Stxbp1/Munc18-1 haploinsufficiency impairs inhibition and mediates key neurological features of STXBP1 encephalopathy

  1. Wu Chen
  2. Zhao-Lin Cai
  3. Eugene S Chao
  4. Hongmei Chen
  5. Colleen M Longley
  6. Shuang Hao
  7. Hsiao-Tuan Chao
  8. Joo Hyun Kim
  9. Jessica E Messier
  10. Huda Y Zoghbi
  11. Jianrong Tang
  12. John W Swann
  13. Mingshan Xue  Is a corresponding author
  1. Department of Neuroscience, Baylor College of Medicine, United States
  2. The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, United States
  3. Program in Developmental Biology, Baylor College of Medicine, United States
  4. Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, United States
  5. Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, United States
  6. Department of Molecular and Human Genetics, Baylor College of Medicine, United States
  7. McNair Medical Institute, The Robert and Janice McNair Foundation, United States
  8. Howard Hughes Medical Institute, Baylor College of Medicine, United States
9 figures, 4 videos, 1 table and 4 additional files

Figures

Figure 1 with 2 supplements
Stxbp1 haploinsufficient mice exhibit reduced Stxbp1 levels, survival, and body weights and develop hindlimb clasping.

(A) Genomic structures of Stxbp1 WT, tm1a (KO-first), and tm1d (KO) alleles. In the tm1a allele, the STOP including the En2SA-IRES-lacZ-pA trapping cassette (see Figure 1—figure supplement 1A) truncates the Stxbp1 mRNA after exon 6. In the tm1d allele, exon 7 is deleted, resulting in a premature stop codon in exon 8. E, exon; FRT, Flp recombination site; loxP, Cre recombination site. (B) Representative Western blots of proteins from different brain regions of 3-month-old WT, Stxbp1tm1d/+, and Stxbp1tm1a/+ mice. Gapdh, a housekeeping protein as loading control. The brain regions are labeled by the same abbreviations as in (C). (C) Summary data of normalized Stxbp1 expression levels from different brain regions. Stxbp1 levels were first normalized by the Gapdh levels and then by the average Stxbp1 levels of all WT mice from the same blot. Each filled (male) or open (female) circle represents one mouse. (D) Stxbp1tm1d/+ and Stxbp1tm1a/+ male mice were crossed with WT female mice. Pie charts show the observed genotypes of the offspring at weaning (i.e., around the age of 3 weeks). Stxbp1tm1d/+ and Stxbp1tm1a/+ mice were significantly less than Mendelian expectations. (E) Stxbp1tm1d/+ and Stxbp1tm1a/+ mice were smaller and showed hindlimb clasping (arrows). (F) Body weights as a function of age. M, male; F, female. (G) The fraction of mice with hindlimb clasping as a function of age. Bar graphs are mean ± s.e.m. **, p<0.01; ***, p<0.001; ****, p<0.0001.

Figure 1—figure supplement 1
Generation of two new Stxbp1 null alleles.

(A) The Stxbp1 WT genomic region was targeted by a multipurpose cassette that contains an Engrailed two splice acceptor site (En2SA), an encephalomyocarditis virus internal ribosomal entry site (IRES), lacZ, SV40 polyadenylation element (pA), and floxed exon 7, resulting in the KO-first allele (tm1a). The restriction enzymes and probes used in the Southern blots are indicated in the diagrams. The KO-first allele was converted to the KO allele (tm1d) by crossing Stxbp1tm1a/+ mice with Rosa26-Flpo and Sox2-Cre mice sequentially. (B) Representative Western blots of Stxbp1 and Gapdh proteins extracted from the brains at embryonic day 17.5. Stxbp1 was detected by an antibody recognizing the C terminus (left two blots) or the N terminus (right two blots). The genotypes are indicated above the samples. Note that Stxbp1 was reduced in heterozygous mutants and absent in homozygous mutants. (C) Summary data of normalized Stxbp1 expression levels. Stxbp1 levels were first normalized by the Gapdh levels and then by the average Stxbp1 levels of all WT mice from the same blot. The data obtained by both Stxbp1 antibodies are combined. Each triangle represents one mouse. Bar graphs are mean ± s.e.m. **, p<0.01; ***, p<0.001; ****, p<0.0001.

Figure 1—figure supplement 2
Reduced survival of Stxbp1 haploinsufficient mice.

(A) Stxbp1tm1d/+ and Stxbp1tm1a/+ male mice were crossed with WT female mice. The observed genotypes of the offspring at weaning (i.e., around the age of 3 weeks) are shown in the pie charts. The male and female Stxbp1tm1d/+ and Stxbp1tm1a/+ mice were significantly less than Mendelian expectations. Note that the genotypes of some female mice were not determined and therefore, they were not included in this analysis. (B) Survival curves of a subset of Stxbp1tm1d/+, Stxbp1tm1a/+, and WT mice that were monitored for 80 weeks. The numbers of observed mice are indicated in the figures. n.s., p>0.05; *, p<0.05; **, p<0.01; ****, p<0.0001.

Figure 2 with 2 supplements
Motor dysfunctions of Stxbp1 haploinsufficient mice.

(A) Representative tracking plots of the mouse positions in the open-field test. Note that Stxbp1tm1d/+ and Stxbp1tm1a/+ mice traveled less in the center (dashed box) than WT mice. (B–E) Summary data showing hyperactivity and anxiety-like behaviors of Stxbp1tm1d/+ and Stxbp1tm1a/+ mice in the open-field test. Stxbp1tm1d/+ and Stxbp1tm1a/+ mice showed an increase in the total moving distance (B) and speed (C), and a decrease in the ratio of center moving distances over total moving distance (D) and vertical activity (E). (F–H) Stxbp1tm1d/+ and Stxbp1tm1a/+ mice had weaker forelimb grip strength (F), made more foot slips per travel distance on a wire grid (G), and took more time to get down from a vertical pole (H). The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.

Figure 2—figure supplement 1
Normal performance of Stxbp1tm1d/+ mice in rotarod, dowel, inverted screen, and wire hang tests.

(A) In the 2 day rotarod test, 6–7 week old Stxbp1tm1d/+ mice performed better than WT mice, as they were able to walk (left panel) and stay (right panel) on the rotating rod for longer time, probably due to their lower body weights or hyperactivity. (B,C) Similar to (A), but for the ages of 26–27 weeks (B) and 61–69 weeks (C). Stxbp1tm1d/+ mice performed similar to WT mice. (D) In the 4 day rotarod test, Stxbp1tm1d/+ mice performed similar to WT mice at the age of 10–11 weeks. (E,F) Stxbp1tm1d/+ mice could stay on the dowel (6.5- or 9.5 mm diameter) for similar amount of time as WT mice. (G,H) Stxbp1tm1d/+ mice could hang on the screen (G) or wire (H) for similar amount of time as WT mice. The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05; *, p<0.05; **, p<0.01.

Figure 2—figure supplement 2
Stxbp1 haploinsufficient mice have normal sensory functions.

(A) Stxbp1tm1d/+ and Stxbp1tm1a/+ mice showed similar acoustic startle responses as WT mice at different sound levels. (B) In the pre-pulse inhibition test, when a weak sound (74, 78, or 82 dB) preceded a loud sound (120 dB), Stxbp1tm1d/+ and Stxbp1tm1a/+ mice showed a similar reduction in the startle responses to the loud sound as WT mice. (C) In the hot plate test, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice showed similar latencies in response to the high temperature as WT mice. The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05.

Figure 3 with 1 supplement
Impaired cognition of Stxbp1 haploinsufficient mice.

(A) In the novel object recognition test with 24 hr testing intervals, the ability of a mouse to recognize the novel object was assessed by the preference index (see Materials and methods). On days 1, 2, 3, and 5, mice were presented with the same two identical objects. In contrast to WT mice, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice did not show a preference for the novel object on day 4 when they were presented with the familiar object and a novel object. (B–E) In the fear conditioning test, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice at two different ages showed a reduction in both context-induced (B,D) and cue-induced (C,E) freezing behaviors 24 hr after training. The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. ***, p<0.001; ****, p<0.0001.

Figure 3—figure supplement 1
Stxbp1 haploinsufficient mice show an impairment in object recognition and fear memory, but not working memory.

(A) In the novel object recognition test with 5 min testing intervals, Stxbp1tm1d/+ mice did not show a preference for the novel object on trial four when they were presented with the familiar and novel objects. (B) In the novel object recognition test with 24 hr testing intervals (same as Figure 3A), Stxbp1tm1d/+ mice spent less time interacting with the familiar and novel objects. (C,D) In the modified novel object recognition test with 24 hr testing intervals, Stxbp1tm1d/+ mice spent similar amount of time interacting with the familiar and novel objects as WT mice (C), but they still failed to show a preference for the novel object on day 4 (D). (E) Stxbp1tm1d/+ mice showed a reduction in both contextual (left panel) and cued (right panel) fear memories 1 hr after training. (F) In the Y maze spontaneous alternation test that evaluates working memory, Stxbp1tm1d/+ mice made similar numbers of choices (left panel) and alternations (right panel) as WT mice. The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05; *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.

Figure 4 with 1 supplement
Stxbp1 haploinsufficient mice show increased anxiety-like and repetitive behaviors.

(A,B) In the elevated plus maze test, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice entered the open arms less frequently (A) and traveled shorter distance in the open arms (B). (C,D) In the light-dark chamber test, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice made less transitions between the light and dark chambers (C) and traveled shorter distance in the light chamber (D). (E–G) In the hole-board test, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice poked similar numbers of holes as WT mice (E) and made similar or more total nose pokes (F). They made more repetitive nose pokes (i.e.,≥2 consecutive pokes) than WT mice across different holes (G). The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05; *, p<0.05; **, p<0.01; ***, p<0.001.

Figure 4—figure supplement 1
The movements of Stxbp1 haploinsufficient mice in elevated plus maze and light-dark chamber tests.

(A,B) In the elevated plus maze test, the total travel distances (A) and travel distances in the closed arms (B) of Stxbp1tm1d/+ and Stxbp1tm1a/+ mice were similar or slightly longer than those of WT mice. (C,D) In the light-dark chamber test, the total travel distances of Stxbp1tm1d/+ mice were reduced (C) due to the reduction of their travel distances in the light chamber and normal travel distances in the dark chamber (D). The total travel distances of Stxbp1tm1a/+ mice were normal (C) and their travel distances in the dark chamber was slightly increased as compared to WT mice (D). The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05; *, p<0.05; **, p<0.01.

Figure 5 with 1 supplement
Stxbp1 haploinsufficient mice show increased aggressive behaviors and reduced nest building and digging behaviors.

(A–C) In the resident-intruder test, male Stxbp1tm1d/+ and Stxbp1tm1a/+ mice showed a reduction in the latency to attack the male intruder mice (A). The total duration (B) and number (C) of their attacks were increased as compared to WT mice. (D) In the tube test, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice won more competitions against their WT littermates. (E) In the three-chamber test, Stxbp1tm1d/+ and Stxbp1tm1a/+ mice showed a preference in interacting with the partner mouse over the object. (F,G) Stxbp1tm1d/+ and Stxbp1tm1a/+ mice built poor quality nests. The quality of the nests was scored according to the criteria in (F) for three consecutive days (G). (H) Stxbp1tm1d/+ and Stxbp1tm1a/+ mice buried fewer marbles than WT mice. The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.

Figure 5—figure supplement 1
Stxbp1 haploinsufficient mice show normal social interactions.

(A) In the partition test, Stxbp1tm1d/+ and WT mice showed a similar preference for the novel partner mouse over the familiar partner mouse. The numbers and ages of tested mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.

Figure 6 with 1 supplement
Stxbp1tm1d/+ mice exhibit cortical hyperexcitability and epileptic seizures.

(A–D) Representative EEG traces of the left frontal cortex (L-FC), left somatosensory cortex (L-SC), and right somatosensory cortex (R-SC), and EMG traces of the neck muscle from WT (A,B) and Stxbp1tm1d/+ mice (C,D). Spike-wave discharges (SWDs, indicated by the blue arrows) occurred frequently and often in a cluster manner in Stxbp1tm1d/+ mice (see Video 1). The gray line-highlighted SWDs from WT and Stxbp1tm1d/+ mice were expanded to show the details of the oscillations (B,C). A long SWD (i.e.,>4 s) during REM sleep from a Stxbp1tm1d/+ mouse is shown in (D) (see Video 2). (E) Summary data showing the overall SWD frequency (left panel), duration (middle panel), and average spike rate (right panel). (F) The numbers of SWDs per hour in WT (left Y axis) and Stxbp1tm1d/+ (right Y axis) mice are plotted as a function of time of day and averaged over 3 days. (G) Video frames showing a myoclonic jump from a Stxbp1tm1d/+ mouse (see Video 3). The mouse was in REM sleep before the jump. (H) Representative EEG and EMG traces showing myoclonic jerks (indicated by the blue arrows) from a Stxbp1tm1d/+ mouse (see Video 4). Two episodes of myoclonic jerks highlighted by the gray lines were expanded to show that the EEG discharges occurred prior to (the first episode) or simultaneously with (the second episode) the EMG discharges. (I,J) Summary data showing the frequencies of two types of myoclonic seizures in different behavioral states. The numbers and ages of recorded mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.

Figure 6—figure supplement 1
The clustering of SWDs in Stxbp1tm1d/+ mice does not result from a random distribution of frequent SWD episodes.

(A–C) In Stxbp1tm1d/+ mice, many SWDs occurred in a cluster manner. A SWD cluster is defined as five or more episodes of SWDs that occur with an inter-episode-interval of 60 s or less. For each Stxbp1tm1d/+ mouse, simulations were performed to determine if the clustering of SWD episodes was simply due to the overall high frequencies of episodes. The recorded episodes of SWDs from a Stxbp1tm1d/+ mouse were randomly distributed in the same period of time for 106 times. The number of SWD clusters was determined from each simulated distribution, and the results of the 106 simulations are shown as the probability distribution of the number of SWD clusters for each mouse. The vertical lines with the same color as the probability distribution curves represent the numbers of the recorded SWD clusters in each mouse. The numbers of simulated SWD clusters are all smaller than that of recorded SWD clusters for each Stxbp1tm1d/+ mouse (p<10−6), demonstrating that a random distribution of the same number of SWD episodes does not result in the same clustering of SWDs in Stxbp1tm1d/+ mice.

Cortical neuron densities are unaltered in Stxbp1tm1d/+ mice.

(A) Representative fluorescent images of coronal sections stained by antibodies against NeuN (blue), Pv (green), and Sst (magenta). Note the similar cytoarchitecture between WT (upper panel) and Stxbp1tm1d/+ (lower panel) mice. (B) Representative fluorescent images of the somatosensory cortices within the boxed regions in (A) for WT (upper panels) and Stxbp1tm1d/+ (lower panels) mice. (C) Summary data showing similar densities of neurons (i.e., NeuN positive cells), Pv, and Sst interneurons in the somatosensory cortices of WT and Stxbp1tm1d/+ mice. (D) Summary data showing that the ratios of Pv and Sst interneurons to all somatosensory cortical neurons are similar between WT and Stxbp1tm1d/+ mice. The numbers and ages of mice are indicated in the figures. Each filled (male) or open (female) circle represents one mouse. Bar graphs are mean ± s.e.m. n.s., p>0.05.

Figure 8 with 2 supplements
Inhibitory synapses from Pv and Sst interneurons are differentially impaired in Stxbp1tm1d/+ mice.

(A) uIPSCs of a layer 2/3 pyramidal neuron (Vm = + 10 mV) in the somatosensory cortex (upper panels) evoked by a train of 10 Hz action potentials in a nearby Pv interneuron (lower panels) from WT and Stxbp1tm1d/+ mice. 50 individual traces (lighter color) and the average trace (darker color) are superimposed. Note smaller uIPSCs in the Stxbp1tm1d/+ neuron. (B) Unitary connectivity rates from Pv interneurons to pyramidal neurons were similar between WT (27 connections out of 33 pairs) and Stxbp1tm1d/+ (26 connections out of 32 pairs) mice. (C) Cumulative frequencies of uIPSC amplitudes evoked by the first action potentials in the trains (median: WT, 217.3 pA; Stxbp1tm1d/+, 127.1 pA). Inset, each filled (male) or open (female) circle represents the uIPSC amplitude of one synaptic connection. (D) uIPSC amplitudes during the trains of action potentials were normalized by the amplitudes of the first uIPSCs. Note the similar synaptic depression between WT and Stxbp1tm1d/+ neurons. (E–H) Similar to (A–D), but for Sst interneurons. Unitary connectivity rates from Sst interneurons to pyramidal neurons (F) in Stxbp1tm1d/+ mice (25 connections out of 36 pairs) were less than WT mice (34 connections out of 36 pairs). The uIPSC amplitudes evoked by the first action potentials in the trains (G, median: 83.5 pA and 68.0 pA, respectively) and synaptic depression (H) were similar between WT and Stxbp1tm1d/+ mice. The ages of mice are indicated in the figures. Bar graphs are mean ± s.e.m. n.s., p>0.05; *, p<0.05; **, p<0.01.

Figure 8—figure supplement 1
Intrinsic neuronal excitability of Stxbp1tm1d/+ mice is slightly increased.

(A) Membrane potentials (upper panels) in response to current injections (lower panels) in layer 2/3 pyramidal neurons of the somatosensory cortex from WT and Stxbp1tm1d/+ mice. (B–D) Summary data showing that Stxbp1tm1d/+ neurons had similar resting membrane potentials and rheobase currents as WT neurons, but their input resistances were 19% larger than WT neurons. The numbers and ages of recorded neurons are indicated in the figures. Each filled (male) or open (female) circle represents one neuron. Bar graphs are mean ± s.e.m. n.s., p>0.05; *, p<0.05.

Figure 8—figure supplement 2
Spontaneous excitatory inputs onto Pv and Sst interneurons are unaltered in Stxbp1tm1d/+ mice.

(A,B) sEPSCs of a Pv (A) or Sst (B) interneuron (Vm = –70 mV) in the layer 2/3 of the somatosensory cortex from WT or Stxbp1tm1d/+ mice. (C,D) Summary data showing that the frequency and amplitude of sEPSCs in Pv (C) and Sst (D) interneurons are similar between WT and Stxbp1tm1d/+ mice. The numbers and ages of recorded neurons are indicated in the figures. Each filled (male) or open (female) circle represents one neuron. Bar graphs are mean ± s.e.m. n.s., p>0.05.

Figure 9 with 1 supplement
Pv and Sst interneurons-mediated quantal IPSCs are isolated by a novel optogenetic method and are unaltered in Stxbp1tm1d/+ mice.

(A) Schematic of slice experiments in (B). ChR2 in Pv interneurons. (B) mIPSCs in a layer 2/3 pyramidal neuron (Vm = + 10 mV) from the somatosensory cortex of WT or Stxbp1tm1d/+ mice. The intensity of blue light is indicated above the mIPSC traces. Note the increase of mIPSC frequency during blue light stimulation. The quantal IPSC trace was computed by subtracting the average mIPSC trace of the baseline period from that of the light stimulation period (bottom row). (C,D) As in (A,B), but for ChR2 in Sst interneurons. (E,F) Summary data showing that the average amplitude, charge, and decay time constant of Pv (E) or Sst (F) interneuron-mediated quantal IPSCs are similar between WT and Stxbp1tm1d/+ mice. The numbers and ages of recorded neurons are indicated in the figures. Each filled (male) or open (female) circle represents one neuron. Bar graphs are mean ± s.e.m. n.s., p>0.05.

Figure 9—figure supplement 1
Ramping down blue light intensity minimizes the tonic currents during optogenetic activation of interneurons.

(A) Schematic of slice experiments in (B,C). ChR2 in Pv interneurons. (B,C) mIPSCs of the same layer 2/3 pyramidal neuron (Vm = + 10 mV) from the somatosensory cortex of a WT mouse. The intensity of blue light is indicated above the mIPSC traces. Note the increase of tonic currents (i.e., the increase of holding currents) caused by the constant blue light stimulation (B), which was reduced by gradually decreasing the blue light intensity (C).

Videos

Video 1
Stxbp1tm1d/+ mice show clusters of SWDs.

A representative video showing a SWD cluster in a Stxbp1tm1d/+ mouse. The top three traces are EEG signals from the left frontal cortex, right somatosensory cortex, and left somatosensory cortex. The bottom trace is the EMG signal from the neck muscle. The vertical line indicates the time of the current video frame. Note that the EEG signal from the left somatosensory cortex (the third channel) is inverted.

Video 2
Stxbp1tm1d/+ mice show long SWDs.

A representative video showing a long SWD during REM sleep in a Stxbp1tm1d/+ mouse. The top three traces are EEG signals from the left frontal cortex, right somatosensory cortex, and left somatosensory cortex. The bottom trace is the EMG signal from the neck muscle. The vertical line indicates the time of the current video frame. Note that the EEG signal from the left somatosensory cortex (the third channel) is inverted.

Video 3
Stxbp1tm1d/+ mice show myoclonic jumps.

A representative video showing a myoclonic jump of a Stxbp1tm1d/+ mouse. The top three traces are EEG signals from the left frontal cortex, right somatosensory cortex, and left somatosensory cortex. The bottom trace is the EMG signal from the neck muscle. The vertical line indicates the time of the current video frame. Note that the EEG signal from the left somatosensory cortex (the third channel) is inverted.

Video 4
Stxbp1tm1d/+ mice show myoclonic jerks.

A representative video showing a myoclonic jerk of a Stxbp1tm1d/+ mouse. The top three traces are EEG signals from the left frontal cortex, right somatosensory cortex, and left somatosensory cortex. The bottom trace is the EMG signal from the neck muscle. The vertical line indicates the time of the current video frame. Note that the EEG signal from the left somatosensory cortex (the third channel) is inverted.

Tables

Key resources table
Reagent type
(species) or resource
DesignationSource or referenceIdentifiersAdditional
information
Cell line
(M. musculus)
Stxbp1tm1a(EUCOMM)Hmgu embryonic stem cell clones (C57BL/6N strain)European Conditional Mouse Mutagenesis Program (EUCOMM)HEPD0510_5_A09, HEPD0510_5_B10
Genetic reagent (M. musculus)Stxbp1tm1a (C57BL/6J strain)This paper
Genetic reagent
(M. musculus)
Stxbp1tm1d (C57BL/6J strain)This paper
Genetic reagent (M. musculus)B6(Cg)-Tyrc-2J/JThe Jackson LaboratoryRRID:IMSR_JAX:000058
Genetic reagent
(M. musculus)
Rosa26-Flpo (C57BL/6J strain)The Jackson LaboratoryRRID:IMSR_JAX:012930
Genetic reagent (M. musculus)Sox2-Cre (C57BL/6J strain)The Jackson LaboratoryRRID:IMSR_JAX:008454
Genetic reagent (M. musculus)C57BL/6JThe Jackson LaboratoryRRID:IMSR_JAX:000664
Genetic reagent (M. musculus)Pv-ires-Cre (C57BL/6J strain)The Jackson
Laboratory
RRID:IMSR_JAX:017320
Genetic reagent (M. musculus)Sst-ires-Cre (C57BL/6;129S4 strain)The Jackson LaboratoryRRID:IMSR_JAX:013044
Genetic reagent (M. musculus)Rosa26-CAG-LSL-tdTomato (C57BL/6J strain)The Jackson
Laboratory
RRID:IMSR_JAX:007914
AntibodyRabbit anti-Munc18-1, polyclonalAbcam, catalog # ab3451RRID:AB_303813(1:2000 or 1:5,000)
AntibodyRabbit anti-Munc18-1, polyclonalSynaptic Systems, catalog # 116002RRID:AB_887736(1:2000 or 1:5,000)
AntibodyRabbit anti-Gapdh, polyclonalSanta Cruz Biotechnology, catalog #sc-25778RRID:AB_10167668(1:300 or 1:1,000)
AntibodyGoat anti-rabbit IgG
conjugated with IRDye 680LT, polyclonal
LI-COR Biosciences, catalog # 925–68021RRID:AB_2713919(1:20,000)
AntibodyRabbit anti-Somatostatin, polyclonalPeninsula Laboratories
International, catalog # T4103.0050
RRID:AB_518614(1:3,000)
AntibodyMouse anti-Parvalbumin, monoclonalEMD Millipore, catalog # MAB1572RRID:AB_2174013(1:1,000)
AntibodyGuinea pig anti-NeuN, polyclonalSigma Millipore, catalog # ABN90RRID:AB_11205592(1:1,000)
AntibodyGoat anti-guinea pig IgG (H+L) conjugated with
Alexa Flour 488, polyclonal
Invitrogen, catalog # A-11073RRID:AB_2534117(1:1,000)
AntibodyGoat anti-mouse IgG (H+L)
conjugated with Alexa Flour 555, polyclonal
Invitrogen, catalog # A-21424RRID:AB_141780(1:1,000)
AntibodyGoat anti-rabbit IgG (H+L) conjugated with Alexa Flour 647, polyclonalInvitrogen, catalog # A-21245RRID:AB_141775(1:1,000)
Recombinant DNA reagentpAAV-EF1α-DIO-hChR2(H134R)-P2A-EYFPThis paperAddgene: 139283This plasmid was used to produce the AAV vector used in Figure 9.
Transfected constructAAV9-EF1α-DIO-hChR2(H134R)-P2A-EYFPThis paper, Baylor College of Medicine Gene Vector CoreAddgene: 139283This AAV vector was used in Figure 9.
Software, algorithmAxograph X 1.5.4AxoGraphRRID:SCR_014284https://axograph.com
Software, algorithmpClamp 10.7Molecular DevicesRRID:SCR_011323https://www.moleculardevices.com
Software, algorithmImage Studio Lite 5.0LI-COR BiosciencesRRID:SCR_013715https://www.licor.com
Software, algorithmMATLAB R2015 to R2017MathWorksRRID:SCR_001622https://www.mathworks.com
Software, algorithmPrism 6.0, 7.0, and 8.0GraphPadRRID:SCR_002798https://www.graphpad.com
Software, algorithmSpyder 3.3.6 with AnacondaSpyderRRID:SCR_017585https://www.spyder-ide.org
Software, algorithmSirenia 1.7.2 to 1.8.3Pinnacle TechnologyRRID:SCR_016183https://www.pinnaclet.com
Software, algorithmImaris 9.2Oxford InstrumentsRRID:SCR_007370https://imaris.oxinst.com

Additional files

Supplementary file 1

EEG phenotypes of individual WT and Stxbp1tm1d/+ mice.

The parameters charactering the SWDs, myoclonic jerks, and myoclonic jumps of each WT and Stxbp1tm1d/+ mouse are presented in the table.

https://cdn.elifesciences.org/articles/48705/elife-48705-supp1-v2.xlsx
Supplementary file 2

Phenotypic comparison of human patients and mouse models.

The phenotyping tests in mouse models (the second column) are grouped based on the clinical features of STXBP1 encephalopathy (the first column). The results of phenotyping tests from different mouse models and studies are presented in the table.

https://cdn.elifesciences.org/articles/48705/elife-48705-supp2-v2.docx
Supplementary file 3

Statistics of experimental results.

The details of all statistical tests, numbers of replicates, and P values are presented for each experiment in the table.

https://cdn.elifesciences.org/articles/48705/elife-48705-supp3-v2.xlsx
Transparent reporting form
https://cdn.elifesciences.org/articles/48705/elife-48705-transrepform-v2.docx

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Wu Chen
  2. Zhao-Lin Cai
  3. Eugene S Chao
  4. Hongmei Chen
  5. Colleen M Longley
  6. Shuang Hao
  7. Hsiao-Tuan Chao
  8. Joo Hyun Kim
  9. Jessica E Messier
  10. Huda Y Zoghbi
  11. Jianrong Tang
  12. John W Swann
  13. Mingshan Xue
(2020)
Stxbp1/Munc18-1 haploinsufficiency impairs inhibition and mediates key neurological features of STXBP1 encephalopathy
eLife 9:e48705.
https://doi.org/10.7554/eLife.48705