Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Mol Pharmacol ; 105(3): 202-212, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38302135

ABSTRACT

Vascular smooth muscle KATP channels critically regulate blood flow and blood pressure by modulating vascular tone and therefore represent attractive drug targets for treating several cardiovascular disorders. However, the lack of potent inhibitors that can selectively inhibit Kir6.1/SUR2B (vascular KATP) over Kir6.2/SUR1 (pancreatic KATP) has eluded discovery despite decades of intensive research. We therefore screened 47,872 chemically diverse compounds for novel inhibitors of heterologously expressed Kir6.1/SUR2B channels. The most potent inhibitor identified in the screen was an N-aryl-N'-benzyl urea compound termed VU0542270. VU0542270 inhibits Kir6.1/SUR2B with an IC50 of approximately 100 nM but has no apparent activity toward Kir6.2/SUR1 or several other members of the Kir channel family at doses up to 30 µM (>300-fold selectivity). By expressing different combinations of Kir6.1 or Kir6.2 with SUR1, SUR2A, or SUR2B, the VU0542270 binding site was localized to SUR2. Initial structure-activity relationship exploration around VU0542270 revealed basic texture related to structural elements that are required for Kir6.1/SUR2B inhibition. Analysis of the pharmacokinetic properties of VU0542270 showed that it has a short in vivo half-life due to extensive metabolism. In pressure myography experiments on isolated mouse ductus arteriosus vessels, VU0542270 induced ductus arteriosus constriction in a dose-dependent manner similar to that of the nonspecific KATP channel inhibitor glibenclamide. The discovery of VU0542270 provides conceptual proof that SUR2-specific KATP channel inhibitors can be developed using a molecular target-based approach and offers hope for developing cardiovascular therapeutics targeting Kir6.1/SUR2B. SIGNIFICANCE STATEMENT: Small-molecule inhibitors of vascular smooth muscle KATP channels might represent novel therapeutics for patent ductus arteriosus, migraine headache, and sepsis; however, the lack of selective channel inhibitors has slowed progress in these therapeutic areas. Here, this study describes the discovery and characterization of the first vascular-specific KATP channel inhibitor, VU0542270.


Subject(s)
KATP Channels , Animals , Mice , Glyburide , KATP Channels/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Sulfonylurea Receptors/antagonists & inhibitors
2.
Assay Drug Dev Technol ; 21(3): 89-96, 2023 04.
Article in English | MEDLINE | ID: mdl-36930852

ABSTRACT

Antagonists of the serotonin receptor 2B (5-HT2B) have shown great promise as therapeutics for the treatment of pulmonary arterial hypertension, valvular heart disease, and related cardiopathies. Herein, we describe a high-throughput screen campaign that led to the identification of highly potent and selective 5-HT2B antagonists. Furthermore, selected compounds were profiled for their predicted ability to cross the blood-brain barrier. Two exemplary compounds, VU0530244 and VU0631019, were predicted to have very limited potential for brain penetration in human subjects, a critical profile for the development of 5-HT2B antagonists devoid of centrally-mediated adverse effects.


Subject(s)
Receptor, Serotonin, 5-HT2B , Serotonin , Humans
3.
Bioorg Med Chem Lett ; 87: 129256, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36966977

ABSTRACT

Kir6.2/SUR1 is an ATP-regulated potassium channel that acts as an intracellular metabolic sensor, controlling insulin and appetite-stimulatory neuropeptides secretion. In this Letter, we present the SAR around a novel Kir6.2/SUR1 channel opener scaffold derived from an HTS screening campaign. New series of compounds with tractable SAR trends and favorable potencies are reported.


Subject(s)
Sulfonylurea Receptors , Sulfonylurea Receptors/metabolism
4.
Proc Natl Acad Sci U S A ; 120(4): e2212338120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36649421

ABSTRACT

To fertilize an oocyte, the membrane potential of both mouse and human sperm must hyperpolarize (become more negative inside). Determining the molecular mechanisms underlying this hyperpolarization is vital for developing new contraceptive methods and detecting causes of idiopathic male infertility. In mouse sperm, hyperpolarization is caused by activation of the sperm-specific potassium (K+) channel SLO3 [C. M. Santi et al., FEBS Lett. 584, 1041-1046 (2010)]. In human sperm, it has long been unclear whether hyperpolarization depends on SLO3 or the ubiquitous K+ channel SLO1 [N. Mannowetz, N. M. Naidoo, S. A. S. Choo, J. F. Smith, P. V. Lishko, Elife 2, e01009 (2013), C. Brenker et al., Elife 3, e01438 (2014), and S. A. Mansell, S. J. Publicover, C. L. R. Barratt, S. M. Wilson, Mol. Hum. Reprod. 20, 392-408 (2014)]. In this work, we identified the first selective inhibitor for human SLO3-VU0546110-and showed that it completely blocked heterologous SLO3 currents and endogenous K+ currents in human sperm. This compound also prevented sperm from hyperpolarizing and undergoing hyperactivated motility and induced acrosome reaction, which are necessary to fertilize an egg. We conclude that SLO3 is the sole K+ channel responsible for hyperpolarization and significantly contributes to the fertilizing ability of human sperm. Moreover, SLO3 is a good candidate for contraceptive development, and mutation of this gene is a possible cause of idiopathic male infertility.


Subject(s)
Infertility, Male , Large-Conductance Calcium-Activated Potassium Channels , Humans , Male , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Membrane Potentials/physiology , Semen , Spermatozoa/physiology
5.
Mol Pharmacol ; 101(5): 357-370, 2022 05.
Article in English | MEDLINE | ID: mdl-35246480

ABSTRACT

Heteromeric Kir4.1/Kir5.1 (KCNJ10/KCNJ16) inward rectifier potassium (Kir) channels play key roles in the brain and kidney, but pharmacological tools for probing their physiology and therapeutic potential have not been developed. Here, we report the discovery, in a high-throughput screening of 80,475 compounds, of the moderately potent and selective inhibitor VU0493690, which we selected for characterization and chemical optimization. VU0493690 concentration-dependently inhibits Kir4.1/5.1 with an IC50 of 0.96 µM and exhibits at least 10-fold selectivity over Kir4.1 and ten other Kir channels. Multidimensional chemical optimization of VU0493690 led to the development of VU6036720, the most potent (IC50 = 0.24 µM) and selective (>40-fold over Kir4.1) Kir4.1/5.1 inhibitor reported to date. Cell-attached patch single-channel recordings revealed that VU6036720 inhibits Kir4.1/5.1 activity through a reduction of channel open-state probability and single-channel current amplitude. Elevating extracellular potassium ion by 20 mM shifted the IC50 6.8-fold, suggesting that VU6036720 is a pore blocker that binds in the ion-conduction pathway. Mutation of the "rectification controller" asparagine 161 to glutamate (N161E), which is equivalent to small-molecule binding sites in other Kir channels, led to a strong reduction of inhibition by VU6036720. Renal clearance studies in mice failed to show a diuretic response that would be consistent with inhibition of Kir4.1/5.1 in the renal tubule. Drug metabolism and pharmacokinetics profiling revealed that high VU6036720 clearance and plasma protein binding may prevent target engagement in vivo. In conclusion, VU6036720 represents the current state-of-the-art Kir4.1/5.1 inhibitor that should be useful for probing the functions of Kir4.1/5.1 in vitro and ex vivo. SIGNIFICANCE STATEMENT: Heteromeric inward rectifier potassium (Kir) channels comprising Kir4.1 and Kir5.1 subunits play important roles in renal and neural physiology and may represent inhibitory drug targets for hypertension and edema. Herein, we employ high-throughput compound library screening, patch clamp electrophysiology, and medicinal chemistry to develop and characterize the first potent and specific in vitro inhibitor of Kir4.1/5.1, VU6036720, which provides proof-of-concept that drug-like inhibitors of this channel may be developed.


Subject(s)
Potassium Channels, Inwardly Rectifying , Animals , Gene Library , High-Throughput Screening Assays , Mice , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/metabolism
6.
Lett Drug Des Discov ; 16(12): 1387-1394, 2019 Dec.
Article in English | MEDLINE | ID: mdl-32201485

ABSTRACT

BACKGROUND: This article describes the challenges in the discovery and optimization of mGlu2/4 heterodimer Positive Allosteric Modulators (PAMs). METHODS: Initial forays based on VU0155041, a PAM of both the mGlu4 homodimer and the mGlu2/4 heterodimer, led to flat, intractable SAR that precluded advancement. Screening of a collection of 1,152 FDA approved drugs led to the discovery that febuxostat, an approved xanthine oxidase inhibitor, was a moderately potent PAM of the mGlu2/4 heterodimer (EC50 = 3.4 µM), but was peripherally restricted (rat Kp = 0.03). Optimization of this hit led to PAMs with improved potency (EC50s <800 nM) and improved CNS penetration (rat Kp >2, an ~100-fold increase). RESULTS: However, these new amide analogs of febuxostat proved to be either GIRK1/2 and GIRK1/4 activators (primary carboxamide congeners) or mGlu2 PAMs (secondary and tertiary amides) and not selective mGlu2/4 heterodimer PAMs. CONCLUSION: These results required the team to develop a new screening cascade paradigm, and exemplified the challenges in developing allosteric ligands for heterodimeric receptors.

7.
Mol Pharmacol ; 94(2): 926-937, 2018 08.
Article in English | MEDLINE | ID: mdl-29895592

ABSTRACT

The inward rectifier potassium (Kir) channel Kir4.1 (KCNJ10) carries out important physiologic roles in epithelial cells of the kidney, astrocytes in the central nervous system, and stria vascularis of the inner ear. Loss-of-function mutations in KCNJ10 lead to EAST/SeSAME syndrome, which is characterized by epilepsy, ataxia, renal salt wasting, and sensorineural deafness. Although genetic approaches have been indispensable for establishing the importance of Kir4.1 in the normal function of these tissues, the availability of pharmacological tools for acutely manipulating the activity of Kir4.1 in genetically normal animals has been lacking. We therefore carried out a high-throughput screen of 76,575 compounds from the Vanderbilt Institute of Chemical Biology library for small-molecule modulators of Kir4.1. The most potent inhibitor identified was 2-(2-bromo-4-isopropylphenoxy)-N-(2,2,6,6-tetramethylpiperidin-4-yl)acetamide (VU0134992). In whole-cell patch-clamp electrophysiology experiments, VU0134992 inhibits Kir4.1 with an IC50 value of 0.97 µM and is 9-fold selective for homomeric Kir4.1 over Kir4.1/5.1 concatemeric channels (IC50 = 9 µM) at -120 mV. In thallium (Tl+) flux assays, VU0134992 is greater than 30-fold selective for Kir4.1 over Kir1.1, Kir2.1, and Kir2.2; is weakly active toward Kir2.3, Kir6.2/SUR1, and Kir7.1; and is equally active toward Kir3.1/3.2, Kir3.1/3.4, and Kir4.2. This potency and selectivity profile is superior to Kir4.1 inhibitors amitriptyline, nortriptyline, and fluoxetine. Medicinal chemistry identified components of VU0134992 that are critical for inhibiting Kir4.1. Patch-clamp electrophysiology, molecular modeling, and site-directed mutagenesis identified pore-lining glutamate 158 and isoleucine 159 as critical residues for block of the channel. VU0134992 displayed a large free unbound fraction (fu) in rat plasma (fu = 0.213). Consistent with the known role of Kir4.1 in renal function, oral dosing of VU0134992 led to a dose-dependent diuresis, natriuresis, and kaliuresis in rats. Thus, VU0134992 represents the first in vivo active tool compound for probing the therapeutic potential of Kir4.1 as a novel diuretic target for the treatment of hypertension.


Subject(s)
Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/chemistry , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology , Animals , Binding Sites , Diuretics/chemistry , Electrolytes , HEK293 Cells , Humans , Male , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Mutagenesis, Site-Directed , Potassium Channels, Inwardly Rectifying/genetics , Rats , Small Molecule Libraries/chemistry , Substrate Specificity
8.
PLoS One ; 12(3): e0174742, 2017.
Article in English | MEDLINE | ID: mdl-28358902

ABSTRACT

AMPA receptors (AMPAR) are ligand gated ion channels critical for synaptic transmission and plasticity. Their dysfunction is implicated in a variety of psychiatric and neurological diseases ranging from major depressive disorder to amyotrophic lateral sclerosis. Attempting to potentiate or depress AMPAR activity is an inherently difficult balancing act between effective treatments and debilitating side effects. A newly explored strategy to target subsets of AMPARs in the central nervous system is to identify compounds that affect specific AMPAR-auxiliary subunit complexes. This exploits diverse spatio-temporal expression patterns of known AMPAR auxiliary subunits, providing means for designing brain region-selective compounds. Here we report a high-throughput screening-based pipeline that can identify compounds that are selective for GluA2-CNIH3 and GluA2-stargazin complexes. These compounds will help us build upon the growing library of AMPAR-auxiliary subunit specific inhibitors, which have thus far all been targeted to TARP γ-8. We used a cell-based assay combined with a voltage-sensitive dye (VSD) to identify changes in glutamate-gated cation flow across the membranes of HEK cells co-expressing GluA2 and an auxiliary subunit. We then used a calcium flux assay to further validate hits picked from the VSD assay. VU0612951 and VU0627849 are candidate compounds from the initial screen that were identified as negative and positive allosteric modulators (NAM and PAM), respectively. They both have lower IC50/EC50s on complexes containing stargazin and CNIH3 than GSG1L or the AMPAR alone. We have also identified a candidate compound, VU0539491, that has NAM activity in GluA2(R)-CNIH3 and GluA2(Q) complexes and PAM activity in GluA2(Q)-GSG1L complexes.


Subject(s)
Receptors, AMPA/metabolism , Biological Transport , Calcium/metabolism , Calcium Channels/metabolism , Electrophysiology , HEK293 Cells , Humans
10.
ACS Chem Neurosci ; 8(3): 558-568, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27805811

ABSTRACT

The two-pore-domain potassium (K2P) channel TREK-2 serves to modulate plasma membrane potential in dorsal root ganglia c-fiber nociceptors, which tunes electrical excitability and nociception. Thus, TREK-2 channels are considered a potential therapeutic target for treating pain; however, there are currently no selective pharmacological tools for TREK-2 channels. Here we report the identification of the first TREK-2 selective activators using a high-throughput fluorescence-based thallium (Tl+) flux screen (HTS). An initial pilot screen with a bioactive lipid library identified 11-deoxy prostaglandin F2α as a potent activator of TREK-2 channels (EC50 ≈ 0.294 µM), which was utilized to optimize the TREK-2 Tl+ flux assay (Z' = 0.752). A HTS was then performed with 76 575 structurally diverse small molecules. Many small molecules that selectively activate TREK-2 were discovered. As these molecules were able to activate single TREK-2 channels in excised membrane patches, they are likely direct TREK-2 activators. Furthermore, TREK-2 activators reduced primary dorsal root ganglion (DRG) c-fiber Ca2+ influx. Interestingly, some of the selective TREK-2 activators such as 11-deoxy prostaglandin F2α were found to inhibit the K2P channel TREK-1. Utilizing chimeric channels containing portions of TREK-1 and TREK-2, the region of the TREK channels that allows for either small molecule activation or inhibition was identified. This region lies within the second pore domain containing extracellular loop and is predicted to play an important role in modulating TREK channel activity. Moreover, the selective TREK-2 activators identified in this HTS provide important tools for assessing human TREK-2 channel function and investigating their therapeutic potential for treating chronic pain.


Subject(s)
Action Potentials/physiology , Calcium/metabolism , Ganglia, Spinal/cytology , Nociceptors/drug effects , Nociceptors/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Action Potentials/drug effects , Animals , Antibodies/pharmacology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Electric Stimulation , Fluoxetine/pharmacology , HEK293 Cells , Humans , Lectins/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/immunology , Protein Synthesis Inhibitors/pharmacology , Tetracycline/pharmacology
11.
Sci Rep ; 6: 36954, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27849039

ABSTRACT

Insecticide resistance is a growing threat to mosquito control programs around the world, thus creating the need to discover novel target sites and target-specific compounds for insecticide development. Emerging evidence suggests that mosquito inward rectifier potassium (Kir) channels represent viable molecular targets for developing insecticides with new mechanisms of action. Here we describe the discovery and characterization of VU041, a submicromolar-affinity inhibitor of Anopheles (An.) gambiae and Aedes (Ae.) aegypti Kir1 channels that incapacitates adult female mosquitoes from representative insecticide-susceptible and -resistant strains of An. gambiae (G3 and Akron, respectively) and Ae. aegypti (Liverpool and Puerto Rico, respectively) following topical application. VU041 is selective for mosquito Kir channels over several mammalian orthologs, with the exception of Kir2.1, and is not lethal to honey bees. Medicinal chemistry was used to develop an analog, termed VU730, which retains activity toward mosquito Kir1 but is not active against Kir2.1 or other mammalian Kir channels. Thus, VU041 and VU730 are promising chemical scaffolds for developing new classes of insecticides to combat insecticide-resistant mosquitoes and the transmission of mosquito-borne diseases, such as Zika virus, without harmful effects on humans and beneficial insects.


Subject(s)
Aedes/drug effects , Anopheles/drug effects , Insecticide Resistance/drug effects , Insecticides/pharmacology , Mosquito Vectors/drug effects , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Bees/drug effects
12.
ACS Chem Neurosci ; 6(6): 871-8, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-25879403

ABSTRACT

Ion channels are critical for life, and they are targets of numerous drugs. The sequencing of the human genome has revealed the existence of hundreds of different ion channel subunits capable of forming thousands of ion channels. In the face of this diversity, we only have a few selective small-molecule tools to aid in our understanding of the role specific ion channels in physiology which may in turn help illuminate their therapeutic potential. Although the advent of automated electrophysiology has increased the rate at which we can screen for and characterize ion channel modulators, the technique's high per-measurement cost and moderate throughput compared to other high-throughput screening approaches limit its utility for large-scale high-throughput screening. Therefore, lower cost, more rapid techniques are needed. While ion channel types capable of fluxing calcium are well-served by low cost, very high-throughput fluorescence-based assays, other channel types such as sodium channels remain underserved by present functional assay techniques. In order to address this shortcoming, we have developed a thallium flux-based assay for sodium channels using the NaV1.7 channel as a model target. We show that the assay is able to rapidly and cost-effectively identify NaV1.7 inhibitors thus providing a new method useful for the discovery and profiling of sodium channel modulators.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sodium Channel Blockers/pharmacology , Thallium/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Patch-Clamp Techniques , Sodium Channel Blockers/chemistry , Veratridine/pharmacology
13.
PLoS One ; 9(11): e110772, 2014.
Article in English | MEDLINE | ID: mdl-25375326

ABSTRACT

Vector-borne diseases such as dengue fever and malaria, which are transmitted by infected female mosquitoes, affect nearly half of the world's population. The emergence of insecticide-resistant mosquito populations is reducing the effectiveness of conventional insecticides and threatening current vector control strategies, which has created an urgent need to identify new molecular targets against which novel classes of insecticides can be developed. We previously demonstrated that small molecule inhibitors of mammalian Kir channels represent promising chemicals for new mosquitocide development. In this study, high-throughput screening of approximately 30,000 chemically diverse small-molecules was employed to discover potent and selective inhibitors of Aedes aegypti Kir1 (AeKir1) channels heterologously expressed in HEK293 cells. Of 283 confirmed screening 'hits', the small-molecule inhibitor VU625 was selected for lead optimization and in vivo studies based on its potency and selectivity toward AeKir1, and tractability for medicinal chemistry. In patch clamp electrophysiology experiments of HEK293 cells, VU625 inhibits AeKir1 with an IC50 value of 96.8 nM, making VU625 the most potent inhibitor of AeKir1 described to date. Furthermore, electrophysiology experiments in Xenopus oocytes revealed that VU625 is a weak inhibitor of AeKir2B. Surprisingly, injection of VU625 failed to elicit significant effects on mosquito behavior, urine excretion, or survival. However, when co-injected with probenecid, VU625 inhibited the excretory capacity of mosquitoes and was toxic, suggesting that the compound is a substrate of organic anion and/or ATP-binding cassette (ABC) transporters. The dose-toxicity relationship of VU625 (when co-injected with probenecid) is biphasic, which is consistent with the molecule inhibiting both AeKir1 and AeKir2B with different potencies. This study demonstrates proof-of-concept that potent and highly selective inhibitors of mosquito Kir channels can be developed using conventional drug discovery approaches. Furthermore, it reinforces the notion that the physical and chemical properties that determine a compound's bioavailability in vivo will be critical in determining the efficacy of Kir channel inhibitors as insecticides.


Subject(s)
Aedes/drug effects , Insecticides/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Drug Interactions , HEK293 Cells , Humans , Oocytes/drug effects , Probenecid/pharmacology , Xenopus
14.
J Med Chem ; 57(23): 10192-7, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25423411

ABSTRACT

A duplexed, functional multiaddition high throughput screen and subsequent iterative parallel synthesis effort identified the first highly selective and CNS penetrant glucagon-like peptide-1R (GLP-1R) positive allosteric modulator (PAM). PAM (S)-9b potentiated low-dose exenatide to augment insulin secretion in primary mouse pancreatic islets, and (S)-9b alone was effective in potentiating endogenous GLP-1R to reverse haloperidol-induced catalepsy.


Subject(s)
Indoles/chemical synthesis , Pyrrolidines/chemical synthesis , Receptors, Glucagon/drug effects , Allosteric Regulation/drug effects , Animals , Catalepsy/chemically induced , Catalepsy/drug therapy , Central Nervous System Agents/therapeutic use , Drug Synergism , Exenatide , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Haloperidol , High-Throughput Screening Assays , Indoles/metabolism , Indoles/pharmacokinetics , Indoles/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Male , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Peptides/pharmacology , Pyrrolidines/metabolism , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Structure-Activity Relationship , Venoms/pharmacology
15.
ACS Chem Neurosci ; 5(12): 1221-37, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25225882

ABSTRACT

Metabotropic glutamate receptor 7 (mGlu7) is a member of the group III mGlu receptors (mGlus), encompassed by mGlu4, mGlu6, mGlu7, and mGlu8. mGlu7 is highly expressed in the presynaptic active zones of both excitatory and inhibitory synapses, and activation of the receptor regulates the release of both glutamate and GABA. mGlu7 is thought to be a relevant therapeutic target for a number of neurological and psychiatric disorders, and polymorphisms in the GRM7 gene have been linked to autism, depression, ADHD, and schizophrenia. Here we report two new pan-group III mGlu positive allosteric modulators, VU0155094 and VU0422288, which show differential activity at the various group III mGlus. Additionally, both compounds show probe dependence when assessed in the presence of distinct orthosteric agonists. By pairing studies of these nonselective compounds with a synapse in the hippocampus that expresses only mGlu7, we have validated activity of these compounds in a native tissue setting. These studies provide proof-of-concept evidence that mGlu7 activity can be modulated by positive allosteric modulation, paving the way for future therapeutics development.


Subject(s)
Excitatory Amino Acid Agents/chemistry , Excitatory Amino Acid Agents/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Acetanilides/chemistry , Acetanilides/pharmacology , Animals , Benzoates/pharmacology , CHO Cells , Calcium/metabolism , Cricetulus , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Glutamic Acid/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , In Vitro Techniques , Male , Mice, Inbred C57BL , Picolinic Acids/chemistry , Picolinic Acids/pharmacology , Propionates/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Rats , Receptors, Metabotropic Glutamate/genetics , Structure-Activity Relationship , Thallium/metabolism , Transfection
16.
J Biomol Screen ; 19(6): 847-58, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24525870

ABSTRACT

Injectable, degradation-resistant peptide agonists for the glucagon-like peptide 1 (GLP-1) receptor (GLP-1R), such as exenatide and liraglutide, activate the GLP-1R via a complex orthosteric-binding site and are effective therapeutics for glycemic control in type 2 diabetes. Orally bioavailable orthosteric small-molecule agonists are unlikely to be developed, whereas positive allosteric modulators (PAMs) may offer an improved therapeutic profile. We hypothesize that allosteric modulators of the GLP-1R would increase the potency and efficacy of native GLP-1 in a spatial and temporally preserved manner and/or may improve efficacy or side effects of injectable analogs. We report the design, optimization, and initial results of a duplexed high-throughput screen in which cell lines overexpressing either the GLP-1R or the glucagon receptor were coplated, loaded with a calcium-sensitive dye, and probed in a three-phase assay to identify agonists, antagonists, and potentiators of GLP-1, and potentiators of glucagon. 175,000 compounds were initially screened, and progression through secondary assays yielded 98 compounds with a variety of activities at the GLP-1R. Here, we describe five compounds possessing different patterns of modulation of the GLP-1R. These data uncover PAMs that may offer a drug-development pathway to enhancing in vivo efficacy of both endogenous GLP-1 and peptide analogs.


Subject(s)
Glucagon-Like Peptide 1/chemistry , High-Throughput Screening Assays/methods , Receptors, Glucagon/chemistry , Allosteric Regulation/drug effects , Allosteric Site , Animals , Binding Sites , CHO Cells , Calcium/chemistry , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Cyclic AMP/chemistry , Disease Progression , Exenatide , Glucose/chemistry , Humans , Liraglutide/chemistry , Peptides/chemistry , Recombinant Proteins/chemistry , Signal Transduction , Venoms/chemistry
17.
Assay Drug Dev Technol ; 11(9-10): 532-43, 2013.
Article in English | MEDLINE | ID: mdl-24266659

ABSTRACT

The inward rectifier potassium (Kir) channel Kir4.1 plays essential roles in modulation of neurotransmission and renal sodium transport and may represent a novel drug target for temporal lobe epilepsy and hypertension. The molecular pharmacology of Kir4.1 is limited to neurological drugs, such as fluoxetine (Prozac(©)), exhibiting weak and nonspecific activity toward the channel. The development of potent and selective small-molecule probes would provide critically needed tools for exploring the integrative physiology and therapeutic potential of Kir4.1. A fluorescence-based thallium (Tl(+)) flux assay that utilizes a tetracycline-inducible T-Rex-HEK293-Kir4.1 cell line to enable high-throughput screening (HTS) of small-molecule libraries was developed. The assay is dimethyl sulfoxide tolerant and exhibits robust screening statistics (Z'=0.75±0.06). A pilot screen of 3,655 small molecules and lipids revealed 16 Kir4.1 inhibitors (0.4% hit rate). 3,3-Diphenyl-N-(1-phenylethyl)propan-1-amine, termed VU717, inhibits Kir4.1-mediated thallium flux with an IC50 of ∼6 µM. An automated patch clamp assay using the IonFlux HT workbench was developed to facilitate compound characterization. Leak-subtracted ensemble "loose patch" recordings revealed robust tetracycline-inducible and Kir4.1 currents that were inhibited by fluoxetine (IC50=10 µM), VU717 (IC50=6 µM), and structurally related calcium channel blocker prenylamine (IC50=6 µM). Finally, we demonstrate that VU717 inhibits Kir4.1 channel activity in cultured rat astrocytes, providing proof-of-concept that the Tl(+) flux and IonFlux HT assays can enable the discovery of antagonists that are active against native Kir4.1 channels.


Subject(s)
Astrocytes/physiology , Flow Cytometry/methods , Patch-Clamp Techniques/methods , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/physiology , Spectrometry, Fluorescence/methods , Animals , Astrocytes/drug effects , Cells, Cultured , Drug Evaluation, Preclinical/methods , Flow Injection Analysis/methods , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
18.
J Med Chem ; 56(18): 7243-59, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23947773

ABSTRACT

Starting from a singleton chromanone high throughput screening (HTS) hit, we describe a focused medicinal chemistry optimization effort leading to the identification of a novel series of phenoxymethyl-dihydrothiazolopyridone derivatives as selective positive allosteric modulators (PAMs) of the metabotropic glutamate 5 (mGlu5) receptor. These dihydrothiazolopyridones potentiate receptor responses in recombinant systems. In vitro and in vivo drug metabolism and pharmacokinetic (DMPK) evaluation allowed us to select compound 16a for its assessment in a preclinical animal screen of possible antipsychotic activity. 16a was able to reverse amphetamine-induced hyperlocomotion in rats in a dose-dependent manner without showing any significant motor impairment or overt neurological side effects at comparable doses. Evolution of our medicinal chemistry program, structure activity, and properties relationships (SAR and SPR) analysis as well as a detailed profile for optimized mGlu5 receptor PAM 16a are described.


Subject(s)
Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacology , Receptor, Metabotropic Glutamate 5/chemistry , Thiazoles/chemistry , Thiazoles/pharmacology , Allosteric Regulation/drug effects , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Humans , Locomotion/drug effects , Male , Rats , Receptor, Metabotropic Glutamate 5/metabolism , Structure-Activity Relationship
19.
ACS Chem Neurosci ; 4(9): 1278-86, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-23730969

ABSTRACT

The G-protein activated, inward-rectifying potassium (K(+)) channels, "GIRKs", are a family of ion channels (Kir3.1-Kir3.4) that has been the focus of intense research interest for nearly two decades. GIRKs are comprised of various homo- and heterotetrameric combinations of four different subunits. These subunits are expressed in different combinations in a variety of regions throughout the central nervous system and in the periphery. The body of GIRK research implicates GIRK in processes as diverse as controlling heart rhythm, to effects on reward/addiction, to modulation of response to analgesics. Despite years of GIRK research, very few tools exist to selectively modulate GIRK channels' activity and until now no tools existed that potently and selectively activated GIRKs. Here we report the development and characterization of the first truly potent, effective, and selective GIRK activator, ML297 (VU0456810). We further demonstrate that ML297 is active in two in vivo models of epilepsy, a disease where up to 40% of patients remain with symptoms refractory to present treatments. The development of ML297 represents a truly significant advancement in our ability to selectively probe GIRK's role in physiology as well as providing the first tool for beginning to understand GIRK's potential as a target for a diversity of therapeutic indications.


Subject(s)
Anticonvulsants/therapeutic use , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , Phenylurea Compounds/therapeutic use , Pyrazoles/therapeutic use , Seizures/drug therapy , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electroshock/adverse effects , HEK293 Cells , High-Throughput Screening Assays , Humans , Injections, Intraperitoneal , Mice , Microsomes, Liver/metabolism , Molecular Structure , Patch-Clamp Techniques , Pentylenetetrazole/toxicity , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Pyrazoles/pharmacology , Rats , Receptors, Metabotropic Glutamate/drug effects , Recombinant Proteins/drug effects , Seizures/etiology , Valproic Acid/therapeutic use
20.
J Biol Chem ; 287(47): 39613-25, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23033485

ABSTRACT

The human Ether-à-go-go-related gene (hERG)-encoded K(+) current, I(Kr) is essential for cardiac repolarization but is also a source of cardiotoxicity because unintended hERG inhibition by diverse pharmaceuticals can cause arrhythmias and sudden cardiac death. We hypothesized that a small molecule that diminishes I(Kr) block by a known hERG antagonist would constitute a first step toward preventing hERG-related arrhythmias and facilitating drug discovery. Using a high-throughput assay, we screened a library of compounds for agents that increase the IC(70) of dofetilide, a well characterized hERG blocker. One compound, VU0405601, with the desired activity was further characterized. In isolated, Langendorff-perfused rabbit hearts, optical mapping revealed that dofetilide-induced arrhythmias were reduced after pretreatment with VU0405601. Patch clamp analysis in stable hERG-HEK cells showed effects on current amplitude, inactivation, and deactivation. VU0405601 increased the IC(50) of dofetilide from 38.7 to 76.3 nM. VU0405601 mitigates the effects of hERG blockers from the extracellular aspect primarily by reducing inactivation, whereas most clinically relevant hERG inhibitors act at an inner pore site. Structure-activity relationships surrounding VU0405601 identified a 3-pyridiyl and a naphthyridine ring system as key structural components important for preventing hERG inhibition by multiple inhibitors. These findings indicate that small molecules can be designed to reduce the sensitivity of hERG to inhibitors.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/drug therapy , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Naphthyridines/chemistry , Naphthyridines/pharmacology , Phenethylamines/adverse effects , Potassium Channel Blockers/adverse effects , Pyridines/chemistry , Pyridines/pharmacology , Sulfonamides/adverse effects , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Dose-Response Relationship, Drug , Drug Discovery , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , HEK293 Cells , Humans , Muscle Proteins/genetics , Myocardium/metabolism , Myocardium/pathology , Phenethylamines/pharmacology , Potassium Channel Blockers/pharmacology , Rabbits , Structure-Activity Relationship , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...