- Research
- Open access
- Published:
Convergent depression of activity-dependent bulk endocytosis in rodent models of autism spectrum disorder
Molecular Autism volume 16, Article number: 26 (2025)
Abstract
Background
The key pathological mechanisms underlying autism spectrum disorder (ASD) remain relatively undetermined, potentially due to the heterogenous nature of the condition. Targeted studies of a series of monogenic ASDs have revealed postsynaptic dysfunction as a central conserved mechanism. Presynaptic dysfunction is emerging as an additional disease locus in neurodevelopmental disorders; however, it is unclear whether this dysfunction drives ASD or is an adaptation to the altered brain microenvironment.
Methods
To differentiate between these two competing scenarios, we performed a high content analysis of key stages of the synaptic vesicle lifecycle in primary neuronal cultures derived from a series of preclinical rat models of monogenic ASD. These five independent models (Nrxn1+/−, Nlgn3−/y, Syngap+/−, Syngap+/Δ−GAP, Pten+/−) were specifically selected to have perturbations in a diverse palette of genes that were expressed either at the pre- or post-synapse. Synaptic vesicle exocytosis and cargo trafficking were triggered via two discrete trains of activity and monitored using the genetically-encoded reporter synaptophysin-pHluorin. Activity-dependent bulk endocytosis was assessed during intense neuronal activity using the fluid phase marker tetramethylrhodamine-dextran.
Results
Both synaptic vesicle fusion events and cargo trafficking were unaffected in all models investigated under all stimulation protocols. However, a key convergent phenotype across neurons derived from all five models was revealed, a depression in activity-dependent bulk endocytosis.
Limitations
The study is exclusively conducted in primary cultures of hippocampal neurons; therefore, the impact on neurons from other brain regions or altered brain microcircuitry was not assessed. No molecular mechanism has been identified for this depression.
Conclusion
This suggests that depression of activity-dependent bulk endocytosis is a presynaptic homeostatic mechanism to correct for intrinsic dysfunction in ASD neurons.
Background
Autism spectrum disorder (ASD) impacts up to 1 in 150 children, with intellectual disability (ID) being a frequent co-morbidity [1]. Despite this high prevalence, the mechanisms that result in brain dysfunction at the molecular, cell and circuit level in these neurodevelopmental disorders are largely undetermined. The study of monogenic ASD provides an opportunity to decipher these key mechanisms via the identification of a molecular locus [2]. Interestingly, a number of the most prevalent genetic causes of these disorders concentrate on genes that control synaptic events [3]. In support, multiple preclinical monogenic ASD models display defects at the synaptic and circuit level, consistent with perturbations observed in humans with similar mutations [4, 5].
The synapse is comprised of the presynapse, which releases chemical neurotransmitters during invasion of action potentials (APs), and the postsynapse which integrates this input and adapts output via a series of plastic changes. Postsynaptic alterations are commonly observed in rodent models of ASD, supporting the view that this subcellular structure is a physical convergence point in the genesis and expression of ASD [6]. Key proteins include synaptic Ras GTPase-activating protein 1 (SynGAP1) and fragile X messenger ribonucleoprotein (FMRP), which control synaptic plasticity via excitatory α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor trafficking and protein translation downstream from postsynaptic metabotropic receptors respectively [7,8,9]. Furthermore, dysfunction in cell adhesion molecules such as presynaptic neurexins and postsynaptic neuroligins also commonly result in ASD via incorrect stabilisation and maintenance of synapse function [10,11,12]. Therefore, dysfunction in a series of genes, including those encoding key postsynaptic molecules is a highly prevalent cause of ASD.
In contrast to the postsynapse, presynaptic dysfunction in neurodevelopmental disorders is still relatively under-researched. This is in spite of a cohort of key genes essential for neurotransmitter release having been identified as causal in conditions such as epilepsy, ID and ASD [13, 14]. Neurotransmitter release occurs in response to the activity-dependent influx of calcium, via voltage-gated channels, which triggers the fusion of neurotransmitter-containing synaptic vesicles (SVs) [15]. The functional pool of SVs is relatively small in typical central nerve terminals, making their efficient regeneration essential for the maintenance of neurotransmission. This is mediated by a series of endocytosis modes, which are recruited in both time and space by different patterns of neuronal activity [16, 17]. SV cargo retrieval is dependent on clathrin-mediated endocytosis (CME), which can occur at either the presynaptic plasma membrane or on intracellular endosomes generated by either ultrafast endocytosis (UFE) or activity-dependent bulk endocytosis (ADBE [18,19,20]. Ultrafast endocytosis saturates rapidly during AP trains [21], whereas ADBE is the dominant endocytosis mode during intense neuronal activity [22, 23].
We recently discovered a selective defect in ADBE in neurons lacking FMRP [24], which translated into a decrease in presynaptic performance during periods of high activity. Since this phenotype could be corrected via agonism of big potassium (BK) channels, we hypothesized that the depression of ADBE was an adaptation to the hyperexcitability observed in Fmr1−/y neurons and circuits [25,26,27]. In this study, we tested this hypothesis by examining SV recycling in a cohort of five independent monogenic rat ASD models the majority of which display hyperexcitability at either the cell or circuit level. We discovered that neurons from all models displayed depressed ADBE, with no obvious deficit in SV exocytosis or cargo trafficking. This therefore supports the hypothesis that a reduction in ADBE is a specific homeostatic adaptation to intrinsic synaptic dysfunction in ASD models.
Methods
Materials
Unless otherwise specified, all cell culture reagents were obtained from Invitrogen (Paisley, UK). Foetal bovine serum was from Biosera (Nuaille, France). Papain was obtained from Worthington Biochemical (Lakewood, NJ, USA). All other reagents were obtained from Sigma-Aldrich (Poole, UK) unless specified. Rabbit anti-SV2A was obtained from Abcam (Cambridge, UK; ab32942 RRID: AB_778192). Anti-rabbit Alexa Fluor 488 (A11008 RRID: AB_143165) was obtained from Invitrogen (Paisley, UK). Synaptophysin-pHluorin (sypHy) was a gift from Prof. L. Lagnado (University of Sussex, UK).
Rat models
Procedures were performed in accordance with the UK Animal (Scientific Procedures) Act 1986, under Project and Personal Licence authority and were approved by the Animal Welfare and Ethical Review Body at the University of Edinburgh (Home Office project licence – 7008878). Similarly, procedures were conducted in accordance with protocols approved by the Institutional Animal Ethics Committee of Institute for Stem Cell Science and Regenerative Medicine, Bangalore. All animals were killed by Schedule 1 procedures in accordance with UK Home Office Guidelines; adults were killed by exposure to CO2 followed by decapitation, whereas embryos were killed by decapitation followed by destruction of the brain. In Edinburgh, rats were housed on a 12/12 h light/dark cycle with a 21 ± 2 °C room temperature and food/water ad libitum. In Bangalore, rats were maintained on a 14 h light/10 h dark cycle with ad-libitum access to diet and water.
All transgenic rats in this study were generated by Horizon Discovery (now Envigo).
Sprague–Dawley Nlgn3−/y transgenic rats were created as previously described [28]. Long Evans-SGem1/PWC, referred to in the manuscript as Syngap+/− rats, were created as described [29]. Colony founders of Long Evans-SGem2/PWC, referred to as Syngap+/Δ−GAP, were produced by zinc finger nuclease-mediated deletion of the GAP domain of Syngap as described [30, 31]. Sprague Dawley Nrxn1tm1sage rats, referred to as Nrxn1+/− rats [32, 33] were purchased from Horizon Discovery. Sprague Dawley Pten+/− rats were purchased from Horizon Discovery and were generated as described previously [34].
Hippocampal cultures
Hippocampi from each embryo (e18.5-e19.5) were processed separately to avoid contamination across genotypes. For rats with an X chromosome mutation (Nlgn3), male embryos were taken for hippocampal dissection. For all other rat lines embryos of both sexes were used.
Dissociated primary hippocampal cultures were prepared from embryos as previously described [24]. Briefly, isolated hippocampi were digested in a 10 U/mL papain solution (Worthington Biochemical, LK003178) at 37 °C for 20 min. The papain was then neutralised using DMEM F12 (ThermoFisher Scientific, 21331-020) supplemented with 10% Foetal bovine serum (BioSera, S1810-500) and 1% penicillin/streptomycin (ThermoFisher Scientific, 15140-122). Cells were triturated to form a single cell suspension and plated at 5 × 104 cells (with the exception of single cell tetramethylrhodamine (TMR)-dextran uptake experiments, 2.5 × 104 cells) per coverslip on laminin (10 µg/ mL; Sigma Aldrich, L2020) and poly-D‐lysine (Sigma Aldrich, P7886) coated 25 mm glass coverslips (VWR International Ltd, Lutterworth, UK). Cultures were maintained in Neurobasal media (ThermoFisher Scientific, 21103-049) supplemented with 2% B-27 (ThermoFisher Scientific, 17504-044), 0.5 mM L‐glutamine (ThermoFisher Scientific, 25030-024) and 1% penicillin/streptomycin. After 2–3 days in vitro (DIV), 1 µM of cytosine arabinofuranoside (Sigma Aldrich, C1768) was added to each well to inhibit glial proliferation. Hippocampal neurons were transfected with sypHy at DIV 7 using Lipofectamine 2000 (ThermoFisher Scientific, 11668027) prior to imaging at DIV 13–15.
High content screening of SV recycling using sypHy
SypHy-transfected neurons were visualised at 500 nm band pass excitation with a 515 nm dichroic filter and a long-pass > 520 nm emission filter on a Zeiss Axio Observer D1 inverted epifluorescence microscope (Cambridge, UK). Images were captured using an AxioCam 506 mono camera (Zeiss) with a Zeiss EC Plan Neofluar 40x/1.30 oil immersion objective. Image acquisition was performed using Zen Pro software (Zeiss). Hippocampal cultures were mounted in a Warner Instruments (Hamden, CT, USA) imaging chamber with embedded parallel platinum wires (RC-21BRFS) while undergoing constant perfusion with imaging buffer (119 mM NaCl, 2.5 mM KCl, 2 mM CaCl2, 2 mM MgCl2, 25 mM HEPES, 30 mM glucose at pH 7.4, supplemented with 10 µM 6-cyano-7-nitroquinoxaline-2,3-dione (Abcam, Cambridge, UK, ab120271) and 50 µM DL-2-Amino-5-phosphonopentanoic acid (Abcam, Cambridge, UK, ab120044). Images were acquired at 4 s intervals. After acquisition of a 1 min baseline, neurons were challenged with an impermeant acidic buffer (69 mM NaCl, 2.5 mM KCl, 2 mM CaCl2, 2 mM MgCl2, 25 mM MES, 30 mM glucose at pH 5.5) for 1 min. After returning to imaging buffer for 2 min, cultures were challenged with two field stimuli (delivered using a Digitimer LTD MultiStim system-D330 stimulator, current output 100 mA, current width 1 ms) separated by 5 min. Neurons were first stimulated at 10 Hz for 30 s (300 APs) then 40 Hz for 10 s (400 APs). Finally, after a 3 min recovery period, alkaline buffer (50 mM NH4Cl substituted for 50 mM NaCl in imaging buffer) was used to reveal the maximal pHluorin response.
Time traces were analysed using the FIJI distribution of Image J (National Institutes of Health). Images were aligned using the Rigid body model of the StackReg plugin (https://imagej.net/StackReg). Nerve terminal fluorescence was measured using the Time Series Analyser plugin (https://imagej.nih.gov/ij/plugins/time-series.html). Regions of interest (ROIs) 5 pixels in diameter were placed over nerve terminals that responded to the electrical stimulus. A response trace was calculated for each cell by averaging the individual traces from each selected ROI. For sypHy time traces, fluorescence decay time constants (tau, τ, s) were calculated by fitting a monoexponential decay curve to data from the time point after the end of electrical stimulation.
Tetramethylrhodamine (TMR)-dextran uptake
TMR-dextran (ThermoFisher Scientific, D1842) uptake was performed as described previously [24]. Neurons were mounted on a Zeiss Axio Observer D1 microscope as described above before challenging with 400 action potentials (40 Hz) in the presence of 50 µM of TMR-dextran (40,000 MW) in imaging buffer. The TMR-dextran solution was immediately washed away after stimulation terminated, and images were acquired using 556/25 nm excitation and 630/98 nm emission bandpass filters (Zeiss) while undergoing constant perfusion. Per coverslip of cells, 3–6 different fields of view were imaged. The TMR-dextran puncta in each image were quantified using the Analyze Particles plugin of Image J (NIH, https://imagej.nih.gov/ij/developer/api/ij/plugin/filter/ParticleAnalyzer.html) to select and count particles of 0.23–0.91 µm2. For all experiments, for each condition, at least one unstimulated coverslip was imaged to correct for the background level of TMR-dextran uptake.
SV2A immunofluorescence staining
Immunofluorescence staining was performed as previously described [24]. Briefly, neurons were fixed with 4% paraformaldehyde (Sigma Aldrich, 47608) in PBS for 15 min. Excess paraformaldehyde was quenched with 50 mM NH4Cl in PBS. Cells were then permeabilized in 1% bovine serum albumin (BSA; Roche Diagnostics GmbH, Germany, 10735078001) in PBS-Triton 0.1% solution for 5 min and blocked in 1% BSA in PBS at room temperature for 1 h. After blocking, cells were incubated in rabbit anti-SV2A (1:200 dilution) for 1 h, after which the cultures were washed with PBS and incubated with fluorescently conjugated secondary antibodies (anti-rabbit Alexa Fluor 488; 1:1000 dilution) for 1 h. The coverslips were mounted on slides for imaging with FluorSave (Millipore, Darmstadt, Germany, 345789). SV2A puncta were visualised at 500 nm band pass excitation with a 515 nm dichroic filter and a long-pass > 520 nm emission filter on a Zeiss Axio Observer D1 inverted epifluorescence microscope (Cambridge, UK). Images were captured using an AxioCam 506 mono camera (Zeiss) with a Zeiss EC Plan Neofluar 40x/1.30 oil immersion objective. SV2A puncta in each image were quantified using the Analyze Particles plugin of Image J to select and count particles of 0.23–3.18 µm2.
Preparation of synaptosomes
Rat brains were sliced in ice cold ACSF (124 mM NaCl, 2.5 mM KCl, 1.2 mM NaH2PO4, 25 mM NaHCO3, 20 mM Glucose, 2 mM CaCl2, 1 mM MgCl2) using a vibrating blade microtome (VT1200S, Leica). Hippocampi were dissected in ice-cold ACSF from P28 Nrxn1+/−, Nlgn3−/y and P60 Pten+/− rats with wild-type male littermate controls. Tissue from three rats was pooled together to produce one preparation of synaptosomes. A total tissue lysate was prepared in a sucrose/EDTA buffer (0.32 M Sucrose, 1 mM EDTA, 5 mM Tris, 4oC, pH 7.4) using a pre-chilled motorized Teflon glass homogenizer, followed by centrifugation at 1075 g for 10 min at 4oC. Pure synaptosomes were isolated by adding supernatant on top of a discontinuous Percoll-density gradient (3% top, 10% middle, and 23% bottom; Percoll, P1644, Sigma-Aldrich, UK) and centrifuged at 47,807 g for 8 min at 4oC. The fraction between 23% and 10% was collected and re-suspended in HEPES-Buffered-Krebs (118.5 mM NaCl, 4.7 mM KCl, 1.18 mM MgSO4, 10 mM Glucose, 1 mM Na2HPO4, 20 mM HEPES, pH 7.4) and synaptosomes were pelleted by centrifugation at 20,198 g for 15 min at 4oC. Synaptosome pellets were dissolved in RIPA buffer (50 mM, Tris-HCl pH 7.4, 150 mM NaCl, 1% Triton, 0.5% Na-deoxycholate, 0.1% SDS, 1 mM sodium-orthovanadate, 1 mM PMSF, 1 mM EDTA) supplemented with protease inhibitors (Roche complete mini EDTA-free protease inhibitor cocktail 4693159001, Sigma-Aldrich, UK) and phosphatase inhibitors (cocktail II P5726, cocktail III P0044, Sigma-Aldrich, UK). Protein levels were estimated by a MicroBCA Assay (Pierce BCA protein estimation kit, Cat # 23225, Thermofisher, UK).
Western blotting
Approximately 10 µg of each protein extract was separated on a precast gradient gel (NuPAGE 4–12% Bis-Tris Protein Gels, NP0336BOX, Thermofisher) and transferred to PVDF membrane (GE10600022, Thermofisher, UK). After protein transfer, membranes were stained with a reversible protein stain kit (memcode 24585, Thermofisher Scientific) according to the manufacturer’s instructions. The exception was for Pten+/−, where nitrocellulose membrane (1620115, Bio-Rad) and reversible protein stain kit (memcode 24580, theromofisher Scientifc) were used. All membranes were blocked with Odyssey Blocking Buffer (Cat #-927-50003, LI-COR Biotech.) for 1-hour at room temperature followed by incubation at 4oC overnight with primary antibodies (PTEN, 1:1000, Cat # sc393186, RRID AB_2923140, Santa Cruz; NLGN3, 1:1000, Cat # 129113, RRID AB_2619816, Synaptic Systems; NRXN1, 1:1000, Cat # 175103, RRID AB_10697816, Synaptic Systems). Membranes were washed with TBST (0.1% Tween 20) followed by a 1-hour incubation with secondary antibodies (IRDye 800CW Goat anti-Rabbit IgG- 1: 10,000, Cat # 925-32211, RRID AB_2651127; IRDye 680LT Goat anti Mouse IgG- 1: 10,000, Cat # 925-68020, RRID AB_2687826, LI-COR Biotechnology) at room temperature. After washing the membranes with TBST, immunoblots were dried and digitally scanned by using a CLx Odyssey Infrared Imaging System, LI-COR, UK Ltd. The density of individual bands was calculated using Licor Image Studio Lite software. Each value was normalised to total protein and then to their control littermates. Data analysis for immunoblotting was done using Graphpad Prism version 6.0e software.
Experimental design and statistical analysis
Microsoft Excel (Microsoft, Washington, USA) and Prism 6 software (GraphPad software Inc., San Diego USA) were used for data processing and analysis. The experimenter was blinded to genotype during data acquisition and analysis. For all figures, results are presented with error bars as ± SEM, and the n for each condition represents the number of coverslips imaged. For all assays, cells were obtained from at least three independent cultures. In sypHy assays, at least 10 ROIs were collected from each coverslip. The number of ROIs examined was comparable for all experiments. Normality was determined using a D’Agostino & Pearson omnibus normality test. For comparison between genotypes, an unpaired Students t test was performed where data followed a normal distribution, with a Mann-Whitney test performed for those that did not. For comparison between > 2 conditions, a one-way ANOVA was performed. Results were corrected for multiple comparisons. Full statistical reporting is provided in Table 1.
High content monitoring of SV recycling. Hippocampal neurons were transfected with synaptophysin-pHluorin (sypHy) on day in vitro (DIV) 7 and imaged at DIV 13–15. Transfected neurons were challenged with a pulse of impermeant acidic buffer for 1 min (to reveal the surface fraction of sypHy, shaded pink). After returning to imaging buffer for 2 min, neurons were stimulated with a train of 300 action potentials (10 Hz) followed by a 5 min rest period. After this rest period neurons were stimulated with a further train of 400 action potentials (40 Hz). Finally, after a further 3 min neurons were exposed to an alkaline buffer (NH4Cl, to reveal the total SV pool, blue shaded region). Stimulation is indicated by bars. Mean trace displays the average sypHy fluorescent response of wild-type neurons ± SEM. Traces are ΔF/F0 and are presented as a fraction of the total SV pool
Nrxn1+/− neurons display depressed ADBE. A-C) Hippocampal synaptosome lysates from either wild-type (WT) or Nrxn1+/− rats were probed for the presence of Neurexin-1 (Nrxn1, A) and total protein (B). (C) Quantification of Nrxn1 levels in both are displayed, normalised to total protein ± SEM, n = 3 independent synaptosome preparations for both WT and Nrxn1+/−, * p = 0.0365, unpaired t test. D-J) Hippocampal neurons derived from either WT or Nrxn1+/− rat embryos were transfected with synaptophysin-pHluorin (sypHy) after 7 days in vitro (DIV) and imaged at DIV 13–15. D, H) Mean sypHy fluorescence traces of WT and Nrxn1+/− hippocampal neurons normalised to either the total SV pool as revealed by NH4Cl (D) or peak fluorescence during electrical stimulation (H) ± SEM. E) Mean sypHy surface fraction presented as a percentage of the total SV pool ± SEM. F, G) Mean peak sypHy response in response to either 10 Hz (F) or 40 Hz (G) action potential trains ± SEM. (I, J) Mean sypHy retrieval time constants (t) in response to either 10 Hz (I) or 40 Hz (J) action potential trains ± SEM. For D-Jn = 9 coverslips for both WT and Nrxn1+/− from 3 independent cultures. (K, L) Primary hippocampal cultures derived either WT or Nrxn1+/− rat embryos were challenged with a train of action potentials (40 Hz, 10 s) in the presence of tetramethylrhodamine (TMR)-dextran (50 µM). TMR-dextran was immediately washed away and the number of TMR-dextran puncta were counted. (K) Representative images of TMR-dextran uptake in WT and Nrxn1+/− cultures. Scale bar = 50 μm. L) Mean number of TMR-dextran puncta per field of view normalised to WT ± SEM (n = 13 coverslips from 3 independent cultures for WT and Nrxn1+/−). (M) Primary hippocampal cultures derived from either WT or Nrxn1+/− rat embryos were fixed at DIV13-15 and stained for the presence of SV2A. (N) Mean number of SV2A puncta per field of view normalised to WT ± SEM (WT n = 12 coverslips, Nrxn1+/−n = 15 coverslips from 3 independent cultures). In all cases an unpaired two-sided students t test was performed, ** p = 0.0037, unpaired t test
Nlgn3−/y neurons display depressed ADBE. A-C) Hippocampal synaptosome lysates from either wild-type (WT) or Nlgn3−/y rats were probed for the presence of Neuroligin-3 (Nlgn3, A) and total protein (B). (C) Quantification of Nlgn3 levels in both are displayed, normalised to total protein ± SEM, n = 4 independent synaptosome preparations for both WT and Nlgn3−/y, *** p < 0.0001, unpaired t test. (D-J) Hippocampal neurons derived from either WT or Nlgn3−/y rat embryos were transfected with synaptophysin-pHluorin (sypHy) after 7 days in vitro (DIV) and imaged at DIV 13–15. (D, H) Mean sypHy fluorescence traces of WT and Nlgn3−/y hippocampal neurons normalised to either the total SV pool as revealed by NH4Cl (D) or peak fluorescence during electrical stimulation (H) ± SEM. (E) Mean sypHy surface fraction presented as a percentage of the total SV pool ± SEM. F, G) Mean peak sypHy response in response to either 10 Hz (F) or 40 Hz (G) action potential trains ± SEM. (I, J) Mean sypHy retrieval time constants (τ) in response to either 10 Hz (I) or 40 Hz (J) action potential trains ± SEM. For D-J WT n = 11 coverslips, Nlgn3−/yn = 14 from 3 independent cultures. (K, L) Primary hippocampal cultures derived either WT or Nlgn3−/y rat embryos were challenged with a train of action potentials (40 Hz, 10 s) in the presence of tetramethylrhodamine (TMR)-dextran (50 µM). TMR-dextran was immediately washed away and the number of TMR-dextran puncta were counted. (K) Representative images of TMR-dextran uptake in WT and Nlgn3−/y cultures. Scale bar = 50 μm. (L) Mean number of TMR-dextran puncta per field of view normalised to WT ± SEM (WT n = 11 coverslips, Nlgn3−/yn = 12 from 3 independent cultures). (M) Primary hippocampal cultures derived from either WT or Nlgn3−/y rat embryos were fixed at DIV13-15 and stained for the presence of SV2A. (N) Mean number of SV2A puncta per field of view normalised to WT ± SEM (WT n = 14 coverslips, Nlgn3−/yn = 15 coverslips from 3 independent cultures). In all cases an unpaired two-sided students t test was performed, except E, F and J, * p = 0.0112, unpaired t test
Syngap−/−neurons display depressed ADBE. Hippocampal neurons derived from either wild-type (WT), Syngap+/− or Syngap−/− rat embryos were transfected with synaptophysin-pHluorin (sypHy) after 7 days in vitro (DIV) and imaged at DIV 13–15. A, E) Mean sypHy fluorescence traces of WT, Syngap+/− or Syngap−/− hippocampal neurons normalised to either the total SV pool as revealed by NH4Cl (A) or peak fluorescence during electrical stimulation (E) ± SEM. B) Mean sypHy surface fraction presented as a percentage of the total SV pool ± SEM. C, D) Mean peak sypHy response in response to either 10 Hz (C) or 40 Hz (D) action potential trains ± SEM. F, G) Mean sypHy retrieval time constants (τ) in response to either 10 Hz (F) or 40 Hz (G) action potential trains ± SEM. For A-G, WT n = 12 coverslips, Syngap+/−n = 11 and Syngap−/−n = 12 from 3 independent cultures. H-I) Primary hippocampal cultures derived either wild-type (WT), Syngap+/− or Syngap−/− rat embryos were challenged with a train of action potentials (40 Hz, 10 s) in the presence of tetramethylrhodamine (TMR)-dextran (50 µM). TMR-dextran was immediately washed away and the number of TMR-dextran puncta were counted. H) Representative images of TMR-dextran uptake in WT, Syngap+/− or Syngap−/− cultures. Scale bar = 50 μm. I) Mean number of TMR-dextran puncta per field of view normalised to WT ± SEM (WT n = 15 coverslips, Syngap+/− and Syngap−/−n = 16 from 3 independent cultures). J) Primary hippocampal cultures derived from either WT, Syngap+/− or Syngap−/− rat embryos were fixed at DIV13-15 and stained for the presence of SV2A. K) Mean number of SV2A puncta per field of view normalised to WT ± SEM (WT and Syngap+/−n = 15 coverslips, Syngap−/−n = 13 from 3 independent cultures). In all cases a one-way ANOVA was performed, ** p = 0.01
SyngapΔ − GAP/Δ−GAP neurons display depressed ADBE. Hippocampal neurons derived from either wild-type (WT), Syngap+/Δ−GAP or SyngapΔ − GAP/Δ−GAP rat embryos were transfected with synaptophysin-pHluorin (sypHy) after 7 days in vitro (DIV) and imaged at DIV 13–15. A, E) Mean sypHy fluorescence traces of WT, Syngap+/Δ−GAP or SyngapΔ − GAP/Δ−GAP hippocampal neurons normalised to either the total SV pool as revealed by NH4Cl (A) or peak fluorescence during electrical stimulation (E) ± SEM. B) Mean sypHy surface fraction presented as a percentage of the total SV pool ± SEM. C, D) Mean peak sypHy response in response to either 10 Hz (C) or 40 Hz (D) action potential trains ± SEM. F, G) Mean sypHy retrieval time constants (τ) in response to either 10 Hz (F) or 40 Hz (G) action potential trains ± SEM. For A-G, WT n = 14 coverslips, Syngap+/Δ−GAPn = 12 and SyngapΔ − GAP/Δ−GAPn = 10 from 3 independent cultures. H-I) Primary hippocampal cultures derived either wild-type (WT), Syngap+/Δ−GAP or SyngapΔ − GAP/Δ−GAP rat embryos were challenged with a train of action potentials (40 Hz, 10 s) in the presence of tetramethylrhodamine (TMR)-dextran (50 µM). TMR-dextran was immediately washed away and the number of TMR-dextran puncta were counted. H) Representative images of TMR-dextran uptake in WT, Syngap+/Δ−GAP or SyngapΔ − GAP/Δ−GAP cultures. Scale bar = 50 μm. I) Mean number of TMR-dextran puncta per field of view normalised to WT ± SEM (WT n = 8 coverslips, Syngap+/Δ−GAP and SyngapΔ − GAP/Δ−GAPn = 9 from 3 independent cultures). J) Primary hippocampal cultures derived from either WT, Syngap+/Δ−GAP or SyngapΔ − GAP/Δ−GAP rat embryos were fixed at DIV13-15 and stained for the presence of SV2A. K) Mean number of SV2A puncta per field of view normalised to WT ± SEM (WT n = 10 coverslips, Syngap+/Δ−GAPn = 11 and SyngapΔ − GAP/Δ−GAPn = 13 from 3 independent cultures). In all cases a one-way ANOVA was performed, * p = 0.017
Results
High content screening of SV recycling
To determine whether the observed depression of ADBE in Fmr1−/y neurons [24] was a wider synaptic signature of ASD, we examined SV recycling using a battery of optical assays in primary cultures of hippocampal neurons derived from monogenic rat ASD models. A high content protocol was designed to capture a series of different SV recycling parameters using the genetically-encoded reporter sypHy (Fig. 1). SypHy consists of the abundant SV cargo protein synaptophysin, that has a pH-sensitive EGFP (pHluorin) inserted into an intralumenal loop [35]. It reports the pH of its immediate environment, with fluorescence quenched in the acidic SV interior and unquenched during SV fusion. Therefore, the extent of SV fusion can be estimated by the evoked increase in fluorescence during stimulation. SypHy is then retrieved from the plasma membrane and packaged to SVs, which are acidified to permit neurotransmitter filling. SV cargo retrieval is rate limiting when compared to acidification [35, 36] but see [37], meaning that the kinetics and extent of the former can be estimated from the fluorescence decrease after stimulation terminates.
Pten+/− neurons display depressed ADBE. A-C) Hippocampal synaptosome lysates from either wild-type (WT) or Pten+/− rats were probed for the presence of PTEN (A) and total protein (B). (C) Quantification of PTEN levels in both are displayed, normalised to total protein ± SEM, n = 5 independent synaptosome preparations for both WT and Pten+/−, * p = 0.0178, unpaired t test. D-J) Hippocampal neurons derived from either WT or Pten+/− rat embryos were transfected with synaptophysin-pHluorin (sypHy) after 7 days in vitro (DIV) and imaged at DIV 13–15. D, H) Mean sypHy fluorescence traces of WT and Pten+/− hippocampal neurons normalised to either the total SV pool as revealed by NH4Cl (D) or peak fluorescence during electrical stimulation (H) ± SEM. E) Mean sypHy surface fraction presented as a percentage of the total SV pool ± SEM. F, G) Mean peak sypHy response in response to either 10 Hz (F) or 40 Hz (G) action potential trains ± SEM. I, J) Mean sypHy retrieval time constants (τ) in response to either 10 Hz (I) or 40 Hz (J) action potential trains ± SEM. For D-J, WT n = 15 coverslips, Pten+/−n = 19 from 3 independent cultures. K, L) Primary hippocampal cultures derived either wild-type (WT) or Pten+/− rat embryos were challenged with a train of action potentials (40 Hz, 10 s) in the presence of tetramethylrhodamine (TMR)-dextran (50 µM). TMR-dextran was immediately washed away and the number of TMR-dextran puncta were counted. K) Representative images of TMR-dextran uptake in WT and Pten+/− cultures. Scale bar = 50 μm. L) Mean number of TMR-dextran puncta per field of view normalised to WT ± SEM (WT n = 24 coverslips, Pten+/−n = 20 from 3 independent cultures)
First, sypHy-expressing neurons were exposed to an impermeant acidic buffer to quench sypHy fluorescence on the presynaptic plasma membrane, since the altered surface fraction of SV cargo can be indicative of chronic dysfunction in either their clustering or retrieval [38, 39]. Second, after recovery of the sypHy response to the acid buffer, neurons were challenged with two sequential AP trains (10 Hz, 30 s or 40 Hz 10 s) to reveal whether SV fusion or cargo retrieval was disrupted during periods of low and high activity. Finally, neurons were exposed to buffer containing NH4Cl, to unquench all sypHy within acidic compartments, to estimate the extent of SV fusion as a proportion of the total SV pool. In parallel, activity-dependent uptake of the large fluid phase marker TMR-dextran (40 kDa), was measured, since it exclusively reports ADBE due to size exclusion from SVs [40].
ADBE is depressed in both Nrxn1 +/− and Nlgn3 −/y neurons
We first tested our high content imaging protocol on two preclinical rodent models - Nrxn1+/− and Nlgn3−/y rats. These rat models replicate the genetic alterations in the human NRXN1 or NLGN3 genes that are primarily responsible for ASD [10, 41], due to haploinsufficiency of the Nrxn1 gene, or deletion of the X-linked Nlgn3 gene respectively. An additional reason for investigating these rat models is that an unbiased screen of molecules on bulk endosomes generated via ADBE revealed that synaptic adhesion molecules were disproportionately represented [42]. Furthermore, the gene products of Nrxn1 and Nlgn3, Neurexin-1 and Neuroligin-3, were present on bulk endosomes purified via two independent approaches [42]. This suggests the potential for a direct mechanistic link between adhesion molecules, ASD and ADBE. Therefore, we first determined whether these construct-valid rat models of ASD displayed dysfunctional SV recycling and ADBE.
Neurexins are presynaptic cell adhesion molecules that stabilise synapses via trans-synaptic interactions with postsynaptic partners such as neuroligins [41]. Their conditional knockout has pleotropic effects on evoked neurotransmitter release, with the primary deficit being an inefficient coupling of voltage-gated calcium influx to SV fusion [43]. Interestingly, patient-derived NRXN1+/− induced pluripotent neurons display an intrinsic hyperexcitability when investigated [44]. Since ADBE is also triggered via activity-dependent calcium influx [45, 46], we first determined the impact of Nrxn1 haploinsufficiency on ADBE and SV recycling in primary hippocampal cultures from the Nrxn1+/− rat [32, 33]. Western blotting confirmed a reduction in Neurexin-1 levels to approximately 50% (Fig. 2A-C). When the high content sypHy assay was performed on Nrxn1+/− neurons and wild-type littermate controls, no significant difference in the extent of the evoked sypHy response was observed to either AP train (10–40 Hz, Fig. 2D, F-H). This suggests that SV fusion was not significantly impacted by the absence of a single Nrxn1 allele. Furthermore, the kinetics and extent of SV cargo retrieval in Nrxn1+/− neurons were not significantly impacted when compared to wild-type neurons (Fig. 2I, J). This absence of effect was corroborated by the comparable extent of sypHy surface fraction between wild-type and Nrxn1+/− neurons (Fig. 2E). However, when ADBE was assessed, a significant reduction in the number of nerve terminals displaying activity-dependent uptake of TMR-dextran was observed in Nrxn1+/− neurons when compared to wild-type littermate controls (Fig. 2K, L). This decrease in TMR-dextran puncta number was not due to a reduction in nerve terminals, since staining with the SV marker SV2A revealed no change in this parameter when Nrxn+/− and wild-type neurons were compared (Fig. 2M, N). Therefore Nrxn1+/− neurons display a selective deficit in ADBE, but not SV fusion or cargo retrieval.
Neurexin-1 forms complexes with postsynaptic adhesion molecules to drive synaptogenesis and synaptic stability [12]. Key proteins in this context are the neuroligin family, of which neuroligin-3 is an important member. Neuroligin-3 is located at both excitatory and inhibitory synapses and performs essential roles in synaptic development, function and maintenance [47]. As stated above, NLGN3 gene mutations are associated with ASD, with the majority of pathogenic mutations resulting in a loss of neuroligin-3 function [10]. Therefore, we next determined whether ADBE was depressed in hippocampal neurons derived from a recently generated model of neuroligin-3 dysfunction, the Nlgn3−/y rat [28]. After validating the absence of Neuroligin-3 using semi-quantitative Western blotting (Fig. 3A-C), we first addressed whether Nlgn3−/y neurons displayed altered SV fusion or cargo retrieval using our high content sypHy screen. Nlgn3−/y neurons displayed no defect in SV fusion in response to either stimulation protocol, when compared to wild-type littermate controls (Fig. 3D, F-H). Furthermore, there was no difference between Nlgn3−/y and wild-type neurons in terms of either the extent or kinetics of sypHy retrieval (Fig. 3I, J). Finally, there was no significant change in the surface fraction of sypHy between the two genotypes (Fig. 3E), suggesting both SV cargo retrieval and clustering are unaffected by the absence of neuroligin-3. In contrast, the number of nerve terminals displaying an activity-dependent accumulation of TMR-dextran was reduced in Nlgn3−/y neurons when compared to wild-type controls (Fig. 3K, L). Since the number of nerve terminals was unchanged in Nlgn3−/y cultures (revealed by SV2A immunostaining, Fig. 3M, N), this meant that, similar to Nrxn1+/− neurons, Nlgn3−/y neurons display reduced ADBE.
Loss of syngap results in depressed ADBE
Neurexin-1 is a presynaptic protein, whereas neuroligin-3 forms essential complexes that modify and maintain presynaptic function [12]. Therefore, the observed depression of ADBE may result from a direct mechanistic involvement of these molecules in this endocytosis mode. To test the hypothesis that depression in ADBE was a consequence of intrinsic synaptic dysfunction in ASD, and not to loss of a key regulatory molecule, we next examined ASD models where the affected gene product is expressed exclusively at the postsynapse. The models chosen for these experiments were the Syngap+/− rat and the Syngap+/Δ−GAP rat [29,30,31]. SynGAP performs key roles at the postsynapse, principally in the control of AMPA receptor trafficking and synaptic plasticity [7]. Furthermore, SYNGAP haploinsufficiency is a highly prevalent cause of ASD, resulting from loss of function mutations in the SYNGAP gene [48, 49]. The Syngap+/− rat model was generated by introducing a frameshift mutation in the Syngap gene, resulting in nonsense mediated decay of mRNA encoding the mutant allele [29]. In contrast, the Syngap+/Δ−GAP rat has a deletion in the exons encoding the calcium/lipid binding C2, and GTPase activating protein (GAP) domain [30, 31]. The level of SynGAP protein expression for both models has been validated previously [29, 31].
As stated above, the exclusive postsynaptic localisation of SynGAP provides an excellent opportunity to test the hypothesis that depression of ADBE is a consequence of the ASD brain microenvironment. We first determined the ability of Syngap+/− hippocampal neurons to perform efficient SV fusion and cargo trafficking using our sypHy assay. In this instance, we also examined the performance of Syngap−/− neurons in this assay, to determine if the complete absence of the protein exacerbated any potential phenotype. When SV fusion was assessed, neither Syngap+/− nor Syngap−/− neurons displayed any deficit when compared to wild-type littermate controls during either the low or high frequency stimulus train (Fig. 4A, C-E). Furthermore, neither genotype displayed dysfunctional SV cargo retrieval in response to these trains, or an alteration in the surface distribution of sypHy (Fig. 4B, F,G). In contrast, Syngap+/− neurons displayed a reduction the number of activity-dependent TMR-dextran puncta, a decrease which became significant in Syngap−/− neurons when compared to wild-type controls (Fig. 4H, I). This was not a result of a decrease in nerve terminals in these primary cultures, since the number of SV2A puncta was unchanged across all genotypes (Fig. 4J, K). Therefore, a depression in ADBE is still observed even in neurons where the affected gene product is exclusively postsynaptic.
SynGAP functions as a GAP that negatively regulates Ras and Rap GTPases to control both F-actin dynamics (RasGAP) and p38 MAPK (RapGAP) activity [50,51,52]. However, SynGAP has other functions outside of its GAP activity. Its C-terminus alters synaptic strength via PDZ interactions with PSD-95, which in turn control AMPA receptor recruitment and synaptogenesis [53,54,55]. While the role of the GAP activity in brain function remains poorly understood, recent work has suggested that this catalytic domain may not be obligatory for synaptic plasticity [56]. To delineate these more structural roles from its GAP activity, we next exploited the Syngap+/Δ-GAP rat model [30, 31], which shares many behavioural traits with the Syngap+/- rat. Primary hippocampal cultures were prepared from both Syngap+/Δ-GAP and SyngapΔ-GAP/Δ-GAP rats in addition to wild-type littermate controls. When the three genotypes were assessed for SV fusion phenotypes using the sypHy assay, there was no significant difference for either stimulus train (Fig. 5A, C-E). There was also no significant effect of genotype on SV cargo retrieval after either stimulus and no impact on the surface fraction of sypHy (Fig. 5B, F,G). When the number of activity-dependent TMR-dextran puncta were determined, Syngap+/Δ-GAP neurons displayed a reduction, which again became significant when SyngapΔ-GAP/Δ-GAP neurons were examined (Fig. 5H, I). Furthermore, the number of nerve terminals in culture, identified via SV2A staining, was unchanged across all genotypes (Fig. 5J, K). This result therefore confirms that loss of postsynaptic SynGAP function in two independent model systems results in depression of ADBE.
Pten+/− neurons display depressed ADBE
The demonstration of depression of ADBE in SynGAP models, which have exclusively postsynaptic deficits, provides strong support for the hypothesis that a reduction in ADBE is a convergent consequence of ASD. As a final proof, we exploited a different rat ASD model that has no discernible presynaptic locus of dysfunction, the Pten+/− rat [34]. PTEN (phosphatase and tensin homologue deleted on chromosome 10) is a tumour suppressor, that negatively regulates the AKT / mTOR signalling pathway [57]. Haploinsufficiency resulting from loss of function mutations in the PTEN gene result in PTEN hamartoma tumor syndrome (PHTS [58]), with common links to ASD, macrocephaly, epilepsy and neurodevelopmental impairment [59,60,61].
Semi-quantitative Western blotting confirmed a reduction in PTEN expression in Pten+/− rats (Fig. 6A-C). Primary hippocampal cultures from either Pten+/− rat embryos or wild-type littermate controls were entered into the sypHy high content assay. Consistent with other models of ASD, Pten+/− neurons displayed no defect in SV fusion, cargo retrieval or surface stranding of sypHy (Fig. 6D-J). Intriguingly, Pten+/− neurons did display a reduction in the number of activity-dependent TMR-dextran puncta when compared to littermate controls (Fig. 6K, L). Therefore, in an ASD model system with no overt presynaptic dysfunction, depression of ADBE still occurs.
Discussion
The complex aetiology of ASD complicate the investigation of their causal mechanisms. Because of this, the study of monogenic ASD, where the genetic locus is known, has provided a series of key insights into potential convergent signalling pathways [3]. In this study we have revealed that neurons derived from a range of rat models of monogenic ASD display a depression of ADBE, a presynaptic endocytosis mode that is dominant during periods of high neuronal activity. This depression was observed regardless of whether the mutated gene was presynaptic, postsynaptic or neither, suggesting it is a convergent strategy to ameliorate disrupted synaptic function.
We initially revealed depression in ADBE in Fmr1−/y neurons, a model for fragile X syndrome [24]. Similar to this study, SV fusion and cargo trafficking were unaffected. A BK channel agonist corrected the depression of ADBE in these neurons, suggesting the alteration could be a result of the direct regulation of BK channels by the Fmr1 gene product, FMRP [62,63,64]. However, a BK channel antagonist did not recapitulate the depression in wild-type neurons, suggesting that FMRP had no direct mechanistic role in ADBE [24]. In this work, we determined whether ADBE was perturbed in other ASD models that did not display overt defects in presynaptic endocytosis. The large fluid phase marker TMR-dextran was used for these studies, since it reports ADBE due to size exclusion from single SVs [22]. TMR-dextran is an excellent reporter of ADBE, since interventions that block this endocytosis mode selectively reduce TMR-dextran uptake [45, 46, 65,66,67].
Specific cell adhesion molecules such as CHL1 and N-cadherins have proposed roles at the presynapse in both activity-dependent SV retrieval and ADBE [68,69,70,71]. In fact, cell adhesion molecules including both neurexin-1 and neuroligin-3 are greatly over-represented on ADBE-generated bulk endosomes [42]. This suggests that they might be required for optimal ADBE. In support, neurexin-1 controls localised calcium channel coupling to SV fusion at the active zone [41, 43] and both neurexin-1 and neuroligin-3 are synaptogenic [12]. However, a direct role via these presynaptic functions is unlikely. This is because ADBE is triggered via delocalised calcium increases, rather than localised calcium influx at the active zone [45] and there was no significant change in synapse number in either Nrxn1+/− or Nlgn3−/y cultures, suggesting synaptogenesis was not modulating this effect (this study). No change in synapse number was also observed in cultured human Nrxn1+/− neurons, however these neurons did display a considerable decrease in both spontaneous and evoked neurotransmitter release [72]. Intriguingly, mouse neurons that were engineered for the same Nrxn1+/− genotype displayed no significant neurotransmitter release defect [72], in a similar manner to the rat Nrxn1+/− neurons in this study. Therefore, it is possible that human neurons are exquisitely sensitive to depletion of neurexin-1 and that rodent neurons are more resilient to this insult.
Two independent models of SYNGAP haploinsufficiency disorder displayed depression of ADBE. The exclusive postsynaptic location of SynGAP provides strong support for the depression of ADBE being a compensatory adjustment in ASD. The expression of specific isoforms of SynGAP are driven via neuronal activity and have downstream effects on mEPSC frequency [73], suggesting a potential presynaptic role. However, it is more likely that these effects are mediated via the nano-organisation of AMPA receptors at the postsynapse. The observation that a depression of ADBE was observed in Syngap+/Δ−GAP neurons suggests that this depression resulted from a loss of enzyme activity, rather than interactions with postsynaptic partners such as PSD-95, LRRTMs and neuroligins [53,54,55]. In this context, key enzymatic roles for SynGAP could include either its RasGAP, which regulates F-actin and spine dynamics [74] or Rap-GAP activity which controls the p38 MAPK signalling pathway ultimately regulating postsynaptic AMPA receptor trafficking [51, 75, 76]. The obligatory role of the GAP domain in SYNGAP function has recently been questioned [56], however the GAP rat displays a series of circuit and behavioural phenotypes that suggest its enzymatic role of required for optimal function [31]. Regardless, the depression of ADBE in two independent models where the gene product is expressed exclusively at the postsynapse provides strong support for this depression to be a homeostatic adaptation to disrupted synaptic function.
The depression of ADBE in a novel Pten+/− model could potentially be explained by the fact that PTEN is expressed at growth cones during axonal navigation and synaptogenesis. However, its expression is restricted to the postsynapse in mature neurons [77]. The synaptic locus of dysfunction in neurons lacking PTEN appears to be due to dysregulation of the AKT/mTOR signalling pathway, specifically mTORC1/RAPTOR [78]. Deficiency in PTEN results in neuronal hypertrophy and hyperexcitability [79,80,81,82], which is corrected in multiple models via inhibition of AKT/mTOR signalling via rapamycin [78, 83, 84]. PTEN deficient neurons display enhanced excitatory neurotransmission, which appears to be due to enhanced postsynaptic function. Intriguingly, an increase in the size of the RRP and mEPSC frequency was also observed, however this was most likely due to an increase in the number of available synapses in these neurons [78, 79, 84]. In agreement with this hypothesis, no change in evoked SV fusion events were detected in this study.
The advent of monogenic rat models of ASD has transformed our understanding of social interaction deficits and cognitive behaviour, especially those that are difficult to replicate in mouse models with the same genetic mutations [85]. The study of neurons derived from these models can provide important insight into the fundamental cellular mechanisms that underpin these subtle alterations. This study reveals one such mechanism, the depression of a single presynaptic endocytosis mode, which is observed in all five independent rat models used. We suggest that the convergence upon depression of ADBE is a conserved and scalable lever through which to sculpt synaptic strength and circuit hyperexcitability regardless of genetic insults, differences in cellular localisation, and mechanisms of pathogenicity of the genes mutated in ASD.
The central question arising from this study is, what the molecular mechanism responsible for the depression of ADBE in ASD model neurons? ADBE is the dominant endocytosis mode during high activity and appear to have a similar molecular mechanism to ultrafast endocytosis, which is triggered by sparse stimulation [22, 46, 86,87,88]. Since both pathways occur with a timescale that is an order of magnitude faster than SV cargo clustering and retrieval via CME, an emerging view is that CME initiates on the presynaptic plasma membrane but completes on endosomes formed via either UFE or ADBE [18,19,20]. Since SV fusion and cargo retrieval are not impacted in ASD models, it suggests that the depression is intrinsic to ADBE or mechanisms that control it. One compelling explanation is that the observed depression is a compensatory mechanism to limit the intrinsic circuit hyperexcitability observed in many ASD models including many of the models used in this study [26, 27, 44, 78,79,80,81,82, 89,90,91] but see [92]. It will be key to determine the impact of elevated excitability in the long-term on presynaptic events such as ADBE.
Out of all potential presynaptic endocytosis modes, a depression in ADBE is the most intriguing, since its depression would be manifested disproportionately across specific cell types. This is because it is triggered during periods of high activity [22], meaning that GABAergic neurons and inhibitory circuits (which typically display higher firing rates [93]), may be excessively impacted. In agreement, reduced ADBE results in an inability to sustain neurotransmission during high activity [24, 67, 94]. Therefore, a depression of ADBE may not be a homogenous adaptation across the brain, but rather a circuit-specific correction in neuronal function. It will be essential in future work to determine how, (1) ADBE is impacted in intact circuits in ASD model system and (2) how circuit-specific modulation of this activity-dependent endocytosis mode may sculpt ASD-like behaviours.
Limitations
There are a number of limitations to the current work. The principal limitation is that the work has been performed exclusively in primary neuronal culture. Therefore, the impact of altered brain microcircuitry observed in these models of ASD cannot be assessed in this experimental system. Furthermore, this work has been performed on embryonic hippocampal neurons, therefore, there is a possibility that these findings do not extend to neurons from distinct brain regions which may display different phenotypes. However, the fact that a convergent depression of ADBE is observed across all hippocampal neurons derived from these models strongly suggests that it is an adaptation to the intrinsic dysfunction due to a specific monogenic insult.
One intrinsic defect observed in almost all of these rat models or in patient-derived neurons is hyperexcitability. However, at this current time Nlgn3−/y circuits and isolated neurons have not been reported to display this phenotype. Therefore, the suggestion that the depression of ADBE is a consequence of hyperexcitability in ASD requires further validation, both in a demonstration of hyperexcitability of all ASD neurons in culture, and in artificially modulating neuronal culture excitability and examining the impact on ADBE.
One final limitation of the study is that the mechanism of ADBE depression in ASD neurons was not determined. This is an ongoing line of enquiry however, and requires a detailed interrogation of molecular alterations at the synapse coupled to targeted interventions to modulate expression of known molecules essential for ADBE.
Conclusions
The depression of ADBE observed across five independent monogenic rat models of ASD in this study, in addition to that previously observed in Fmr1−/y neurons, suggests that it is a homeostatic mechanism to correct for some aspect of intrinsic dysfunction. Intrinsic hyperexcitability is a potential instigator of this depression and will be the focus of future investigations both in vitro and in vivo.
Data availability
The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.
Abbreviations
- ADBE:
-
Activity-dependent bulk endocytosis
- AMPA:
-
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
- AP:
-
Action potential
- ASD:
-
Autism spectrum disorder
- BK channels:
-
Big potassium channels
- CME:
-
Clathrin-mediated endocytosis
- DIV:
-
Days in vitro
- FMRP:
-
Fragile X messenger ribonucleoprotein
- GAP:
-
GTPase activating protein
- ID:
-
Intellectual disability
- SV:
-
Synaptic vesicle
- sypHy:
-
Synaptophysin-pHluorin
- TMR-dextran:
-
Tetramethylrhodamine-dextran
- UFE:
-
Ultrafast endocytosis
References
Mefford HC, Batshaw ML, Hoffman EP. Genomics, intellectual disability, and autism. N Engl J Med. 2012;366(8):733–43.
Krol A, Feng G. Windows of opportunity: timing in neurodevelopmental disorders. Curr Opin Neurobiol. 2018;48:59–63.
Yuen MD, Bookman M, Thiruvahindrapuram JLH, Patel B. Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat Neurosci. 2017;20(4):602–11.
Sahin M, Sur M. Genes, circuits, and precision therapies for autism and related neurodevelopmental disorders. Science. 2015;350(6263).
Contractor A, Ethell IM, Portera-Cailliau C. Cortical interneurons in autism. Nat Neurosci. 2021;24(12):1648–59.
Bonsi P, De Jaco A, Fasano L, Gubellini P. Postsynaptic autism spectrum disorder genes and synaptic dysfunction. Neurobiol Dis. 2022;162:105564.
Gamache TR, Araki Y, Huganir RL. Twenty years of syngap research: from synapses to cognition. J Neurosci. 2020;40(8):1596–605.
Bear MF, Huber KM, Warren ST. The mGluR theory of fragile X mental retardation. Trends Neurosci. 2004;27(7):370–7.
Sidorov MS, Auerbach BD, Bear MF. Fragile X mental retardation protein and synaptic plasticity. Mol Brain. 2013;6:15.
Uchigashima M, Cheung A, Futai K. Neuroligin-3: A Circuit-Specific synapse organizer that shapes normal function and autism spectrum Disorder-Associated dysfunction. Front Mol Neurosci. 2021;14:749164.
Cao X, Tabuchi K. Functions of synapse adhesion molecules neurexin/neuroligins and neurodevelopmental disorders. Neurosci Res. 2017;116:3–9.
Craig AM, Kang Y. Neurexin-neuroligin signaling in synapse development. Curr Opin Neurobiol. 2007;17(1):43–52.
Bonnycastle K, Davenport EC, Cousin MA. Presynaptic dysfunction in neurodevelopmental disorders: insights from the synaptic vesicle life cycle. J Neurochem. 2021;157(2):179–207.
Verhage M, Sørensen JB, SNAREopathies. Diversity in mechanisms and symptoms. Neuron. 2020;107(1):22–37.
Südhof TC. Neurotransmitter release: the last millisecond in the life of a synaptic vesicle. Neuron. 2013;80(3):675–90.
Chanaday NL, Cousin MA, Milosevic I, Watanabe S, Morgan JR. The synaptic vesicle cycle revisited: new insights into the modes and mechanisms. J Neurosci. 2019;39(42):8209–16.
Kononenko NL, Haucke V. Molecular mechanisms of presynaptic membrane retrieval and synaptic vesicle reformation. Neuron. 2015;85(3):484–96.
Kononenko NL, Puchkov D, Classen GA, Walter AM, Pechstein A, Sawade L, et al. Clathrin/AP-2 mediate synaptic vesicle reformation from endosome-like vacuoles but are not essential for membrane retrieval at central synapses. Neuron. 2014;82(5):981–8.
Watanabe S, Trimbuch T, Camacho-Perez M, Rost BR, Brokowski B, Sohl-Kielczynski B, et al. Clathrin regenerates synaptic vesicles from endosomes. Nature. 2014;515(7526):228–33.
Cousin MA. Integration of synaptic vesicle cargo retrieval with endocytosis at central nerve terminals. Front Cell Neurosci. 2017;11:234.
Soykan T, Kaempf N, Sakaba T, Vollweiter D, Goerdeler F, Puchkov D, et al. Synaptic vesicle endocytosis occurs on multiple timescales and is mediated by Formin-Dependent actin assembly. Neuron. 2017;93(4):854–66. e4.
Clayton EL, Evans GJ, Cousin MA. Bulk synaptic vesicle endocytosis is rapidly triggered during strong stimulation. J Neurosci. 2008;28(26):6627–32.
Kokotos AC, Cousin MA. Synaptic vesicle generation from central nerve terminal endosomes. Traffic. 2015;16(3):229–40.
Bonnycastle K, Kind PC, Cousin MA. FMRP sustains presynaptic function via control of Activity-Dependent bulk endocytosis. J Neurosci. 2022;42(8):1618–28.
Zhang Y, Bonnan A, Bony G, Ferezou I, Pietropaolo S, Ginger M, et al. Dendritic channelopathies contribute to neocortical and sensory hyperexcitability in Fmr1(-/y) mice. Nat Neurosci. 2014;17(12):1701–9.
Booker SA, Domanski APF, Dando OR, Jackson AD, Isaac JTR, Hardingham GE, et al. Altered dendritic spine function and integration in a mouse model of fragile X syndrome. Nat Commun. 2019;10(1):4813.
Das Sharma S, Pal R, Reddy BK, Selvaraj BT, Raj N, Samaga KK, et al. Cortical neurons derived from human pluripotent stem cells lacking FMRP display altered spontaneous firing patterns. Mol Autism. 2020;11(1):52.
Anstey NJ, Kapgal V, Tiwari S, Watson TC, Toft AKH, Dando OR, et al. Imbalance of flight-freeze responses and their cellular correlates in the Nlgn3(-/y) rat model of autism. Mol Autism. 2022;13(1):34.
Mastro TL, Preza A, Basu S, Chattarji S, Till SM, Kind PC, Kennedy MB. A sex difference in the response of the rodent postsynaptic density to syngap haploinsufficiency. Elife. 2020;9:e52656.
Buller-Peralta I, Maicas-Royo J, Lu Z, Till SM, Wood ER, Kind PC, et al. Abnormal brain state distribution and network connectivity in a SYNGAP1 rat model. Brain Commun. 2022;4(6):fcac263.
Katsanevaki D, Till SM, Buller-Peralta I, Nawaz MS, Louros SR, Kapgal V, et al. Key roles of C2/GAP domains in SYNGAP1-related pathophysiology. Cell Rep. 2024;43(9):114733.
Kight KE, Argue KJ, Bumgardner JG, Bardhi K, Waddell J, McCarthy MM. Social behavior in prepubertal neurexin 1α deficient rats: A model of neurodevelopmental disorders. Behav Neurosci. 2021;135(6):782–803.
Achterberg EJM, Biemans B, Vanderschuren L. Neurexin1α knockout in rats causes aberrant social behaviour: relevance for autism and schizophrenia. Psychopharmacology (Berl). 2024 Feb 29. Online ahead of print. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00213-024-06559-z
Rowley PA, Guerrero-Gonzalez J, Alexander AL, Yu JJ. Convergent microstructural brain changes across genetic models of autism spectrum disorder-A pilot study. Psychiatry Res Neuroimaging. 2019;283:83–91.
Granseth B, Odermatt B, Royle SJ, Lagnado L. Clathrin-mediated endocytosis is the dominant mechanism of vesicle retrieval at hippocampal synapses. Neuron. 2006;51(6):773–86.
Atluri PP, Ryan TA. The kinetics of synaptic vesicle reacidification at hippocampal nerve terminals. J Neurosci. 2006;26(8):2313–20.
Egashira Y, Takase M, Takamori S. Monitoring of vacuolar-type H + ATPase-mediated proton influx into synaptic vesicles. J Neurosci. 2015;35(8):3701–10.
Gordon SL, Leube RE, Cousin MA. Synaptophysin is required for synaptobrevin retrieval during synaptic vesicle endocytosis. J Neurosci. 2011;31(39):14032–6.
Pan PY, Marrs J, Ryan TA. Vesicular glutamate transporter 1 orchestrates recruitment of other synaptic vesicle cargo proteins during synaptic vesicle recycling. J Biol Chem. 2015;290(37):22593–601.
Harper CB, Smillie KJ. Current molecular approaches to investigate pre-synaptic dysfunction. J Neurochem. 2021;157(2):107–29.
Südhof TC. Synaptic neurexin complexes: A molecular code for the logic of neural circuits. Cell. 2017;171(4):745–69.
Kokotos AC, Peltier J, Davenport EC, Trost M, Cousin MA. Activity-dependent bulk endocytosis proteome reveals a key presynaptic role for the monomeric GTPase Rab11. Proc Natl Acad Sci U S A. 2018;115(43):E10177–86.
Luo F, Sclip A, Jiang M, Südhof TC. Neurexins cluster Ca(2+) channels within the presynaptic active zone. Embo J. 2020;39(7):e103208.
Avazzadeh S, Quinlan LR, Reilly J, McDonagh K, Jalali A, Wang Y, et al. NRXN1α(+/-) is associated with increased excitability in ASD iPSC-derived neurons. BMC Neurosci. 2021;22(1):56.
Morton A, Marland JR, Cousin MA. Synaptic vesicle exocytosis and increased cytosolic calcium are both necessary but not sufficient for activity-dependent bulk endocytosis. J Neurochem. 2015;134(3):405–15.
Clayton EL, Anggono V, Smillie KJ, Chau N, Robinson PJ, Cousin MA. The phospho-dependent dynamin-syndapin interaction triggers activity-dependent bulk endocytosis of synaptic vesicles. J Neurosci. 2009;29(24):7706–17.
Varoqueaux F, Aramuni G, Rawson RL, Mohrmann R, Missler M, Gottmann K, et al. Neuroligins determine synapse maturation and function. Neuron. 2006;51(6):741–54.
Vlaskamp DRM, Shaw BJ, Burgess R, Mei D, Montomoli M, Xie H, et al. SYNGAP1 encephalopathy: A distinctive generalized developmental and epileptic encephalopathy. Neurology. 2019;92(2):e96–107.
Mignot C, von Stülpnagel C, Nava C, Ville D, Sanlaville D, Lesca G, et al. Genetic and neurodevelopmental spectrum of SYNGAP1-associated intellectual disability and epilepsy. J Med Genet. 2016;53(8):511–22.
Chen HJ, Rojas-Soto M, Oguni A, Kennedy MB. A synaptic Ras-GTPase activating protein (p135 SynGAP) inhibited by cam kinase II. Neuron. 1998;20(5):895–904.
Krapivinsky G, Medina I, Krapivinsky L, Gapon S, Clapham DE. SynGAP-MUPP1-CaMKII synaptic complexes regulate p38 MAP kinase activity and NMDA receptor-dependent synaptic AMPA receptor potentiation. Neuron. 2004;43(4):563–74.
Pena V, Hothorn M, Eberth A, Kaschau N, Parret A, Gremer L, et al. The C2 domain of syngap is essential for stimulation of the Rap GTPase reaction. EMBO Rep. 2008;9(4):350–5.
Kim JH, Liao D, Lau LF, Huganir RL. SynGAP: a synaptic RasGAP that associates with the PSD-95/SAP90 protein family. Neuron. 1998;20(4):683–91.
Opazo P, Sainlos M, Choquet D. Regulation of AMPA receptor surface diffusion by PSD-95 slots. Curr Opin Neurobiol. 2012;22(3):453–60.
Walkup WG, Mastro TL, Schenker LT, Vielmetter J, Hu R, Iancu A et al. A model for regulation by SynGAP-α1 of binding of synaptic proteins to PDZ-domain ‘Slots’ in the postsynaptic density. Elife. 2016;5:e16813.
Araki Y, Rajkovich KE, Gerber EE, Gamache TR, Johnson RC, Tran THN, et al. SynGAP regulates synaptic plasticity and cognition independently of its catalytic activity. Science. 2024;383(6686):eadk1291.
Waite KA, Eng C. Protean PTEN: form and function. Am J Hum Genet. 2002;70(4):829–44.
Busch RM, Srivastava S, Hogue O, Frazier TW, Klaas P, Hardan A, et al. Neurobehavioral phenotype of autism spectrum disorder associated with germline heterozygous mutations in PTEN. Transl Psychiatry. 2019;9(1):253.
Conti S, Condò M, Posar A, Mari F, Resta N, Renieri A, et al. Phosphatase and tensin homolog (PTEN) gene mutations and autism: literature review and a case report of a patient with Cowden syndrome, autistic disorder, and epilepsy. J Child Neurol. 2012;27(3):392–7.
Satterstrom FK, Kosmicki JA, Wang J, Breen MS, De Rubeis S, An JY, et al. Large-Scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell. 2020;180(3):568–e8423.
Klein S, Sharifi-Hannauer P, Martinez-Agosto JA. Macrocephaly as a clinical indicator of genetic subtypes in autism. Autism Res. 2013;6(1):51–6.
Deng PY, Rotman Z, Blundon JA, Cho Y, Cui J, Cavalli V, et al. FMRP regulates neurotransmitter release and synaptic information transmission by modulating action potential duration via BK channels. Neuron. 2013;77(4):696–711.
Myrick LK, Deng PY, Hashimoto H, Oh YM, Cho Y, Poidevin MJ, et al. Independent role for presynaptic FMRP revealed by an FMR1 missense mutation associated with intellectual disability and seizures. Proc Natl Acad Sci U S A. 2015;112(4):949–56.
Deng PY, Klyachko VA. Genetic upregulation of BK channel activity normalizes multiple synaptic and circuit defects in a mouse model of fragile X syndrome. J Physiol. 2016;594(1):83–97.
Clayton EL, Sue N, Smillie KJ, O’Leary T, Bache N, Cheung G, et al. Dynamin I phosphorylation by GSK3 controls activity-dependent bulk endocytosis of synaptic vesicles. Nat Neurosci. 2010;13(7):845–51.
Smillie KJ, Pawson J, Perkins EM, Jackson M, Cousin MA. Control of synaptic vesicle endocytosis by an extracellular signalling molecule. Nat Commun. 2013;4:2394.
Blumrich EM, Nicholson-Fish JC, Pronot M, Davenport EC, Kurian D, Cole A, et al. Phosphatidylinositol 4-kinase IIα is a glycogen synthase kinase 3-regulated interaction hub for activity-dependent bulk endocytosis. Cell Rep. 2023;42(6):112633.
Leshchyns’ka I, Sytnyk V, Richter M, Andreyeva A, Puchkov D, Schachner M. The adhesion molecule CHL1 regulates uncoating of clathrin-coated synaptic vesicles. Neuron. 2006;52(6):1011–25.
Vitureira N, Letellier M, White IJ, Goda Y. Differential control of presynaptic efficacy by postsynaptic N-cadherin and β-catenin. Nat Neurosci. 2011;15(1):81–9.
van Stegen B, Dagar S, Gottmann K. Release activity-dependent control of vesicle endocytosis by the synaptic adhesion molecule N-cadherin. Sci Rep. 2017;7:40865.
Dagar S, Teng Z, Gottmann K. Transsynaptic N-Cadherin adhesion complexes control presynaptic vesicle and bulk endocytosis at physiological temperature. Front Cell Neurosci. 2021;15:713693.
Pak C, Danko T, Mirabella VR, Wang J, Liu Y, Vangipuram M, et al. Cross-platform validation of neurotransmitter release impairments in schizophrenia patient-derived NRXN1-mutant neurons. Proc Natl Acad Sci U S A. 2021;118:22.
McMahon AC, Barnett MW, O’Leary TS, Stoney PN, Collins MO, Papadia S, et al. SynGAP isoforms exert opposing effects on synaptic strength. Nat Commun. 2012;3:900.
Bär J, Kobler O, van Bommel B, Mikhaylova M. Periodic F-actin structures shape the neck of dendritic spines. Sci Rep. 2016;6:37136.
Rumbaugh G, Adams JP, Kim JH, Huganir RL. SynGAP regulates synaptic strength and mitogen-activated protein kinases in cultured neurons. Proc Natl Acad Sci U S A. 2006;103(12):4344–51.
Zhu JJ, Qin Y, Zhao M, Van Aelst L, Malinow R. Ras and Rap control AMPA receptor trafficking during synaptic plasticity. Cell. 2002;110(4):443–55.
Kreis P, Leondaritis G, Lieberam I, Eickholt BJ. Subcellular targeting and dynamic regulation of PTEN: implications for neuronal cells and neurological disorders. Front Mol Neurosci. 2014;7:23.
Tariq K, Cullen E, Getz SA, Conching AKS, Goyette AR, Prina ML, et al. Disruption of mTORC1 rescues neuronal overgrowth and synapse function dysregulated by Pten loss. Cell Rep. 2022;41(5):111574.
Williams MR, DeSpenza T Jr., Li M, Gulledge AT, Luikart BW. Hyperactivity of newborn Pten knock-out neurons results from increased excitatory synaptic drive. J Neurosci. 2015;35(3):943–59.
Southwell DG, Seifikar H, Malik R, Lavi K, Vogt D, Rubenstein JL, Sohal VS. Interneuron transplantation rescues social behavior deficits without restoring Wild-Type physiology in a mouse model of autism with excessive synaptic Inhibition. J Neurosci. 2020;40(11):2215–27.
Skelton PD, Frazel PW, Lee D, Suh H, Luikart BW. Pten loss results in inappropriate excitatory connectivity. Mol Psychiatry. 2019;24(11):1627–40.
Santos VR, Pun RYK, Arafa SR, LaSarge CL, Rowley S, Khademi S, et al. PTEN deletion increases hippocampal granule cell excitability in male and female mice. Neurobiol Dis. 2017;108:339–51.
Getz SA, DeSpenza T Jr., Li M, Luikart BW. Rapamycin prevents, but does not reverse, aberrant migration in Pten knockout neurons. Neurobiol Dis. 2016;93:12–20.
Weston MC, Chen H, Swann JW. Multiple roles for mammalian target of Rapamycin signaling in both glutamatergic and GABAergic synaptic transmission. J Neurosci. 2012;32(33):11441–52.
Till SM, Hickson RDL, Kind PC. Cross-species considerations in models of neurodevelopmental disorders. Trends Neurosci. 2022;45(3):171–2.
Watanabe S, Rost BR, Camacho-Perez M, Davis MW, Sohl-Kielczynski B, Rosenmund C, Jorgensen EM. Ultrafast endocytosis at mouse hippocampal synapses. Nature. 2013;504(7479):242–7.
Imoto Y, Raychaudhuri S, Ma Y, Fenske P, Sandoval E, Itoh K et al. Dynamin is primed at endocytic sites for ultrafast endocytosis. Neuron. 2022;110:2815–35.
Watanabe S, Mamer LE, Raychaudhuri S, Luvsanjav D, Eisen J, Trimbuch T, et al. Synaptojanin and endophilin mediate neck formation during ultrafast endocytosis. Neuron. 2018;98(6):1184–e976.
Modi B, Pimpinella D, Pazienti A, Zacchi P, Cherubini E, Griguoli M. Possible implication of the CA2 hippocampal circuit in social cognition deficits observed in the neuroligin 3 Knock-Out mouse, a Non-Syndromic animal model of autism. Front Psychiatry. 2019;10:513.
Ozkan ED, Creson TK, Kramár EA, Rojas C, Seese RR, Babyan AH, et al. Reduced cognition in Syngap1 mutants is caused by isolated damage within developing forebrain excitatory neurons. Neuron. 2014;82(6):1317–33.
Clement JP, Aceti M, Creson TK, Ozkan ED, Shi Y, Reish NJ, et al. Pathogenic SYNGAP1 mutations impair cognitive development by disrupting maturation of dendritic spine synapses. Cell. 2012;151(4):709–23.
Michaelson SD, Ozkan ED, Aceti M, Maity S, Llamosas N, Weldon M, et al. SYNGAP1 heterozygosity disrupts sensory processing by reducing touch-related activity within somatosensory cortex circuits. Nat Neurosci. 2018;21(12):1–13.
Bartos M, Vida I, Jonas P. Synaptic mechanisms of synchronized gamma oscillations in inhibitory interneuron networks. Nat Rev Neurosci. 2007;8(1):45–56.
Nicholson-Fish JC, Kokotos AC, Gillingwater TH, Smillie KJ, Cousin MA. VAMP4 is an essential cargo molecule for Activity-Dependent bulk endocytosis. Neuron. 2015;88(5):973–84.
Funding
This work was supported by grants from the Simons Foundation (529508), Epilepsy Research UK (P2003) and the RS McDonald Fund.
Author information
Authors and Affiliations
Contributions
KB and MAC were responsible for the conception and design of the experiments. KB and MSN were responsible for the collection and assembly of data. KB was responsible for the analysis and interpretation of data. KB, PCK, MAC wrote the manuscript, and all authors had the opportunity to contribute to its editing. All persons designated as authors qualify for authorship, and all those who qualify for authorship are listed. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Procedures were performed in accordance with the UK Animal (Scientific Procedures) Act 1986, under Project and Personal Licence authority and were approved by the Animal Welfare and Ethical Review Body at the University of Edinburgh (Home Office project licence – 7008878). Similarly, procedures were conducted in accordance with protocols approved by the Institutional Animal Ethics Committee of Institute for Stem Cell Science and Regenerative Medicine, Bangalore.
Consent for publication
Not applicable.
Competing interests
Peter Kind is an Associate Editor for Molecular Autism.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Bonnycastle, K., Nawaz, M.S., Kind, P.C. et al. Convergent depression of activity-dependent bulk endocytosis in rodent models of autism spectrum disorder. Molecular Autism 16, 26 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13229-025-00660-6
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13229-025-00660-6