Main

One reason why it is difficult to maintain constant synaptic strength is that during high-frequency presynaptic activation there is little time to replenish vesicles, and synaptic strength decreases as vesicle depletion becomes more severe (Extended Data Fig. 1a, c). Consequently, many synapses depress to a greater extent as activation frequency increases8,9,10. This property can be used computationally as a dynamic gain control mechanism1. Despite the challenges posed by vesicle depletion, some synapses transmit faithfully across a wide range of activation frequencies3,4,5 (Extended Data Fig. 1b, d–f). In the vestibular system, such frequency-invariant transmission effectively conveys the absolute firing rate of vestibular afferents, and contributes to the linearity of occulomotor reflexes4. Several mechanisms have been proposed to explain how synapses could maintain frequency-invariant transmission3,5,11, but the molecular tools to address these models directly have been lacking. Here we test the hypothesis that facilitation, a short-lived increase in the probability of release (PR), counteracts vesicle depletion to produce frequency-invariant transmission (Extended Data Fig. 1d). We take advantage of the recent finding that genetic knockout of the slow synaptotagmin isoform Syt7 selectively eliminates facilitation at some synapses7.

We began by studying synapses made by Purkinje cells (PCs), the sole output neurons of the cerebellar cortex, which fire spontaneously at 10–120 Hz in vivo12. Purkinje cells form GABA (γ-aminobutyric acid)-releasing synapses in the deep cerebellar nuclei (DCN, Fig. 1a), and express Syt7 (Fig. 1b). The presence of Syt7 was intriguing because, unlike many synapses with prominent Syt7 expression, PC–DCN synapses depress. We found that Syt7 expression at PC–DCN synapses is age-dependent, unlike expression of the PC marker calbindin and Syt2 (Extended Data Fig. 2). The onset of Syt7 expression is correlated with the development of frequency-invariant transmission at PC–DCN synapses (Fig. 1c–f and Extended Data Figs 3, 4). In young animals (postnatal day (P)13–15), Syt7 expression is low, and steady-state transmission is frequency-dependent in both wild-type and Syt7 knockout mice (Fig. 1c–e, left). Syt7 expression increases in juvenile wild-type mice (P21–32), and transmission becomes more frequency-invariant with age. However, in juvenile Syt7 knockout mice, frequency-invariant transmission does not develop (Fig. 1c–e, middle). The differences between wild-type and knockout mice become more pronounced in adulthood (Fig. 1c–e, right). As a consequence, synaptic charge transfer (the product of inhibitory postsynaptic current (IPSC) amplitude and activation frequency) becomes more linear during development in wild-type mice, but remains sublinear in Syt7 knockout mice (Fig. 1f). These findings indicate that Syt7 is required for frequency-invariant transmission at PC–DCN synapses.

Figure 1: Syt7 is required for frequency-invariant transmission at PC–DCN synapses.
figure 1

a, PC axons were stimulated and responses were recorded from DCN projection neurons. b, Sagittal cerebellum from a wild-type mouse at postnatal day 25 (P25) immunolabelled for calbindin (left) and Syt7 (right). Scale bar, 0.5 mm. c, High-power images of the DCN immunolabelled for calbindin and Syt7 from young (left, P13–15), juvenile (middle, P21–32), and adult (right, P80–110) wild-type (WT) and Syt7 knockout (KO) mice. Scale bar, 10 μm. d, Representative IPSCs for wild-type (black) and Syt7 knockout mice (red) for young (left), juvenile (middle) and adult (right) mice. Stimulus artefacts have been blanked for clarity. Vertical scale bars, 1 nA. e, Normalized steady-state amplitudes (IPSCSS) as a function of stimulation frequency for young (left), juvenile (middle) and adult (right) mice. f, Charge transfer calculated as the product of IPSCSS amplitude and stimulation frequency for young (left), juvenile (middle) and adult (right) mice. Data are mean ± s.e.m. Number of experiments shown in Extended Data Table 1.

PowerPoint slide

To determine what caused the loss of frequency-invariant transmission in Syt7 knockout mice, we examined recovery from depression, the initial PR, and facilitation. Syt7 mediates rapid calcium-dependent recovery from depression in cultured hippocampal synapses13. One possibility is that Syt7 contributes to frequency-independent transmission by accelerating recovery from depression during high-frequency activity (Extended Data Fig. 1e). Recovery from depression would then be faster in wild-type mice than in Syt7 knockout mice; however, this was not the case (Extended Data Fig. 5). Syt7 also mediates asynchronous release during high-frequency stimulation at some synapses14,15, but asynchronous release is not prominent at PC–DCN synapses (Extended Data Fig. 6).

We also tested the hypothesis that PR is elevated in Syt7 knockout mice. If PR were increased, stronger depletion could more effectively mask facilitation and lead to a loss of frequency-invariant transmission6. An increase in PR would be expected to increase the size of PC inputs, but we found no significant difference in single fibre size between wild-type and Syt7 knockout mice (Extended Data Fig. 7). We lowered external calcium (Cae) to decrease PR and reduce the masking effects of vesicle depletion, and found that at PC–DCN synapses facilitation was prominent in wild-type mice, but weak or absent in Syt7 knockout mice (Fig. 2a). If the loss of frequency-independent transmission is a consequence of an increase in PR, then reducing PR by lowering Cae should lead to frequency-invariant transmission in Syt7 knockout mice. However, when Cae was lowered from 1.5 mM (control) to 1 or 0.5 mM to reduce IPSC amplitudes to 42% and 12% of control, respectively5, transmission in Syt7 knockout mice remained frequency-dependent (Fig. 2b, c). These results indicate that the loss of frequency invariance in Syt7 knockout mice does not result from an increase in initial PR.

Figure 2: Syt7 is required for a hidden form of facilitation at PC–DCN synapses.
figure 2

a, Experiments were conducted in P21–32 mice in low Cae (0.3 mM) to reduce the initial probability of release. Responses are shown for representative experiments (top) and summarized across experiments (bottom). b, Average of 40th–50th IPSCs as a function of stimulation frequency. c, Ratio of IPSCSS induced by 100-Hz and 10-Hz (IPSC100/10 Hz) stimulation in different Cae. d, PC synapses were stimulated in 1.5 mM Cae at 10 Hz to reach steady-state, followed by 100 Hz stimulation in young (left), juvenile (middle) and adult (right) mice. Example responses (top) and summaries (bottom) are shown. Stimulus artefacts have been blanked for clarity. Vertical scale bars, 0.25 nA. *P < 0.05, **P < 0.01, one-way ANOVA with Tukey’s post-hoc test. Data are mean ± s.e.m. Number of experiments shown in Extended Data Table 1.

PowerPoint slide

We next looked for evidence of facilitation at physiological levels of Cae (1.5 mM). Facilitation was measured by first using stimulation at 10 Hz to induce baseline depletion, and then increasing the frequency to 100 Hz5,16. In wild-type mice, frequency steps revealed a transient enhancement with a magnitude that was correlated with the development of frequency-invariant transmission (Fig. 2d). These results are consistent with facilitation leading to sustained increases in PR that are transiently revealed until they are masked by vesicle depletion. In Syt7 knockout mice, facilitation was not observed at any age, and increases in stimulus frequency depressed IPSCs. Thus, Syt7-dependent facilitation is prominent when transmission is frequency independent (juvenile and adult wild-type mice), and is weak when transmission is frequency dependent (young wild-type mice and Syt7 knockout mice of all ages).

To determine whether impaired frequency-invariant transmission was due specifically to presynaptic loss of Syt717, we performed rescue experiments in global Syt7 knockout mice. We used adeno-associated viruses (AAVs) to express yellow-fluorescent protein-tagged channelrhodopsin 2 (ChR2–YFP) alone, or to bicistronically express both Syt7 and ChR2–YFP presynaptically7 (Fig. 3a). However, it is not possible to obtain expression in all PCs, and it is impractical to optically stimulate axons at high frequencies for prolonged trains18. We therefore used optical stimulation to identify ChR2-expressing fibres that could be isolated electrically (Fig. 3b, c). These axons were then electrically stimulated at a high frequency. In Syt7 knockout mice, facilitation was absent in axons expressing ChR2–YFP alone, but was prominent in fibres expressing both ChR2–YFP and Syt7 (Fig. 3d). Steady-state transmission was nearly constant when Syt7 was expressed, but remained frequency-dependent when only ChR2–YFP was present (Fig. 3e, f). Consequently, charge transfer in Syt7-expressing PC axons was linear, but remained sub-linear when only ChR2–YFP was expressed (Fig. 3g-h). Viral expression of Syt7 allowed PCs in Syt7 knockout mice to become as frequency invariant as in wild-type mice (Fig. 3h), indicating that presynaptic Syt7 mediates frequency-invariant transmission.

Figure 3: Presynaptic expression of Syt7 restores facilitation and frequency-invariant transmission at PC– DCN synapses in Syt7 knockouts.
figure 3

a, ChR2–YFP fluorescence and Syt7 immunolabelling in the DCN of Syt7 knockout mice following AAV-driven expression of either ChR2–YFP alone (top) or ChR2–YFP and Syt7 (bottom) in PCs. Scale bar, 10 μm. b, c, The ability of IPSCs evoked electrically and optically to occlude each other was used to identify labelled fibres. b, If PC fibres express ChR2, the algebraic sum of electrical (red) and optical (blue) stimulation (grey) exceeds responses evoked by simultaneous optical and electrical stimulation (black). c, If electrically stimulated axons do not express ChR2, the sum of optical and electrical responses (grey) matches responses evoked by simultaneous optical and electrical stimulation (black). Vertical scale bars, 0.5 nA (b, c). d, PC axons expressing ChR2–YFP alone (red) or ChR2–YFP and Syt7 (purple) were electrically stimulated at 10 Hz and then 100 Hz. eg, PC inputs were stimulated at 5–150 Hz for ChR2–YFP alone (red) or ChR2–YFP and Syt7 (purple). e, Responses evoked by trains. Stimulus artefacts have been blanked for clarity. Vertical scale bars, 1 nA (d, e). f, Average steady-state responses versus stimulation frequency. g, Charge transfer as a function of stimulation frequency. h, Summary of ratios of IPSCSS amplitudes at 100 Hz and 10 Hz. **P < 0.01, one-way ANOVA with Tukey’s post-hoc test. Data are mean ± s.e.m. Number of experiments shown in Extended Data Table 1.

PowerPoint slide

Having established that Syt7 is required for frequency invariance in PC synapses, we investigated whether Syt7 plays a similar role at vestibular synapses. Afferents from the vestibular ganglion project to the magnocellular medial vestibular nucleus (MVNm, Fig. 4a). Syt7 is expressed in the neuropil of the MVNm, where vestibular afferents form glutamatergic synapses (Fig. 4b). Although previous studies did not report facilitation at this synapse11, we found that vestibular synapses show facilitation at low levels of Cae in wild-type but not Syt7 knockout mice (Fig. 4c). At physiological levels of Cae, step changes in stimulation frequency produced transient enhancement in wild-type but not Syt7 knockout mice (Fig. 4d). These responses suggest that Syt7-mediated facilitation is also present at vestibular synapses, but is masked by depletion. We also found that steady-state transmission was frequency-invariant and charge transfer was linear in wild-type but not Syt7 knockout mice (Fig. 4e–g, Extended Data Fig. 8).

Figure 4: Syt7 is also required for frequency-invariant transmission at vestibular synapses.
figure 4

a, Schematic showing stimulus electrode placement to activate vestibular afferents while recording from MVNm neurons. b, Immunofluorescence for VGLUT2 (left) and Syt7 (right) surrounding MVNm cells in P25 mice. Scale bar, 20 μm. c, Responses evoked by stimulating vestibular afferents in 0.5 mM Cae. d, Vestibular inputs were stimulated at 10 Hz, followed by 100 Hz in 1.5 mM Cae. eg, Vestibular inputs were stimulated with 5–150-Hz trains in 1.5 mM Cae in wild-type and Syt7 knockout mice. e, Representative (left) and average (right) EPSCs evoked by trains. f, Average steady-state responses against stimulation frequency. g, Charge transfer as a function of stimulation frequency. Stimulus artefacts have been blanked for clarity. Vertical scale bars, 0.1 nA. Data are mean ± s.e.m. Number of experiments shown in Extended Data Table 1.

PowerPoint slide

We have shown that Syt7 mediates a hidden component of facilitation that counteracts partial vesicle depletion to produce linear charge transfer. Synaptic properties in wild-type and Syt7 knockout mice conform to a model in which the loss of frequency-invariance in Syt7 knockout mice is accounted for by the absence of facilitation5 (Extended Data Fig. 9). Linear charge transfer and Syt7 expression at PC synapses emerge during development, but for technical reasons most synapses have been studied only in young animals19. The prevalence of frequency-invariant transmission in adults is unknown, but indirect measurements in vivo suggest that it may be widespread20,21. Moreover, Syt7 is developmentally regulated22,23 and widely expressed in the adult brain, often in cells that make depressing synapses22,24. The kinetics of Syt7 make it well suited to operate within the physiological firing range of PCs and vestibular afferents4,12,25,26. While our results show that linear charge transfer is mediated by Syt7 at PC and vestibular synapses, other synapses may use different mechanisms (Extended Data Fig. 1).

At frequency-invariant synapses, the amount of neurotransmitter release scales linearly with the firing frequency, and can thus faithfully encode presynaptic spike rates. Sensorimotor processing in the vestibular system and intensity discrimination in the auditory system have been proposed to operate through a linear rate code4,27,28. Understanding the mechanisms involved in frequency-invariant transmission will enable genetic manipulations that could yield fundamental insights into how synaptic computations contribute to circuit function and behaviour.

Methods

Animals and viruses

All mice were handled in accordance with NIH guidelines and protocols approved by the Harvard Medical Area Standing Committee on Animals. Syt7 knockout mice29 (Jackson Laboratory) and wild-type littermates of either sex were used. Statistical tests were not used to predetermine sample size. Unless stated otherwise, the experiments were not randomized. In Figs 3a–g, 4d–g and Extended Data Figs 2, 7, 8, all experiments were performed blind to genotype and virus identity. In Figs 1, 2 and Extended Data Figs 3, 4, 5, 6, experiments were initially performed blind but blinding was abandoned after the initial rounds of experiments because genotypes could easily be identified from physiology alone. AAV2/1-hSyn-hChR2(H134R)–EYFP was obtained from the University of Pennsylvania Vector Core. AAV2/1-hSyn-hChR2(H134R)–EYFP–2A-Syt7 was obtained from the Boston Children’s Virus Core. Plasmid sequences are available upon request.

Stereotaxic surgeries were performed on P16–18 Syt7 knockout mice anaesthetized with ketamine/xylazine (100/10 mg/kg) supplemented with isoflurane (1–4%). Viruses were injected through glass capillary needles using a Nanoject II (Drummond) mounted on a stereotaxic apparatus (Kopf). Three injections were made in the right cerebellar cortex: 1.0, 1.5, 2.0 mm lateral, 1 mm posterior from lambda, 0.2–0.3 mm depth; 300–500 nl virus suspension was delivered to each site at a rate of 100 nl per min, and the needle was retracted 5–10 min after injection. Analgesic (buprenorphine 0.05 mg/kg) was administered subcutaneously for 48 h post-surgery.

Slice preparation

Acute slices were prepared from mice of both sexes (P21–32 unless otherwise indicated). Mice were killed 12–14 days after AAV injections. Animals were anaesthetized with ketamine/xylazine (100/10 mg/kg) and transcardially perfused with solution composed of (in mM): 110 choline Cl, 2.5 KCl, 1.25 NaH2PO4, 25 NaHCO3, 25 glucose, 0.5 CaCl2, 7 MgCl2, 3.1 Na pyruvate, 11.6 Na ascorbate, 0.002 (R)-CPP, 0.005 NBQX, oxygenated with 95% O2/5% CO2, kept at 35 °C. For DCN recordings, a cut was made down the midline of the hindbrain, and the cut face of each side was glued to the slicing chamber to generate sagittal slices. For the vestibular nuclei, a cut was made down the midbrain between the cerebellum and cortex and glued to the slicing chamber to generate coronal slices. Two hundred and fifty micrometre thick sections were made on a Leica 1200S vibratome and were then transferred to a holding chamber with ACSF containing (in mM): 127 NaCl, 2.5 KCl, 1.25 NaH2PO4, 25 NaHCO3, 25 glucose, 1.5 CaCl2, 1 MgCl2, and allowed to recover at 35 °C for at least 20 min before cooling to room temperature.

Electrophysiology

All experiments were performed at 34–35 °C with a flow rate of 3–5 ml/min. The recording ACSF had the same composition as the incubation ACSF unless otherwise stated. Recordings were made primarily in the dentate and interposed nuclei of the DCN. Large diameter (>15 μm) neurons were selected for recording as previously described5. For DCN recordings, borosilicate electrodes (1–2 MΩ) were filled with internal solution consisting (in mM) of: 110 CsCl, 10 HEPES, 10 TEA-Cl, 1 MgCl2, 4 CaCl2, 5 EGTA, 20 Cs-BAPTA, 2 QX314, 0.2 D600, pH 7.3. Cells were held at −30 to −40 mV in the presence of 5 μM NBQX to block AMPA receptors, 2.5 μM (R)-CPP to block NMDA receptors, 1 μM strychnine to block glycine receptors, and 1 μM CGP 55845 to block GABAB receptors. A glass monopolar stimulus electrode (2–3 MΩ) filled with ACSF was placed in the white matter surrounding the DCN to activate PC axons.

To study vestibular afferents, recordings were made from cells in the magnocellular medial vestibular nucleus. Recording electrodes (1.5–2 MΩ) were filled with (in mM): 150 Cs-gluconate, 3 CsCl, 10 HEPES, 0.5 EGTA, 3 MgATP, 0.5 NaGTP, 5 Tris-phosphocreatine and 5 Na-phosphocreatine; pH 7.2. Cells were held at −60 mV in the presence of 100 μM picrotoxin or 5 μM SR-95531 to block GABAA receptors, 2.5 μM (R)-CPP to block NMDA receptors, 1 μM strychnine to block glycine receptors, and 1 μM CGP 55845 to block GABAB receptors. A concentric bipolar electrode was placed in the expansion of the vestibulocochlear nerve as it reaches the dorsal brainstem, adjacent to the lateral vestibular nucleus. To prevent poor fibre recruitment or entrainment of local circuit elements, trains with EPSCs composed of multi-phasic rise or decay phases were excluded from analysis. For trains in low Cae, EPSCs became very small. In some cases, 5 μM NBQX was washed in at the end of experiments and traces in the presence of NBQX were subtracted to remove stimulus artefacts.

Trains of 100 stimuli (5–150 Hz, randomized), followed by 100 stimuli at 100 Hz, were applied every 20–30 s for vestibular and PC synapses. For 500-stimulus trains at 100 Hz, trials were collected every 60 s. Extracellular stimulation of axons became unreliable after 500 stimuli. For all recordings, only the capacitance roughly equivalent to the cell body was compensated (5 pF), and series resistance (1–6 MΩ) was compensated up to 80%. Experiments were discarded if series resistance changed by 30%. Experiments were discarded if fibres could not be consistently recruited, as assessed by major discrete changes observed in PSC amplitude during trains, or if the PSC amplitude never reached steady state during trains. Liquid junction potentials were left unsubtracted. In low Cae experiments, total divalents were kept constant by elevating external Mg. Experiments using the low affinity GABAA receptor antagonist TPMPA were performed as previously described5.

Analysis

Recordings were collected using a Multiclamp 700B (Molecular Devices) in Igor Pro (Wavemetrics) sampled at 20 kHz and filtered at 4 kHz. All data were analysed using custom-written scripts in Matlab (Mathworks). Stimulus artefacts were blanked for clarity. IPSC amplitudes during trains were measured from averaged traces of 3–10 trials as the peak evoked current, with a baseline measured 2 ms before stimulus onset. During high-frequency trains, IPSCs did not fully decay before subsequent stimuli. Baselines in this case were measured by extrapolating a single exponential fit from the previous IPSC (Extended Data Fig. 6). Trains from rest typically reached steady-state after 10–20 stimuli, and the steady-state was measured as the average IPSC size of the 40th to 60th stimuli. Charge transfer was calculated as the product of steady-state IPSC size and stimulation frequency, and normalized to the charge transfer of 10 Hz stimulation. All data are presented as means ± s.e.m. unless otherwise indicated. In some cases error bars are occluded by markers. Statistical significance was assessed using unpaired two-tailed Student’s t-test or one-way ANOVA with Tukey’s post-hoc test unless otherwise noted. A subset of the data from wild-type Syt7 littermates (~50%) in Fig. 1 and Fig. 2d (middle) has been presented previously5.

Optogenetics

It was impossible to reliably stimulate PC axons with prolonged trains at high frequency (>50 Hz) using optical stimulation alone (ChR2–H134R or Chronos). We therefore used ChR2–YFP to identify labelled fibres that were then stimulated electrically. In this approach, optical stimulation of axons expressing ChR2–YFP could occlude electrical stimulation of the same fibres, presumably because of the refractory period of the action potential. By contrast, if electrically stimulated fibres did not express ChR2–YFP, electrically evoked responses could not be occluded by optical stimulation, and combined stimulation was equivalent to the linear sum of optical and electrical activation alone. PC boutons expressing ChR2–YFP were stimulated by pulses of 473 nm light (0.5–1 ms, 160 mW/mm2) from an LED (Thorlabs) through a 60× objective producing an 80-μm diameter spot of light over the cell body. A stimulus electrode was placed in the white matter and converging PC axons were located as in other experiments. Randomized trials of single optical, electrical, or closely timed (0–2 ms) paired optical and electrical stimuli were applied every 5 s. When more than 70% of the electrically evoked component could be occluded by optical stimulation, and could be consistently recruited, those fibres were then activated using high-frequency electrical stimulation. Following electrical trains, occlusion trials were performed again to confirm that the same set of fibres was stimulated throughout all trials. At the end of experiments, a low concentration of SR-95531 (300–800 nM) was washed in and occlusion trials were again repeated to ensure occlusion was not the result of poor voltage clamp. In a subset of experiments, light pulses were also delivered over the stimulus electrode instead of the cell body, producing similar results.

Immunohistochemistry

Mice of both sexes (P21–32 unless otherwise indicated) were anaesthetized with ketamine/xylazine (100/10 mg/kg) and transcardially perfused with PBS followed by 4% paraformaldehyde (PFA) in PBS. To determine the expression of Syt7, Syt2, calbindin and VGAT specifically in PCs, we also perfused one mouse (at P43) in which PC synapses were labelled with TdTomato (Pcp2-Cre(Jdhu) × Synaptophysin–TdTomato, Extended Data Fig. 2). Brains were removed and post-fixed in PFA overnight. Sagittal sections of the cerebellum or coronal sections of the brainstem (50 μm) were then permeabilized (0.2% triton X-100 in PBS) for 10 min and blocked for 1 h (4% normal goat serum in 0.1% triton X-100) at room temperature. Slices were then incubated overnight at 4 °C with primary antibodies (mouse anti-Syt7 targeting the C2A domain, UC Davis/NIH NeuroMab Facility, clone N275/14, RRID: AB_11030371, 1 μg/ml, 1:100; rabbit anti-calbindin D28K, Millipore Ab1778, 1:200; mouse anti-Syt2, Zirc znp-1, 1:200; guinea-pig anti-VGAT, Synaptic Systems 131004, 1 μg/ml, 1:500; guinea-pig anti-VGLUT2, Synaptic Systems 135404, 1 μg/ml, 1:1,200). To prevent background when co-staining with VGLUT2 and Syt7, Syt7 primary antibodies were applied alone overnight at 4 °C, then VGLUT2 primary antibodies alone for 2 h at room temperature. Following primary antibodies, slices were incubated with secondary antibodies for 2 h at room temperature (anti-rabbit-AlexaFluor488, Abcam ab150077; anti-guinea-pig-AlexaFluor488, Abcam ab150185; anti-mouse-AlexaFluor647, Abcam ab150115). For experiments comparing ages or genotypes, all tissue was stained and processed in parallel. Z-stacks of each sample were collected using an Olympus Fluoview1000 confocal microscope using the same settings across ages and genotypes, and processed identically in ImageJ. For each genotype and age, identical anatomical locations and tissue depth were selected for presentation.

Modelling

In a previous study we modelled the PC–DCN synapse5. Models used previously to explain data in young animals30 were reproduced, but could not fit the data from P21–32 mice5. Several other types of models were also attempted but failed to accurately account for data observed in juvenile wild-type mice. We found that a two-pool model fit all experimental observations5. We extended this model to Syt7 knockout mice (Extended Data Fig. 9). In this model there are two pools of vesicles, consisting of, respectively, N1 and N2 vesicles, with initial probability of release P1 and P2 and time constants of recovery τR1 and τR2. P1 stays constant whereas P2 increases as a result of facilitation, F2, which has the frequency dependence that was determined experimentally. All of these parameters were constrained by extensive experimental studies for wild-type mice5. Once parameters for wild-type data were determined, F2 was set to 0 to model Syt7 knockout mice. Minor adjustments (<15% change) were made to some parameters to fit the data more accurately, with the exception of τR1, which was reduced by 55% to conform to the observed recovery from depression in Syt7 knockouts (Extended Data Fig. 5). This change in τR1 had a negligible effect on steady-state IPSC amplitudes because Pool 1 still depleted rapidly. Parameters for modelling wild-type mice were: N1/N2 = 0.35; P1 = 0.20; τR1 = 7.7 s; P2 = 0.025; τR2 = 0.2 s; F2 = 0.04; τFdeact = 0.1 s. Parameters for modelling Syt7 knockout mice were: N1/N2 = 0.36; P1 = 0.23; τR1 = 3.5 s; P2 = 0.025; τR2 = 0.2 s; F2 = 0.

Data availability

The data, computational models and analysis scripts that support the findings are available upon reasonable request from the corresponding author.