Heterozygous expression of a Kcnt1 gain-of-function variant has differential effects on SST- and PV-expressing cortical GABAergic neurons

  1. Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, VA, USA
  2. Department of Neurological Sciences, University of Vermont, Burlington, VT, USA
  3. Translational Biology, Medicine, and Health Graduate Program, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
  4. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
  5. School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
  6. Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
  7. Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
  8. Department of Chemistry, Vanderbilt University, Nashville, TN, USA
  9. Institute for Genomic Medicine, Columbia University, New York, NY, USA
  10. Department of Neurology, Columbia University, New York, NY, USA
  11. School of Neuroscience, Virginia Tech, Blacksburg, VA, USA

Peer review process

Revised: This Reviewed Preprint has been revised by the authors in response to the previous round of peer review; the eLife assessment and the public reviews have been updated where necessary by the editors and peer reviewers.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Michelle Antoine
    National Institute on Alcohol Abuse and Alcoholism, Bethesda, United States of America
  • Senior Editor
    Sacha Nelson
    Brandeis University, Waltham, United States of America

Reviewer #1 (Public Review):

Summary:

This manuscript reports the effects of a heterozygous mutation in the KCNT1 potassium channels on the properties of ion currents and firing behavior of excitatory and inhibitory neurons in the cortex of mice expressing KCNT1-Y777H. In humans, this mutation as well as multiple other heterozygotic mutations produce very severe early-onset seizures and produce a major disruption of all intellectual function. In contrast, in mice, this heterozygous mutation appears to have no behavioral phenotype or any increased propensity to seizures. A relevant phenotype is, however, evident in mice with the homozygous mutation, and the authors have previously published the results of similar experiments with the homozygotes. As perhaps expected, the neuronal effects of the heterozygous mutation presented in this manuscript are generally similar but markedly smaller than the previously published findings on homozygotes. There are, however, some interesting differences, particularly on PV+ interneurons, which appear to be more excitable than wild type in the heterozygotes but more excitable in the heterozygotes. This raises the interesting question, which has been explicitly discussed by the authors in the revised manuscript, as to whether the reported changes represent homeostatic events that suppress the seizure phenotype in the mouse heterozygotes or simply changes in excitability that do not reach the threshold for behavioral outcomes.

Strengths and Weaknesses:

(1) The authors find that the heterozygous mutation in PV+ interneurons increases their excitability, a result that is opposite from their previous observation in neurons with the corresponding homozygous mutation. They propose that this results from the selective upregulation of a persistent sodium current INaP in the PV+ interneurons. These observations are very interesting ones, and they raised some issues in the original submission:

A) The protocol for measuring the INaP current could potentially lead to results that could be (mis)interpreted in different ways in different cells. First, neither K currents nor Ca currents are blocked in these experiments. Instead, TTX is applied to the cells relatively rapidly (within 1 second) and the ramp protocol is applied immediately thereafter. It is stated that, at this time, Na currents and INaP are fully blocked but that any effects on Na-activated K currents are minimal. In theory this would allow the pre- to post- difference current to represent a relatively uncontaminated INaP. This would, however, only work if activation of KNa currents following Na entry is very slow, taking many seconds. A good deal of literature has suggested that the kinetics of activation of KNa currents by Na influx vary substantially between cell types, such that single action potentials and single excitatory synaptic events rapidly evoke KNa currents in some cell types. This is, of course, much faster than the time of TTX application. Most importantly, the kinetics of KNa activation may be different in different neuronal types, which would lead to errors that could produce different estimates of INaP in PV+ interneurons vs other cell types.

In their revised manuscript, the authors have provided good data demonstrating that, at least for the PV and SST neurons, loss of KNa currents after TTX application is slow relative to the time course of loss of INaP, justifying the use of this protocol for these neuronal types.

B) As the authors recognize, INaP current provides a major source of cytoplasmic sodium ions for the activation. An expected outcome of increased INaP is, therefore, further activation of KNa currents, rather than a compensatory increase in an inward current that counteracts the increase in KNa currents, as is suggested in the discussion.

The authors comment in the rebuttal that, despite the fact that sodium entry through INaP is known to activate KNa channels, an increase in INaP does not necessarily imply increased KNa current. This issue should be addressed directly somewhere in the text, perhaps most appropriately in the discussion.

C) The numerical simulations, in general, provide a very useful way to evaluate the significance of experimental findings. Nevertheless, while the in-silico modeling suggests that increases in INaP can increase firing rate in models of PV+ neurons, there is as yet insufficient information on the relative locations of the INaP channels and the kinetics of sodium transfer to KNa channels to evaluate the validity of this specific model.

The authors have now put in all of the appropriate caveats on this very nicely in the revised manuscript.

(2) The effects of the KCNT1 channel blocker VU170 on potassium currents are somewhat larger and different from those of TTX, suggesting that additional sources of sodium may contribute to activating KCNT1, as suggested by the authors. Because VU170 is, however, a novel pharmacological agent, it may be appropriate to make more careful statements on this. While the original published description of this compound reported no effect on a variety of other channels, there are many that were not tested, including Na and cation channels that are known to activate KCNT1, raising the possibility of off-target effects.

In the revised version, the authors have added more to the manuscript on this issue and have added a very clear discussion of this to the text (in the discussion section).

This is a very clear and thorough piece of work, and the authors are to be congratulated on this. My one remaining suggestion would be to make an explicit statement about whether increased sodium influx through INaP channels, which is thought to activate KNa channels, would be likely to increase KNa current in these neurons (see comment 1B).

Reviewer #2 (Public Review):

Summary:

In this manuscript, Shore et al. investigate the consequent changes in excitability and synaptic efficacy of diverse neuronal populations in an animal model of juvenile epilepsy. Using electrophysiological patch-clamp recordings from dissociated neuronal cultures, the authors find diverging changes in two major populations of inhibitory cell types, namely somatostatin (SST)- and parvalbumin (PV)-positive interneurons, in mice expressing a variant of the KCNT1 potassium channel. They further suggest that the differential effects are due to a compensatory increase in the persistent sodium current in PV interneurons in pharmacological and in silico experiments. It remains unclear why this current is selectively enhanced in PV-interneurons.

Strengths:

(1) Heterozygous KCNT1 gain of function variant was used which more accurately models the human disorder.

(2) The manuscript is clearly written, and the flow is easy to follow. The authors explicitly state the similarities and differences between the current findings and the previously published results in the homozygous KCNT1 gain of function variant.

(3) This study uses a variety of approaches including patch clamp recording, in silico modeling and pharmacology that together make the claims stronger.

(4) Pharmacological experiments are fraught with off-target effects and thus it bolsters the authors' claims when multiple channel blockers (TTX and VU170) are used to reconstruct the sodium-activated potassium current.

Weaknesses:

(1) This study mostly relies on recordings in dissociated cortical neurons. Although specific WT interneurons showed intrinsic membrane properties like those reported for acute brain slices, it is unclear whether the same will be true for those cells expressing KCNT1 variants, especially when the excitability changes are thought to arise from homeostatic compensatory mechanisms. The authors do confirm that mutant SST-interneurons are hypoexcitable using an ex vivo slice preparation which is consistent with work for other KCTN1 gain of function variants (e.g. Gertler et al., 2022). However, the key missing evidence is the excitability state of mutant PV-interneurons, given the discrepant result of reduced excitability of PV cells reported by Gertler et al in acute hippocampal slices.

Reviewer #3 (Public Review):

Summary:

The present manuscript by Shore et al. entitled Reduced GABAergic Neuron Excitability, Altered Synaptic Connectivity, and Seizures in a KCNT1 Gain-of-Function Mouse Model of Childhood Epilepsy" describes in vitro and in silico results obtained in cortical neurons from mice carrying the KCNT1-Y777H gain-of-function (GOF) variant in the KCNT1 gene encoding for a subunit of the Na+-activated K+ (KNa) channel. This variant corresponds to the human Y796H variant found in a family with Autosomal Dominant Nocturnal Frontal lobe epilepsy. The occurrence of GOF variants in potassium channel encoding genes is well known, and among potential pathophysiological mechanisms, impaired inhibition has been documented as responsible for KCNT1-related DEEs. Therefore, building on a previous study by the same group performed in homozygous KI animals, and considering that the largest majority of pathogenic KCNT1 variants in humans occur in heterozygosis, the Authors have investigated the effects of heterozygous Kcnt1-Y777H expression on KNa currents and neuronal physiology among cortical glutamatergic and the 3 main classes of GABAergic neurons, namely those expressing vasoactive intestinal polypeptide (VIP), somatostatin (SST), and parvalbumin (PV), crossing KCNT1-Y777H mice with PV-, SST- and PV-cre mouse lines, and recording from GABAergic neurons identified by their expression of mCherry (but negative for GFP used to mark excitatory neurons).

The results obtained revealed heterogeneous effects of the variant on KNa and action potential firing rates in distinct neuronal subpopulations, ranging from no change (glutamatergic and VIP GABAergic) to decreased excitability (SST GABAergic) to increased excitability (PV GABAergic). In particular, modelling and in vitro data revealed that an increase in persistent Na current occurring in PV neurons was sufficient to overcome the effects of KCNT1 GOF and cause an overall increase in AP generation.

Strengths:

The paper is very well written, the results clearly presented and interpreted, and the discussion focuses on the most relevant points.
The recordings performed in distinct neuronal subpopulations (both in primary neuronal cultures and, for some subpopulations, in cortical slices, are a clear strength of the paper. The finding that the same variant can cause opposite effects and trigger specific homeostatic mechanisms in distinct neuronal populations is very relevant for the field, as it narrows the existing gap between experimental models and clinical evidence.

Weaknesses:

My main concern regarding the epileptic phenotype of the heterozygous mice investigated has been clarified in the revision, where the infrequent occurrence of seizures is more clearly stated. Also, a more detailed statistical analysis of the modeled neurons has been added in the revision.

Author response:

The following is the authors’ response to the original reviews.

Introduction to the revised manuscript:

We thank all three reviewers for their time and insightful comments on our original submission. We are submitting a substantially revised manuscript that includes several new experiments, analyses, discussion points, and clarifications that we believe address all of the main concerns of the reviewers.

To address the request of Reviewers 2 and 3 to reinforce key findings in a more physiologically intact preparation, we performed recordings of YH-HET SST neurons in brain slices and found that these neurons show impairments in AP generation similar to those observed in YH-HET SST cultured neurons. These data are summarized in a new figure (Fig. 9). Along these lines, we performed additional recordings in cultured neurons at room temperature compared with physiological temperature and found that WT and YH-HET PV neuronal properties were similarly altered by temperature increases, suggesting that our YH variant-induced neuronal phenotypes are not temperature dependent. These data are shown in a new supplemental figure (Supplemental Fig. 4-3). To address concerns of Reviewer 1 regarding our KNa and NaP current recordings, we performed new experiments to further assess the specificity of the VU170 blocker in KNa KO neurons (summarized in Supplemental Fig. 5-2) and to better characterize the time course over which TTX blocks the persistent Na+ current and the KNa current (summarized in Supplemental Fig. 7-1). These latter two experiments provide further clarity and confidence in the accuracy of our measurements of both KNa and NaP currents. Lastly, to address the concern of Reviewer 3 regarding statistical analyses of the modeling data, we’ve added a new table with the results of a repeated measures ANOVA analysis (Supplemental Table 6), and two new figures illustrating the relative changes in each neuron group compared to their controls (Supplemental Figures 6-2 and 7-2).

In addition to the new experiments and analyses, we’ve added three new paragraphs to the Discussion section. As the hyperexcitability phenotype in YH-HET PV neurons is somewhat unexpected, we’ve added a paragraph comparing our findings with those found in PV neurons in another KCNT1 GOF model. We’ve also added a paragraph to speculate on the contribution of YH-HET variant-induced alterations in SST and PV neurons to network behavior and seizure propensity. Lastly, we’ve added a paragraph to include the additional limitations and caveats of our study requested by the reviewers.

Public Reviews:

Reviewer #1 (Public Review):

Summary:

This manuscript reports the effects of a heterozygous mutation in the KCNT1 potassium channels on the properties of ion currents and the firing behavior of excitatory and inhibitory neurons in the cortex of mice expressing KCNT1-Y777H. In humans, this mutation as well as multiple other heterozygotic mutations produce very severe early-onset seizures and produce a major disruption of all intellectual function. In contrast, in mice, this heterozygous mutation appears to have no behavioral phenotype or any increased propensity to seizures.

Regarding the last sentence above, we wanted to clarify a point that we neglected to emphasize in the initial submission. In the Results section from our previous paper (Shore et al., 2020), we failed to observe seizures in 14 heterozygous mice, whereas 23/25 homozygous mice showed seizures by video-EEG. However, in the fifth paragraph of the Discussion section from that paper, we further stated that “during the preparation and review of [that] article, we observed seizures in two Kcnt1-Y777H heterozygous mice, one during a widefield Ca2+ imaging experiment and the other during a video-EEG experiment”. Thus, we concluded that “heterozygous expression can result in seizures in a rodent model, but apparently at a much lower frequency than that observed with homozygous expression”. To emphasize these findings, we’ve added a sentence to the Introduction in this manuscript about the occurrence of infrequent seizures in Kcnt1-Y777H heterozygous mice, along with a reference to the Discussion of our previous paper.

A relevant phenotype is, however, evident in mice with the homozygous mutation, and the authors have previously published the results of similar experiments with the homozygotes. As perhaps expected, the neuronal effects of the heterozygous mutation presented in this manuscript are generally similar but markedly smaller than the previously published findings on homozygotes. There are, however, some interesting differences, particularly on PV+ interneurons, which appear to be more excitable than wild type in the heterozygotes but more excitable in the heterozygotes. This raises the interesting question (which could be more explicitly discussed by the authors) as to whether the reported changes represent homeostatic events that suppress the seizure phenotype in the mouse heterozygotes or simply changes in excitability that do not reach the threshold for behavioral outcomes.

That is an interesting question. We have added a new paragraph to the Discussion speculating about whether the alterations in SST and PV excitability suppress seizures or do not reach the threshold for behavioral outcomes. This seems to be requested by the second reviewer as well in Weaknesses point #2.

Strengths and Weaknesses:

(1) The authors find that the heterozygous mutation in PV+ interneurons increases their excitability, a result that is opposite from their previous observation in neurons with the corresponding homozygous mutation.

We would like to provide a minor clarification to the above statement that, in this manuscript, we show that “the heterozygous mutation in PV+ interneurons increases their excitability, a result that is opposite from their previous observation in neurons with the corresponding homozygous mutation”. In our previous manuscript, we assessed YH-HOM phenotypes in NFS and FS GABAergic neurons, but did not specifically mark PV neurons. Although the YH-HOM FS neurons showed an increase in rheobase and a decrease in AP firing, the magnitudes of these effects were far less than those observed in the NFS population. More importantly, the FS GABAergic population likely consists of PV- and SST-expressing neurons; thus, we can not directly compare the results from the NFS and FS groups to the PV and SST groups, respectively (please see our response to Weaknesses point #3, Reviewer #2). We apologize for the confusion.

They propose that this results from the selective upregulation of a persistent sodium current INaP in the PV+ interneurons. While the observations are very interesting, there are three issues concerning this interpretation that should be addressed:

A) The protocol for measuring the INaP current could potentially lead to results that could be (mis)interpreted in different ways in different cells. First, neither K currents nor Ca currents are blocked in these experiments. Instead, TTX is applied to the cells relatively rapidly (within 1 second) and the ramp protocol is applied immediately thereafter. It is stated that, at this time, Na currents and INaP are fully blocked but that any effects on Na-activated K currents are minimal. In theory, this would allow the pre- to post-difference current to represent a relatively uncontaminated INaP. This would, however, only work if activation of KNa currents following Na entry is very slow, taking many seconds. A good deal of literature has suggested that the kinetics of activation of KNa currents by Na influx vary substantially between cell types, such that single action potentials and single excitatory synaptic events rapidly evoke KNa currents in some cell types. This is, of course, much faster than the time of TTX application. Most importantly, the kinetics of KNa activation may be different in different neuronal types, which would lead to errors that could produce different estimates of INaP in PV+ interneurons vs other cell types.

First, we’d like to point out that we did not want to block K+ currents (which would also block KNa) when measuring INaP for these experiments, because our hypothesis was that the increased KNa current in YH-HET PV neurons was somehow causing an increase in INaP, and it is possible that this increase depends on an intact KNa. Thus, we decided to use a method based on the observation in our experiments, and previously made by others (Budelli et al., 2009), that the reduction of outward current after TTX addition is slow relative to the rapid reduction in Na+ current. We understand and agree with the reviewer that, if KNa currents were blocked more quickly by TTX in some neuron types than others, then our estimate of INaP using this method would be contaminated in these neuron types, which would lead to inaccurate measurements. To assess this possibility among the main neuron types used in this study, we performed new experiments in which we monitored the time course of INaP block and subsequent IKNa loss following TTX application in PV and SST neurons during slow voltage ramps. We note that action potentials are not present in the slow voltage ramps due to inactivation of the transient Na+ current. These new experiments show that, in SST and PV (both WT and Het) neurons, the block of INaP is nearly complete at the 6s time point, whereas the decay in IKNa is far slower (V50 of ≈ 25s), and importantly, these results do not differ substantially by cell type or genotype. These data suggest that our measurements of INaP are not significantly contaminated by IKNa, and that this method allows for the effective separation of these two currents. These data have been added as a supplemental figure (Supplemental Fig. 7-1) and are briefly described and referenced in the Results section.

B) As the authors recognize, INaP current provides a major source of cytoplasmic sodium ions for the activation. An expected outcome of increased INaP is, therefore, further activation of KNa currents, rather than a compensatory increase in an inward current that counteracts the increase in KNa currents, as is suggested in the discussion.

We agree that the increase in INaP could theoretically further increase IKNa, as veratridine was previously shown to increase IKNa (Hage & Salkoff, 2012). However, we do not believe that this would necessarily be the case, because as the reviewer notes in their next comment, there is insufficient information on the relative locations of the INaP and KCNT1 channels, as well as the kinetics of sodium transfer to KCNT1 channels, and even less is known in the context of KCNT GOF neurons. Thus, there are a couple of plausible reasons that increased INaP may not alter KNa currents in YH-HET PV neurons: (1) In YH-HET PV neurons, the particular sodium channels that are responsible for the increased INaP may not be located within close proximity to the KCNT1 channels. (2) Homeostatic mechanisms that alter the AIS length, or move the AIS further from the soma, in response to altered neuronal excitability are well described (Grubb & Burrone, 2010; Kuba et al., 2010); thus, it is possible that in YH-HET PV neurons, the length or location of the AIS is altered, leading to uncoupling of the sodium channels that are responsible for the increased INaP to the KCNT1 channels.

C) Numerical simulations, in general, provide a very useful way to evaluate the significance of experimental findings. Nevertheless, while the in-silico modeling suggests that increases in INaP can increase firing rate in models of PV+ neurons, there is as yet insufficient information on the relative locations of the INaP channels and the kinetics of sodium transfer to KNa channels to evaluate the validity of this specific model.

We completely agree; thus, we have described each of these limitations in the Discussion. We state that the model neurons may “lack more detailed features of ion channels, such as post-translational modifications and subcellular localizations”, and that our KCNT1 model conductance is “hampered by an incomplete understanding of the relationship between Na+ influx, membrane voltage, and channel gating in neurons”.

(2) The greatest effect of TTX application would be expected to be the elimination of large transient inward sodium currents. Why are no such currents visible in the control (pre-TTX) or the difference currents (Fig. 2)? Is it possible I missed something in the methods?

We apologize for the confusion and our mistake in failing to mention this important feature of the displayed traces. To include all of the representative traces in the figures, and prevent overlap of the traces, we removed the large inward sodium currents using the masking tool in Adobe Illustrator in Figure 2 and Supplemental Figure 5-1. We have added that information to the relevant figure legends. We have also provided unmasked images of the representative traces from Figure 2 and Supplemental Figure 5-1 to illustrate the large transient inward sodium currents, and the significant reduction of these currents with TTX treatment.

(3) As expected, the changes in many of the measured parameters are smaller in the present study with heterozygotes than those previously reported for the homozygous mutation. Some of the statements on the significance of some of the present findings need to be stated more clearly. For example, in the results section describing Fig. 2, it is stated that "In glutamatergic and NFS GABAergic YH-HET neurons, the overall KNa current was increased ...as measured by a significant effect of genotype ...." Later in the same paragraph it is stated that the increases in KNa current are not significant. Apparently, different tests lead to different conclusions. Both for the purpose of understanding the pathophysiological effects of changes in KNa current and for making further numerical simulations, more explicit clarifying statements should be made.

We apologize for the confusion on the description of these statistics. The results come from the same test, which is a Generalized Linear Mixed Model (GLMM). The factors in our GLMM were voltage step, genotype, and a voltage step x genotype interaction term. The overall effect of genotype is significant in glutamatergic neurons, but pairwise tests at each voltage step show no significant effect of genotype at any given voltage. This is somewhat analogous to running a traditional ANOVA on multiple groups and finding a significant ANOVA p-value but no significant post-hoc multiple comparisons tests, and is not uncommon. Our interpretation of this is that heterozygous expression of the YH variant in glutamatergic neurons likely increases KNa currents across positive potentials (as was seen with the YH-HOM glutamatergic neurons), but only a small amount at each positive step; thus, we lack the statistical power to determine any particular voltage step where this occurs.

(4) The effects of the KCNT1 channel blocker VU170 on potassium currents are somewhat larger and different from those of TTX, suggesting that additional sources of sodium may contribute to activating KCNT1, as suggested by the authors. Because VU170 is, however, a novel pharmacological agent, it may be appropriate to make more careful statements on this. While the original published description of this compound reported no effect on a variety of other channels, there are many that were not tested, including Na and cation channels that are known to activate KCNT1, raising the possibility of off-target effects.

We agree and thank the reviewer for making this point. To address this question, we measured KNa currents in WT vs. Kcnt1/Kcnt2-dKO neurons using VU170 to illustrate the extent of outward current due to off-target effects of the drug. These data have been included as a supplemental figure (Supplemental Fig. 5-2). We have also added several sentences to the Results section referencing this figure. Interestingly, in Kcnt1/Kcnt2-dKO neurons, VU170 seems to be quite specific across the negative potentials, as no outward currents are apparent until approximately -10 mV onward, whereas across positive potentials, there is a VU170-senstive outward current reaching ~1 nA by +50 mV. We have also included a note of caution in interpreting these data and added the possibility of off-target effects of VU170 as an alternative explanation for the differences observed on KNa currents between TTX and VU170 to the Discussion section.

(5) The experiments were carried out at room temperature. Is it possible that different effects on firing patterns in heterozygotes and homozygotes would be observed at more physiological temperatures?

Yes, it is reasonable to assume that an increased temperature would affect neuronal firing patterns in cultured neurons, as temperature differences have been shown to alter synaptic transmission and neuronal function, as assessed in both cultured neuron and slice recordings. All of our recordings were performed at room temperature in this study, and although they are valid with regard to between-group comparisons, this additional caveat is worth mentioning. We have added this to the paragraph describing study limitations in the Discussion section.

To better understand the effects of temperature in our recordings, we have now compared membrane and AP generation parameters at room temperature (~22°C) and at a more physiological temperature (35°C) in a before-after study of 16 WT neurons, including both glutamatergic and GABAergic neuron types. Not surprisingly, we found robust alterations in all parameters assessed, excluding resting membrane potential and capacitance. We further assessed the effect of temperature on WT and YH-HET PV neurons, as the PV neurons expressing the YH variant showed the most unexpected phenotypes in our study. In our room temperature recordings, we showed that the YH-HET variant decreased the rheobase current, increased the AP amplitude, and increased the AP firing. In our before-after comparison (22°C-35°C) of PV neurons (WT; n=11, YH-Het; n=10), the WT and YH-HET neurons showed the same temperature-dependent effects on these parameters, including increased rheobase, decreased AP amplitude, and a higher maximal firing rate, at 35°C compared to those at 22°C. These data have been added to the manuscript as a supplemental figure (Supplemental Fig. 4-3) and are briefly referenced and described in the Results section.

Moreover, in our original manuscript, we showed that the effects of the homozygous YH variant on glutamatergic and NFS GABAergic neuron excitability were highly similar between cultured recordings at room temperature (~22°C) and slice recordings at 32°C. Taken together, these data suggest that the reported neurophysiological phenotypes downstream of the YH variant are likely not temperature dependent.

Reviewer #2 (Public Review):

Summary:

In this manuscript, Shore et al. investigate the consequent changes in excitability and synaptic efficacy of diverse neuronal populations in an animal model of juvenile epilepsy. Using electrophysiological patch-clamp recordings from dissociated neuronal cultures, the authors find diverging changes in two major populations of inhibitory cell types, namely somatostatin (SST)- and parvalbumin (PV)-positive interneurons, in mice expressing a variant of the KCNT1 potassium channel. They further suggest that the differential effects are due to a compensatory increase in the persistent sodium current in PV interneurons in pharmacological and in silico experiments.

Strengths:

(1) Heterozygous KCNT1 gain of function variant was used which more accurately models the human disorder.

(2) The manuscript is clearly written, and the flow is easy to follow. The authors explicitly state the similarities and differences between the current findings and the previously published results in the homozygous KCNT1 gain of function variant.

(3) This study uses a variety of approaches including patch clamp recording, in silico modeling, and pharmacology that together make the claims stronger.

(4) Pharmacological experiments are fraught with off-target effects and thus it bolsters the authors' claims when multiple channel blockers (TTX and VU170) are used to reconstruct the sodium-activated potassium current. Having said that, it would be helpful to see the two drug manipulations used in the same experiment. Notably, does the more selective blocker VU170 mimic the results of TTX for NFS GABAergic cells in Figure 2? And does it unmask a genotype difference for FS GABAergic cells like the one seen in PV interneurons in Figure 5C3.

To illustrate the two drug manipulations in the same experiment, we recorded from WT SST and PV neurons (5 neurons/group) and blocked KNa currents first using TTX and then VU170, following wash out between the two drugs, in the same neurons. Below, we have plotted the points at each voltage step for each SST and PV neuron, and for each drug treatment, on the same graph to show how they vary directly. At each voltage step, lines connect the points representing the TTX-sensitive and VU-sensitive currents for each neuron to show the individual effects (left-most graphs). Summary data are shown across all voltages (middle graphs) and across negative voltages (right-most graphs).

Author response image 1.

We have not used VU170 on FS and NFS populations of GABAergic neurons. However, for reasons that are explained more extensively below in response to Weaknesses #3, we would not predict KNa currents recorded from SST- and PV-GABAergic neurons to mimic those of NFS- and FS-GABAergic neurons, respectively.

Weaknesses:

(1) This study relies on recordings in dissociated cortical neurons. Although specific WT interneurons showed intrinsic membrane properties like those reported for acute brain slices, it is unclear whether the same will be true for those cells expressing KCNT1 variants. This reviewer highly recommends confirming some of the key findings using an ex vivo slice preparation. This is especially important given the discrepant result of reduced excitability of PV cells reported by Gertler et al., 2022 (cited here in the manuscript but not discussed in this context) in acute hippocampal slices for a different KCNT1 gain of function variant.

We thank the reviewer for this suggestion. To test whether SST-expressing YH-HET neurons show similar impairments to those observed in culture, we crossed the FVB-Tg(GadGFP)45704Swn/J transgenic mouse line (Jackson Labs #003718), also known as the GIN line, to the Kcnt1-YH line. Mice from the GIN line express eGFP in a subpopulation of SST-expressing neurons in the hippocampus and cortex. We performed slice recordings of cortical layer 2/3, GFP-expressing neurons from P21-30, WT and YH-HET GIN mice. Although the input resistance was not significantly decreased, the rheobase was higher in the YH-HET neurons, and they fired fewer APs across increasing current steps, than WT neurons, supporting the main findings from the SST-expressing neurons in culture. These data have been added to the manuscript in a new figure (Fig. 9).

Regarding the previously published results on the effect of KCNT1 GOF on PV neuron excitability by Gertler et al., we have written a new paragraph in the Discussion section (last paragraph of the section, “Neuron-type-dependent KCNT1 GOF effects”) that discusses the differences between the findings by Gertler et al. and the current study.

To further investigate the effects of heterozygous YH variant expression on SST- vs. PV-expressing neuron excitability in ex vivo slice recordings, we are now crossing a cre-inducible, Td-Tomato Red reporter line (Ai9) to the Kcnt1-YH line. After obtaining Ai9Tg/Tg; Kcnt1m/+ mice, we will cross these to Sst-Cre and Pvalb-Cre lines to be able to record from marked SST and PV, WT and YH-HET neurons in slice. We plan on submitting results from these recordings as an eLife Research Advances article linked to this article.

(2) It is unclear how different pieces of results fit together to form a story about the disease pathophysiology.

We have added a paragraph to the Discussion to speculate on how these various GABAergic subtype-specific effects downstream of the YH variant may contribute to overall network/brain pathology and seizure propensity in heterozygous mice.

For example, hyperexcitability of PV cells would suggest more inhibition which would counter seizure propensity. However, spontaneous inhibitory postsynaptic currents show no change in pyramidal neurons. Moreover, how do the authors reconcile that the reductions in synaptic inputs onto interneurons in Figure 3B with the increases in Figure 8? This should be discussed.

Generally, network and synaptic alterations downstream of the heterozygous variant were quite minimal compared with those of the homozygous variant. Although there were reductions in the frequency of synaptic inputs onto inhibitory neurons, the changes were relatively small. Thus, we concluded that the neuronal effects downstream of the heterozygous YH variant were below some threshold to result in broader network effects on synaptic activity and connectivity similar to those in the homozygous YH model. The discrepancies between our GABAergic vs. FS/NFS vs. VIP/SST/PV data will be discussed in more detail in response to Weakness #3.

(3) Similarly, the results in this work are not entirely internally consistent. For example, given the good correspondence between FS and NFS GABAergic cells with PV and SST expression, why are FS GABAergic cells hyperexcitable in Figure 1? If anything, there is a tendency to show reduced excitability like the NFS GABAergic cells.

In our neuron cultures, 76-80% of Neu-N-expressing neurons are GFP+ (from the CamKII-eGFP virus used to mark glutamatergic neurons), and of the remaining ~20-24%, the majority are GABAergic (verified using the Dlx5/6-mRuby virus to mark GABAergic neurons and using electrophysiology to assess AP parameters and analyze evoked responses). In our original experiments, recordings sampled from this larger GABAergic population were used (Fig. 3), or this population was sorted almost equally into FS and NFS (Figs. 1 and 2).

In later experiments, we isolated and cultured neurons from VIP-Cre, SST-Cre, and PV-Cre mouse lines and marked these neuron types in vitro with a Cre-inducible mCherry virus. In the VIP-Cre cultures, ~6% of the GFP- population, or 1.2% of the Neu-N-population, was mCherry+. In the SST-Cre cultures, ~20.5% of the GFP- population, or 4.7% of the Neu-N-population, was mCherry+. In the PV-Cre cultures, less than 1% of the Neu-N-population was mCherry+, which is not surprising considering the relatively late onset of PV expression compared with those of VIP and SST. Thus, we would estimate that we are marking and recording from less than 30% of the total GABAergic population in these in vitro experiments, rather than the 80-90% that these three populations would sum to in vivo.

Furthermore, using our original criteria for sorting GABAergic neurons into FS and NFS subtypes, all VIP recorded neurons were of the NFS type, PV of the FS type, whereas SST were of the FS (38%) and NFS (62%) types, which is not far off from the significant fraction of SST neurons that have been shown to be fast-spiking in slice experiments (Kvitsiani et al., 2013; Urban-Ciecko & Barth, 2016). Therefore, the FS group consists of both PV and SST neurons, and the NFS group consists of both VIP and SST neurons, and likely also contains immature PV neurons that have not yet developed a fast-spiking phenotype. Taken together, this suggests that the data from these two sets of experiments (FS/NFS vs. VIP/SST/PV) are not directly comparable.

Also, why do the WT I-V curves look so different between Figures 2 and 5? This reviewer suggests at least a brief explanation in the discussion.

As to the differences in appearance between the WT I-V curves in Figures 2 and 5, those plots are from different neuron types (Fig. 2: Glutamatergic, FS GABAergic, and NFS GABAergic vs. Fig. 5: VIP-, SST-, and PV-expressing), and the KNa currents are isolated using different methods (Fig. 2: TTX-subtraction vs. Fig. 5: VU170-subtraction). TTX blocks an inward Na+ current, which is apparent across subthreshold voltages in Fig. 2C1-3, whereas VU170 does not block this current, making it not apparent in Fig. 5C1-3. Also, the bottom three panels in Fig. 2C1-3 show the KNa current from -80 to 0 mV, whereas those in Fig. 5C1-3 show from -80 to -30 mV, to better illustrate the areas spanning KNa current increases, so their appearance is not directly comparable.

(4) Given the authors' claim that the KCNT1 activation curve is a major contributor to the observed excitability differences in specific GABA cell subtypes, it would be helpful to directly measure the activation curve in the variants experimentally as was done for WT KCNT1 in Figure 6A and use the derived kinetics in the compartmental model.

We apologize for the confusion. Although the activation curves among different GABAergic subtypes from WT KCNT1 are distinct, and we believe that these varying kinetics contribute to the neuron-type-specific phenotypes of KCNT1 GOF, we didn’t intend to suggest that the heterozygous Y777H variant itself causes neuron-type-specific alterations to the activation curves of the GABAergic subtypes. To clarify this point, below, we show the high similarity of the activation curves between WT KCNT1 and YH-HET KCNT1 in each of the GABAergic subtypes.

Author response image 2.

Reviewer #3 (Public Review):

Summary:

The present manuscript by Shore et al. entitled Reduced GABAergic Neuron Excitability, Altered Synaptic Connectivity, and Seizures in a KCNT1 Gain-of-Function Mouse Model of Childhood Epilepsy" describes in vitro and in silico results obtained in cortical neurons from mice carrying the KCNT1-Y777H gain-of-function (GOF) variant in the KCNT1 gene encoding for a subunit of the Na+-activated K+ (KNa) channel. This variant corresponds to the human Y796H variant found in a family with Autosomal Dominant Nocturnal Frontal lobe epilepsy. The occurrence of GOF variants in potassium channel encoding genes is well known, and among potential pathophysiological mechanisms, impaired inhibition has been documented as responsible for KCNT1-related DEEs. Therefore, building on a previous study by the same group performed in homozygous KI animals, and considering that the largest majority of pathogenic KCNT1 variants in humans occur in heterozygosis, the Authors have investigated the effects of heterozygous Kcnt1-Y777H expression on KNa currents and neuronal physiology among cortical glutamatergic and the 3 main classes of GABAergic neurons, namely those expressing vasoactive intestinal polypeptide (VIP), somatostatin (SST), and parvalbumin (PV), crossing KCNT1-Y777H mice with PV-, SST- and PV-cre mouse lines, and recording from GABAergic neurons identified by their expression of mCherry (but negative for GFP used to mark excitatory neurons).

The results obtained revealed heterogeneous effects of the variant on KNa and action potential firing rates in distinct neuronal subpopulations, ranging from no change (glutamatergic and VIP GABAergic) to decreased excitability (SST GABAergic) to increased excitability (PV GABAergic). In particular, modelling and in vitro data revealed that an increase in persistent Na current occurring in PV neurons was sufficient to overcome the effects of KCNT1 GOF and cause an overall increase in AP generation.

Strengths:

The paper is very well written, the results clearly presented and interpreted, and the discussion focuses on the most relevant points.

The recordings performed in distinct neuronal subpopulations are a clear strength of the paper. The finding that the same variant can cause opposite effects and trigger specific homeostatic mechanisms in distinct neuronal populations is very relevant for the field, as it narrows the existing gap between experimental models and clinical evidence.

Weaknesses:

My main concern is in the epileptic phenotype of the heterozygous mice investigated. In fact, in their previous paper the Authors state that "...Kcnt1-Y777H heterozygous mice did not exhibit any detectable epileptiform activity" (first sentence on page 4). However, in the present manuscript, they indicate twice in the discussion section that these mice exhibit "infrequent seizures". This relevant difference needs to be clarified to correctly attribute to the novel pathophysiological mechanism a role in seizure occurrence. Were such infrequent seizures clearly identified on the EEG, or were behavioral seizures? Could the authors quantify this "infrequent" value? This is crucial also to place in the proper perspective the Discussion statement regarding "... the increased INaP contribution to ... network hyperexcitability and seizures".

We apologize for the confusion. Indeed, in the Results section from our previous paper, we failed to observe seizures in 14 heterozygous mice, whereas 23/25 homozygous mice showed seizures by video-EEG. However, in the fifth paragraph of the Discussion section from that paper, we further stated that “during the preparation and review of [that] article, we observed seizures in two Kcnt1-Y777H heterozygous mice, one during a widefield Ca2+ imaging experiment and the other during a video-EEG experiment”. Thus, we concluded that “heterozygous expression can result in seizures in a rodent model, but apparently at a much lower frequency than that observed with homozygous expression”. To emphasize these findings, we’ve added a sentence to the Introduction in this manuscript about the occurrence of infrequent seizures in Kcnt1-Y777H heterozygous mice, along with a reference to the Discussion of our previous paper.

Of the two observed seizures, one seizure was captured in the Weston Lab at the University of Vermont from a Kcnt1-Y777H heterozygous mouse expressing a calcium indicator (after it was bred to the Snap25-GCaMP6s line) during a Ca2+ widefield imaging experiment, and it was accompanied by a time-locked video of the seizure event. The other seizure was recorded as a control during a drug study using video-EEG. This Kcnt1-Y777H heterozygous mouse had multiple tonic seizures, as evidenced by EEG traces and the accompanying video, which were recorded and analyzed in the Frankel Lab at Columbia University. The seizures from heterozygous mice have not been officially quantified, as they have only been rarely observed across multiple different experiments using heterozygous mice at multiple institutions, making quantification quite difficult.

Lastly, regarding attributing the role of the identified pathological mechanisms to seizure occurrence mentioned by the reviewer, we have added a paragraph to the Discussion to speculate on how the various GABAergic subtype-specific effects downstream of the YH variant may contribute to the general lack of network/brain pathology and seizure generation in heterozygous mice.

Also, some statistical analysis seems to be missing. For example, I could not find any for the data shown in Fig. 6. Thus, the following statement: "the model PV neurons responded to KCNT1 GOF with decreased AP firing and an increased rheobase" requires proper statistical evaluation.

We thank the reviewer for this suggestion. We were initially hesitant to apply a formal statistical analysis to the modeling data because it differs in important ways from the experimental data. However, we have now provided statistical analyses of these data, with some caveats. Because we applied each KCNT1 GOF level (40, 35, and 30 mM) to the same set of neurons for each data set, we performed repeated measures ANOVA analyses to assess differences due to GOF in each subtype. We note that some changes are statistically significant, but may not be physiologically relevant. For example, there are changes in input resistance and rheobase in VIP neurons only at the higher GOF level (30 mM), but the magnitude of each change is quite small relative to those in SST neurons (Rin: 1.7 MΩ in VIP vs. 23.2 MΩ in SST, rheo: 1.7 pA in VIP vs. 52.5 pA in SST), and likely as a consequence, there are no downstream effects on the AP firing rate at either GOF level in VIP neurons. It is important to examine the magnitude of the effects and interpret them in the context of the changes in other neuron types and in the experimental data, thus, we’ve provided two new figures to better illustrate the relative changes in each neuron type (Supplemental Figures 6-2 and 7-2). We have also added these statistical results to Figures 6E2, 6F2, 6G2, and 7E, and Supplemental Fig. 6-1, and we have described them in the Results section. A summary of the statistics has also been added in Supplemental Table 6.

Recommendations for the authors:

Reviewer #2 (Recommendations For The Authors):

In addition to addressing the weaknesses highlighted in the public review, this reviewer recommends using a KCNT1 agonist such as loxapine to see if activating the potassium channel mimics the KCNT1 GOF in SST and PV cells.

Although we appreciate this suggestion, we’re not sure whether treating GABAergic subtypes with loxapine would provide much clarity in the absence of many supporting experiments. First, the amount of channel activation and any changes in kinetics caused by loxapine would need to be measured and compared to the YH-HET GOF effects in order to interpret the results. In addition, the aforementioned caveat about off-target effects of small molecules would also have to be considered, as loxapine inhibits many other channels at nanomolar concentrations.

More importantly, we hypothesize that several of the GABAergic subtype-specific effects of KCNT1 GOF result from homeostatic or adaptive mechanisms due to long-term increases in KNa currents. For instance, PV-expressing YH-HET neurons had a lower rheobase, increased AP amplitude, and increased AP firing frequency, effects that we believe are due, not to increased KNa currents themselves, but to a compensatory increase in a persistent Na+ current. For the SST neurons, we hypothesize that the increased capacitance and soma size, together with the increased electrical coupling, exacerbate the hypoexcitability phenotype downstream of the YH variant. Thus, we would not necessarily expect that opening KCNT1 channels by acute loxapine treatment would mimic many of these effects.

Indeed, in a previous study using a different KCNT1 GOF mouse model, loxapine treatment mimics KCNT1 GOF effects in some neuron types (reduced AP firing frequency in loxapine-treated, WT PV neurons mimics that observed in heterozygous KCNT1 GOF PV neurons), but not in others (reduced AP firing frequency in loxapine-treated, WT pyramidal neurons does not mimic the unaltered AP firing frequency observed in heterozygous and homozygous KCNT1 GOF pyramidal neurons) (Gertler et al., 2022).

Related to this suggestion by the reviewer, we are currently performing studies using a KCNT1 blocker in WT and Kcnt1-KO neurons to better understand the role of KCNT1 among cortical neuronal subtypes that will be published in a future manuscript.

Reviewer #3 (Recommendations For The Authors):

Though I realize that primary cultures allow for efficient identification of neuronal subclasses, it would have been useful to show that similar changes also occur in neurons with conserved in vivo connectivity, such as those recorded from brain slices.

We thank the reviewer for this suggestion. We have added an additional figure (Fig. 9) showing that the hypoexcitability phenotype observed in SST neurons in culture recordings is conserved in SST neurons in slice recordings from GIN mice, which express GFP predominately in SST-expressing neurons.

In addition, further experiments in PV neurons from Kcnt1-Y777H homozygous mice would provide evidence for a gene-dosage role in the changes found in heteros.

For this manuscript, we chose to focus our efforts on understanding the effects of heterozygous Kcnt1 variant expression in various neuronal subtypes with the goal of better modeling GOF variant effects in human disease. However, we’re very interested in investigating the effects of homozygous expression of the YH variant on each of the GABAergic subtypes to compare with those found in this study, but this requires more rounds of breeding to get homozygous mice with GABAergic subtype-specific expression of cre recombinase. We look forward to reporting the results from these experiments in a future manuscript.

Also, when addressing the issue regarding the different effects of the same GOF variant on the excitability of distinct neuronal populations in the Discussion or Introduction sections, the authors may want to cite the recent work on KCNQ2 and KCNQ3 by the Tzingounis group (https://pubmed.ncbi.nlm.nih.gov/37607817/).

We thank the reviewer for bringing this manuscript to our attention. We have added this citation to a new paragraph in the Discussion section regarding neuron-type specific effects of ion channel variants (the last paragraph focusing on the effects in PV neurons).

Budelli, G., Hage, T. A., Wei, A., Rojas, P., Jong, Y. J., O'Malley, K., & Salkoff, L. (2009). Na+-activated K+ channels express a large delayed outward current in neurons during normal physiology. Nat Neurosci, 12(6), 745-750. https://doi.org/10.1038/nn.2313

Gertler, T. S., Cherian, S., DeKeyser, J. M., Kearney, J. A., & George, A. L., Jr. (2022). K(Na)1.1 gain-of-function preferentially dampens excitability of murine parvalbumin-positive interneurons. Neurobiol Dis, 168, 105713. https://doi.org/10.1016/j.nbd.2022.105713

Grubb, M. S., & Burrone, J. (2010). Activity-dependent relocation of the axon initial segment fine-tunes neuronal excitability. Nature, 465(7301), 1070-1074. https://doi.org/10.1038/nature09160

Hage, T. A., & Salkoff, L. (2012). Sodium-activated potassium channels are functionally coupled to persistent sodium currents. J Neurosci, 32(8), 2714-2721. https://doi.org/10.1523/JNEUROSCI.5088-11.2012

Kuba, H., Oichi, Y., & Ohmori, H. (2010). Presynaptic activity regulates Na(+) channel distribution at the axon initial segment. Nature, 465(7301), 1075-1078. https://doi.org/10.1038/nature09087

Kvitsiani, D., Ranade, S., Hangya, B., Taniguchi, H., Huang, J. Z., & Kepecs, A. (2013). Distinct behavioural and network correlates of two interneuron types in prefrontal cortex. Nature, 498(7454), 363-366. https://doi.org/10.1038/nature12176

Shore, A. N., Colombo, S., Tobin, W. F., Petri, S., Cullen, E. R., Dominguez, S., Bostick, C. D., Beaumont, M. A., Williams, D., Khodagholy, D., Yang, M., Lutz, C. M., Peng, Y., Gelinas, J. N., Goldstein, D. B., Boland, M. J., Frankel, W. N., & Weston, M. C. (2020). Reduced GABAergic neuron excitability, altered synaptic connectivity, and seizures in a KCNT1 gain-of-function mouse model of childhood epilepsy. Cell Rep.

Urban-Ciecko, J., & Barth, A. L. (2016). Somatostatin-expressing neurons in cortical networks. Nat Rev Neurosci, 17(7), 401-409. https://doi.org/10.1038/nrn.2016.53

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation