Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
1.
J Phys Org Chem ; 35(11)2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36568026

RÉSUMÉ

The glutamine transporter ASCT2 is highly overexpressed in cancer cells. Block of glutamine uptake by ASCT2 is a potential strategy to inhibit growth of cancer cells. However, pharmacology of the ASCT2 binding site is not well established. In this work, we report the computational docking to the binding site, and the synthesis of a new class of ASCT2 inhibitors based on the novel L-hydroxyhomoserine scaffold. While these compounds inhibit the ASCT2 leak anion conductance, as expected for competitive inhibitors, they did not block leak conductance in glutamate transporters (EAAT1-3 and EAAT5). They were also ineffective with respect to subtype ASCT1, which has >57% amino acid sequence similarity to ASCT2. Molecular docking studies agree very well with the experimental results and suggest specific polar interactions in the ASCT2 binding site. Our findings add to the repertoire of ASCT2 inhibitors and will aid in further studies of ASCT2 pharmacology.

2.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article de Anglais | MEDLINE | ID: mdl-34507995

RÉSUMÉ

ASCT2 (SLC1A5) is a sodium-dependent neutral amino acid transporter that controls amino acid homeostasis in peripheral tissues. In cancer, ASCT2 is up-regulated where it modulates intracellular glutamine levels, fueling cell proliferation. Nutrient deprivation via ASCT2 inhibition provides a potential strategy for cancer therapy. Here, we rationally designed stereospecific inhibitors exploiting specific subpockets in the substrate binding site using computational modeling and cryo-electron microscopy (cryo-EM). The final structures combined with molecular dynamics simulations reveal multiple pharmacologically relevant conformations in the ASCT2 binding site as well as a previously unknown mechanism of stereospecific inhibition. Furthermore, this integrated analysis guided the design of a series of unique ASCT2 inhibitors. Our results provide a framework for future development of cancer therapeutics targeting nutrient transport via ASCT2, as well as demonstrate the utility of combining computational modeling and cryo-EM for solute carrier ligand discovery.


Sujet(s)
Système ASC de transport d'acides aminés/antagonistes et inhibiteurs , Fixation compétitive , Chimie computationnelle , Cryomicroscopie électronique/méthodes , Glutamine/métabolisme , Préparations pharmaceutiques/administration et posologie , Système ASC de transport d'acides aminés/métabolisme , Sites de fixation , Conception de médicament , Humains , Antigènes mineurs d'histocompatibilité/métabolisme , Simulation de docking moléculaire , Préparations pharmaceutiques/composition chimique , Liaison aux protéines , Domaines protéiques , Structure tertiaire des protéines , Relation structure-activité
3.
Trends Biochem Sci ; 46(1): 28-40, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32828650

RÉSUMÉ

The solute carrier 16 (SLC16) family represents a diverse group of membrane proteins mediating the transport of monocarboxylates across biological membranes. Family members show a variety of functional roles ranging from nutrient transport and intracellular pH regulation to thyroid hormone homeostasis. Changes in the expression levels and transport function of certain SLC16 transporters are manifested in severe health disorders including cancer, diabetes, and neurological disorders. L-Lactate-transporting SLC16 family members play essential roles in the metabolism of certain tumors and became validated drug targets. This review illuminates the SLC16 family under a new light using structural information obtained from a SLC16 homolog. Furthermore, the role of these transporters in cancer metabolism and how their inhibition can contribute to anticancer therapy are discussed.


Sujet(s)
Transporteurs d'acides monocarboxyliques/composition chimique , Symporteurs/composition chimique , Transport biologique , Humains , Transporteurs d'acides monocarboxyliques/génétique , Conformation des protéines , Symporteurs/génétique , Diffraction des rayons X
4.
Biochem J ; 477(8): 1443-1457, 2020 04 30.
Article de Anglais | MEDLINE | ID: mdl-32242892

RÉSUMÉ

Glutamine transport across cell membranes is performed by a variety of transporters, including the alanine serine cysteine transporter 2 (ASCT2). The substrate-binding site of ASCT2 was proposed to be specific for small amino acids with neutral side chains, excluding basic substrates such as lysine. A series of competitive inhibitors of ASCT2 with low µM affinity were developed previously, on the basis of the 2,4-diaminobutyric acid (DAB) scaffold with a potential positive charge in the side chain. Therefore, we tested whether basic amino acids with side chains shorter than lysine can interact with the ASCT2 binding site. Molecular docking of L-1,3-diaminopropionic acid (L-DAP) and L-DAB suggested that these compounds bind to ASCT2. Consistent with this prediction, L-DAP and L-DAB, but not ornithine, lysine or D-DAP, elicited currents when applied to ASCT2-expressing cells. The currents were carried by anions and showed the hallmark properties of ASCT2 currents induced by transported substrates. The L-DAP response could be eliminated by a competitive ASCT2 inhibitor, suggesting that binding occurs at the substrate binding site. The KM for L-DAP was weakly voltage dependent. Furthermore, the pH dependence of the L-DAP response showed that the compound can bind in several protonation states. Together, these results suggest that the ASCT2 binding site is able to recognize L-amino acids with short, basic side chains, such as the L-DAP derivative ß-N-methylamino-l-Alanine (BMAA), a well-studied neurotoxin. Our results expand the substrate specificity of ASCT2 to include amino acid substrates with positively charged side chains.


Sujet(s)
Système ASC de transport d'acides aminés/métabolisme , Acides aminés basiques/métabolisme , Antigènes mineurs d'histocompatibilité/métabolisme , Système ASC de transport d'acides aminés/composition chimique , Système ASC de transport d'acides aminés/génétique , Acides aminés basiques/composition chimique , Amino-butyrates/composition chimique , Amino-butyrates/métabolisme , Animaux , Sites de fixation , Antienzymes/composition chimique , Antienzymes/métabolisme , Cellules HEK293 , Humains , Cinétique , Antigènes mineurs d'histocompatibilité/composition chimique , Antigènes mineurs d'histocompatibilité/génétique , Simulation de docking moléculaire , Liaison aux protéines , Rats , Spécificité du substrat
5.
Trends Pharmacol Sci ; 40(10): 790-800, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31519459

RÉSUMÉ

There are over 420 human solute carrier (SLC) transporters from 65 families that are expressed ubiquitously in the body. The SLCs mediate the movement of ions, drugs, and metabolites across membranes and their dysfunction has been associated with a variety of diseases, such as diabetes, cancer, and central nervous system (CNS) disorders. Thus, SLCs are emerging as important targets for therapeutic intervention. Recent technological advances in experimental and computational biology allow better characterization of SLC pharmacology. Here we describe recent approaches to modulate SLC transporter function, with an emphasis on the use of computational approaches and computer-aided drug design (CADD) to study nutrient transporters. Finally, we discuss future perspectives in the rational design of SLC drugs.


Sujet(s)
Conception de médicament , Protéines transporteurs de solutés/composition chimique , Système ASC de transport d'acides aminés/composition chimique , Système ASC de transport d'acides aminés/métabolisme , Humains , Ligands , Antigènes mineurs d'histocompatibilité/composition chimique , Antigènes mineurs d'histocompatibilité/métabolisme , Modèles moléculaires , Récepteur somatostatine/composition chimique , Récepteur somatostatine/métabolisme , Protéines transporteurs de solutés/métabolisme , Relation structure-activité
6.
J Gen Physiol ; 151(3): 357-368, 2019 03 04.
Article de Anglais | MEDLINE | ID: mdl-30718375

RÉSUMÉ

The neutral amino acid transporter alanine serine cysteine transporter 2 (ASCT2) belongs to the solute carrier 1 (SLC1) family of transport proteins and transports neutral amino acids, such as alanine and glutamine, into the cell in exchange with intracellular amino acids. This amino acid transport is sodium dependent, but not driven by the transmembrane Na+ concentration gradient. Glutamine transport by ASCT2 is proposed to be important for glutamine homoeostasis in rapidly growing cancer cells to fulfill the energy and nitrogen demands of these cells. Thus, ASCT2 is thought to be a potential anticancer drug target. However, the pharmacology of the amino acid binding site is not well established. Here, we report on the synthesis and characterization of a novel class of ASCT2 inhibitors based on an amino acid scaffold with a sulfonamide/sulfonic acid ester linker to a hydrophobic group. The compounds were designed based on an improved ASCT2 homology model using the human glutamate transporter hEAAT1 crystal structure as a modeling template. The compounds were shown to inhibit with a competitive mechanism and a potency that scales with the hydrophobicity of the side chain. The most potent compound binds with an apparent affinity, K i, of 8 ± 4 µM and can block the alanine response with a K i of 40 ± 23 µM at 200 µM alanine concentration. Computational analysis predicts inhibitor interactions with the binding site through molecular docking. In conclusion, the sulfonamide/sulfonic acid ester scaffold provides facile synthetic access to ASCT2 inhibitors with a potentially large variability in chemical space of the hydrophobic side chain. These inhibitors will be useful chemical tools to further characterize the role of ASCT2 in disease as well as improve our understanding of inhibition mechanisms of this transporter.


Sujet(s)
Système ASC de transport d'acides aminés/antagonistes et inhibiteurs , Modulateurs du transport transmembranaire/pharmacologie , Simulation de docking moléculaire , Sulfonamides/pharmacologie , Acides sulfoniques/pharmacologie , Système ASC de transport d'acides aminés/composition chimique , Système ASC de transport d'acides aminés/métabolisme , Sites de fixation , Esters/composition chimique , Cellules HEK293 , Humains , Modulateurs du transport transmembranaire/composition chimique , Antigènes mineurs d'histocompatibilité/composition chimique , Antigènes mineurs d'histocompatibilité/métabolisme , Liaison aux protéines , Sulfonamides/composition chimique , Acides sulfoniques/composition chimique
7.
Front Chem ; 6: 279, 2018.
Article de Anglais | MEDLINE | ID: mdl-30137742

RÉSUMÉ

The Alanine-Serine-Cysteine transporter (SLC1A5, ASCT2), is a neutral amino acid exchanger involved in the intracellular homeostasis of amino acids in peripheral tissues. Given its role in supplying glutamine to rapidly proliferating cancer cells in several tumor types such as triple-negative breast cancer and melanoma, ASCT2 has been identified as a key drug target. Here we use a range of computational methods, including homology modeling and ligand docking, in combination with cell-based assays, to develop hypotheses for structure-function relationships in ASCT2. We perform a phylogenetic analysis of the SLC1 family and its prokaryotic homologs to develop a useful multiple sequence alignment for this protein family. We then generate homology models of ASCT2 in two different conformations, based on the human EAAT1 structures. Using ligand enrichment calculations, the ASCT2 models are then compared to crystal structures of various homologs for their utility in discovering ASCT2 inhibitors. We use virtual screening, cellular uptake and electrophysiology experiments to identify a non-amino acid ASCT2 inhibitor that is predicted to interact with the ASCT2 substrate binding site. Our results provide insights into the structural basis of substrate specificity in the SLC1 family, as well as a framework for the design of future selective and potent ASCT2 inhibitors as cancer therapeutics.

8.
Oncotarget ; 7(20): 28765-82, 2016 May 17.
Article de Anglais | MEDLINE | ID: mdl-27144834

RÉSUMÉ

The evolutionarily conserved Hippo inhibitory pathway plays critical roles in tissue homeostasis and organ size control, while mutations affecting certain core components contribute to tumorigenesis. Here we demonstrate that proliferation of Hippo pathway mutant human tumor cells exhibiting high constitutive TEAD transcriptional activity was markedly inhibited by dominant negative TEAD4, which did not inhibit the growth of Hippo wild-type cells with low levels of regulatable TEAD-mediated transcription. The tankyrase inhibitor, XAV939, identified in a screen for inhibitors of TEAD transcriptional activity, phenocopied these effects independently of its other known functions by stabilizing angiomotin and sequestering YAP in the cytosol. We also identified one intrinsically XAV939 resistant Hippo mutant tumor line exhibiting lower and less durable angiomotin stabilization. Thus, angiomotin stabilization provides a new mechanism for targeting tumors with mutations in Hippo pathway core components as well as a biomarker for sensitivity to such therapy.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Antienzymes/pharmacologie , Régulation de l'expression des gènes tumoraux/physiologie , Composés hétérocycliques 3 noyaux/pharmacologie , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines membranaires/métabolisme , Protéines du muscle/métabolisme , Protein-Serine-Threonine Kinases/génétique , Facteurs de transcription/métabolisme , Angiomotines , Lignée cellulaire tumorale , Prolifération cellulaire/physiologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Voie de signalisation Hippo , Humains , Protéines des microfilaments , Mutation , Facteurs de transcription à domaine TEA , Tankyrases/antagonistes et inhibiteurs
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...