Abstract
Experience- and activity-dependent transcription is a candidate mechanism to mediate development and refinement of specific cortical circuits. Here, we demonstrate that the activity-dependent transcription factor myocyte enhancer factor 2C (MEF2C) is required in both presynaptic layer (L) 4 and postsynaptic L2/3 mouse (male and female) somatosensory (S1) cortical neurons for development of this specific synaptic connection. While postsynaptic deletion of Mef2c weakens L4 synaptic inputs, it has no effect on inputs from local L2/3, contralateral S1, or the ipsilateral frontal/motor cortex. Similarly, homozygous or heterozygous deletion of Mef2c in presynaptic L4 neurons weakens L4 to L2/3 excitatory synaptic inputs by decreasing presynaptic release probability. Postsynaptic MEF2C is specifically required during an early postnatal, experience-dependent, period for L4 to L2/3 synapse function, and expression of transcriptionally active MEF2C (MEF2C-VP16) rescues weak L4 to L2/3 synaptic strength in sensory-deprived mice. Together, these results suggest that experience- and/or activity-dependent transcriptional activation of MEF2C promotes development of L4 to L2/3 synapses. Additionally, MEF2C regulates the expression of many pre- and postsynaptic genes in postnatal cortical neurons. Interestingly, MEF2C was necessary for activity-dependent expression of many presynaptic genes, including those that function in transsynaptic adhesion and neurotransmitter release. This work provides mechanistic insight into the experience-dependent development of specific cortical circuits.
Significance Statement
Experience-driven neuronal activity is necessary for development of synaptic connectivity of specific cortical circuits. Here, we demonstrate that the activity-dependent transcription factor myocyte enhancer factor 2C (MEF2C) is necessary for development of a specific synaptic connection between layer (L) 4 and L2/3 neurons in the mouse somatosensory cortex. MEF2C is required in both presynaptic L4 and postsynaptic L2/3 neurons during an early postnatal and experience-dependent period for development of their connection. Our results suggest that sensory experience drives transcriptional activation of MEF2C to promote development of the L4 to L2/3 synaptic connection. Additionally, we identify activity-dependent, MEF2C-regulated presynaptic genes that promote development of specific connections. This work provides insight into the mechanisms by which sensory experience determines development of cortical circuit connectivity.
Introduction
Sensory experience drives and refines development and connectivity of neocortical circuits (Katz and Shatz, 1996; Feldman and Brecht, 2005). For example, in the rodent primary somatosensory “barrel” cortex (S1), whisker experience is necessary during a critical postnatal period for development of the synaptic connection between layer (L) 4, the primary thalamic recipient layer, and L2/3 neurons, which integrate sensory input. Sensory deprivation by whisker trimming, from postnatal day (P) 10 to P14, results in weak L4 to L2/3 synaptic inputs and weak whisker-evoked sensory responses in L2/3 (Fox, 1992; Stern et al., 2001). Conversely, single whisker experience, induced by trimming adjacent whiskers during this time window, rapidly strengthens whisker-evoked responses and L4 synaptic inputs onto L2/3 neurons from the experienced, but not deprived, barrel column (Glazewski and Fox, 1996; Clem and Barth, 2006; Wen and Barth, 2011).
A candidate molecule to mediate experience-dependent cortical synapse development and refinement is myocyte enhancer factor 2C (MEF2C). MEF2C belongs to the MEF2 (A-D) family of activity-dependent transcription factors and is enriched in L4 and L2/3 neurons during early postnatal ages through adulthood (G. E. Lyons et al., 1995; Allen Brain Atlas). MEF2 acts as a transcriptional activator or repressor depending on neuronal activity and its interactions with cofactors (A. K. Shalizi and Bonni, 2005; Potthoff and Olson, 2007). Neuronal depolarization and Ca2+ influx stimulate posttranslational modifications of MEF2 and cofactors to promote transcription of target genes (A. K. Shalizi and Bonni, 2005; Flavell et al., 2006; A. Shalizi et al., 2006). MEF2 transcriptional activity is also regulated by experience, including sleep, learning, and drugs of abuse (Flavell et al., 2008; Pulipparacharuvil et al., 2008; Cole et al., 2012; Bjorness et al., 2020). Previous studies in the hippocampus and cerebellum implicate postsynaptic MEF2A/C/D in activity-induced elimination of excitatory synapses (Flavell et al., 2006; A. Shalizi et al., 2006; Barbosa et al., 2008; Pfeiffer et al., 2010; Chang et al., 2017). Very little is known about the function of presynaptic MEF2 and its role in activity-dependent synapse development or strengthening (Yamada et al., 2013).
In the neocortex, we demonstrated that MEF2C differentially regulates excitatory synaptic inputs depending on their origin. Postnatal deletion of Mef2c in L2/3 neurons in S1 weakens synaptic inputs from L4 but strengthens inputs from contralateral S1 (Rajkovich et al., 2017). Furthermore, sensory deprivation by whisker trimming prevents or occludes the effects of Mef2c deletion on inputs from both L4 and contralateral S1 (Rajkovich et al., 2017). From these results, we hypothesized that sensory experience drives MEF2C-dependent transcriptional activation in L2/3 neurons to selectively promote the development and strengthening of inputs from L4. Importantly, loss-of-function mutations in MEF2C are associated with neurodevelopmental disorders in humans, including MEF2C haploinsufficiency syndrome (MCHS) characterized by intellectual disability, autism, and seizures (Mitchell et al., 2018; Assali et al., 2019). Conversely, MEF2C transcriptional activation is associated with cognitive function and resilience with aging (Barker et al., 2021). Studies support similar roles for MEF2C in mice (Assali et al., 2019; Fahey et al., 2023). Therefore, the study of MEF2C regulation of synaptic connectivity across the lifespan will be relevant to understanding human cognitive function and disease.
Here, we demonstrate that MEF2C is required in both presynaptic L4 and postsynaptic L2/3 neurons for experience-dependent strengthening of their synaptic connections. In contrast, postsynaptic MEF2C is not required for development of long-range inputs from ipsilateral or contralateral cortices. Postnatal expression of a transcriptionally active MEF2C in L2/3 neurons rescues weak L4 to L2/3 synaptic inputs in sensory-deprived mice. This result supports a model where experience drives transcriptional activation of MEF2C target genes that specifically strengthen L4 to L2/3 inputs. To identify MEF2C- and activity-dependent genes, we performed RNA-seq in cortical slice cultures from mice with Mef2c deletion in excitatory neurons. MEF2C bidirectionally regulates many pre- and postsynaptic genes. Interestingly, MEF2C is required for activity-dependent expression of presynaptic genes that promote synapse development and function. Our results provide knowledge of activity-dependent mechanisms that mediate development of specific synaptic connections.
Materials and Methods
Animals
Conditional Mef2c (Mef2cfl/fl; RRID:MGI:3719006; Arnold et al., 2007) mice were obtained from Dr. Eric Olson (University of Texas Southwestern Medical Center), and CaMKII-iCre (BAC; Casanova et al., 2001) mice were obtained from Dr. Joe Takahashi (University of Texas Southwestern Medical Center). Ai14-tdTomato Cre-reporter mice (RRID:IMSR_JAX:007914) and Scnn1a-Tg3-Cre mice (RRID:IMSR_JAX:009613) were obtained from the Jackson Laboratory. All mice were maintained on a C57/B6J strain. Animals were given ad libitum access to food and water and were reared on a 12 h light/dark cycle. All animal experiments were conducted in accordance with the Institutional Animal Care and Use Committee at the University of Texas Southwestern Medical Center.
Virus injections
To sparsely delete Mef2c in postsynaptic L2/3 neurons, AAV9.eGFP-Cre (Addgene #105545) was injected into the lateral ventricle of postnatal day (P) 1–2 Mef2cfl/fl mice as described previously (Rajkovich et al., 2017). Unless otherwise indicated, all virus injections were administered at a volume, per mouse, of 0.4–0.6 µl of a ∼1012 vg/ml titer virus diluted in saline. To label long-range axons, AAV9.ChR2-mCherry (Addgene #100054) was delivered to superficial layers (0.3–0.5 mm underneath the skull) of the somatosensory cortex in the right hemisphere for callosal projections as described previously (Rajkovich et al., 2017; Zhang et al., 2021) or the prefrontal cortex in the left hemisphere, immediately posterior to the coronal suture and ∼1 mM lateral to the sagittal suture, for ipsilateral motor cortex projections. For paired-pulse experiments, Scnn1a-Cre mice were bilaterally injected with 0.7–0.8 µl of AAV9.EF1a-double floxed-hChR2(H134R)-mCherry-WPRE-HGHpA (Addgene #20297-AAV9; titer, ∼1013 vg/ml) into each lateral ventricle. AAV-MEF2C-VP16 bi-GFP vector was a generous gift from Dr. Chris Cowan (Pulipparacharuvil et al., 2008) and was packaged into AAV9 at the Baylor College of Medicine Gene Vector Core.
For juvenile cortical injection, P14 pups were anesthetized using an isoflurane–oxygen mixture and fixed on a stereotaxic frame with nonrupture cuff ear bars. A small hole was made on the exposed skull using a micro drill with 0.5 mm tip diameter burrs. The coordinate used to target S1 was 0.5–0.8 posterior and 2.9–0.3 lateral from bregma (Mao et al., 2011). AAV9.eGFP-Cre was delivered through a pulled glass pipette using a syringe pump at a speed of 100 nl/min into the cortex (0.4–0.7 mm deep under pia).
Acute cortical slices
Slices were prepared as described (Rajkovich et al., 2017). Briefly, P18–P32 mice, male and female, were anesthetized by intraperitoneal injection of ketamine/xylazine until unresponsive to a toe pinch and then decapitated. Mice older than postnatal day (P) 25 were transcardially perfused with cold dissection buffer prior to decapitation. Acute coronal slices (300 µm thickness) of somatosensory barrel cortex were sliced using a Leica VT1200S Vibratome. Brains were fixed to tissue block and submerged in ice-cold dissection buffer containing the following (in mM): 110 choline chloride, 25 NaHCO3, 25 d-glucose, 11.6 ascorbic acid, 2.5 KCl, 1.25 Na2H2PO4, 3.1 Na pyruvate, 1 kynurenate, 7 MgCl2, and 0.5 CaCl2, continuously aerated with 95% CO2/5% O2. After slicing, brain slices were transferred to a recovery chamber of aerated artificial cerebrospinal fluid (ACSF) containing the following (in mM): 125 NaCl, 25 NaHCO3, 10 d-glucose, 2.5 KCl, 1.25 NaH2PO4, 2 MgCl2, and 2 CaCl2, for 30 min at 37°C followed by 30 min at room temperature (RT).
Electrophysiology
Recordings were performed at RT submerged and perfused with oxygenated ACSF (1–2 ml/min) as described previously (Rajkovich et al., 2017; Zhang et al., 2021). Slices were visualized by infrared differential interference contrast (IR-DIC) optics (Fig. 1B; Olympus BX51W1). Whole-cell recordings of L2/3 pyramidal neurons were achieved using borosilicate pipettes (5–8 MΩ) and MultiClamp 700A amplifier (Molecular Devices). Pipette internal solution contained the following (in mM): 130 K-gluconate, 10 HEPES, 6 KCl, 3 NaCl, 0.2 EGTA, 14 phosphocreatine-Tris, 4 Mg-ATP, and 0.4 Na-GTP. Unless stated otherwise, all recordings were conducted in a voltage clamp, holding at −70 mV, and collected using custom LabVIEW programs (LabVIEW 8.6, National Instruments). L2/3 pyramidal neurons were identified by their laminar location, apical dendrites, and burst spiking patterns in response to depolarizing current injection. For experiments performed in TTX, rise time to hyperpolarizing current (>50 ms) was used as a criterion (Povysheva et al., 2006). Only L2/3 neurons located directly above L4 barrels (i.e., within the home column) were included for analysis; neurons located within barrel septa were discarded. Excitatory neurons with resting membrane potential <−50 mV and series resistance <35 MΩ were included in the analysis. Voltages were not corrected for junction potential. For simultaneous patch-clamp recordings, the distance between the pair of cells (center-to-center) was 10–40 µm. Recordings were conducted in brain slices with <10% local virus infection efficiency to maintain sparse AAV-Cre-GFP infection.
Table 1-1
Intrinsic properties of neurons used for experiments in Figures 1-5. Download Table 1-1, DOCX file.
Table 1-2
Frequency vs current injection data for Figures 1 and 3. Download Table 1-2, DOCX file.
Optogenetic stimulation of long-range axonal inputs
P0–P1 Mef2cfl/fl mice received a unilateral ventricular injection of AAV9.eGFP-Cre. At the same time, AAV9.ChR2-mCherry was injected into the contralateral S1 cortex to label callosal projections as described (Zhang et al., 2021). For labeling of prefrontal/motor cortex inputs, at P3 AAV9.ChR2- mCherry was injected into the prefrontal cortex ipsilateral to the AAV9.eGFP-Cre injection at P0–P1. This delay was to minimize the spreading of ChR2 virus and optimize viral expression (Fig. 1H). The fluorescence of GFP-positive soma and mCherry-labeled axons was visualized using a fluorescent mercury lamp (Excelitas Technologies). Only slices containing clearly labeled mCherry-positive axons were used for recording. Slices with somatic infection of ChR2-mCherry due to virus spillover were discarded to avoid contamination from local inputs. The infection quantity of recorded Cre-GFP–positive neurons in the barrel cortex was ∼3–5% (Zhang et al., 2021). Only neurons residing in a cortical area with densely labeled ChR2-mCherry(+) axons were subject to recording to achieve a reliable magnitude and reduced variability of light-induced responses. For all LED experiments, responses were evoked by a 2 ms flash from a digitally controlled blue LED (final beam diameter, 350 µm; power, 0.1–4.6 mW; wavelength, 470 nm; M470L4-C1, Thorlabs) through a 40×water-immersed objective. The LED flash was centered on the soma and proximal apical dendrites of recorded neurons. To measure LED-evoked EPSCs, each cell (or cell pair) was stimulated 3–10 times with 20–30 s intervals. LED power was adjusted to obtain an EPSC amplitude of 100–1,000 pA in WT neurons (for cell pairs). The external solution for both LED and subcellular channelrhodopsin-assisted circuit mapping (sCRACM) experiments contained ACSF with 1 µM TTX, 100 µM 4-aminopyridine (4-AP), 10 µM (±)-3-(2-carboxypiperazin-4-yl)propyl-1-phosphonic acid (CPP), and 100 µM picrotoxin to isolate monosynaptic AMPAR-mediated excitatory inputs. Callosal input strength was measured as the peak amplitude of an average EPSC of 3–10 evoked EPSCs for each neuron.
Subcellular channelrhodopsin-assisted circuit mapping (sCRACM)
The microscope objective was set to 4× to visualize a broad area. ChR2-expressing axons were stimulated by a blue laser (1 ms; wavelength, 473 nm; power range, 0.7–16 mW; final beam diameter, 25 µm; CrystaLaser) scanning across a 12 × 12 grid (50 µm spacing) in a pseudorandom order to avoid repeated activation of neighboring locations. The grid was aligned along the pia and centered in the mediolateral position on the recorded somas. Each cell was stimulated within the grid 2–4 times at 40 s intervals, and the laser-evoked EPSCs were collected at each location. Most neuron pairs (>90%) were homogeneously distributed between 150 and 300 µm from the pia surface.
Laser scanning photostimulation with glutamate uncaging
Laser scanning photostimulation (LSPS) experiments were conducted as described previously (Rajkovich et al., 2017; Zhang et al., 2021). Acute coronal brain slices with L2/3 apical dendrites parallel to the slice surface were used to preserve the planar barrel cortical geometry of cross-layer synaptic pathways spanning at least three barrel columns of the primary somatosensory cortex. ACSF perfused during experiments included high-divalent cations (4 mM of both MgCl2 and CaCl2) and the NMDA receptor antagonist CPP (10 µM) to reduce polysynaptic local circuit activity and 4-methoxy-7-nitroindolinyl-caged-L-glutamate (MNI glutamate, Hello Bio, HB0423; 0.2–0.3 mM). Once an L2/3 neuron was patched in whole-cell, a 1 ms, 2 Hz UV laser flash (wavelength, 355 nm; power range, 30–40 mW; final beam diameter, 20 μm; DPSS Lasers) was delivered at individual points within a 16 × 16 grid (50 × 60 μm x–y spacing) in a pseudorandom order, and elicited EPSCs were recorded. The grid was aligned along the pia surface at low-power 4×magnification, centered mediolaterally with the soma location in L2/3. Each neuron included in the analysis consisted of 2–4 maps, scanned sequentially with a 40 s interval between maps. An IR-DIC image of a brain slice with an in-place patch pipette was acquired with stimulation grid overlay prior to LSPS to mark the soma location and reference slice anatomy (i.e., barrels).
LSPS analysis
A single average EPSC map was calculated from acquired LSPS maps for each neuron using LabVIEW software, where at each stimulation point on the grid the averaged UV laser-evoked area of the EPSC was measured within a 5–80 ms time window immediately following the laser pulse. A response was removed and not included in the map average when “direct” and nonsynaptic due to being elicited within 5 ms of LSPS and glutamate uncaging onto the recorded neuron. For responses with the majority of monosynaptic transmission but minor contamination from “direct” activation, the “direct” response component was subtracted from the EPSC trace by fitting a double-exponential decay equation. A color map for each neuron depicting the average EPSC area was generated. Within each genotype, all individual color maps were combined through spatial alignment with focus on the center of the L4 “home” barrel directly under the recorded L2/3 neurons. Individual color maps were superimposed by transposing each map such that the L4 home barrel center was located at the origin of the alignment grid, the mediolateral orientation of the brain slice was preserved, and the home barrel in x and y dimensions was stretched to normalize the barrel size. Pixel size for final average genotype color maps was halved through pixel interpolation (25 × 30 μm) to provide higher spatial resolution for soma alignment. The black pixels within an averaged color map indicate deleted “direct” responses or pixels that did not meet the minimum sampling threshold (minimum of 60% of neurons per stimulation point). Analysis of L4 home barrel responses was calculated by exporting the average responses around the origin in a 200 × 200 μm area encompassing the L4 barrel. Adjacent L4 home barrel responses were calculated using the same 200 × 200 μm area measurements but by averaging the two-barrel regions on the left and right sides of the home barrel. L2/3 and L5 responses were calculated using a 200 µm horizontal section across the relevant layer, and average EPSCs were reported. Average region-specific analyses for each neuron are represented in the averaged color maps and quantified and compared by genotype in bar graphs. CaMKII-Mef2c and Scnn1a-Mef2c LSPS experiments compared cells across animals, and statistics done were one-way ANOVA with multiple comparisons to Cre(−) controls. Experiments with paired-cell recordings (KO and neighboring untransfected or Cre(−) neurons were compared within animals, and statistics calculated were paired t tests.
Loose-seal cell-attached patch LSPS
Acute brain slices were prepared as described above for electrophysiology. High-divalent (4 mM of both MgCl2 and CaCl2) ACSF was similarly used to perfuse slices during recording and was used as the internal solution for patching pipettes. A loose-seal patch was obtained on an L4 excitatory neuron within a barrel, with a seal resistance of between 30 and 250 MΩ. The loose seal was maintained in current-clamp mode with no holding current. LSPS stimulation and glutamate uncaging were used to elicit action potentials (AP) from direct uncaging of glutamate onto the L4 neuron. The same LSPS laser settings and glutamate concentration were used for synaptic mapping (above), however with an 8 × 8 µm grid with 50 × 60 µm spacing. Most spiking occurred immediately surrounding the soma location. For each neuron, 2–3 maps were collected. The number of spikes per map was quantified for each map, averaged per neuron, and combined per genotype.
Optogenetic paired-pulse stimulation of layer 4 neurons
P1 mice expressing L4 Scnn1a-Tg3-Cre either on a Mef2cfl/fl, Mef2cfl/+, or WT background underwent bilateral intraventricular injection of AAV9-EF1a-double floxed-hChR2(H134R)-mCherry-WPRE-HGHpA as described above. Fluorescence of mCherry-positive L4 S1 barrels was visualized in acute coronal slices using a fluorescent mercury lamp. Slices containing at least two brightly fluorescent barrels with >50% of fluorescent L4 neurons were used for experiments. Whole-cell recordings from L2/3 neurons above mCherry(+) barrels were obtained. For evoked responses from ChR2-containing L4 neurons, slices were stimulated with paired pulses (50–600 ms) of 5, 10, or 15 ms blue LED flash through a 4× objective focused and centered over L2/3 and L4 (final beam diameter, 6 mm; power, 3.5–10.3 mW; wavelength, 470 nm; M470L4-C1, Thorlabs). TTX and 4-AP were included in recording ACSF (1 and 100 µm, respectively) as described to isolate EPSCs evoked from ChR2-expressing L4 axons (Petreanu et al., 2007). The strength and duration of LED pulse were adjusted to evoke an EPSC to the first LED pulse with an amplitude between 50 and 600 pA. For each cell, five responses at each interpulse interval were averaged. For each average trace, paired-pulse ratios were calculated as the ratio of peak amplitudes of the second pulse to the first pulse.
Statistics: electrophysiology
All statistical tests and graphs were performed and generated using GraphPad Prism 8 (GraphPad Software). The normality of data was determined with normality tests in GraphPad Prism. Equivalent nonparametric statistical tests were used to compare non-normally distributed data. Experiments with paired-cell recordings [KO and neighboring untransfected or Cre(−) neurons were compared within animals, and statistics calculated were paired t tests]. Data comparing control Cre(−), Het, and KO conditions were analyzed with one-way ANOVAs and Dunnett's multiple-comparison tests, unless stated otherwise. Datasets with two conditions were analyzed with t tests. In all figures, *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.
Slice culture activity paradigm
Organotypic cortical slice cultures were prepared as described previously (Pfeiffer et al., 2010) from P6 Cre(−) Mef2cfl/fl or CaMKII-Cre(+)-Mef2cfl/fl (CamKII-Mef2c KO) mice littermates. At 8 d in vitro (DIV), slices were treated with slice culture media containing either 1% DMSO and 1% ddH2O (vehicle; basal activity) or 100 μM picrotoxin and 100 μM 4-AP (treated; high activity). For qPCR validation of immediate-early gene induction experiments, only Cre(−) cultures were used. A biological replicate was one culture made from one mouse. Three biological replicates per condition per genotype were used for RNA-seq libraries.
Quantitative RT-PCR of activity-dependent genes
RNA was extracted from flash-frozen slice culture samples from CaMKII-Cre(−) mice using the Macherey–Nagel NucleoSpin RNA Plus Kit (catalog #740984). RNA concentration was quantified using a NanoDrop, and cDNA was synthesized using an equal volume of the total RNA and the High-Capacity cDNA Reverse Transcription Kit from Applied Biosystems (catalog #4368814). Quantitative real-time PCR (qRT-PCR) was conducted using Applied Biosystems QuantStudio 6. For qPCR primers, see Extended Data Table 7-1. Expression levels are based on the ΔΔCt with normalization to Gapdh.
cDNA library preparation
After treatment, slice culture samples were flash-frozen. RNA was extracted using the Macherey–Nagel NucleoSpin RNA Plus Kit (catalog #740984). RNA quantification was conducted using Qubit, and RNA integrity was validated using the Agilent TapeStation (Agilent, catalog #5067-5579, catalog #5067-5581, catalog #5067-5580). We prepared 150 bp long pair-end cDNA libraries using the NEB kit for positive selection of poly(A) mRNA (catalog #E7765S, catalog #7490S, catalog #E6440S). cDNA quality was assessed using the Agilent TapeStation (catalog #5067-5584 and catalog #5067-5585), and DNA quantification was conducted using Qubit. All the samples were randomized to reduce batch effects and submitted to the Next Generation Sequencing Core at the University of Texas Southwestern Medical Center sequenced using a NextSeq 2000 giving a total of 80–100 million reads per sample.
Read mapping and differential gene expression analysis
Raw BCL files were demultiplexed into fastq files. Using FastQC, basic statistics for percent duplicates, GC content, and read count were collected for each biological sample (Andrews, 2010). The data were trimmed using Trimmomatic (Bolger et al., 2014), aligned to the mm10 genome using Spliced Transcripts Alignment to a Reference (Dobin et al., 2012). PCA and hierarchical clustering were performed to detect outliers. Differential gene expression analysis was performed using DESeq2 (Anders et al., 2014). DESeq2 analysis was conducted with raw counts per million (CPM) values, normalizing both Cre(−) and KO “Activity” treatment slices to their respective Cre(−) and KO “Basal” controls. Differentially expressed genes (DEGs) were identified using a log2 fold change (FC) cutoff of ±0.3 and an adjusted p-value of <0.05. Four comparisons were conducted using DESeq.
Gene ontology analysis
Gene ontology (GO) analysis was conducted on all lists of DEGs and unique and shared gene sets between groups using ToppGene (https://toppgene.cchmc.org/; J. Chen et al., 2009) and SynGO (https://syngoportal.org/; Koopmans et al., 2019). GO categories were considered significant if they contained at least three genes and if they had a Benjamini–Hochberg (B–H)-corrected p-value of <0.05.
DEG enrichment analysis with other datasets
The SuperExactTest R package (Wang et al., 2015) was used to assess the significant overlap between groups of DEGs from our dataset and other groups of genes. The datasets used were a single-cell dataset from the S1 cortex (Zeisel et al., 2015), neurodevelopmental disorder gene sets (https://gene.sfari.org/; Gandal et al., 2018; Zhou et al., 2023), and genes related to cognitive resilience (Barker et al., 2021). Gene overlap figures were obtained by inputting lists of DEGs for groups of interest into Venny. Counts per million (CPM) for genes of interest were normalized to the average Cre(−) basal CPM values and plotted into a heatmap using the dplyr R package (Wickham et al., 2023).
Synaptosomes and cortical lysates
Synaptosomes were prepared as described previously (Mendoza et al., 2022). Briefly, slice culture samples were homogenized in 0.3 ml homogenization buffer {320 mM sucrose, 5 mM sodium pyrophosphate, 1 mM EDTA, 10 mM HEPES, pH 7.4, phosphatase inhibitors [including 200 nM okadaic acid, containing protease inhibitor mixture (Sigma-Aldrich P8340)], and phosphatase inhibitor mixtures 2 and 3 [Sigma-Aldrich P5726 and P0040, respectively]} by passage through a 26 g needle, 12 times. P1 pellets and crude membrane/synaptosomes were obtained through sequential centrifugation. The pellets were then lysed in lysis buffer [50 mm Tris, pH 7.4, 120 mm NaCl, 1 mM EDTA and 1.0% SDS and 1.0% sodium deoxycholate, containing protease inhibitor mixture (Sigma-Aldrich P8340) and phosphatase inhibitor mixtures 2 and 3 (Sigma-Aldrich P5726 and P0040, respectively)].
Whole-cell lysates of the S1 cortex were prepared from P18 CaMKII-Mef2c KO or Cre(−) mice in a lysis buffer. Samples were further homogenized using brief (1–2 s) pulses of sonication until lysates were clear. Lysates were then centrifuged at 10,000 × g for 10 min at 4°C to remove insoluble material.
Western blots
Protein concentration was determined using a BCA Protein Assay Kit (Pierce). Aliquots (5–15 μg) were run on SDS–PAGE gels and transferred to a PVDF membrane. After blocking with 5% BSA or 5% milk in 1× TBS, 0.05% Tween 20 for 1 h, membranes were incubated with the following primary antibodies in blocking buffer overnight at 4°C: FLRT2 (AF2877-SP, Proteintech), SLIT2 (20217-1-AP, Proteintech), synapsin 1 (SYN1; 106011, Synaptic Systems), and LRRTM1 (AF4897-S, R&D Systems). Following incubation in primary antibody overnight, immunoblots were incubated with HRP-conjugated secondary antibodies (Jackson ImmunoResearch) in 5% milk in 1× TBS, 0.05% Tween 20 for 1 h at room temperature. Membranes were washed and developed using the SuperSignal West Pico PLUS Chemiluminescent Substrate (34580, Thermo Fisher Scientific). Signal was acquired using Chemidoc MP (Bio-Rad) and analyzed using Image Lab (Bio-Rad). Membranes were stained with No-Stain (Thermo Fisher Scientific, catalog #A44449) for visualization and quantitation of total protein. Fold change (FC) of expression between genotypes was determined by normalizing experimental protein expression to total protein and then normalizing the KO and control [Cre(−)] samples to the average normalized value from all Cre(−) samples from that blot. Pairwise or ANOVA statistical tests were run to determine significant differences in quantified protein expression levels.
Results
Early postnatal cell-autonomous and postsynaptic deletion of Mef2c selectively weakens L4 to L2/3 synaptic inputs
We previously reported that early postnatal deletion of Mef2c from postsynaptic L2/3 neurons weakened excitatory synaptic inputs from local columnar circuits but not inputs from the contralateral barrel cortex (S1; Rajkovich et al., 2017). Here, we set out to determine if MEF2C generally promotes local synaptic inputs but not inputs from long-range projections, including those from the ipsilateral cortex. To address this question, we deleted Mef2c in a sparse population (∼3–5%) of early postnatal S1 L2/3 neurons by injecting an AAV expressing a GFP-tagged Cre (AAV9.GFP-Cre) into the left lateral ventricle of postnatal day (P) 1 Mef2c homozygous floxed (Mef2cfl/fl) mouse pups (Fig. 1A; Kim et al., 2013). We first confirmed that Mef2c deletion in L2/3 neurons weakened excitatory synaptic inputs from local columnar cortical circuits. To do this, we prepared acute coronal slices of the S1 cortex from the AAV-injected hemisphere of Mef2cfl/fl mice at P18–P25 and performed simultaneous whole-cell patch-clamp recordings from pairs of GFP-Cre(+) (“Mef2c KO”) and neighboring GFP-Cre(−) or “Cre(−)” L2/3 neurons (Fig. 1B). We then mapped the strength of local excitatory synaptic inputs onto recorded cells using laser scanning photostimulation (LSPS) with glutamate uncaging as we have described previously (Rajkovich et al., 2017; Zhang et al., 2021; Fig. 1B). Briefly, slices were bathed in MNI-caged glutamate and stimulated with pseudorandom flashes of a UV laser beam at individual locations within a 16-by-16 grid spanning L1–L5. Laser stimulation focally released glutamate in the slice to evoke action potentials and synaptic transmission at that location. Synaptic transmission was measured through EPSCs recorded from an L2/3 neuron as each location on the map was stimulated. Amplitudes of monosynaptic EPSCs were converted into a color-coded map representing the strength of synaptic inputs from each location, and individual maps were aligned based on the position of the L4 “home,” or within column, barrel to generate an average map for each genotype (Fig. 1C). Responses from direct glutamate activation onto recorded neurons, as described in methods, were excluded from analysis and represented by black pixels. Compared to neighboring Cre(−) neurons, Mef2c KO neurons had a ∼50% decrease in the amplitude of EPSCs evoked from the L4 home barrel and from adjacent barrels in L4, consistent with previous findings (Rajkovich et al., 2017). EPSCs evoked from horizontal L2/3 or from L5 were not different between Cre(−) and Mef2c KO neurons (Fig. 1D). Additionally, Mef2c KO neurons did not differ from neighboring Cre(−) neurons with respect to resting membrane potential, input resistance or intrinsic excitability (Extended Data Tables 1-1, 1-2). In contrast to evoked EPSCs, the frequency of spontaneous miniature EPSCs (mEPSCs) measured in the presence of TTX was increased in Mef2c KO L2/3 neurons [Cre(−), 0.51 ± 0.03 Hz; Mef2c KO, 0.87 ± 0.07 Hz; p < 0.0001; paired t test], as previously shown (Rajkovich et al., 2017); and mEPSC amplitude was unchanged [Cre(−), 15.64 ± 0.42 pA; Mef2c KO, 16.08 ± 0.51 pA; n.s.]. This result indicates that MEF2C in postsynaptic L2/3 neurons selectively promotes evoked excitatory synaptic transmission from L4 neurons.
To measure synaptic inputs from long-range projections, such as the contralateral S1, we performed channelrhodopsin-assisted circuit mapping (CRACM) of labeled callosal axons. Mef2cfl/fl mice were injected at P1 with AAV expressing mCherry-tagged channelrhodopsin into the right S1 and AAV9.GFP-Cre into the left lateral ventricle to sparsely delete Mef2c in the left S1 (Fig. 1E). At P18–P25, pairs of Cre(−) and Cre(+) Mef2c KO L2/3 pyramidal neurons surrounded by strong, visible ChR2-mCherry–labeled callosal axons were recorded in slices of the left S1 (Fig. 1F). Blue LED flashes centered over recorded somas evoked monosynaptic EPSCs directly from callosal axon terminals in the presence of tetrodotoxin and 4-AP (Petreanu et al., 2009; Zhang et al., 2021). Mef2c KO neurons had a trending increase in the strength of callosal synaptic inputs, as measured by EPSC amplitudes and compared with neighboring Cre(−) neurons (Fig. 1G; p = 0.08; n = 41 cell pairs). To determine if there is a callosal synaptic strength difference localized within a specific subcellular region, we mapped the spatial distribution of callosal synaptic inputs onto postsynaptic L2/3 neurons using blue laser stimulation (sCRACM; Petreanu et al., 2009). Using this method, we observed no difference between Cre(−) and Mef2c KO L2/3 neurons when we plotted the callosal input strength against vertical positions relative to the soma (Fig. 1H).
To determine if MEF2C-regulated ipsilateral long-range synaptic connections, we measured the input strength from the prefrontal/motor cortex to the barrel cortex. AAV9.ChR2-mCherry was injected into the prefrontal cortex ipsilateral to the ventricle injected with AAV9.GFP-Cre (Fig. 1I). At P18–P25, a strong and homogeneous labeling of axons in L1 was observed in the ipsilateral barrel cortex with some labeling in deep layers (Fig. 1J), which is characteristic of long-range projections from the prefrontal/motor cortex within the same hemisphere (Mao et al., 2011). A lack of ChR2-mCherry expression in L2/3 and L4 also indicated that there was no local transfection of ChR2 in the barrel cortex. To recruit more long-range axons, LED stimulation was centered on L1 above the recorded cell pair. Similar to callosal inputs from contralateral S1, we observed normal synaptic strength of ipsilateral long-range cortical inputs onto Mef2c KO L2/3 pyramidal neurons (Fig. 1K). Overall, these results indicate that postsynaptic MEF2C in L2/3 pyramidal neurons functions to selectively promote development and/or strengthening of synaptic inputs from L4.
Postsynaptic MEF2C is not required for maintenance of L4 to L2/3 synaptic strength
The development of local L4 to L2/3 synaptic connections in the barrel cortex depends on normal whisker sensory experience during a critical period that occurs in the second postnatal week for rodents (P10–P14; Lendvai et al., 2000; Stern et al., 2001; Shepherd et al., 2003; Wen and Barth, 2011). Based on previous results (Rajkovich et al., 2017), we hypothesized that postsynaptic MEF2C promotes experience-dependent development of L4 to L2/3 synapses. To further test this idea, we examined if postsynaptic MEF2C is required only during the experience-dependent critical period or is also necessary for the maintenance of L4 to L2/3 synapses after P14. To do this, we deleted Mef2c in postsynaptic L2/3 neurons by injecting AAV9.GFP-Cre into S1 barrel cortex of Mef2cfl/fl mice at P14. Based on our previous study (Rajkovich et al., 2017), we expect Mef2c to be deleted by P20–P21. At P27–P32, we prepared acute slices and measured L4 synaptic input strengths onto neighboring pairs of Cre(−) and postsynaptic Mef2c KO L2/3 neurons with LSPS (Fig. 2A). To isolate the cell-autonomous role of postsynaptic MEF2C, we recorded from L2/3 neurons distanced from the injection site, where only sporadically transfected neurons can be found within the region, using LSPS mapping. With the late deletion of Mef2c, local L4 synaptic inputs onto postsynaptic Mef2c KO L2/3 neurons were of normal strength as compared with Cre(−) neighbors (Fig. 2B,C). This result demonstrates that postsynaptic MEF2C is required for the development, but not maintenance, of L4 to L2/3 synapses during an age that coincides with the experience-dependent critical period.
Postnatal hetero- and homozygous deletion of Mef2c in excitatory neurons results in weak synaptic inputs from L4 to L2/3 neurons but does not affect L4 excitability
To study the molecular mechanisms by which MEF2C regulates cortical circuit development, we created a mouse with early postnatal deletion of Mef2c in a large population of cortical excitatory neurons. To do this, we crossed BAC-CaMKIIa-iCre mice (Casanova et al., 2001) to Mef2cfl/fl mice. CaMKIIa-iCre is expressed beginning approximately P2–P3 and expresses in excitatory neurons in cortical layers 2–4 and 6 (Fig. 3A). Western blots of cortical lysates from CaMKII-Cre-Mef2c KO mice showed loss of full-length MEF2C at P21 (Fig. 3B). To assess the function of local cortical excitatory circuits, we performed LSPS on L2/3 pyramidal neurons in the S1 cortex from CaMKII-Cre-Mef2cfl/fl (CaMKII-Mef2c KO) or CaMKII-Cre-Mef2cfl/+ (CaMKII-Mef2c-Het) mice. Controls were Mef2cfl/fl or Mef2cfl/+ littermates without Cre [Cre(−); Fig. 3C]. Like postsynaptic, sparse deletion of Mef2c in L2/3 neurons, CaMKII-Mef2c KO mice had weak (∼50% reduction) EPSCs evoked from the L4 home barrel and adjacent barrels (Fig. 3D,E). Surprisingly, CaMKII-Mef2c-Het mice also had weak L4 home barrel inputs (Fig. 3D,E). The strength of synaptic inputs from adjacent L2/3 were unchanged onto CaMKII-Mef2c-Het or KO L2/3 neurons [Cre(−), 3.83 ± 0.71 pA; CaMKII-Mef2c-Het, 3.95 ± 0.77pA; CaMKII-Mef2c KO, 4.1 ± 0.40 pA; n = 20, 20, and 24 cells, respectively; n.s.).
Because CaMKII-iCre is expected to delete Mef2c in L4 neurons, the weak L4 to L2/3 inputs in the CaMKII- Mef2c KO and Het barrel cortex may be due to reduced LSPS glutamate-driven excitability of L4 neurons. To test this possibility, we performed loose-seal cell-attached recordings in L4 neurons from CaMKII-Mef2c KO, Het, and Cre(−) slices and compared action potential (AP) firing in response to LSPS-induced glutamate uncaging (Fig. 3F–I). The number of APs elicited with LSPS averaged between 4 and 6 spikes per cell, and there were no differences between CaMKII-Mef2c KO, Het, and Cre(−) L4 neurons. Consistent with this result, the intrinsic excitability of CaMKII-Mef2c KO and Het L2/3 neurons was unchanged (Extended Data Table 1-2). These results suggest that the reduced L4 to L2/3 LSPS-evoked EPSCs in CaMKII-Mef2c-Het and KO neurons are likely due to the weak synaptic strength of L4 inputs to L2/3.
Presynaptic loss of Mef2c in layer 4 neurons weakens L4 to L2/3 synaptic inputs through decreased probability of neurotransmitter vesicular release
The observation of weak L4 to L2/3 synaptic inputs in the CaMKII-Mef2c-Het mice is in contrast to our previous results that sparse, postsynaptic, and heterozygous deletion of Mef2c in L2/3 neurons had no effect on L4 synaptic input strength (Rajkovich et al., 2017). Thus, we hypothesized that heterozygous deletion of Mef2c in presynaptic L4 neurons results in weak synaptic connections to L2/3. To test this idea, we deleted Mef2c selectively in L4 neurons by crossing Mef2cfl/fl mice with the Scnn1a-Tg3-Cre (Scnn1a-Cre) driver line, conferring Cre expression in the L4 cortex (Madisen et al., 2010). Scnn1a-Cre is reported to express between P3 and P14 in the cortex (Allen Brain Atlas; https://connectivity.brain-map.org/transgenic/experiment/127170789). To confirm this, we crossed Scnn1a-Cre mice to Ai14-tdTomato Cre-reporter mice (Madisen et al., 2010) or injected P1 pups with AAV9 expressing a Cre-dependent mCherry-tagged channelrhodopsin (ChR2-mCherry). Consistent with reported expression data, there was little to no Cre-reporter expression at P2–P6 in the S1 cortex (data not shown), but robust expression was observed in L4 barrels of S1 at P10 and P22 as visualized by ChR2-mCherry (Fig. 4A) or tdTomato (Fig. 4B), respectively. LSPS was used to map the strength of L4 to L2/3 synaptic inputs in slices from Cre(−), Scnn1a-Mef2c-Het, and Scnn1a-Mef2c KO mice. As in CaMKII-Mef2c-Het and KO mice, weak LSPS-evoked EPSCs from the L4 home barrel were observed in both Scnn1a-Mef2c KO and Scnn1a-Mef2c-Het as compared with Cre(−) mice (Fig. 4C,D). No changes were detected in intrinsic measures (Extended Data Table 1-1). This result suggests that MEF2C functions in presynaptic L4 neurons to promote development and/or strength of synapses onto L2/3.
To determine if MEF2C in presynaptic L4 neurons strengthened synapses through enhancing presynaptic release probability, we measured short-term synaptic plasticity or paired-pulse ratio (PPR; see Materials and Methods) of evoked EPSCs from L4 neurons. PPR is inversely proportional to presynaptic glutamate release probability. For example, if weak EPSCs are mediated in part by decreases in release probability, we would expect a corresponding increase in PPR. To evoke PPR of EPSCs from L4 neurons, we injected AAV9 expressing a Cre-dependent ChR2-mCherry intraventricularly into the cortex of P1 Scnn1a-Cre mice. ChR2-mCherry expression was robust at P18 and confined to L4 in the cortex (Fig. 4E). At P18–P25, slices were prepared from injected Scnn1a-Mef2c KO, Het, and “WT” [Scnn1a-Cre(+); Mef2c+/+] mice. EPSCs were evoked onto L2/3 pyramidal neurons with pulses of blue LED stimulation of L4 at two interpulse intervals (200 and 400 ms). At both intervals, EPSCs in all genotypes exhibited paired-pulse depression. However, Scnn1a-Mef2c KO neurons exhibited reduced paired-pulse depression, or increased PPR, compared with WT Cre(+) mice, which is consistent with a lower presynaptic release probability (Fig. 4F,G, Extended Data Table 1-1). An intermediate trend toward increased PPR was observed in Scnn1a-Mef2c-Het neurons, and a two-way ANOVA revealed a main effect of genotype on PPR (F(1,83) = 7.35; p = 0.008). These results suggest MEF2C in presynaptic L4 neurons strengthens synaptic inputs onto L2/3 neurons by increasing presynaptic release probability.
Germline heterozygous deletion of Mef2c does not affect L4 and callosal synaptic input strength onto L2/3 neurons
Germline haploinsufficiency of MEF2C in humans is associated with MEF2C haploinsufficiency syndrome (MCHS), characterized by intellectual disability, autism, and epilepsy. A mouse model of MCHS was established from a germline deletion of Mef2c (Mef2c-Het) and displays autism-relevant behaviors (Harrington et al., 2020). Because we observe weak L4 to L2/3 synaptic inputs with postnatal, heterozygous deletion of Mef2c, with either CamKII-Cre or Scnn1a-Cre (Figs. 3, 4), we hypothesized that a similar phenotype may occur with germline haploinsufficiency of Mef2c and may be relevant to MCHS. To test this possibility, we measured L4 to L2/3 synaptic strength in P18–P25 WT and Mef2c-Het littermates with LSPS. In contrast to postnatal, cortical deletion of Mef2c, the strength of LSPS-evoked EPSCs from L4 was unchanged in the Mef2c-Het (Fig. 5A,B). This result agrees with previous findings of normal L4 to L2/3 synaptic transmission in Mef2c-Het mice evoked with electrical stimulation. Similar to L4 inputs, callosal input strengths onto L2/3 neurons were normal in Mef2c-Het mice as measured using blue light LED to evoke EPSCs from ChR2-mCherry expressing callosal axons (Fig. 5C–F). Taken together, these results suggest that compensatory mechanisms may function to maintain L4 synaptic strength with germline heterozygous deletion of Mef2c.
Postsynaptic expression of MEF2C-VP16 rescues weak L4 to L2/3 inputs in response to sensory deprivation
The development of L4 to L2/3 synaptic connections in the barrel cortex depends on normal whisker sensory experience as well as MEF2C (Lendvai et al., 2000; Stern et al., 2001; Shepherd et al., 2003; Wen and Barth, 2011). We previously demonstrated that sensory deprivation by whisker trimming prevents or occludes weakening of L4 to L2/3 synaptic inputs by deletion of Mef2c in L2/3 neurons (Rajkovich et al., 2017). These results suggested that MEF2C and experience function in a common signaling pathway to promote development and strengthening of L4 to L2/3 synapses. Because sensory experience and neuronal activity trigger MEF2C transcriptional activation (Rashid et al., 2014; Assali et al., 2019), we hypothesized that transcriptional activation of MEF2C promotes L4 to L2/3 synaptic input strength and functions downstream of sensory experience. To test this hypothesis, we injected WT mice intraventricularly at P1 with AAV expressing a constitutively transcriptionally active MEF2C (MEF2C-VP16) bicistronically with GFP (Pulipparacharuvil et al., 2008; Fig. 6A). At P17–P21, we performed dual recordings of MEF2C-VP16 [GFP(+)] and neighboring untransfected [GFP(−)] L2/3 pyramidal neurons and LSPS mapping. Surprisingly, we observed no difference in the strength of home barrel L4 synaptic inputs between nontransfected and postsynaptic MEF2-VP16–transfected L2/3 neurons in WT mice (Fig. 6A,B). Thus, overexpression of active MEF2C on a WT background does not strengthen L4 to L2/3 synaptic inputs. This may be due to MEF2C transcriptional activation and/or L4 synaptic input strengths being saturated in sensory-experienced WT mice. If so, then sensory deprivation by whisker trimming may reduce MEF2C transcriptional activity and/or L4 synaptic strength and allow strengthening of L4 inputs in response to MEF2C-VP16. To test this idea, we compared the effects of MEF2C-VP16 on L4 to L2/3 synaptic strength in sensory deprived (unilateral whisker trimming; P9–P17), or contralateral S1, to that in spared, or ipsilateral, S1 of the same WT mice (Fig. 6C). LSPS maps were collected from pairs MEF2C-VP16 [GFP(+)] and neighboring GFP(−) L2/3 pyramidal neurons in slices from deprived or spared hemispheres. In the deprived hemisphere, MEF2C-VP16–expressing L2/3 neurons had increased strength of L4 inputs as compared to their untransfected neighbors, whereas in the spared hemisphere, there was no difference in L4 input strength between MEF2C-VP16–expressing and GFP(−) L2/3 neurons (Fig. 6D–F). A comparison of L4 synaptic input strength across hemispheres revealed that sensory deprivation weakened L4 synaptic inputs onto untransfected neurons, but not in MEF2C-VP16–expressing neurons (Fig. 6D–F). These results indicate that overexpression of transcriptionally active MEF2C can prevent or rescue the weakening of L4 to L2/3 synaptic connections due to sensory deprivation and supports the hypothesis that transcriptional activation of MEF2C promotes L4 synaptic input development downstream of sensory experience.
To test whether postsynaptic MEF2C-VP16 is sufficient to rescue weak L4 to L2/3 inputs in the CaMKII-Mef2c KO, we sparsely transfected MEF2C-VP16 in the cortex of CamKII-Mef2c KO mice with P1 AAV injection and performed LSPS mapping on pairs of untransfected Mef2c KO [GFP(−)] and transfected MEF2C-VP16 [GFP(+)] L2/3 pyramidal neurons. Surprisingly, L4 synaptic inputs were unaffected by MEF2C-VP16 expression in L2/3 neurons of CaMKII-Mef2c KO mice (Fig.6G,H). Based on the expression pattern of CaMKII-Cre (Fig. 3A), Mef2c is expected to be deleted in L4 and L2/3 neurons. Postsynaptic expression of MEF2C-VP16 may not rescue weak L4 to L2/3 inputs in CaMKII-Mef2c KO neurons because MEF2C is also required in presynaptic L4 neurons. Together these results reinforce the results that MEF2C is necessary in both L4 and L2/3 neurons for development of their synaptic connections.
MEF2C regulates expression of pre- and postsynaptic genes under basal and high activity conditions in cortical slice cultures
Based on our functional results, we hypothesize that neuronal activity stimulates MEF2C-dependent transcription of genes that promote development and function of L4 to L2/3 excitatory synapses. Because MEF2C is necessary for both presynaptic L4 and postsynaptic L2/3 neurons, we hypothesize that MEF2C regulates transcription of genes that promote both pre- and postsynaptic function. To identify activity-dependent genes in the postnatal developing cortex and the regulation of these genes by MEF2C, we established an activity-induced gene expression paradigm in ex vivo cortical slice cultures. S1-containing cortical slices from wildtype mice were prepared at P6–P7 and cultured in vitro for 8 d or equivalent postnatal day (ED) 14–15 (Fig. 7A). We first performed a time course of activity-dependent gene expression to identify a time point for robust gene expression using quantitative (q) RT-PCR for known activity-induced genes or MEF2 target genes (Fos, Egr2, Arc, Nr4a1; Extended Data Table 7-1). At ED14–ED15, cultures were treated with media containing the GABAA receptor blocker (picrotoxin) and K+ channel blocker (4-aminopyridine, 4-AP), for 15 min, 1, or 3 h to increase neuronal activity (Fig. 7B–E). We observed the most robust activity-dependent gene expression at the 3 h time point and used this treatment time in subsequent experiments. To identify activity-induced genes and their regulation by MEF2C, slice cultures were prepared from Cre negative [Cre(−)] and CaMKII-Mef2c KO (or “KO”) mice and treated with either vehicle (“Basal”) or picrotoxin/4-AP (“Activity”) for 3 h. mRNA from cultures were then processed for bulk RNA-seq analysis.
Table 7-1
Download Table 7-1, XLSX file.
Table 7-2
Download Table 7-2, XLSX file.
Table 7-3
Download Table 7-3, XLSX file.
Table 7-4
Download Table 7-4, XLSX file.
Processed tissue samples expressed, on average, ∼13,000 genes [with counts per million reads (CPM) ≥ 1.0] in Cre(−) samples (Extended Data Table 7-2). Differentially expressed genes (DEGs) were identified using DESeq2 conducted on raw CPMs with a log2 FC cutoff of ±0.3 and an adjusted p-value of <0.05 (Extended Data Table 7-3). We compared four conditions: (1) “Cre(−) Basal” (vehicle treated), (2) “Cre(−) Activity” (picrotoxin/4-AP treated), (3) “KO Basal,” and (4) “KO Activity.” To identify activity-driven genes in the Cre(−) cortex, we normalized the “Cre(−) Activity” condition to those in “Cre(−) Basal” using DESeq2 and identified 714 upregulated and 1,061 downregulated genes in “Cre(−) Activity” (Fig. 7F; Extended Data Tables 7-3, 7-4). Gene ontology (GO) analysis of both up- and downregulated DEGs using ToppFun (J. Chen et al., 2009) revealed enrichment for molecular functions and biological processes related to transcription regulation, DNA, and chromatin binding and the nuclear cellular compartment (Extended Data Table 8-1). Many transcription factors upregulated by activity were canonical activity-dependent immediate-early genes (Fos, Jun, Npas4, Egr1) as well as other known activity-induced genes (Homer1, Bdnf, Arc; Hrvatin et al., 2018; Bach et al., 2023; Pollina et al., 2023). A comparison of gene expression in “KO Activity” to “KO Basal” conditions found 800 genes upregulated and 1,041 genes downregulated by activity in KO (Fig. 7G). GO analysis revealed enrichment for genes related to transcription, DNA, and chromatin binding (Extended Data Table 8-1), many of those similar to what we observed in Cre(−) cultures. Thus, the activity-induced expression of many transcription factors and regulators of transcription does not require MEF2C.
Table 8-1
Download Table 8-1, XLSX file.
To identify MEF2C-regulated genes in the different activity conditions, we first compared gene expression in the “KO Activity” versus “Cre(−) Activity” conditions. This comparison found 621 upregulated genes and 557 downregulated genes (Fig. 8A; Extended Data Tables 7-3, 7-4), which are both enriched for genes expressed in S1 pyramidal neurons (Fig. 8B; Zeisel et al., 2015). Upregulated genes were also associated with inhibitory neurons suggesting non-cell-autonomous effects of Mef2c deletion in excitatory neurons. GO analysis of upregulated genes revealed the most significant enrichment for biological processes of transsynaptic signaling, synaptic transmission, organization and assembly, and cellular compartments of pre- and postsynapse and dendrites (Extended Data Table 8-1). To refine the GO analysis for synaptic terms, we used an evidence-based, expert-curated database, SynGO (Koopmans et al., 2019). Upregulated genes in “KO Activity” showed enrichment for biological processes of synapse assembly, organization, and transsynaptic signaling in pre- and postsynaptic compartments (Fig. 8C,D). Interestingly, analysis of downregulated genes in the “KO Activity” condition with SynGO found enrichment for genes that function in transsynaptic signaling, synapse adhesion, and organization within the pre- and postsynaptic compartments (Fig. 8E,F; Extended Data Table 8-1). Thus, under high activity conditions, MEF2C primarily regulates expression of genes that function in synaptic processes in pre- and postsynaptic compartments and both suppresses and promotes their expression.
Comparison of gene expression in CaMKII-Mef2c KO and Cre(−) cultures under basal activity conditions revealed 1,452 upregulated and 1,552 downregulated genes in the KO (Fig. 9A; Extended Data Tables 7-3, 7-4). Upregulated genes were enriched for transsynaptic signaling and synaptic transmission in both pre- and postsynaptic compartments and in genes associated with cortical pyramidal neurons and interneurons (Fig. 9C; Extended Data Table 8-1). In contrast to the high activity condition, downregulated genes in KO cultures under basal activity conditions were enriched for processes of cell adhesion, extracellular matrix compartment, and translation at synapses as well as genes associated with astrocytes and microglia (Fig. 9D). In summary, MEF2C suppresses expression of genes that function at synapses and regulate synaptic function under both basal and high activity conditions. However, only under high activity conditions does MEF2C promote the expression of genes involved in synaptic function, assembly, and organization. Up- and downregulated genes in KO cultures overlapped with up- and downregulated genes, respectively, observed in the cortex with embryonic deletion of Mef2c in excitatory neurons (Emx1 Cre; Harrington et al., 2016) or germline Mef2c-Het mice (Harrington et al., 2020; Fig. 9E,F). This overlap was significant under both basal and high activity conditions and suggests that there are similar MEF2C-regulated genes in the cortex in slice cultures and in vivo and may be relevant to MCHS.
While loss of function mutations in MEF2C are associated with autism and intellectual disability (Assali et al., 2019), MEF2C transcriptional activity is positively associated with cognitive function in mice and humans and cognitive resilience with aging (Mitchell et al., 2018; Barker et al., 2021). In support of these associations, upregulated genes in the “KO/Cre(−) Basal” condition significantly overlapped with SFARI ASD genes (p < 0.01; https://gene.sfari.org/), and downregulated genes significantly overlapped with genes associated with cognitive resilience in humans (Barker et al., 2021; Fig. 9G). These results suggest that MEF2C suppresses genes associated with ASD and promotes expression of genes associated with cognitive function in the cortex. Under both basal and high activity conditions, MEF2C-regulated genes overlap with genes associated with human attention-deficit hyperactivity disorder (Zhou et al., 2023) and genes containing broad enhancer-like chromatin domains (BELD; Fig. 9G). BELD genes are highly transcribed during early postnatal brain development and are enriched for genes involved in synapse development and ASD (Zhao et al., 2018). Taken together, these results implicate MEF2C in expression of genes associated with neurodevelopment, cognition, and neuropsychiatric disorders.
Under high activity conditions, MEF2C promotes expression of genes encoding presynaptic proteins that promote presynaptic development and function
Based on our electrophysiology results, we hypothesize that MEF2C transcriptional activation functions downstream of experience and neuronal activity to promote L4 to L2/3 synapse development. Thus, we would predict that there is a set of activity-induced genes that require MEF2C and function to promote excitatory synaptic development and/or function. Genes that are induced by activity in Cre(−), but not in CaMKII-Mef2c KO cultures, would be candidate genes to support our hypothesis. To identify such genes, we compared genes that are induced by activity in Cre(−) cultures [Cre(−) Activity/Basal; 714 genes] with those induced by activity in KO cultures (KO Activity/Basal; 800 genes; Fig. 10A). Of the 714 activity-induced genes in Cre(−) cultures, 245 genes were not induced by activity in KO cultures. Interestingly, GO analysis of these 245 genes with SynGO found enrichment for genes that functioned primarily in presynaptic compartments, including the active zone and synaptic vesicle membrane and in processes of presynaptic assembly (Fig. 10B,C; Extended Data Table 8-1). Of these genes, some encode voltage-sensitive ion channels (Scn1a, Cacnb4, Kcnh4) or regulate synaptic vesicle fusion (Snap25, Synpr, Rims3). Interestingly, many genes in this group encode presynaptic cell adhesion proteins that mediate synapse formation and maturation (Flrt2, Nrxn1, Tenm1, Cbln2, Cbln3) and proteins that regulate cell–cell adhesion and/or axon guidance (EphB2, Pcdh19, Sema3e, Sema4b, Sema7a; Fig. 10D). Therefore, MEF2C is required for the activity-dependent expression of many genes that regulate presynaptic function and development that may contribute to the experience-dependent development of L4 to L2/3 synaptic inputs (Fig. 11E,F). A comparison of the 245 activity- and MEF2C-dependent genes with a MEF2C ChIP-seq dataset (Ma and Telese, 2015) found a significant overlap of genes with MEF2C bound at the promoter (p = 0.0044; Extended Data Table 10-1). This result suggests that many of these genes are direct targets of MEF2C. Of the 469 genes commonly induced by activity in Cre(−) and KO cultures, SynGO analysis only found enrichment in the extrasynaptic compartment and BDNF signaling (Extended Data Table 8-1). Interestingly, these 469 genes also overlapped with MEF2C promoter-bound genes suggesting that many are also direct targets, but MEF2C is dispensable for their induction by activity (Extended Data Table 10-1).
Table 10-1
Download Table 10-1, XLSX file.
To determine if changes in expression of activity and MEF2C-dependent genes resulted in altered protein levels, we performed Western blots for selected presynaptic proteins from Cre(−) and KO cultures under basal and high activity conditions (Fig. 10E,H). We selected SLIT2 and FLRT2 for their presynaptic and transsynaptic role in input-specific synapse development (Blockus et al., 2021; Südhof, 2021) and blotted a synaptosome fraction to enrich for synaptic proteins. FLRT2 and SLIT2 protein levels were decreased in KO cultures, as compared with Cre(−), in high activity, but not basal activity conditions (Fig. 10E,H). To determine if FLRT2 and SLIT2 were regulated by MEF2C in vivo, we blotted S1 cortical lysates from P18 CaMKII-Mef2c KO and Cre(−) littermates. We observed decreases in FLRT2, but not SLIT2, in CaMKII-Mef2c KO cortices (Fig. 10F,I). To determine if there were more general decreases in presynaptic proteins, we blotted for synapsin 1 (SYN1) in the culture synaptosomes and cortical lysates and found that SYN1 was unaffected by CaMKII-Mef2c KO in both preparations (Fig. 10G; data not shown). The protein changes are consistent with gene expression results that under conditions of increased neuronal activity, MEF2C promotes expression of select presynaptic proteins that promote synapse development.
MEF2C promotes expression of postsynaptic genes under conditions of high activity
While the analysis in Figure 10 primarily implicated genes that function in the presynaptic compartment, our electrophysiology results also support a role for MEF2C in postsynaptic L2/3 neurons in development of L4 to L2/3 synapses. Since we were able to rescue L4 to L2/3 synaptic strength with postsynaptic expression of MEF2C-VP16, we hypothesized that MEF2C transcriptional activation of genes that functioned in the postsynaptic compartment promotes L4 synaptic input strength. Therefore, we looked more closely at MEF2C-dependent postsynaptic genes. Under high activity conditions, MEF2C promoted expression of genes that were either expressed in the postsynaptic membrane and/or functioned to regulate postsynaptic strength (Fig. 8E,F). Interestingly, some of these genes functioned in synapse adhesion, formation, or maturation such as Lrrtm1, Pcdh8, and Nrg1 (Fig. 11A; Li et al., 2007; Yasuda et al., 2007; Südhof, 2021). To validate changes in protein expression of a candidate postsynaptic adhesion molecule, we performed Western blots for LRRTM1 in S1 cortical homogenates of P21 CaMKII-Mef2c KO and Cre(−) littermates (Fig. 11B), or whole homogenates or synaptosomes from KO and Cre(−) slice cultures under both basal and high activity conditions (Fig. 11C,D). In contrast to changes in Lrrtm1 gene expression, we were unable to detect changes in LRRTM1 protein in the MEF2C KO cortex. Because changes in LRRTM1 may be synapse specific, for example, only at L4 to L2/3 synaptic inputs, we may be unable to resolve these changes with bulk methods, and cell- and input-specific methods may be required.
Discussion
Development and strengthening of cortical circuits require normal sensory experience and patterned neuronal activity (Wen and Barth, 2011). Here, we demonstrate a requirement for the activity-regulated transcription factor, MEF2C, in both pre- and postsynaptic neurons during a postnatal, experience-dependent period for strengthening the specific synaptic connection between L4 and L2/3 neurons. In contrast, MEF2C is not required in L2/3 neurons for the function of long-range synaptic inputs originating from either contralateral S1 or ipsilateral frontal or motor cortices. The requirement for presynaptic MEF2C in L4 neurons suggests a model where sensory experience drives patterned neural activity of pre- and postsynaptic neurons to stimulate a MEF2C-dependent transcriptional program for coordinated expression of pre- and postsynaptic proteins to strengthen specific synaptic connections (Fig. 11E,F). Candidate genes that could serve such a function are the transsynaptic, cell adhesion molecules that direct and promote specific synaptic connections in both the pre- and postsynaptic compartments (Südhof, 2021). In support of this model, we observed that under high activity conditions, MEF2C promotes expression of genes that function in pre- and postsynaptic compartments, transsynaptic signaling, and synapse organization and assembly. Specifically, MEF2C is necessary for activity-induced expression of FLRT2 and SLIT2 that function in the assembly of specific presynaptic circuits (Sando et al., 2019; Blockus et al., 2021). These findings suggest that MEF2C contributes to experience-dependent development of specific cortical circuits through regulation of genes that function in synaptic cell adhesion and transsynaptic signaling.
In the barrel cortex, L2/3 acquires whisker-evoked responses during the second postnatal week (P12–P14), which is also a critical period for experience-dependent development of L4 to L2/3 synapses (Stern et al., 2001; Wen and Barth, 2011). Postnatal deletion of Mef2c before, but not after P14, leads to decreased L4 to L2/3 input strength. Sensory deprivation by whisker trimming or postsynaptic deletion of Mef2c during this postnatal period results in weak L4 to L2/3 synapses. Furthermore, sensory deprivation occludes or prevents further weakening by postsynaptic deletion of Mef2c suggesting that sensory experience and MEF2C function in a common signaling pathway to promote L4 to L2/3 synaptic strength (Rajkovich et al., 2017). Because experience and neuronal activity drive MEF2 transcriptional activity (Flavell et al., 2008; Cole et al., 2012; M. R. Lyons et al., 2012; L. F. Chen et al., 2020), we hypothesize that sensory experience-driven neuronal activity drives MEF2C-dependent transcriptional activation of genes that selectively promote L4 to L2/3 synaptic inputs. In support of this idea, postsynaptic expression of a constitutively transcriptionally active MEF2C (MEF2C-VP16) strengthens L4 to L2/3 synapses in sensory-deprived cortex suggesting that active MEF2C functions downstream of sensory experience.
Previous work in the cortex and other brain regions has implicated MEF2 transcriptional activation in elimination of excitatory synapses (Flavell et al., 2006; A. Shalizi et al., 2006; Pfeiffer et al., 2010; Assali et al., 2019). For example, in cultured cortical neurons or cerebellar granule cells, deletion or knockdown of MEF2C or MEF2A reduces excitatory synapse number, and this effect is rescued by expression of a transcriptional repressing form of MEF2 (MEF2-engrailed) and mimicked by expression of MEF2-VP16 in WT neurons (A. Shalizi et al., 2006; Harrington et al., 2016). Therefore, an alternative interpretation of our findings is that loss of MEF2C derepresses transcription of genes that promote elimination of L4 to L2/3 synapses. However, we observe that MEF2C-VP16 rescues weak L4 to L2/3 synapses in a sensory-deprived cortex and has no effect in spared or normally experienced WT mice supporting a function of MEF2C transcriptional activation in synapse strengthening and/or development. Differences in our results and others may be because we are examining the specific L4 synaptic input pathways in vivo, whereas studies in cultures cannot distinguish between specific inputs. Furthermore, there may be experience-dependent and/or in vivo regulation of MEF2C or its interactors that reveal a synapse-strengthening effect of MEF2C (Telese et al., 2015; Zhang et al., 2016). A splice variant of Mef2c, lacking the γ domain, is highly expressed in cortical neurons (M. R. Lyons et al., 2012). The γ domain contains a phosphorylation site (Ser396) that promotes sumoylation of MEF2C and inhibits its transcriptional activity (Zhu and Gulick, 2004; Kang et al., 2006). Mef2c variants lacking the γ domain are highly activated by neuronal depolarization (M. R. Lyons et al., 2012) and may be particularly sensitive to sensory experience to promote L4 to L2/3 synaptic strengthening.
Previous work has focused on the postsynaptic function of MEF2 in synapse development and refinement (A. Shalizi et al., 2006; Pfeiffer et al., 2010; Rajkovich et al., 2017). Our work reveals both pre- and postsynaptic roles for MEF2C. Deletion of Mef2c postnatally in presynaptic L4 neurons weakened synaptic inputs onto L2/3 neurons likely through decreases in release probability. Sensory deprivation by whisker trimming similarly weakens L4 synaptic inputs via reductions in release probability (Bender et al., 2006), suggesting that sensory experience and presynaptic MEF2C strengthen L4 to L2/3 synaptic connections through a similar presynaptic mechanism. Because we used ChR2 to evoke synaptic transmission from L4 neurons in the paired-pulse ratio experiments, this may not reflect the effects of presynaptic MEF2C on action potential–evoked release mechanisms. Scnn1a-Cre and Mef2c are also expressed in the somatosensory thalamus (VPM; Allen Brain Atlas), and Mef2c is likely deleted in thalamic inputs to L4, which may have indirect effects on L4 to L2/3 synaptic transmission. Consistent with a role for presynaptic MEF2C in experience-dependent synaptic strengthening, we observed that activity-induced, MEF2C-dependent genes encode proteins expressed in the presynaptic compartment and functioned in neurotransmitter release, synaptic cell adhesion, assembly, and axon guidance. This suggests that MEF2C activates a transcriptional program in response to activity to promote formation and/or maturation of presynaptic function. In cerebellar granule cell cultures, MEF2A eliminates orphan presynaptic sites, or those without an associated postsynaptic site, through transcriptional repression of synaptotagmin 1 (Syt1; Yamada et al., 2013), which, in turn, promotes maturation of neighboring presynaptic boutons. Such a mechanism may also function in L4 neurons. We also observe a distinct dose-dependent requirement for pre- and postsynaptic Mef2c. Heterozygous deletion of Mef2c in presynaptic, but not postsynaptic, neurons weakened L4 to L2/3 inputs (Rajkovich et al., 2017). In contrast, germline heterozygous Mef2c deletion did not affect L4 to L2/3 in agreement with previous studies (Harrington et al., 2020) suggesting compensatory mechanisms to maintain L4 synaptic inputs with germline MEF2C loss.
MEF2C is required postsynaptically in L2/3 neurons for development of L4 inputs, but not for long-range inputs from the contralateral hemisphere or ipsilateral frontal/motor cortex (Rajkovich et al., 2017). There is also evidence for input-specific roles of MEF2C in Purkinje cells (Kamath and Chen, 2019). A candidate mechanism by which MEF2C regulates input-specific development is through the regulation of transsynaptic cell adhesion genes (Südhof, 2021). MEF2C promotes activity-dependent expression of genes encoding both presynaptic (Nrxn1, Flrt2, Tenm1) and postsynaptic (Lrrtm1, 2, and 4 Pcdh8) adhesion proteins and those that interact with them such as Slit2 and Cbln2/3 (Blockus et al., 2021). We show that MEF2C promotes FLRT2 and SLIT2 protein levels under high activity conditions in cultures and in the cortex in vivo. MEF2C-dependent induction of presynaptic cell adhesion molecules in L4 neurons may interact with MEF2C-regulated processes or postsynaptic binding partners in L2/3 neurons to form or stabilize synapses.
In postnatal cortical slice cultures, activity-induced genes that functioned in transcriptional control and chromatin regulation were similarly induced in both Cre(−) and CaMKII-Mef2c KO cortical slices. Notably, under “high activity” conditions, MEF2C bidirectionally regulated genes with synaptic functions, both pre- and postsynaptically. This reinforces functional studies that the MEF2 family of transcription factors bidirectionally regulates many synaptic processes (Assali et al., 2019). In “Basal” activity conditions, many synaptic genes were upregulated, which suggests that MEF2C may derepress synaptic genes to enhance the function of some synaptic inputs (Harrington et al., 2016). As discussed, we observe distinct effects of postsynaptic Mef2c deletion on excitatory synapses depending on the origin of the input; L4 to L2/3 inputs are weak, but inputs from long-range connections are either potentiated (Rajkovich et al., 2017) or unchanged (Fig. 1). Furthermore, the frequency of spontaneous, miniature EPSCs (mEPSCs) is increased (Rajkovich et al., 2017). Therefore, the bidirectional regulation of synaptic genes by MEF2C may function at select synaptic inputs and/or evoked or spontaneous transmission (Guzikowski and Kavalali, 2021). Going forward, it will be important to identify the direct gene targets of MEF2C under different activity conditions to understand its complex effects on synaptic connectivity. In agreement with previous reports, upregulated genes in Mef2c KO cultures overlapped with autism-risk genes in humans and downregulated genes overlapped with cognitive resilience (Fig. 9G; Harrington et al., 2016, 2020; Mitchell et al., 2018; Barker et al., 2021). The bidirectional regulation of MEF2C-regulated gene targets and their effects on synaptic function across the lifespan likely contribute to human cognitive development and function.
Footnotes
This work was supported by the National Institute of Health Grants (R01 HD052731 to K.M.H. and J.R.G.; R01 MH126481 to G.K.; and T32HL139438 to J.N.P.). We would like to thank Patricia Hahn, Jacob Eli Bowles, and Christopher Williams for their technical assistance with genotyping mice and Dr. Genevieve Konopka and David Atkinson for their assistance and advice with RNA-seq analysis.
↵*J.N.P., S.D.W., and Z.Z. contributed equally to this work.
The authors declare no competing financial interests.
- Correspondence should be addressed to Kimberly M. Huber at kimberly.huber{at}utsouthwestern.edu.