Drug Transporters. Группа авторов

Чтение книги онлайн.

Читать онлайн книгу Drug Transporters - Группа авторов страница 40

Drug Transporters - Группа авторов

Скачать книгу

and pharmacogenetic studies (see below).

      3.3.3 Inhibitors

      MATE transport can be inhibited by a number of chemicals. Structural examples are shown in Fig. 3.2. The antiparasitic drug, pyrimethamine, is considered one of the most potent and specific inhibitors. Inhibition of mMate1/hMATE1 is achieved at nanomolar concentrations of pyrimethamine, whereas micromolar concentrations are required to block mOct1/hOCT2 and mOct2/hOCT2 [52]. Cis‐inhibition of TEA transport by hMATE1 is observed with cimetidine, quinidine, procainamide, and verapamil and to a lesser degree nicotine, serotonin, and choline [5, 6]. Likewise, a number of hormones can inhibit TEA transport in hMATE1‐ and mMate1‐expressing cells including corticosterone, testosterone, progesterone, and androstenedione [15]. Notably, no effects on hMATE1 or hMATE2‐K activity by organic anions such as p‐aminohippurate, uric acid, and beta‐lactam antibiotics were observed [5, 6].

      3.4.1 Modeling of Ligand Interactions

      A variety of experimental approaches, including Bayesian machine learning, binary classification modeling, molecular docking and pharmacophore modeling, have been employed to define physicochemical and structural properties of MATE ligands. Inhibitors of MATE1 have been defined by their cationic charge, high molecular weight, and lipophilicity [53]. Using a combination of in vitro and computational approaches, a pharmacophore for hMATE1 and hMATE2‐K inhibitors was defined that favors shared features of high‐affinity inhibitors (such as pyrimethamine and quinidine) and avoids structures observed in low‐affinity inhibitors (such as histamine, caffeine, and chloramphenicol) [54]. Refinement of the model resulted in a pharmacophore for MATE inhibition that included two hydrophobes, a hydrogen‐bond acceptor, and an ionizable feature [54]. A subsequent combinatorial pharmacophore model for hMATE1 inhibition predicted multiple sites for ligand interaction. These included two regions for competitive inhibition by smaller molecules, as well as accommodation of large inhibitors within the central cavity of the transporter where they act noncompetitively to prevent the conformational changes needed for organic cation transport [55].

      3.4.2 Secondary Structure

      Using the X‐ray structure of the NorM transporter (3.65 Å) [64], a homology model for hMATE1 was developed [63]. This model positions hMATE1 in two bundles of six transmembrane helices (N lobe: transmembrane domains 1–6, C lobe: transmembrane domains 7–12) with an internal cavity of ~4,000 Å that is open to the extracellular space [63]. A relatively short cytoplasmic loop between domains 6 and 7 is positioned to connect the two halves and is consistent with hydropathy plots for N or M. Evaluation of the crystal structure of a MATE transporter from Arabidopsis thaliana (2.6 Å), known as AtDTX14, has also provided insights into hMATE1 structure and function [65]. The amino acid sequence identity between hMATE1 and AtDTX14 is 32%. A key hydrogen bonding network in the C‐lobe demonstrated in AtDTX14 is conserved in hMATE1 and is considered to be the substrate‐binding site [65]. While key insights into the likely structure of hMATE1 (and orthologs) have been made, there is little insight into similarities and differences for MATE2/2‐K, as well as a need for a MATE structure from crystallography or cryo‐electron microscopy.

Schematic illustration of predicted structure of the human MATE1 transporter.

      3.4.3 Structural Features

      Treatment of cells expressing rMate1 with p‐chloromercuribenzene sulfonate, an organic mercurial chemical, significantly reduced uptake of TEA. This inhibition of transport could be rescued by dithiothreitol suggesting that reduced sulfhydryl groups are important for the activity of rMate1 [17]. Subsequent site‐directed mutagenesis identified key residues [66]. One histidine (rMate1: His‐385, hMATE1: His‐386, hMATE2‐K: His‐382) and two cysteine residues (rMate1: Cys‐62 and Cys‐126, hMATE1: Cys‐63 and Cys‐127, hMATE2‐K: Cys‐59 and Cys‐123) were essential for transport activity. The impaired function of mutants in these residues was not due to improper trafficking or reduced expression as all localized to the plasma membrane to similar degrees as wild‐type proteins [66]. Interestingly, unlabeled TEA was able to protect against the transport inhibition achieved by the sulfhydryl reagent PCMBS suggesting that rMate1 substrates interact directly with sulfhydryl‐containing Cys‐62 and Cys‐126 [66]. By comparison, unlabeled TEA had no effect on the ability of the histidine residue modifier DEPC to block rMate1 activity in vitro.

Скачать книгу