scispace - formally typeset
Open AccessJournal ArticleDOI

AR-C155858 is a potent inhibitor of monocarboxylate transporters MCT1 and MCT2 that binds to an intracellular site involving transmembrane helices 7–10

Reads0
Chats0
TLDR
Measurement of the inhibitor sensitivity of several chimaeric transporters combining different domains of MCT1 and MCT4 revealed that the binding site for AR-C155858 is contained within the C-terminal half of M CT1, and involves TM (transmembrane) domains 7–10, consistent with previous data identifying Phe360 and Asp302 plus Arg306 as key residues in substrate binding and translocation by MCT 1.
Abstract
In the present study we characterize the properties of the potent MCT1 (monocarboxylate transporter 1) inhibitor AR-C155858. Inhibitor titrations of L-lactate transport by MCT1 in rat erythrocytes were used to determine the Ki value and number of AR-C155858-binding sites (Et) on MCT1 and the turnover number of the transporter (kcat). Derived values were 2.3±1.4 nM, 1.29±0.09 nmol per ml of packed cells and 12.2±1.1 s−1 respectively. When expressed in Xenopus laevis oocytes, MCT1 and MCT2 were potently inhibited by AR-C155858, whereas MCT4 was not. Inhibition of MCT1 was shown to be time-dependent, and the compound was also active when microinjected, suggesting that AR-C155858 probably enters the cell before binding to an intracellular site on MCT1. Measurement of the inhibitor sensitivity of several chimaeric transporters combining different domains of MCT1 and MCT4 revealed that the binding site for AR-C155858 is contained within the C-terminal half of MCT1, and involves TM (transmembrane) domains 7–10. This is consistent with previous data identifying Phe360 (in TM10) and Asp302 plus Arg306 (TM8) as key residues in substrate binding and translocation by MCT1. Measurement of the Km values of the chimaeras for L-lactate and pyruvate demonstrate that both the C- and N-terminal halves of the molecule influence transport kinetics consistent with our proposed molecular model of MCT1 and its translocation mechanism that requires Lys38 in TM1 in addition to Asp302 and Arg306 in TM8 [Wilson, Meredith, Bunnun, Sessions and Halestrap (2009) J. Biol. Chem. 284, 20011–20021].

read more

Content maybe subject to copyright    Report

Citations
More filters
Journal ArticleDOI

Targeting hypoxia in cancer therapy

TL;DR: The two main approaches, namely bioreductive prodrugs and inhibitors of molecular targets upon which hypoxic cell survival depends are reviewed, and the particular challenges and opportunities these overlapping strategies present are addressed.
Journal ArticleDOI

Targeting lactate metabolism for cancer therapeutics

TL;DR: The mechanisms of lactate production and transport are reviewed and emerging evidence indicating that targeting lactate metabolism is a promising approach for cancer therapeutics is highlighted.
Journal ArticleDOI

The Science and Translation of Lactate Shuttle Theory

TL;DR: Clinical studies are utilizing lactate to treat pro-inflammatory conditions and to deliver optimal fuel for working muscles in sports medicine and "Lactate shuttle" concepts describe the roles of lactate in delivery of oxidative and gluconeogenic substrates as well as in cell signaling.
Journal ArticleDOI

The SLC16 gene family – Structure, role and regulation in health and disease☆

TL;DR: It is suggested that the development of other drugs specifically targeting different MCT isoforms may provide a novel approach to cancer chemotherapy.
Journal ArticleDOI

Lactate Regulates Metabolic and Pro-inflammatory Circuits in Control of T Cell Migration and Effector Functions.

TL;DR: A novel role of lactate is established in control of proinflammatory T cell motility and effector functions, which provides a potential molecular mechanism for T cell entrapment and functional changes in inflammatory sites that drive chronic inflammation and offer targeted therapeutic interventions for the treatment of chronic inflammatory disorders.
References
More filters
Journal ArticleDOI

Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice

TL;DR: In this paper, the authors identified monocarboxylate transporter 1 (MCT1) as the prominent path for lactate uptake by a human cervix squamous carcinoma cell line that preferentially utilized lactate for oxidative metabolism.
Journal ArticleDOI

The proton-linked monocarboxylate transporter (MCT) family: structure, function and regulation

TL;DR: There is still much work to be done to characterize the properties of the different MCT isoforms and their regulation, which may have wide-ranging implications for health and disease.
Journal ArticleDOI

The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond.

TL;DR: The monocarboxylate cotransporter family now comprises 14 members, of which only the first four (MCT1–MCT4) have been demonstrated experimentally to catalyse the proton-linked transport of metabolically important monoccarboxylates such as lactate, pyruvate and ketone bodies.
Journal ArticleDOI

The Plasma Membrane Lactate Transporter MCT4, but Not MCT1, Is Up-regulated by Hypoxia through a HIF-1α-dependent Mechanism

TL;DR: It is concluded that MCT4, like other glycolytic enzymes, is up-regulated by hypoxia through a HIF-1α-mediated mechanism, which allows the increased lactic acid produced during Hypoxia to be rapidly lost from the cell.
Journal ArticleDOI

Transport of lactate and other monocarboxylates across mammalian plasma membranes

TL;DR: There are distinct Na(+)-monocarboxylate cotransporters on the luminal surface of intestinal and kidney epithelia, which enable active uptake of lactate, pyruvate, and ketone bodies in these tissues.
Related Papers (5)