Introduction

BDNF/TrkB signaling is required for a variety of neuronal functions including neurotransmission and synaptic plasticity. Upon binding to BDNF, BDNF/TrkB is internalized via pinocytosis1,2, or in a clathrin-dependent manner3 to vesicular compartments called signaling endosomes that might allow for local signaling. Signaling endosomes are predominantly generated at axon terminals by different mechanisms and constitute long-lived organelles that persist while undergoing transport from nerve terminals to the neuronal soma4,5,6. In neurons, retrograde trafficking of TrkB signaling endosomes constitutes an important long-range signaling mechanism that conveys information on presynaptic activity to the cell soma7. How endosomal sorting is accomplished in these organelles and how they escape from the degradative pathway remains still unclear.

A key consequence of TrkB signaling is sustained ERK activation, a process achieved via activation of the small GTPase Rap1 (Supplementary Fig. 1a)8. Accordingly, prenylated Rap1 is associated with TrkB signaling endosomes and regulation of its GTPase activity is, therefore, a likely mechanism to control local TrkB signaling.

A regulator of Rap1 is SIPA1L2 (also known as SPAR2), a member of the SIPA1L family of neuronal RapGAPs (Supplementary Fig. 1b–d)9. The protein is most abundant in granule cells of the dentate gyrus (DG) and cerebellum and shows RapGAP activity for Rap1 and 210. This RapGAP activity promotes the intrinsic GTPase activity of Rap1/2 that catalyzes the hydrolysis of GTP to GDP and inactivates Rap1/2 and consequently, ERK signaling. Here, we report that sipa1l2 knockout (ko) mice show impaired long-term potentiation (LTP) at mossy fiber (MF) synapses and spatial pattern separation, which requires MF plasticity. MF-LTP is an NMDA receptor-independent form of LTP that is expressed presynaptically and depends upon local BDNF/TrkB signaling11. Accordingly, we found that SIPA1L2 directly binds to TrkB and application of a TAT-peptide encompassing the binding region for TrkB in SIPA1L2 induces a similar phenotype in vivo and in vitro in wild-type (wt) mice like those observed in sipa1l2 ko mice. We found that SIPA1L2 links the receptor tyrosine kinase to a dynein motor via a direct interaction with the adaptor protein Snapin which allows retrograde transport. Interestingly, SIPA1L2 concurrently interacts with Light chain 3 (LC3), a marker for autophagosomes that is involved in substrate selection, and this interaction promotes SIPA1L2 RapGAP activity. While autophagosomes are continuously generated at axon terminals, very little is known about the synaptic role of autophagy. Here we show that SIPA1L2 associates with amphisomes, organelles from the autophagic pathway that result from the fusion of autophagosomes with late endosomes, that are positive for the late endosome marker Rab7 as well as LC3 and TrkB. This configuration allows LC3 to tightly control TrkB signaling via interaction with SIPA1L2, which increases the RapGAP activity and promotes Rap1/ERK inactivation. We show that these amphisomes traffick retrogradely along axons, stop at presynaptic boutons and both motility and signaling are controlled by SIPA1L2’s RapGAP activity that reduces the velocity of amphisome transport. Presynaptic LTP induces a protein kinase A (PKA)-dependent dissociation of the SIPA1L2/Snapin complex from dynein intermediate chain (DIC). This increases dwelling time of the amphisome at presynaptic boutons and PKA phosphorylation of SIPA1L2 reduces RapGAP activity, therefore, enabling local TrkB signaling at boutons, which in turn promotes neurotransmitter release. Collectively, the data suggest that retrograde axonal transport of BDNF/TrkB occurs in neuronal amphisomes that allow local control of TrkB signaling and are involved in plasticity-relevant local signaling at presynaptic boutons.

Results

MF-LTP and pattern separation deficits in sipa1l2−/− mice

To study the neuronal function of SIPA1L2, we generated sipa1l2 ko mice (Supplementary Fig. 2a–d). No major morphological abnormalities were observed in the cerebellum and DG (Supplementary Fig. 2e–k), motor learning or coordination (Supplementary Fig. 2l–m). The number of adult-born granule cells and measures of general DG excitability and postsynaptic function were all normal in sipa1l2−/− mice (Supplementary Figs. 2n–o and 3). However, a significant deficit was found when we assessed MF LTP, a form of plasticity presynaptically expressed at MF boutons of DG granule cells (Fig. 1a, b). Accordingly, we also observed a strong impairment in spatial pattern separation, a cognitive process associated with proper DG function and MF LTP12,13,14,15,16, that is responsible for the disambiguation and independent storage of similar memories (Fig. 1c–e). No changes were found in novel object location and recognition paradigms that are sensitive to perturbation of synaptic function in hippocampal CA1 neurons (Fig. 1f). Spatial pattern separation as well as MF LTP have been shown to depend on BDNF/TrkB signaling in the DG11,13 and we indeed found a reduction in MF LTP when we chelated endogenous BDNF during perfusion of slices with TrkB-Fc bodies (Fig. 1g, h), resembling the decline in late phase LTP found in sipa1l2−/− mice.

Fig. 1
figure 1

sipa1l2−/− mice exhibit impaired MF plasticity and deficits in pattern separation. a MF-LTP was induced in acute slices using a high-frequency stimulus (HFS) protocol. The NMDA receptor antagonist D-APV (50 μM) was present during baseline recordings and LTP induction (shaded blue). Bath application of the group II mGluR agonist DCG-IV (2 μM) that suppresses synaptic transmission at the MF pathway was performed during the last 10 min (shaded gray) of each experiment to control input specificity. Left, the average values of fEPSP amplitudes upon MF-LTP induction. Right, fEPSP amplitudes during the last 45–70 min following MF-LTP induction. b Averaged fEPSP amplitudes recorded during the last 10 min of a before DCG-IV application (Mann-Whitney U test). c Timeline (upper panel) and schematic representation of the object distribution (lower panel) of the spatial pattern separation test. Gray bars indicate 10-min intervals. During the sample phase (red-shaded) objects (A1–3) in the similar location recognition group (SLR) were placed closer while objects in the dissimilar location recognition group (dSLR) (A1–3) were placed farther away from each other. During choice phase (gray-shaded), a new object (A4) was introduced. Animals from the SLR find A4 closer to positions A2–A3 and have a higher demand for pattern separation than those from the dSLR. Filled circles (A1–4) represent object location. Open circles indicate the absence of objects. d Exploration time of sipa1l2 wt and ko animals in A1–3 during the sample phase (two-way ANOVA). e Discrimination index during choice phase in the SLR and dSLR groups (unpaired Student’s t test). f Discrimination index of sipa1l2 wt and ko animals during the novel object location recognition and object recognition test (unpaired Student’s t test). g, h Left, average fEPSP amplitudes upon MF-LTP induction performed as explained in a in control and BDNF-depleted slices (TrkB-Fc, 5 µg/mL). Right, a close-up representation from the last 45–70 min. In h, averaged fEPSP amplitudes obtained during the last 10 min of g prior DCG-IV application (Mann-Whitney U test). Bars and error bars depict mean ± SEM in all graphs. Circles represent mean values from individual subjects (df) or slices (a, b, g, h). n.s. not significant. "**" indicates P ≤ 0.01; "***" indicates P ≤ 0.001.

The cytoplasmic domain of TrkB directly binds to SIPA1L2

These experiments raised the question of whether SIPA1L2 might be involved in presynaptic BDNF-TrkB signaling. BDNF/TrkB are internalized at distal axons and transported retrogradely in a dynein-dependent manner as signaling endosomes7,17,4a). Coimmunoprecipitation experiments from extracts of rat hippocampi revealed that the protein is indeed present in immunoprecipitates generated with a TrkB-specific antibody and vice versa, TrkB could be precipitated with a SIPA1L2 antibody (Fig. 2c; Supplementary Fig. 4b). Moreover, full-length TrkB and SIPA1L2 are present in transport complexes immunoprecipitated by an antibody directed against DIC (Fig. 2d) and both proteins are localized to axon terminals in hippocampal primary neurons (Fig. 2e) where they were found in very close association as revealed by STED imaging (Fig. 2f, g).

Fig. 2
figure 2

SIPA1L2/TrkB interaction at presynapses is crucial for the function of the DG. a Confocal images of rat hippocampal neurons immunostained against SIPA1L2, Synaptophysin 1 and Tau. Scale bar is 5 μm. b Pearson’s correlation coefficient calculated for SIPA1L2 and Synaptophysin 1 from a. Circles represent averaged values per region of interest (ROI) analyzed from three independent images. c Endogenous TrkB coimmunoprecipitates SIPA1L2 from rat hippocampal lysates. The two bands in TrkB correspond to the full length (TrkB-FL) and the truncated form (TrkBt). Goat anti-TrkB antibody (R&D) is used for precipitation and detection. d DIC coimmunoprecipitates SIPA1L2 and TrkB but not GM130 from mouse whole brain crude light membrane fraction. Note that the band revealing TrkB corresponds to the full-length form. e Confocal images from rat hippocampal neurons immunostained against SIPA1L2, TrkB and Synaptophysin 1 (Sphy1). Scale bar is 10 μm. f STED images from hippocampal neurons immunostained against SIPA1L2, TrkB and Synaptophysin 1 (confocal). Line profiles (g) indicate relative intensities for STED channels along 1 µm. Scale bar is 1 µm. h Pull-down assay between the juxtamembrane region of TrkB and the 14aa of the binding interface in ActI-SIPA1L2. i Pull-down assay between TrkB and ActI-SIPA1L2 in the presence of 10× TAT binding peptide (TAT) or 10× scrambled peptide (TAT-scr). j Cartoon representing the timeline used for the pattern separation test performed in wt animals infused with TAT peptides and the location of the infusion (red dot). k Exploring time from mice injected with TAT-SIPA1L2 or TAT-scr during the sample phase (two-way ANOVA). l Discrimination indexes obtained during the choice phase in SLR and dSLR groups injected with either TAT-SIPA1L2 or TAT-scr. The number of subjects is depicted in k (unpaired Student’s t test). m fEPSP amplitudes recorded during the last 45–70 min after MF-LTP upon bath perfusion of TAT-SIPA1L2 or TAT-scr peptides. Right, averaged values of fEPSP amplitudes during last 10 min of LTP recording before DCG IV application (Mann-Whitney U test). Bars and error bars represent data as mean ± SEM in all graphs. Circles represent mean values of individual subjects (kl) or slices (m). "*" indicates P ≤ 0.05; "***" indicates P ≤ 0.001.

Next, we mapped the binding region in both proteins with Yeast-Two Hybrid (YTH) and found that the ActI-domain of SIPA1L2 interacts in the TrkB cytosolic domain with the first 12 juxtamembraneous amino acids (aa) (454–465) and the first 23 aa of the tyrosine kinase domain (537–559) (Supplementary Fig. 4c–d). Bacterially expressed GST- and MBP-fusion proteins revealed a direct interaction of cytoplasmic TrkB with the ActI but not the ActII domain of SIPA1L2 in pull-down assays (Supplementary Fig. 4e). We isolated 14 aa in SIPA1L2-ActI that are crucial for the association with TrkB (Fig. 2h; Supplementary Fig. 4f–h) and that happen to be unique to SIPA1L2 when compared to other SIPA1L family members (Supplementary Fig. 1c) and designed a TAT-peptide containing this region. This TAT-peptide competed for binding to TrkB in a pull-down assay (Fig. 2i) and when infused into the DG of wt animals (Fig. 2j; Supplementary Fig. 4i) induced a deficit in spatial pattern separation (Fig. 2k, l) similar to the one observed in sipa1l2−/− mice (Fig. 1e). In addition, the bath perfusion of the TAT-peptide also reduced the amplitude of MF-LTP in acute slices (Fig. 2m). Thus, interruption of the SIPA1L2-TrkB interaction in wt mice mimics the deficits of sipa1l2−/− mice.

SIPA1L2 interacts with Snapin and enables TrkB trafficking

Dynein-dependent retrograde trafficking of TrkB-signaling endosomes in axons requires Snapin, an adaptor protein that recruits dynein by interacting with DIC

Fig. 5
figure 5

The RapGAP activity of SIPA1L2 controls the motility of SIPA1L2-amphisomes. a Time-lapse representation of GFP-SIPA1L2/tRFP-LC3b visiting presynaptic boutons from rat primary hippocampal neurons live-labeled by Stgm-1Oyster650 (yellow ROIs). Note that only ROIs in red are within the axon of interest and considered for analysis. Imaging was performed in conditioned neuronal media and three-channel time-lapse acquired for 5 min. Scale bar = 5 μm. b Representative kymographs from the experiment described in a. Neurons expressed tRFP-LC3b and GFP-SIPA1L2 or GFP-SIPA1L2-N705A. Below, stationary Stgm-1Oyster650 indicates presynaptic boutons. Sketch drawing represents traces of cotrajectories aligned with positions of presynaptic boutons (shaded blue lines). c, d Relative motility of SIPA1L2/LC3b and SIPA1L2-N705A/LC3b cotrajectories as percentage of total cotrajectories shows predominantly retrograde (R) movement (S, stationary; A, anterograde). In d, cotrajectories showed as a percentage of total LC3b or SIPA1L2 trajectories. Circles in bar graphs show values per analyzed kymograph (GFP-SIPA1L2: n = 20 axons, GFP-SIPA1L2-N705A: n = 17 axons). One-way ANOVA with Bonferroni’s posthoc test. e Instant velocity (μm/s) of GFP-SIPA1L2/tRFP-LC3b and GFP-SIPA1L2-N705/tRFP-LC3b cotrajectories. GFP-SIPA1L2: n = 102 instant measures from 20 axons; GFP-SIPA1L2-N705A: n = 77 instant measures from 14 axons. Mann−Whitney U test for the number of instant measures. f, g Quantification (f) and cumulative distribution (g) of the run-length; GFP-SIPA1L2: n = 114 measures from 20 axons; GFP-SIPA1L2-N705A: n = 53 measures from 14 axons (Mann-Whitney U test for the number of measures). h Percentage of LC3b/SIPA1L2 stopovers occurring in presynaptic boutons labeled by a Synaptotagmin 1 (Stgm1) antibody shows the preferential occurrence of these stopovers at boutons. Stops occurring outside Stgm1 labeling are depicted as non-Stgm1. Numbers of stops from 13 axons is depicted under the graph. i Average number of visited boutons in 60 μm axon lengths of SIPA1L2/LC3b and SIPA1L2-N705A/LC3b vesicles. GFP-SIPA1L2: n vesicles = 21 from 20 axons; GFP-SIPA1L2-N705A: n vesicles = 21 from 14 axons cultures (Mann-Whitney U test for number of vesicles). j, k Mean synaptic dwelling time (GFP-SIPA1L2: n = 78 stops from 20 axons; GFP-SIPA1L2-N705A: n = 67 stops from 15 axons) calculated from imaging of neurons in conditioned neuronal media (Mann-Whitney U test for n of stops). Data in bar graphs are depicted as mean ± SEM. n.s. - not significant. "**" indicates P ≤ 0.01; "***" indicates P ≤ 0.001.

cLTP prolongs the stopovers of TrkB-amphisomes at boutons

Presynaptic LTP results in activation of PKA at MF boutons and enhanced synaptic function that is largely mediated by PKA-dependent phosphorylation of different components of the presynaptic release machinery26,27,28,29,30. PKA-mediated phosphorylation of Snapin at Ser50 is crucial for its dissociation from the DIC and a phosphomimetic Snapin-S50D mutant is largely immobile31. Heterologous coimmunoprecipitation experiments revealed that both phosphomimetic (S50D) as well as phospho-deficient (S50A) Snapin interact with tRFP tagged SIPA1L2-470-1025 (Fig. 6a). However, in contrast to the phospho-deficient protein, DIC did not coimmunoprecipitate with phosphomimetic Snapin (Fig. 6a), indicating that PKA-dependent phosphorylation induces the dissociation of the protein from DIC. Consistent with previous work31, time-lapse imaging revealed that phosphomimetic Snapin is largely stationary and was often found in proximity to immobile fl-SIPA1L2-mCherry at axon terminals (Fig. 6b, c). We therefore next wondered whether changes in synaptic activity could affect amphisome trafficking among presynaptic terminals. Trafficking of SIPA1L2/LC3b among presynaptic terminals labeled by anti-Synaptotagmin 1Oyster650 antibodies revealed an enhancement in dwelling time upon induction of chemical LTP (cLTP) (Fig. 6d–f). The PKA inhibitor H89 prevented this effect (Fig. 6e, f).

Fig. 6
figure 6

cLTP prolongs dwelling time of SIPA1L2-amphisomes at presynaptic boutons. a Heterologous coimmunoprecipitation experiments using SIPA1L2-470-1025-tRFP (RapGAP-PDZ) and a phospho-deficient (GFP-Snapin-S50A) or a phosphomimetic Snapin mutant (GFP-Snapin-S50D). Both forms of Snapin coimmunoprecipitates with SIPA1L2, but only the phospho-deficient form of Snapin coimmunoprecipitates with DIC. b, c Snapshots in b obtained from time-lapse imaging in rat hippocampal cells overexpressing GFP-Snapin-S50D together with fl-SIPA1L2-mCherry where mostly immobile (b, arrows) vesicles were found at presynaptic terminals labeled with anti-Synaptotagmin 1-Oyster650 (black arrows). Scale bar is 10 μm. In c quantification of the percentage of stationary cotrajectories (note the difference with Fig. 3k). df Kymographs generated from axons coexpressing GFP-SIPA1L2 and tRFP-LC3b and labeled in vivo with Stgm-1Oyster650 after control or chemical long-term potentiation (cLTP) induction. Imaging was performed in nonconditioned, extracellular imaging buffer. Below, merge images represent traces of cotrajectories aligned with positions of presynaptic boutons (shaded blue lines). Dwelling time of the SIPA1L2-LC3b-amphisomes at boutons is represented in e and corresponding cumulative distribution diagram in f. Data represented as mean ± SEM. N numbers in e, f correspond to vesicles analyzed from 11 axons (control), 14 axons (cLTP), 8 axons (H89) and 6 axons (H89 + cLTP) (one-way ANOVA on ranks with Dunn’s multiple comparison test for vesicles). gi In g, the timeline for the RapGAP activity assays performed in h, i. Recombinant intein-tagged SIPA1L2-470-1025 as well as SIPA1L2-470-1025-S990D (phosphomimetic SIPA1L2 mutant in a potential PKA phosphosite) were used to hydrolize recombinant Rap1b loaded with GTP in the presence of LC3b. Quantification of four independents experiments in i (Mann-Whitney U test). Bar graphs depict data as mean ± SEM. "*" indicates P ≤ 0.05; "***" indicates P ≤ 0.001.

It has been reported that PKA-dependent phosphorylation of Ser499 of Rap1GAP negatively regulates RapGAP activity32. Sequence analysis of SIPA1L2 revealed a high scoring PKA phosphorylation motif (S990-RxxpS motif/ 0,757/ Supplementary Fig. 1d) that is located close to the region that corresponds to the regulatory PKA sites (pSer 499) of Rap1GAP and that is well preserved between SIPA1L family members (Supplementary Fig. 1c). When we tested the hypothesis that PKA in analogy to Rap1GAP might negatively regulate RapGAP activity of SIPA1L2, we found that phosphomimetic SIPA1L2-470-1025-S990D indeed hydrolyzed very little recombinant Rap1b-GTP (Fig. 6g–i), indicating a negative regulation of RapGAP activity by PKA. Collectively these data suggest that following induction of presynaptic plasticity, PKA phosphorylation of Snapin induces the dissociation of the amphisome from Dynein and enhances its residing time at presynaptic boutons. Concomitant phosphorylation of SIPA1L2 diminishes its RapGAP activity and thereby potentially facilitates ERK signaling.

Amphisomes activate ERK at boutons and enhance release

Amphisomes are believed to be transient intermediate organelles that in nonneuronal cells rapidly enter a degradative lysosomal pathway. However, lysosomes are not abundant if at all present in distal axons33. In primary cultures, STED imaging revealed that the lysosomal marker LAMP1 was indeed mostly absent from presynaptic boutons, rendering unlikely that SIPA1L2-TrkB-LC3b associates with a degradative organelle at axon terminals (Supplementary Fig. 6a). A recent study showed that TrkB is transported on autophagosomes to the soma where it regulates gene expression34. This transport requires association of the Clathrin adaptor AP2 with LC3 and DIC34. Therefore, we speculated that amphisomes could constitute more persistent entities in axons where they could serve signaling functions at presynaptic boutons. Accordingly, we found that SIPA1L2/LC3/TrkB complexes are positive for the late endosome marker Rab7 (Fig. 7a; Supplementary Fig. 6b–c). Moreover, kinase-active pTrkBY515 showed a significant degree of colocalization with SIPA1L2 at presynaptic boutons in line with the high association of LC3, SIPA1L2 and TrkB observed before (Fig. 7b, c). This is expected to be higher when assessing colocalization in only those boutons in which SIPA1L2 is present. Accordingly, intensities of pTrkBY515 are higher in SIPA1L2-positive boutons (Fig. 7d–f). This tight association of SIPA1L2 with pTrkBY515 as well as TrkB with LC3 (Fig. 7g, h) and Rap1 with SIPA1L2 and LC3 (Supplementary Fig. 6d) at presynapses indicate that SIPA1L2 might be part of a signaling TrkB amphisome. We also found an extensive colocalization with AP2 in axons and synaptic boutons (Supplementary Fig. 6e–g), suggesting that SIPA1L2-TrkB amphisomes are long-range organelles that will traffic retrogradely to reach the soma34. Moreover, TrkB and SIPA1L2 associate with an autophagosome enriched fraction from rat brain lysates that is also positive for LC3, Rab7 and Snapin (Fig. 7i, j). Of note, SIPA1L2 was not found in the lysosome fraction (Fig. 7i, j).

Fig. 7
figure 7

TrkB-LC3b-Rab7-containing amphisomes are positive for SIPA1L2. a Confocal images of quadruple immunofluorescence performed in rat primary hippocampal neurons stained for SIPA1L2, LC3, TrkB with Rab7. Scale bar is 1 μm. b Confocal images of primary neurons treated with BDNF and stained for pTrkBY515, SIPA1L2, and Bassoon show colocalization of SIPA1L2 and pTrkB Y515 in presynaptic boutons. Scale bar is 5 µm. c Mander’s coefficient calculated for pTrkBY515 and SIPA1L2 in boutons detected by Bassoon staining. For control measurements, the same images were rotated 90º to the right (paired Student’s t test). d, e Representative images in d of rat hippocampal neurons treated with BDNF and stained for pTrkBY515, SIPA1L2 and Synaptophysin1. Arrows indicate boutons where SIPA1L2 is present (black) or absent (red). Note that in the presence of SIPA1L2, pTrkBY515 intensity is higher compared to those from boutons where SIPA1L2 is absent. Quantification is shown in e, circles represent averaged intensities per image (paired Student’s t test for average per image). Averaged intensities are normalized to images acquired from Fc-TrkB-treated neurons. The cumulative frequency distribution is shown in f (n = synaptic boutons). Scale bar is 2 μm. g, h Super-resolution STED imaging performed in rat hippocampal neurons revealed association of TrkB with LC3 at the presynaptic boutons. In h, line profiles from the line in the ROI2 in g. Scale bars 5 µm (overview) and 1 µm (inserts). i, j In i, scheme depicting fractionation protocol performed from rat brain that results in autophagosome (A1), autophagolysosome (A2) and lysosome (L) fraction. Note that Rab7 and SIPA1L2 are only present in the total and A1 fraction (j) according to their presence in amphisomes but not in later stages of the autophagosomal pathway. Lines in dot plot graphs depict data as mean ± SEM. "**" indicates P ≤ 0.01; "***" indicates P ≤ 0.001.

What could be the consequence of amphisome stopover at axon terminals? Local ERK activation has been linked to enhanced neurotransmitter release and presynaptic plasticity35,36. We took advantage of a FRET-based ERK sensor37 that we targeted to the presynaptic compartment following fusion to Synaptophysin-1 (Fig. 8a–e; Supplementary Fig. 6h). We then imaged changes in the GFP fluorescence lifetime, the donor of the FRET pair, coinciding with the stopover of amphisomes, which we identified using either tagged versions of SIPA1L2 or LC3b. Synaptic visits were determined by kymograph analysis (Fig. 8a). Quantification of ∆LifetimeGFP upon the arrival of the amphisome at the bouton (time 0) indeed correlated with a significant decrease in the GFP lifetime (Fig. 8b–e), indicating a higher FRET efficiency due to ERK activation and subsequent phosphorylation of the sensor37. Importantly, this decrease was not observed when the amphisome passed by a bouton without stop** (nonvisited terminals) or in axons where no amphisomal trafficking was observed during the recording time (Fig. 8c–e).

Fig. 8
figure 8

SIPA1L2-amphisomes activate ERK at boutons and potentiate presynaptic function. a Kymographs represent the visit of SIPA1L2-amphisome labeled by SNAP-SIPA1L2 (+SiR647) to a bouton identified by Sy-EKAR. b Representative heat maps depicting GFP lifetime (n.s.) over time. Lifetime0 represents the frame when the amphisome reaches the bouton. Scale bar is 5 μm. c, d Quantification of GFP-∆Lifetime over time in visited and nonvisited boutons. Amphisomal arrival is considered as time 0. Control in d represents data from axons where no trajectories were found. N numbers represent analyzed boutons from n > 3 independent experiments. e Averaged GFP-∆Lifetimes from c, d. One-way ANOVA with Bonferroni’s posthoc test; n = number of boutons. f Representative confocal images from wt and sipa1l2−/ primary neurons overexpressing fl-SIPA1L2-mCherry, SIPA1L2-Δ14-mCherry or mCherry, treated with TrkB-Fc or BDNF and immunostained for Synapsin1,2 (Syn) and phospho-SynS62. White arrows show nontransfected boutons. Insets depict boutons labeled with the asterisk. Scale bar is 5 µm. g Percentage of enhancement of the pSynS62/Syn ratio in BDNF-treated as compared to Trk-Fc-treated neurons. P values correspond to comparisons with Fc-TrkB-treated cells for the corresponding condition (Kruskall-Wallis test with Dunn’s correction; wt-tRFP-Fc-TrkB = 20 axons; ko-tRFP-Fc-TrkB = 13 axons; ko-SIPA1L2-Fc-TrkB = 10 axons; ko-SIPA1L2d14-Fc-TrkB = 15 axons; two independent experiments). h Scheme showing the timeline of the experiment: FM4-64 (10 µM) was loaded in the terminals by a train of pulses (30 s @ 20 Hz) and washed out for 10 min. First FM4-64 unloading was done by delivering 900 pulses@10 Hz. Trafficking of TrkB-GFP or GFP-LC3b was imaged for 10 min before a second unloading protocol was applied. i Representative images showing an axon overexpressing GFP-LC3b after loading with FM4-64 (red). Scale bar is 10 μm. j Kymographs prepared from an axonal segment overexpressing GFP-LC3b and loaded with FM4-64. km Unloading rate after visits of TrkB-GFP (k) or GFP-LC3b (l) and from nonvisited boutons (m). Dots indicate boutons. Right panels show the frequency distribution of unloading rates (paired Student’s t test in k, l and Wilcoxon rank test in m). Shaded lines in kymographs (a, j) represent visited (red) and nonvisited (green) boutons. Bar graphs show mean ± SEM. n.s. stands for not significant. "*", "**", "***" indicate P ≤ 0.05, P≤0.01, P ≤ 0.001, respectively.

ERK activation in presynaptic plasticity results in phosphorylation of Synapsin I (Syn-I), a protein known to play a role in BDNF-mediated increase of neurotransmitter release36. Ultrastructural analysis revealed no major alterations in presynaptic bouton organization and vesicle content in sipa1l2−/− as compared to wt mice (Supplementary Fig. 6i–j). Quantification of ERK-dependent phosphorylation of Syn-I in hippocampal primary neurons from wt mice revealed a significant enhancement of the pSyn/Syn ratio at boutons following BDNF application (Fig. 8f, g) without changes in their number (Supplementary Fig. 6k). The BDNF-induced enhancement of the pSyn/Syn ratio was absent in sipa1l2−/− neurons and could be rescued by re-expression of fl-SIPA1L2 in ko neurons. However, no rescue was observed when we re-expressed the SIPA1L2-Δ14 mutant lacking the TrkB binding region (Fig. 8f, g).

To directly assess the effects of the activation of these molecular pathways in transmitter release, we combined unloading of FM 4-64 dyes to monitor synaptic vesicle fusion with live imaging of LC3b/TrkB trafficking and compared unloading rates at visited and nonvisited boutons (Fig. 8h–j). Analysis of the unloading rates before and after a stopover revealed a significant enhancement in the majority of the analyzed boutons visited by both TrkB-GFP (Fig. 8k) and GFP-LC3b (Fig. 8l). Importantly, this increase was not observed in nonvisited neighboring boutons (Fig. 8m).