interactions of a key class of antiviral drugs, acyclic nucleoside phosphonates (ANPs), with organic anion transporter 1 [(OAT1) SLC22A6] and identified a key amino acid residue responsible for these differences. In OAT1 stably transfected human embryonic kidney 293 cells, the Km value of tenofovir for human OAT1 (hOAT1) was significantly lower than for OAT1 orthologs from common preclinical animals, including cynomolgus monkey, mouse, rat, and dog. Chimeric and site-directed mutagenesis studies along with comparative structure modeling identified serine at position 203 (S203) in hOAT1 as a determinant of its lower Km value. Furthermore, S203 is conserved in apes, and in contrast alanine at the equivalent position is conserved in preclinical animals and Old World monkeys, the most related primates to apes. Intriguingly, transport efficiencies are significantly higher for OAT1 orthologs from apes with high serum uric acid (SUA) levels than for the orthologs from species with low serum uric acid levels. In conclusion, our data provide a molecular mechanism underlying species differences in renal accumulation of nephrotoxic ANPs and a novel insight into OAT1 transport function in primate evolution.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/metabolism , Kidney/drug effects , Kidney/metabolism , Organic Anion Transport Protein 1/metabolism , Adenine/adverse effects , Adenine/analogs & derivatives , Amino Acids/metabolism , Animals , Antiviral Agents/adverse effects , Cell Line , Cercopithecidae , Dogs , HEK293 Cells , Humans , Kinetics , Macaca fascicularis , Mice , Organophosphonates/adverse effects , Organophosphonates/metabolism , Rats , Species Specificity , Uric Acid/blood
14.
PLoS Biol ; 16(4): e2002907, 2018 04.
Article in English | MEDLINE | ID: mdl-29659562

ABSTRACT

A constellation of metabolic disorders, including obesity, dysregulated lipids, and elevations in blood glucose levels, has been associated with cardiovascular disease and diabetes. Analysis of data from recently published genome-wide association studies (GWAS) demonstrated that reduced-function polymorphisms in the organic cation transporter, OCT1 (SLC22A1), are significantly associated with higher total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride (TG) levels and an increased risk for type 2 diabetes mellitus, yet the mechanism linking OCT1 to these metabolic traits remains puzzling. Here, we show that OCT1, widely characterized as a drug transporter, plays a key role in modulating hepatic glucose and lipid metabolism, potentially by mediating thiamine (vitamin B1) uptake and hence its levels in the liver. Deletion of Oct1 in mice resulted in reduced activity of thiamine-dependent enzymes, including pyruvate dehydrogenase (PDH), which disrupted the hepatic glucose-fatty acid cycle and shifted the source of energy production from glucose to fatty acids, leading to a reduction in glucose utilization, increased gluconeogenesis, and altered lipid metabolism. In turn, these effects resulted in increased total body adiposity and systemic levels of glucose and lipids. Importantly, wild-type mice on thiamine deficient diets (TDs) exhibited impaired glucose metabolism that phenocopied Oct1 deficient mice. Collectively, our study reveals a critical role of hepatic thiamine deficiency through OCT1 deficiency in promoting the metabolic inflexibility that leads to the pathogenesis of cardiometabolic disease.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Longevity/genetics , Obesity/genetics , Octamer Transcription Factor-1/genetics , Thiamine Deficiency/genetics , Thiamine/metabolism , Animals , Blood Glucose/metabolism , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fatty Acids/metabolism , Gene Expression Regulation , Gluconeogenesis/genetics , Humans , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Octamer Transcription Factor-1/deficiency , Signal Transduction , Thiamine Deficiency/metabolism , Thiamine Deficiency/pathology , Triglycerides/blood
15.
Hum Mutat ; 39(6): 811-815, 2018 06.
Article in English | MEDLINE | ID: mdl-29524275

ABSTRACT

Isolated hand syndactyly is a common limb malformation with limited known genetic etiology. We used exome sequencing to discover two novel variants, chr11 g.46896373C>G; p.D1403H and chr11 g.46893078G>T; p.Q1564K, in LRP4 in a child with isolated bilateral syndactyly of the third and fourth fingers. Each variant was inherited from a different parent and neither parent was affected. Variants in LRP4 have been previously associated with syndactyly in Cenani-Lenz syndactyly syndrome and Sclerosteosis 2, but have not been reported in individuals with isolated syndactyly. LRP4 inhibits LRP6/LRP5-mediated activation of canonical Wnt signaling and mediates sclerostin-dependent inhibition of bone formation. p.D1403H and p.Q1564K are located within the fourth ß-propeller of the extracellular protein domain that has yet to be associated with human disease. Functional analyses of p.D1403H and p.Q1564K show that they significantly decrease LRP4's inhibition of Wnt signaling. These results suggest that variants in the fourth ß-propeller of the extracellular protein domain may cause a phenotype distinct from previously characterized LRP4 variants.


Subject(s)
Exome Sequencing , LDL-Receptor Related Proteins/genetics , Limb Deformities, Congenital/genetics , Syndactyly/genetics , Homozygote , Humans , Limb Deformities, Congenital/physiopathology , Mutation , Mutation, Missense/genetics , Pedigree , Phenotype , Syndactyly/physiopathology
16.
Science ; 356(6339): 757-759, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28522536

ABSTRACT

Insulin-like growth factor 2 (IGF2) is the major fetal growth hormone in mammals. We identify zinc finger protein 568 (ZFP568), a member of the rapidly evolving Kruppel-associated box-zinc finger protein (KRAB-ZFP) family linked primarily to silencing of endogenous retroelements, as a direct repressor of a placental-specific Igf2 transcript (designated Igf2-P0) in mice. Loss of Zfp568, which causes gastrulation failure, or mutation of the ZFP568-binding site at the Igf2-P0 promoter causes inappropriate Igf2-P0 activation. Deletion of Igf2 can completely rescue Zfp568 gastrulation phenotypes through late gestation. Our data highlight the exquisite selectivity with which members of the KRAB-ZFP family repress their targets and identify an additional layer of transcriptional control of a key growth factor regulating fetal and placental development.


Subject(s)
Embryo, Mammalian/metabolism , Insulin-Like Growth Factor II/deficiency , Insulin-Like Growth Factor II/genetics , Nuclear Proteins/metabolism , Animals , Female , Gastrulation/genetics , Gene Expression Regulation , Mice , Mice, Knockout , Mutation , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Placenta/metabolism , Pregnancy , Promoter Regions, Genetic/genetics , Repressor Proteins/metabolism
17.
J Med Chem ; 60(7): 2685-2696, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28230985

ABSTRACT

Organic cation transporter 1 (OCT1) plays a critical role in the hepatocellular uptake of structurally diverse endogenous compounds and xenobiotics. Here we identified competitive and noncompetitive OCT1-interacting ligands in a library of 1780 prescription drugs by combining in silico and in vitro methods. Ligands were predicted by docking against a comparative model based on a eukaryotic homologue. In parallel, high-throughput screening (HTS) was conducted using the fluorescent probe substrate ASP+ in cells overexpressing human OCT1. Thirty competitive OCT1 ligands, defined as ligands predicted in silico as well as found by HTS, were identified. Of the 167 ligands identified by HTS, five were predicted to potentially cause clinical drug interactions. Finally, virtual screening of 29 332 metabolites predicted 146 competitive OCT1 ligands, of which an endogenous neurotoxin, 1-benzyl-1,2,3,4-tetrahydroisoquinoline, was experimentally validated. In conclusion, by combining docking and in vitro HTS, competitive and noncompetitive ligands of OCT1 can be predicted.


Subject(s)
Organic Cation Transporter 1/antagonists & inhibitors , Organic Cation Transporter 1/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Drug Discovery , HEK293 Cells , Humans , Ligands , Molecular Docking Simulation , Organic Cation Transporter 1/chemistry
18.
Bioorg Med Chem Lett ; 26(20): 5000-5006, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27624080

ABSTRACT

Large neutral amino acid transporter 1 (LAT1) is a solute carrier protein located primarily in the blood-brain barrier (BBB) that offers the potential to deliver drugs to the brain. It is also up-regulated in cancer cells, as part of a tumor's increased metabolic demands. Previously, amino acid prodrugs have been shown to be transported by LAT1. Carboxylic acid bioisosteres may afford prodrugs with an altered physicochemical and pharmacokinetic profile than those derived from natural amino acids, allowing for higher brain or tumor levels of drug and/or lower toxicity. The effect of replacing phenylalanine's carboxylic acid with a tetrazole, acylsulfonamide and hydroxamic acid (HA) bioisostere was examined. Compounds were tested for their ability to be LAT1 substrates using both cis-inhibition and trans-stimulation cell assays. As HA-Phe demonstrated weak substrate activity, its structure-activity relationship (SAR) was further explored by synthesis and testing of HA derivatives of other LAT1 amino acid substrates (i.e., Tyr, Leu, Ile, and Met). The potential for a false positive in the trans-stimulation assay caused by parent amino acid was evaluated by conducting compound stability experiments for both HA-Leu and the corresponding methyl ester derivative. We concluded that HA's are transported by LAT1. In addition, our results lend support to a recent account that amino acid esters are LAT1 substrates, and that hydrogen bonding may be as important as charge for interaction with the transporter binding site.


Subject(s)
Carboxylic Acids/metabolism , Hydroxamic Acids/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Blood-Brain Barrier , Carboxylic Acids/chemistry , Chromatography, High Pressure Liquid , HEK293 Cells , Humans , Hydroxamic Acids/chemistry , Magnetic Resonance Spectroscopy , Structure-Activity Relationship
19.
Nat Genet ; 48(9): 1055-1059, 2016 09.
Article in English | MEDLINE | ID: mdl-27500523

ABSTRACT

Metformin is the first-line antidiabetic drug with over 100 million users worldwide, yet its mechanism of action remains unclear. Here the Metformin Genetics (MetGen) Consortium reports a three-stage genome-wide association study (GWAS), consisting of 13,123 participants of different ancestries. The C allele of rs8192675 in the intron of SLC2A2, which encodes the facilitated glucose transporter GLUT2, was associated with a 0.17% (P = 6.6 × 10(-14)) greater metformin-induced reduction in hemoglobin A1c (HbA1c) in 10,577 participants of European ancestry. rs8192675 was the top cis expression quantitative trait locus (cis-eQTL) for SLC2A2 in 1,226 human liver samples, suggesting a key role for hepatic GLUT2 in regulation of metformin action. Among obese individuals, C-allele homozygotes at rs8192675 had a 0.33% (3.6 mmol/mol) greater absolute HbA1c reduction than T-allele homozygotes. This was about half the effect seen with the addition of a DPP-4 inhibitor, and equated to a dose difference of 550 mg of metformin, suggesting rs8192675 as a potential biomarker for stratified medicine.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucose Transporter Type 2/genetics , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Polymorphism, Single Nucleotide/genetics , Quantitative Trait, Heritable , Blood Glucose/analysis , Body Mass Index , Diabetes Mellitus, Type 2/drug therapy , Genome-Wide Association Study , Glycated Hemoglobin/analysis , Humans , White People
20.
Bioorg Med Chem Lett ; 26(11): 2616-2621, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27106710

ABSTRACT

The transporter protein Large-neutral Amino Acid Transporter 1 (LAT-1, SLC7A5) is responsible for transporting amino acids such as tyrosine and phenylalanine as well as thyroid hormones, and it has been exploited as a drug delivery mechanism. Recently its role in cancer has become increasingly appreciated, as it has been found to be up-regulated in many different tumor types, and its expression levels have been correlated with prognosis. Substitution at the meta position of aromatic amino acids has been reported to increase affinity for LAT-1; however, the SAR for this position has not previously been explored. Guided by newly refined computational models of the binding site, we hypothesized that groups capable of filling a hydrophobic pocket would increase binding to LAT-1, resulting in improved substrates relative to parent amino acid. Tyrosine and phenylalanine analogs substituted at the meta position with halogens, alkyl and aryl groups were synthesized and tested in cis-inhibition and trans-stimulation cell assays to determine activity. Contrary to our initial hypothesis we found that lipophilicity was correlated with diminished substrate activity and increased inhibition of the transporter. The synthesis and SAR of meta-substituted phenylalanine and tyrosine analogs is described.


Subject(s)
Large Neutral Amino Acid-Transporter 1/metabolism , Phenylalanine/pharmacology , Tyrosine/pharmacology , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Structure-Activity Relationship , Tyrosine/analogs & derivatives , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL