Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters








Publication year range
1.
Molecules ; 29(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38893312

ABSTRACT

Gain-of-function mutations in the KCNT1 gene, which encodes the sodium-activated potassium channel known as SLACK, are associated with the rare but devastating developmental and epileptic encephalopathy known as epilepsy of infancy with migrating focal seizures (EIMFS). The design of small molecule inhibitors of SLACK channels represents a potential therapeutic approach to the treatment of EIMFS, other childhood epilepsies, and developmental disorders. Herein, we describe a hit optimization effort centered on a xanthine SLACK inhibitor (8) discovered via a high-throughput screen. Across three distinct regions of the chemotype, we synthesized 58 new analogs and tested each one in a whole-cell automated patch-clamp assay to develop structure-activity relationships for inhibition of SLACK channels. We further evaluated selected analogs for their selectivity versus a variety of other ion channels and for their activity versus clinically relevant SLACK mutants. Selectivity within the series was quite good, including versus hERG. Analog 80 (VU0948578) was a potent inhibitor of WT, A934T, and G288S SLACK, with IC50 values between 0.59 and 0.71 µM across these variants. VU0948578 represents a useful in vitro tool compound from a chemotype that is distinct from previously reported small molecule inhibitors of SLACK channels.


Subject(s)
Potassium Channel Blockers , Structure-Activity Relationship , Humans , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels, Sodium-Activated , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Xanthine/chemistry , Xanthine/pharmacology , Patch-Clamp Techniques , HEK293 Cells , Molecular Structure , Xanthines/chemistry , Xanthines/pharmacology
2.
Bioorg Med Chem ; 95: 117487, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37812884

ABSTRACT

Malignant migrating partial seizure of infancy (MMPSI) is a devastating and pharmacoresistant form of infantile epilepsy. MMPSI has been linked to multiple gain-of-function (GOF) mutations in the KCNT1 gene, which encodes for a potassium channel often referred to as SLACK. SLACK channels are sodium-activated potassium channels distributed throughout the central nervous system (CNS) and the periphery. The investigation described here aims to discover SLACK channel inhibitor tool compounds and profile their pharmacokinetic and pharmacodynamic properties. A SLACK channel inhibitor VU0531245 (VU245) was identified via a high-throughput screen (HTS) campaign. Structure-activity relationship (SAR) studies were conducted in five distinct regions of the hit VU245. VU245 analogs were evaluated for their ability to affect SLACK channel activity using a thallium flux assay in HEK-293 cells stably expressing wild-type (WT) human SLACK. Selected analogs were tested for metabolic stability in mouse liver microsomes and plasma-protein binding in mouse plasma. The same set of analogs was tested via thallium flux for activity versus human A934T SLACK and other structurally related potassium channels, including SLICK and Maxi-K. In addition, potencies for selected VU245 analogs were obtained using whole-cell electrophysiology (EP) assays in CHO cells stably expressing WT human SLACK through an automated patch clamp system. Results revealed that this scaffold tolerates structural changes in some regions, with some analogs demonstrating improved SLACK inhibitory activity, good selectivity against the other channels tested, and modest improvements in metabolic clearance. Analog VU0935685 represents a new, structurally distinct small-molecule inhibitor of SLACK channels that can serve as an in vitro tool for studying this target.


Subject(s)
Potassium Channels , Thallium , Animals , Cricetinae , Humans , Mice , Cricetulus , HEK293 Cells , Nerve Tissue Proteins/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels, Sodium-Activated/genetics , Potassium Channels, Sodium-Activated/metabolism , Seizures , Thallium/metabolism , Oxadiazoles/chemistry , Oxadiazoles/metabolism
3.
Bioorg Med Chem Lett ; 76: 129013, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36184030

ABSTRACT

In this Letter we describe structure-activity relationship (SAR) studies conducted in five distinct regions of a new 2-amino-N-phenylacetamides series of Slack potassium channel inhibitors exemplified by recently disclosed high-throughput screening (HTS) hit VU0606170 (4). New analogs were screened in a thallium (Tl+) flux assay in HEK-293 cells stably expressing wild-type human (WT) Slack. Selected analogs were screened in Tl+ flux versus A934T Slack and other Slo family members Slick and Maxi-K and evaluated in whole-cell electrophysiology (EP) assays using an automated patch clamp system. Results revealed the series to have flat SAR with significant structural modifications resulting in a loss of Slack activity. More minor changes led to compounds with Slack activity and Slo family selectivity similar to the HTS hit.


Subject(s)
Potassium Channels , Thallium , Humans , HEK293 Cells , Nerve Tissue Proteins/metabolism , Potassium Channels, Sodium-Activated , Structure-Activity Relationship
4.
Pharm Pat Anal ; 11(2): 45-56, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35369761

ABSTRACT

Slack channels are sodium-activated potassium channels that are encoded by the KCNT1 gene. Several KCNT1 gain of function mutations have been linked to malignant migrating partial seizures of infancy. Quinidine is an anti-arrhythmic drug that functions as a moderately potent inhibitor of Slack channels; however, quinidine use is limited by its poor selectivity, safety and pharmacokinetic profile. Slack channels represent an interesting target for developing novel therapeutics for the treatment of malignant migrating partial seizures of infancy and other childhood epilepsies; thus, ongoing efforts are directed toward the discovery of small-molecules that inhibit Slack currents. This review summarizes patent applications published in 2020-2021 that describe the discovery of novel small-molecule Slack inhibitors.


Subject(s)
Epilepsy , Nerve Tissue Proteins , Potassium Channel Blockers , Potassium Channels, Sodium-Activated , Child , Epilepsy/drug therapy , Humans , Mutation , Nerve Tissue Proteins/antagonists & inhibitors , Potassium Channel Blockers/therapeutic use , Potassium Channels, Sodium-Activated/antagonists & inhibitors , Quinidine/therapeutic use , Seizures/drug therapy
5.
ACS Chem Neurosci ; 11(21): 3658-3671, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33143429

ABSTRACT

Malignant migrating partial seizures of infancy is a rare, devastating form of epilepsy most commonly associated with gain-of-function mutations in the potassium channel, Slack. Not only is this condition almost completely pharmacoresistant, there are not even selective drug-like tools available to evaluate whether inhibition of these overactivated, mutant Slack channels may represent a viable path forward toward new antiepileptic therapies. Therefore, we used a high-throughput thallium flux assay to screen a drug-like, 100 000-compound library in search of inhibitors of both wild-type and a disease-associated mutant Slack channel. Using this approach, we discovered VU0606170, a selective Slack channel inhibitor with low micromolar potency. Critically, VU0606170 also proved effective at significantly decreasing the firing rate in overexcited, spontaneously firing cortical neuron cultures. Taken together, our data provide compelling evidence that selective inhibition of Slack channel activity can be achieved with small molecules and that inhibition of Slack channel activity in neurons produces efficacy consistent with an antiepileptic effect. Thus, the identification of VU0606170 provides a much-needed tool for advancing our understanding of the role of the Slack channel in normal physiology and disease as well as its potential as a target for therapeutic intervention.


Subject(s)
Calcium Signaling , Nerve Tissue Proteins , Potassium Channels, Sodium-Activated , Cells, Cultured , HEK293 Cells , Humans , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Potassium Channels, Sodium-Activated/antagonists & inhibitors , Potassium Channels, Sodium-Activated/metabolism
6.
J Neurosci ; 39(37): 7438-7449, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31350261

ABSTRACT

Mutations in the KCNT1 (Slack, KNa1.1) sodium-activated potassium channel produce severe epileptic encephalopathies. Expression in heterologous systems has shown that the disease-causing mutations give rise to channels that have increased current amplitude. It is not known, however, whether such gain of function occurs in human neurons, nor whether such increased KNa current is expected to suppress or increase the excitability of cortical neurons. Using genetically engineered human induced pluripotent stem cell (iPSC)-derived neurons, we have now found that sodium-dependent potassium currents are increased several-fold in neurons bearing a homozygous P924L mutation. In current-clamp recordings, the increased KNa current in neurons with the P924L mutation acts to shorten the duration of action potentials and to increase the amplitude of the afterhyperpolarization that follows each action potential. Strikingly, the number of action potentials that were evoked by depolarizing currents as well as maximal firing rates were increased in neurons expressing the mutant channel. In networks of spontaneously active neurons, the mean firing rate, the occurrence of rapid bursts of action potentials, and the intensity of firing during the burst were all increased in neurons with the P924L Slack mutation. The feasibility of an increased KNa current to increase firing rates independent of any compensatory changes was validated by numerical simulations. Our findings indicate that gain-of-function in Slack KNa channels causes hyperexcitability in both isolated neurons and in neural networks and occurs by a cell-autonomous mechanism that does not require network interactions.SIGNIFICANCE STATEMENTKCNT1 mutations lead to severe epileptic encephalopathies for which there are no effective treatments. This study is the first demonstration that a KCNT1 mutation increases the Slack current in neurons. It also provides the first explanation for how this increased potassium current induces hyperexcitability, which could be the underlining factor causing seizures.


Subject(s)
Epilepsy/genetics , Induced Pluripotent Stem Cells/physiology , Mutation/physiology , Nerve Tissue Proteins/genetics , Neurons/physiology , Potassium Channels, Sodium-Activated/genetics , Action Potentials/physiology , Cell Differentiation/physiology , Epilepsy/physiopathology , HEK293 Cells , Humans
7.
Brain Dev ; 40(3): 229-232, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29037447

ABSTRACT

Epilepsy of infancy with migrating focal seizures {a.k.a malignant migrating partial seizures of infancy (MMPSI)} is an uncommon epileptic encephalopathy with a poor prognosis. Migrating focal seizures with autonomic features, developmental stagnation and refractoriness to treatment are its key features. It is caused by genetic defects in various ion channels, most common being sodium activated potassium channel (KCNT1), found in up to 50% of cases. With advent of genetic diagnosis and precision medicine, many targeted therapies have been identified. Antagonist of KCNT1 coded ion channel like Quinidine has shown promising results in MMPSI. Here we report first mutation proven case of MMPSI from India. This child had a novel heterozygous missense mutation in exon10 of the KCNT1 gene (chr9:138650308; c.808C > C/G (p.Q270E)) which was pathogenic. Neither quinidine nor ketogenic diet could control his seizures. Ultimately, the child succumbed to his illness at nine months of age.


Subject(s)
Enzyme Inhibitors/therapeutic use , Mutation/genetics , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Quinidine/therapeutic use , Seizures , Spasms, Infantile , Electroencephalography , Humans , India , Infant , Male , Potassium Channels, Sodium-Activated , Seizures/complications , Seizures/drug therapy , Seizures/genetics , Spasms, Infantile/complications , Spasms, Infantile/drug therapy , Spasms, Infantile/genetics
8.
Mol Cell Neurosci ; 72: 54-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26784557

ABSTRACT

The KCNT1 gene encodes for subunits contributing to the Na(+)-activated K(+) current (KNa), expressed in many cell types. Mutations in KCNT1 have been found in patients affected with a wide spectrum of early-onset epilepsies, including Malignant Migrating Partial Seizures in Infancy (MMPSI), a severe early-onset epileptic encephalopathy characterized by pharmacoresistant focal seizures migrating from one brain region or hemisphere to another and neurodevelopment arrest or regression, resulting in profound disability. In the present study we report identification by whole exome sequencing (WES) of two de novo, heterozygous KCNT1 mutations (G288S and, not previously reported, M516V) in two unrelated MMPSI probands. Functional studies in a heterologous expression system revealed that channels formed by mutant KCNT1 subunits carried larger currents when compared to wild-type KCNT1 channels, both as homo- and heteromers with these last. Both mutations induced a marked leftward shift in homomeric channel activation gating. Interestingly, the KCNT1 blockers quinidine (3-1000µM) and bepridil (0.03-10µM) inhibited both wild-type and mutant KCNT1 currents in a concentration-dependent manner, with mutant channels showing higher sensitivity to blockade. This latter result suggests two genotype-tailored pharmacological strategies to specifically counteract the dysfunction of KCNT1 activating mutations in MMPSI patients.


Subject(s)
Mutation, Missense , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Spasms, Infantile/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Exome , Humans , Infant , Ion Channel Gating , Male , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Potassium Channels, Sodium-Activated , Spasms, Infantile/diagnosis
9.
Epilepsy Res ; 108(2): 340-4, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24315024

ABSTRACT

Sequencing of the KCNT1, PLCB1, SCN1A and TBC1D24 loci was performed in six children with typical features of malignant migrating partial seizures of infancy (MMPSI), to verify the presence of potential disease-causing mutations, including those already reported to be associated with the disease. Sanger sequencing failed to identify in these genes the previously reported pathogenic mutations in these patients, while a comprehensive mutational scanning analysis of these four loci by targeted re-sequencing led to detection of both intronic and exonic new variants. Based on the current knowledge, the sequence variants identified here do not allow to predict functional phenotypes that might explain, at least in part, MMPSI symptoms.


Subject(s)
Epilepsies, Partial/diagnosis , Epilepsies, Partial/genetics , Mutation/genetics , Carrier Proteins/genetics , Female , GTPase-Activating Proteins , Genetic Variation , Humans , Infant , Male , Membrane Proteins , NAV1.1 Voltage-Gated Sodium Channel/genetics , Nerve Tissue Proteins/genetics , Phenotype , Phospholipase C beta/genetics , Potassium Channels/genetics , Potassium Channels, Sodium-Activated , Sequence Deletion
10.
Gene ; 531(2): 467-71, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24029078

ABSTRACT

We performed analysis of KCNT1 in two unrelated patients with malignant migrating partial seizures in infancy. Both patients had intractable focal seizures since two months of age. Their seizures were characterized by a shift of epileptic focus during a single seizure and were resistant to most antiepileptic drugs but responded to vagus nerve stimulation in one and clorazepate in the other. Bidirectional sequencing for KCNT1 was analyzed by standard Sanger sequencing method. A de novo c.862G>A (p.Gly288Ser) missense mutation was identified at the pore region of KCNT1 channel in both patients, whereas all KCNT1 mutations in the previous reports were identified mostly in the intracellular C-terminal region. Computational analysis suggested possible changes in the molecular structure and the ion channel property induced by the Gly288Ser mutation. Because the G-to-A transition was located at CG dinucleotide sequences as previously reported for KCNT1 mutations, the recurrent occurrence of de novo KCNT1 mutations indicated the hot spots of these locations.


Subject(s)
Epilepsies, Partial/genetics , Mutation, Missense , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Age of Onset , Child , Epilepsies, Partial/complications , Epilepsies, Partial/epidemiology , Female , Humans , Infant , Models, Biological , Mutation, Missense/physiology , Pedigree , Potassium Channels, Sodium-Activated , Psychomotor Disorders/complications , Psychomotor Disorders/genetics
SELECTION OF CITATIONS
SEARCH DETAIL