Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34507995

RESUMO

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.


Assuntos
Sistema ASC de Transporte de Aminoácidos/antagonistas & inibidores , Ligação Competitiva , Química Computacional , Microscopia Crioeletrônica/métodos , Glutamina/metabolismo , Preparações Farmacêuticas/administração & dosagem , Sistema ASC de Transporte de Aminoácidos/metabolismo , Sítios de Ligação , Desenho de Fármacos , Humanos , Antígenos de Histocompatibilidade Menor/metabolismo , Simulação de Acoplamento Molecular , Preparações Farmacêuticas/química , Ligação Proteica , Domínios Proteicos , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
2.
Biochem J ; 477(8): 1443-1457, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32242892

RESUMO

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.


Assuntos
Sistema ASC de Transporte de Aminoácidos/metabolismo , Aminoácidos Básicos/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Sistema ASC de Transporte de Aminoácidos/química , Sistema ASC de Transporte de Aminoácidos/genética , Aminoácidos Básicos/química , Aminobutiratos/química , Aminobutiratos/metabolismo , Animais , Sítios de Ligação , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Células HEK293 , Humanos , Cinética , Antígenos de Histocompatibilidade Menor/química , Antígenos de Histocompatibilidade Menor/genética , Simulação de Acoplamento Molecular , Ligação Proteica , Ratos , Especificidade por Substrato
3.
ACS Chem Neurosci ; 14(23): 4252-4263, 2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-37994790

RESUMO

Glutamate transporters are responsible for active transport of the major excitatory neurotransmitter glutamate across the cell membrane, regulating the extracellular glutamate concentration in the mammalian brain. Extracellular glutamate levels in the brain are usually in the submicromolar range but can increase by exocytosis, inhibition of cellular uptake, or through glutamate release by reverse transport, as well as other mechanisms, which can lead to neurodegeneration and neuronal cell death. Such conditions can be encountered upon energy deprivation during an ischemic stroke. Here, we developed acetoxymethyl (AM) ester prodrug-like derivatives of excitatory amino acid transporter (EAAT) inhibitors that permeate the cell membrane and are activated, most likely through hydrolysis by endogenous cellular esterases, to form the active EAAT inhibitor. Upon increase in external K+ concentration, the inhibitors block glutamate efflux by EAAT reverse transport. Using a novel high-affinity fluorescent prodrug-like inhibitor, dl-threo-9-anthracene-methoxy-aspartate (TAOA) AM ester, we demonstrate that the precursor rapidly accumulates inside cells. Electrophysiological methods and fluorescence assays utilizing the iGluSnFR external glutamate sensor were used to demonstrate the efficacy of AM ester-protected inhibitors in inhibiting K+-mediated glutamate release. Together, our results provide evidence for a novel method to potentially prevent glutamate release by reverse transport under pathophysiological conditions in a model cell system, as well as in human astrocytes, while leaving glutamate uptake under physiological conditions operational. This method could have wide-ranging applications in the prevention of glutamate-induced neuronal cell death.


Assuntos
Ácido Glutâmico , Pró-Fármacos , Animais , Humanos , Ácido Glutâmico/metabolismo , Pró-Fármacos/farmacologia , Transporte Biológico , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Ésteres , Mamíferos/metabolismo
4.
J Phys Org Chem ; 35(11)2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36568026

RESUMO

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.

5.
Front Physiol ; 12: 777050, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867484

RESUMO

SLC6A14 (solute carrier family 6 member 14) is an amino acid transporter, driven by Na+ and Cl- co-transport, whose structure, function, and molecular and kinetic mechanism have not been well characterized. Its broad substrate selectivity, including neutral and cationic amino acids, differentiates it from other SLC6 family members, and its proposed involvement in nutrient transport in several cancers suggest that it could become an important drug target. In the present study, we investigated SLC6A14 function and its kinetic mechanism after expression in human embryonic kidney (HEK293) cells, including substrate specificity and voltage dependence under various ionic conditions. We applied rapid solution exchange, voltage jumps, and laser photolysis of caged alanine, allowing sub-millisecond temporal resolution, to study SLC6A14 steady state and pre-steady state kinetics. The results highlight the broad substrate specificity and suggest that extracellular chloride enhances substrate transport but is not required for transport. As in other SLC6 family members, Na+ binding to the substrate-free transporter (or conformational changes associated with it) is electrogenic and is likely rate limiting for transporter turnover. Transient current decaying with a time constant of <1ms is also observed after rapid amino acid application, both in forward transport and homoexchange modes, indicating a slightly electrogenic, but fast and not rate-limiting substrate translocation step. Our results, which are consistent with kinetic modeling, suggest rapid transporter turnover rate and substrate translocation with faster kinetics compared with other SLC6 family members. Together, these results provided novel information on the SLC6A14 transport cycle and mechanism, expanding our understanding of SLC6A14 function.

6.
ACS Chem Biol ; 14(5): 1002-1010, 2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-31026143

RESUMO

Plasma-membrane glutamate transporters of the excitatory amino acid transporter (EAAT) family are important for maintaining a low glutamate concentration in the extracellular space of the mammalian brain. Glutamate is believed to be transported in its negatively charged form and energetically driven by the cotransport of three sodium ions, at least two of which are bound within the dielectric of the membrane. It was hypothesized that binding of substrates and competitive inhibitors is also electrogenic because the binding site is located near the center of the membrane. To test this hypothesis, we rapidly applied a low-affinity competitive inhibitor, kainate, to the glutamate transporter subtype EAAT2, resulting in outward transient current caused by movement of net negative charge of the inhibitor into the low dielectric of the protein/membrane. Consistent with these data, rate constants for inhibitor dissociation and binding were also voltage dependent. Our results are supported by electrostatic calculations and molecular dynamics simulations of spontaneous substrate dissociation, showing that the substrate and inhibitor binding site is located within the membrane environment of low dielectric constant. Charge movement caused by binding of negatively charged amino acid substrate is compensated by the charge of cotransported Na+ ion(s), thus preventing inhibition of substrate binding at negative membrane potentials. This charge compensation mechanism may be relevant for other Na+-driven transporters which recognize negatively charged substrates.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Fenômenos Biofísicos , Encéfalo/metabolismo , Ácido Caínico/metabolismo , Cinética , Mamíferos , Potenciais da Membrana , Simulação de Dinâmica Molecular , Especificidade por Substrato
7.
J Gen Physiol ; 151(3): 357-368, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718375

RESUMO

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.


Assuntos
Sistema ASC de Transporte de Aminoácidos/antagonistas & inibidores , Moduladores de Transporte de Membrana/farmacologia , Simulação de Acoplamento Molecular , Sulfonamidas/farmacologia , Ácidos Sulfônicos/farmacologia , Sistema ASC de Transporte de Aminoácidos/química , Sistema ASC de Transporte de Aminoácidos/metabolismo , Sítios de Ligação , Ésteres/química , Células HEK293 , Humanos , Moduladores de Transporte de Membrana/química , Antígenos de Histocompatibilidade Menor/química , Antígenos de Histocompatibilidade Menor/metabolismo , Ligação Proteica , Sulfonamidas/química , Ácidos Sulfônicos/química
8.
Front Chem ; 6: 279, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30137742

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa