KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons

KCNT1 encodes the sodium-activated potassium channel KNa1.1, expressed preferentially in the frontal cortex, hippocampus, cerebellum, and brainstem. Pathogenic missense variants in KCNT1 are associated with intractable epilepsy, namely epilepsy of infancy with migrating focal seizures (EIMFS), and s...

Full description

Bibliographic Details
Main Authors: Tracy S. Gertler, Suraj Cherian, Jean-Marc DeKeyser, Jennifer A. Kearney, Alfred L. George, Jr
Format: Article
Language:English
Published: Elsevier 2022-06-01
Series:Neurobiology of Disease
Subjects:
Online Access:http://www.sciencedirect.com/science/article/pii/S096999612200105X
_version_ 1811302264625692672
author Tracy S. Gertler
Suraj Cherian
Jean-Marc DeKeyser
Jennifer A. Kearney
Alfred L. George, Jr
author_facet Tracy S. Gertler
Suraj Cherian
Jean-Marc DeKeyser
Jennifer A. Kearney
Alfred L. George, Jr
author_sort Tracy S. Gertler
collection DOAJ
description KCNT1 encodes the sodium-activated potassium channel KNa1.1, expressed preferentially in the frontal cortex, hippocampus, cerebellum, and brainstem. Pathogenic missense variants in KCNT1 are associated with intractable epilepsy, namely epilepsy of infancy with migrating focal seizures (EIMFS), and sleep-related hypermotor epilepsy (SHE). In vitro studies of pathogenic KCNT1 variants support predominantly a gain-of-function molecular mechanism, but how these variants behave in a neuron or ultimately drive formation of an epileptogenic circuit is an important and timely question. Using CRISPR/Cas9 gene editing, we introduced a gain-of-function variant into the endogenous mouse Kcnt1 gene. Compared to wild-type (WT) littermates, heterozygous and homozygous knock-in mice displayed greater seizure susceptibility to the chemoconvulsants kainate and pentylenetetrazole (PTZ), but not to flurothyl. Using acute slice electrophysiology in heterozygous and homozygous Kcnt1 knock-in and WT littermates, we demonstrated that CA1 hippocampal pyramidal neurons exhibit greater amplitude of miniature inhibitory postsynaptic currents in mutant mice with no difference in frequency, suggesting greater inhibitory tone associated with the Kcnt1 mutation. To address alterations in GABAergic signaling, we bred Kcnt1 knock-in mice to a parvalbumin-tdTomato reporter line, and found that parvalbumin-expressing (PV+) interneurons failed to fire repetitively with large amplitude current injections and were more prone to depolarization block. These alterations in firing can be recapitulated by direct application of the KNa1.1 channel activator loxapine in WT but are occluded in knock-in littermates, supporting a direct channel gain-of-function mechanism. Taken together, these results suggest that KNa1.1 gain-of-function dampens interneuron excitability to a greater extent than it impacts pyramidal neuron excitability, driving seizure susceptibility in a mouse model of KCNT1-associated epilepsy.
first_indexed 2024-04-13T07:24:58Z
format Article
id doaj.art-a281ef78d17b40329578f8004652cca5
institution Directory Open Access Journal
issn 1095-953X
language English
last_indexed 2024-04-13T07:24:58Z
publishDate 2022-06-01
publisher Elsevier
record_format Article
series Neurobiology of Disease
spelling doaj.art-a281ef78d17b40329578f8004652cca52022-12-22T02:56:31ZengElsevierNeurobiology of Disease1095-953X2022-06-01168105713KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneuronsTracy S. Gertler0Suraj Cherian1Jean-Marc DeKeyser2Jennifer A. Kearney3Alfred L. George, Jr4Division of Pediatric Neurology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, United States of America; Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America; Corresponding authors at: Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Searle 8-510 320 E. Superior St, Chicago, IL 60611, United States of America.Division of Pediatric Neurology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, United States of America; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of AmericaDepartment of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of AmericaDepartment of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of AmericaDepartment of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States of America; Corresponding authors at: Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Searle 8-510 320 E. Superior St, Chicago, IL 60611, United States of America.KCNT1 encodes the sodium-activated potassium channel KNa1.1, expressed preferentially in the frontal cortex, hippocampus, cerebellum, and brainstem. Pathogenic missense variants in KCNT1 are associated with intractable epilepsy, namely epilepsy of infancy with migrating focal seizures (EIMFS), and sleep-related hypermotor epilepsy (SHE). In vitro studies of pathogenic KCNT1 variants support predominantly a gain-of-function molecular mechanism, but how these variants behave in a neuron or ultimately drive formation of an epileptogenic circuit is an important and timely question. Using CRISPR/Cas9 gene editing, we introduced a gain-of-function variant into the endogenous mouse Kcnt1 gene. Compared to wild-type (WT) littermates, heterozygous and homozygous knock-in mice displayed greater seizure susceptibility to the chemoconvulsants kainate and pentylenetetrazole (PTZ), but not to flurothyl. Using acute slice electrophysiology in heterozygous and homozygous Kcnt1 knock-in and WT littermates, we demonstrated that CA1 hippocampal pyramidal neurons exhibit greater amplitude of miniature inhibitory postsynaptic currents in mutant mice with no difference in frequency, suggesting greater inhibitory tone associated with the Kcnt1 mutation. To address alterations in GABAergic signaling, we bred Kcnt1 knock-in mice to a parvalbumin-tdTomato reporter line, and found that parvalbumin-expressing (PV+) interneurons failed to fire repetitively with large amplitude current injections and were more prone to depolarization block. These alterations in firing can be recapitulated by direct application of the KNa1.1 channel activator loxapine in WT but are occluded in knock-in littermates, supporting a direct channel gain-of-function mechanism. Taken together, these results suggest that KNa1.1 gain-of-function dampens interneuron excitability to a greater extent than it impacts pyramidal neuron excitability, driving seizure susceptibility in a mouse model of KCNT1-associated epilepsy.http://www.sciencedirect.com/science/article/pii/S096999612200105XEpilepsyPotassium channelKCNT1InterneuronMouse
spellingShingle Tracy S. Gertler
Suraj Cherian
Jean-Marc DeKeyser
Jennifer A. Kearney
Alfred L. George, Jr
KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons
Neurobiology of Disease
Epilepsy
Potassium channel
KCNT1
Interneuron
Mouse
title KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons
title_full KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons
title_fullStr KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons
title_full_unstemmed KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons
title_short KNa1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons
title_sort kna1 1 gain of function preferentially dampens excitability of murine parvalbumin positive interneurons
topic Epilepsy
Potassium channel
KCNT1
Interneuron
Mouse
url http://www.sciencedirect.com/science/article/pii/S096999612200105X
work_keys_str_mv AT tracysgertler kna11gainoffunctionpreferentiallydampensexcitabilityofmurineparvalbuminpositiveinterneurons
AT surajcherian kna11gainoffunctionpreferentiallydampensexcitabilityofmurineparvalbuminpositiveinterneurons
AT jeanmarcdekeyser kna11gainoffunctionpreferentiallydampensexcitabilityofmurineparvalbuminpositiveinterneurons
AT jenniferakearney kna11gainoffunctionpreferentiallydampensexcitabilityofmurineparvalbuminpositiveinterneurons
AT alfredlgeorgejr kna11gainoffunctionpreferentiallydampensexcitabilityofmurineparvalbuminpositiveinterneurons