Inwardly rectifying potassium (Kir) channels are key regulators of diverse physiological processes and may represent novel drug targets for diseases. Their therapeutic potential has not been tested directly, however, due to the lack of drug-like compounds targeting inward rectifiers. The lack of selective """"""""probes"""""""" has also hindered efforts to define the physiological functions of some Kir channels. To overcome this formidable barrier and create new opportunities for studying inward rectifier physiology, the investigators performed a high- throughput screen (HTS) of more than 200,000 compounds for small-molecule modulators of ROMK (Kir1.1), a putative target for a novel class of diuretic. One compound, termed VU590, inhibits ROMK at nanomolar concentrations and Kir7.1 in the low micromolar range, making it the first small-molecule inhibitor of both channels. The investigators went on to use medicinal chemistry to rationally design a nanomolar-affinity probe, termed VU591, which is highly selective for ROMK over more than 60 potential off targets, including inward rectifiers and BK channels.
In Aim 1, the investigators will employ state-of-the-art molecular modeling techniques, atomic structure-guided mutagenesis and electrophysiology to define the VU590/591 binding sites in ROMK and Kir7.1. VU591 is remarkably selective for ROMK and therefore represents a promising candidate for further development for use in animal studies.
In Aim 2, the investigators will first determine if VU591 is active in the native tissue by assessing its effects on K and Na transport in isolated perfused cortical collecting ducts under low- and high-flow conditions. The investigators also discovered a nanomolar-affinity inhibitor of a G-protein regulated inward rectifier (GIRK), a putative therapeutic target for atrial fibrillation.
In Aim 3, the investigators will use medicinal chemistry, structure-guided mutagenesis and electrophysiology to define the molecular binding sites for this novel compound termed VU592. These studies will provide important new insights into the atomic structures of inward rectifiers and generate critically needed probes with which to define the integrative physiology and therapeutic potential of these channels. Lay summary: The investigators will combine medicinal chemistry, advanced computational techniques and classical physiological methods to develop drug-like compounds targeting potassium channels that could be therapeutic targets for hypertension, edema and cardiac arrhythmia.

Public Health Relevance

Inward rectifying potassium (Kir) channels play key physiological roles in diverse cellular functions and may represent novel drug targets. However, the lack of selective pharmacological """"""""probes"""""""" has hindered efforts to explore the integrative physiology and therapeutic potential of most Kir channels. Here we propose to employ medicinal chemistry, atomic structure-guided mutagenesis, kidney tubule microperfusion and electrophysiology to develop the small-molecule pharmacology for Kir1.1, Kir7.1 and Kir3 channels.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK082884-04
Application #
8539781
Study Section
Cellular and Molecular Biology of the Kidney Study Section (CMBK)
Program Officer
Ketchum, Christian J
Project Start
2010-09-01
Project End
2015-08-31
Budget Start
2013-09-01
Budget End
2014-08-31
Support Year
4
Fiscal Year
2013
Total Cost
$248,097
Indirect Cost
$85,915
Name
Vanderbilt University Medical Center
Department
Anesthesiology
Type
Schools of Medicine
DUNS #
004413456
City
Nashville
State
TN
Country
United States
Zip Code
37212
Kharade, Sujay V; Kurata, Haruto; Bender, Aaron M et al. (2018) Discovery, Characterization, and Effects on Renal Fluid and Electrolyte Excretion of the Kir4.1 Potassium Channel Pore Blocker, VU0134992. Mol Pharmacol 94:926-937
Kharade, Sujay V; Sheehan, Jonathan H; Figueroa, Eric E et al. (2017) Pore Polarity and Charge Determine Differential Block of Kir1.1 and Kir7.1 Potassium Channels by Small-Molecule Inhibitor VU590. Mol Pharmacol 92:338-346
Swale, Daniel R; Kurata, Haruto; Kharade, Sujay V et al. (2016) ML418: The First Selective, Sub-Micromolar Pore Blocker of Kir7.1 Potassium Channels. ACS Chem Neurosci 7:1013-23
Swale, Daniel R; Engers, Darren W; Bollinger, Sean R et al. (2016) An insecticide resistance-breaking mosquitocide targeting inward rectifier potassium channels in vectors of Zika virus and malaria. Sci Rep 6:36954
Kharade, Sujay V; Nichols, Colin; Denton, Jerod S (2016) The shifting landscape of KATP channelopathies and the need for 'sharper' therapeutics. Future Med Chem 8:789-802
Piermarini, Peter M; Dunemann, Sonja M; Rouhier, Matthew F et al. (2015) Localization and role of inward rectifier K(+) channels in Malpighian tubules of the yellow fever mosquito Aedes aegypti. Insect Biochem Mol Biol 67:59-73
Ghamari-Langroudi, Masoud; Digby, Gregory J; Sebag, Julien A et al. (2015) G-protein-independent coupling of MC4R to Kir7.1 in hypothalamic neurons. Nature 520:94-8
Swale, Daniel R; Sheehan, Jonathan H; Banerjee, Sreedatta et al. (2015) Computational and functional analyses of a small-molecule binding site in ROMK. Biophys J 108:1094-103
Raphemot, Rene; Estévez-Lao, Tania Y; Rouhier, Matthew F et al. (2014) Molecular and functional characterization of Anopheles gambiae inward rectifier potassium (Kir1) channels: a novel role in egg production. Insect Biochem Mol Biol 51:10-9
Rouhier, Matthew F; Hine, Rebecca M; Park, Seokhwan Terry et al. (2014) Excretion of NaCl and KCl loads in mosquitoes. 2. Effects of the small molecule Kir channel modulator VU573 and its inactive analog VU342. Am J Physiol Regul Integr Comp Physiol 307:R850-61

Showing the most recent 10 out of 24 publications