Main

Since the discovery that the hippocampal dCA2 subregion is criticalΒ for social memory3, research on this topic has focused on identifying additional storage sites in the hippocampus5,6,10 and their respective output regions5,6,7,11. Little is known about the input regions to dCA2 or vCA1, which carry information about social encounters and are required for social memory formation. To identify hippocampal-projecting brain regions that are specifically involved in social memory, we exposed TRAP2;Ai14 mice after injection with 4-hydroxytamoxifen (4-OHT) to a novel mouse, a novel object or an empty behavioural chamber to label neurons activated by these experiences, and examined brain regions that are not associated with object memory and project to dCA2 or vCA1 (Extended Data Fig. 1a). Increased numbers of tdTomato-positive cells were found in the MS and nucleus of the diagonal band (NDB) when comparing novel mouse to novel object and control conditions (Extended Data Fig. 1b). Interaction with a novel mouse also increased tdTomato-positive cells in dCA2 (Extended Data Fig. 1c, d). To determine whether, as previously suggested3,12, neurons in different parts of the septum (lateral septum (LS), MS, NDB) form monosynaptic connections on hippocampal neurons, we used monosynaptic rabies tracing, which generated labelled cells in the MS and NDB, but not the LS (Extended Data Fig. 1e, f). Consistent with these and previous results12, anterograde tracing revealed strong innervation of dorsal and ventral hippocampus (Extended Data Fig. 1g, h). Thus, MS and NDB neurons show increased activity during social encounters and project to dCA2 and vCA1.

Modulation of social memory by MS manipulations

The MS and NDB are implicated in hippocampal theta-rhythm13 and arousal14, but little is known about their potential roles in social behaviour. To determine whether the MS influences social memory, we expressed an inhibitory designer receptor exclusively activated by designer drugs (DREADD) fused to mCherry (hM4Di) or mCherry alone (mCh) in the MS of mice and administered the DREADD activator clozapine-N-oxide (CNO) or saline before a short-term social memory assay6,7 (Fig. 1a, b). Two cohorts of control mice, expressing mCh and receiving CNO or expressing hM4Di and receiving saline, exhibited short-term (10-min interval) social memory as evidenced by their preference for the chamber containing a novel mouse compared to that containing a familiar mouse (Fig. 1c, Extended Data Fig. 2a). By contrast, hM4Di mice that received CNO exhibited no short-term memory of the familiar mouse (Fig. 1c, left). Calculating a discrimination score for each subject mouse, which enables more direct comparisons between treatments, confirmed the absence of social memory in mice in which MS neuronal activity was inhibited by hM4Di (Fig. 1c, right). Baseline sociability assessed during the initial, three-chamber sociability assay, as well as inanimate object memory, were not affected by hM4Di-mediated inhibition of the MS (Extended Data Fig. 2b, c).

Fig. 1: Chemogenetic manipulation of the MS bidirectionally regulates social memory.
figure 1

a, Experimental timeline of three-chamber tests. IP, intraperitoneal injection. b, Schematic and representative image of MS injection site. Scale bar, 500 μm. c, d, Left, duration that mice spent in the chamber with a familiar mouse (fm) or a novel mouse (nm). Right, discrimination scores (c: F2, 40 = 4.666, P = 0.0151; mCh, n = 19; hM4Di, n = 12; d: F2, 28 = 8.585, P = 0.0012; mCh, n = 10; hM3Dq, n = 11). e, Timeline of TRAP2 experiment. f, Left, duration in chamber with a familiar mouse or a novel mouse. Right, discrimination scores (t7 = 2.594, P = 0.0357; CNO, n = 8; saline, n = 10). g, Schematic and hM4Di expression in the NDB. Scale bar, 500 μm. h, Left, duration in chamber with a familiar mouse or a novel mouse. Right, discrimination scores (t17 = 1.61, P = 0.1259; mCh, n = 10; hM4Di, n = 9). i, Timeline of five-trial social memory test. j, Duration of direct interaction (F27, 108 = 2.478, P = 0.0005; n = 10). Statistical tests: c, hM4Di + CNO, mCh + CNO duration; d, f, h, duration; f, discrimination scores: two-tailed paired Student’s t-test. c, hM4Di + saline duration: two-tailed Wilcoxon signed rank test. c, d, Discrimination scores: one-way ANOVA with Tukey’s post-hoc test. h, Discrimination scores: two-tailed unpaired Student’s t-test. j, Two-way ANOVA with Tukey’s post-hoc test. NS, not significant; *P < 0.05, **P < 0.01, ***P < 0.001. Error bars denote s.e.m.

Source data

We next addressed whether activation of MS neurons by the excitatory DREADD hM3Dq enhances social memory (Extended Data Fig. 2d). There was no effect of CNO administration in hM3Dq mice before the initial social interaction when social memory was assessed at 10 min (Extended Data Fig. 2e). However, this manipulation prolonged the duration of social memory to 2 h in the hM3Dq mice that received CNO but not in the two cohorts of control mice (Fig. 1d). Indeed, hM3Dq activation of MS neurons during the social interaction maintained social memory for 24 h (Extended Data Fig. 2e) but had no effects on baseline sociability (Extended Data Fig. 2f) or inanimate object memory (Extended Data Fig. 2g). To address whether the MS neurons that were activated during the initial social interaction are specifically necessary for social memory formation, we expressed Cre-dependent hM4Di in the MS of TRAP2 mice and exposed these mice to a novel mouse after administering 4-OHT (Extended Data Fig. 2h). Two weeks later, TRAP2 mice that received saline before the social interaction exhibited normal social memory, which was blocked when the mice received CNO (Fig. 1e, f). Thus, the MS neurons that are activated by a novel social interaction are necessary for normal social memory formation.

To address whether the NDB is also required for social memory, we expressed hM4Di in the NDB but found that administration of CNO did not reduce social memory (Fig. 1g, h). As a final test of the role of the MS in social memory, we performed a five-trial social memory test3. As expected3, by trials 3 and 4, the control mice spent less time interacting with the now familiar intruder mouse, but on trial 5 the time spent interacting with a novel mouse increased to baseline levels (Fig. 1i,Β j, Extended Data Fig. 2i). By contrast, the mice in which MS activity was inhibited (hM4Di + CNO) maintained equal interaction times for all five trials (Fig. 1j, Extended Data Fig. 2i).

To address whether MS activity is necessary for memory acquisition and/or memory recall, we performed optogenetic manipulations by expressing the inhibitory halorhodopsin NpHR3.0 fused to eYFP (NpHR) or eYFP alone in the MS and providing light stimulation at various times during the social memory assay (Fig. 2a). Activation of NpHR throughout the procedure and during acquisition only disrupted social memory, whereas applying light during the recall phase had no effect (Fig. 2b). Social memory was also normal when NpHR mice were not exposed to light (Fig. 2b) and in eYFP mice that received light stimulation throughout the procedure (Fig. 2c). Similar to chemogenetic manipulations, optogenetic inhibition of the MS did not affect baseline sociability or inanimate object memory (Extended Data Fig. 3a–c).

Fig. 2: Inhibition of MS to dorsal CA2 projection disrupts social memory.
figure 2

a, Schematic of experiment. b, c, Left, duration that NpHR mice (b) or eYFP mice (c) spent in the chamber with a familiar mouse or a novel mouse. Right, discrimination scores (b: F3,35 = 5.227, P = 0.0044; n = 10; recall, n = 9; c: t9 = 0.1045, P = 0.919; n = 10). Acq, acquisition phase; rec., recall phase. d, f, Schematic of CNO infusion into dCA2 (d) or vCA1 (f). e, g, Left, duration in chamber with a familiar mouse or a novel mouse. Right, discrimination scores (e: t25 = 2.522, P = 0.0184; mCh, n = 15; hM4Di, n = 12; g: P = 0.4232; mCh, n = 9; hM4Di, n = 8). h, Left, timeline of experiment. Right, hM4Di expression. Scale bar, 500 μm. i, Left, duration in chamber with a familiar mouse or a novel mouse. Right, discrimination scores (P = 0.002; n = 15). Statistical tests: b, c, e, duration; c, discrimination scores; g, i, mCh duration: two-tailed paired Student’s t-test. b, Discrimination scores: one-way ANOVA with Tukey’s post-hoc test. e, Discrimination scores: two-tailed unpaired Student’s t-test. g,Β i, hM4Di duration: two-tailed Wilcoxon signed rank test. g, Discrimination scores: two-tailed Mann–Whitney test. i, Discrimination scores: Kruskal–Wallis with post-hoc Dunn’s test. NS, not significant; *P < 0.05, **P < 0.01. Error bars denote s.e.m.

Source data

Regulation of social memory by MS–dCA2 projections

Having established a critical role for the MS in social memory, we examined whether MS neuron projections to specific hippocampal subregions are required. We expressed hM4Di–mCh or mCh in the MS and implanted cannula above dCA2 or vCA1 to infuse CNO before the sociability assay (Extended Data Fig. 3d). Infusion of CNO into dCA2 of hM4Di mice, but not into dCA2 of mCh mice, blocked social memory (Fig. 2d, e) and had no effects on baseline sociability and inanimate object memory (Extended Data Fig. 3e, f). By contrast, CNO infusion into vCA1 of hM4Di mice had no effect on social memory (Fig. 2f, g).

CNO may have spread to dCA1 and dCA3 and influenced MS projections to these regions. To manipulate the activity of MS neurons that specifically synapse on dCA2 pyramidal neurons, we applied an intersectional genetic strategy in Amigo2-Cre mice, a dCA2-specific Cre-driver line3. We injected adeno-associated viruses (AAVs) expressing Cre-dependent TVA and rabies glycoprotein into dCA2 and an AAV expressing Flp-dependent hM4Di–mCh or mCh into the MS (Fig. 2h). Two weeks later, we injected a rabies virus expressing Flp (ref. 15) into dCA2 (Fig. 2h). Normal social memory was present in mCh mice administered CNO and in hM4Di mice administered saline but was absent in hM4Di mice administered CNO (Fig. 2i), with no differences between male and female mice (Extended Data Fig. 4a). DREADD-mediated inhibition of dCA2-projecting MS cells did not influence baseline sociability or inanimate object memory (Extended Data Fig. 4b, c). Because the MS is implicated in contextual memory16 and aversion17,18, which could influence social memory, we tested whether inhibition of this subset of MS cells influenced contextual fear conditioning or resulted in conditioned place preference or aversion. Fear conditioning was not affected by CNO administration in these hM4Di-expressing mice (Extended Data Fig. 4d) and had no effect in a conditioned place assay (Extended Data Fig. 4e).

To assess whether dCA2-projecting MS neurons send collaterals elsewhere, we used a marker to distinguish fibres of passage from axon terminals. We injected CAV-Flp into the dCA2 and Flp-dependent mGFP-synaptophysin-mRuby into the MS (Extended Data Fig. 4f). dCA2-projecting MS cells exhibited collaterals in the vCA1 and a smaller number in the supramammillary nucleus (Extended Data Fig. 4f). However, the MS collaterals innervating vCA1 are unlikely to be important for social memory as inhibiting the MS to vCA1 projection had no effect (Fig. 2g).

Plasticity at MS–dCA2 synapses

The MS contains a heterogeneous population of neurons14,19. To identify the MS cell types that synapse on dCA2 pyramidal neurons, we performed in situ hybridization to probe for cholinergic (Chat), glutamatergic (Slc17a6, gene name for VGLUT2) and GABAergic (Ξ³-aminobutyric-acid-producing) (Gad2) markers in MS cells that were labelled by GFP through monosynaptic rabies tracing in dCA2 of Amigo2-Cre mice (Extended Data Fig. 5a, b). Quantification of Gfp-positive cells revealed that around 90% of dCA2-projecting MS cells are Chat-positive, around 30% are Slc17a6-positive and around 5% are Gad2-positive, with approximately 80% of Slc17a6-positive cells and 50% of Gad2-positive cells also being positive for Chat (Extended Data Fig. 5c, d). Immunohistochemistry confirmed that the majority of dCA2-projecting MS neurons express CHAT (around 60%), whereas a smaller proportion (around 40%) express calcium/calmodulin-dependent protein kinase II (CaMKII), a marker for glutamatergic neurons20 (Extended Data Fig. 5e). To interrogate the functional role of cholinergic and glutamatergic inputs to dCA2 in social memory, we infused into dCA2 a mixture of muscarinic and nicotinic receptor antagonists or the NMDA receptor antagonist D-AP5 (Fig. 3a). Infusion of D-AP5 prevented social memory, which was not influenced by the blockade of cholinergic synaptic transmission or control infusion of saline (Fig. 3b). Baseline sociability was not influenced by any of these manipulations (Extended Data Fig. 5f, g) but inanimate object memory was prevented by infusion of the antagonists (Extended Data Fig. 5h), perhaps because of leakage into dCA1.

Fig. 3: Glutamatergic inputs from the MS to dCA2 have a crucial role in social memory formation.
figure 3

a, Timeline for dCA2 drug infusions. AP5, D-AP5; Mec, mecamylamine; Sco, scopolamine. b, Left, duration that mice spent in the chamber with a familiar mouse or a novel mouse. Right, discrimination scores (F2,36 = 4.017, P = 0.0266; n = 13). c, Schematic of ex vivo electrophysiology (ephys). d, Current-clamp recordings from dCA2 neurons. Input–output graph of peak postsynaptic potential (PSP) (n = 5/1, cells/mice). Scale bars, 10 mV, 50 ms. e, Sample responses to stimulation at different membrane potentials. Scale bars, 20 mV, 50 ms. f, Left, percentage of cells in which the ChETA (n = 20/9) and ChR2 (n = 22/6) induced PSCs (n = 42/15) were more than 60% reduced by NBQX (10 μM), picrotoxin (50 μM) or mecamylamine (5 μM). Right, representative traces. Scale bars, 50 pA, 100 ms. g, Schematic of experiment. h, Bottom, AMPAR/NMDAR ratios in dCA2 neurons (P = 0.0009) of mice that interacted with a novel object (n = 29/11, cells/mice) or a novel mouse (n = 27/7). Top, representative traces. Scale bars, 100 pA, 100 ms. i, Schematic of in vivo optogenetic LTD induction. j, k, Left, duration in chamber with a familiar mouse or a novel mouse. Right, discrimination scores (j: t9 = 2.538, P = 0.0318; n = 10; k: t9 = 1.155, P = 0.278; n = 10). Statistical tests: b, duration; j, k: two-tailed paired Student’s t-test. b, Discrimination scores: one-way ANOVA with Tukey’s post-hoc test. h, Two-tailed Mann–Whitney test. NS, not significant; *P < 0.05, ***P < 0.001. Error bars denote s.e.m.

Source data

To investigate whether glutamatergic inputs from the MS elicit synaptic responses in dCA2 pyramidal neurons, we expressed channelrhodopsin-2 (ChR2) or a ChR2 variant (ChR2(E123T) accelerated; ChETA) in the MS of Amigo2-Cre mice and Cre-dependent tdTomato in dCA2 (Fig. 3c). Current-clamp recordings from visually identified, tdTomato+ CA2 pyramidal neurons in ex vivo slices in response to optogenetic stimulation of MS inputs showed that excitatory postsynaptic potentials (EPSPs) were generated in all cells (n = 5). Increasing stimulation intensity increased EPSP amplitude (Fig. 3d) and generated action potentials (Fig. 3e). During voltage-clamp recordings, sequential application of the AMPAR antagonist NBQX, the GABA-A receptor antagonist picrotoxin and mecamylamine hydrochloride revealed that around 80% of neurons exhibitedΒ MS input-evoked postsynaptic currents (PSCs) that were substantially (more than 60%) reduced by NBQX, with less than 10% of neurons expressing PSCs that were reduced by picrotoxin or mecamylamine (Fig. 3f, Extended Data Fig. 6a). These results suggest that the major functionally important inputs from MS to dCA2 pyramidal neurons are glutamatergic.

To investigate the role of synaptic strength changes at MS to dCA2 pyramidal cell synapses in social memory, we exposed Amigo2-Cre mice that express ChR2 in the MS and Cre-dependent tdTomato in dCA2 to a novel mouse or novel object and prepared slices immediately after the interaction (Fig. 3g). AMPAR/NMDAR ratiosβ€”a surrogate measure for synaptic strength21β€”in dCA2 neurons were increased after social interaction compared to object interaction (Fig. 3h), whereas paired-pulse ratios were unchanged (Extended Data Fig. 6b). If an increase in strength at MS to dCA2 pyramidal cell excitatory synapses during novel social interactions is required for social memory, reducing this increase should impair social memory. We first confirmed that low frequency (1 Hz for 5 min) optogenetic stimulation of MS inputs elicited a long-term depression (LTD) at synapses on dCA2 pyramidal neurons, which was not influenced by the application of muscarinic, nicotinic and GABA-A receptor antagonists (Extended Data Fig. 6c). Applying the same stimulation protocol (1 Hz for 5 min) in vivo to MS inputs in dCA2 (Fig. 3i, Extended Data Fig. 6d) immediately after memory acquisition to reduce the increase in synaptic strength generated by the novel social interaction prevented the expression of social memory (Fig. 3j). Similarly, applying low frequency stimulation (1 Hz for 10 min) in vivo before the initial social interaction also prevented social memory formation (Extended Data Fig. 7a). Notably, applying the same protocols in control mice expressing eYFP had no effect on social memory (Fig. 3k, Extended Data Fig. 7b) and no effects on baseline sociability or inanimate object memory in either ChR2-or eYFP-expressing mice (Extended Data Fig. 7c–h). These findings suggest that strengthening glutamatergic synapses from MS neurons onto dCA2 pyramidal neurons during a novel social encounter is required for normal social memory.

Social memory modulation by MS 5-HT1B receptors

How might MS neurons be influenced during a novel social interaction? MS neurons express receptors for oxytocin and 5-HT22,23,24, neuromodulators that have roles in social behaviours8,9. To explore the potential role of oxytocin and 5-HT action in the MS in social memory, we infused oxytocin receptor (OXTR) or 5-HT receptor (5-HTR) antagonists into the MS before the social memory assay (Fig. 4a). Social memory was not influenced by infusion of saline or an OXTR antagonist (L-368,899 hydrochloride) but was prevented by infusion of the promiscuous 5-HTR antagonist, methiothepin maleate (Fig. 4b). Infusion of the broad 5-HT2R antagonist methysergide or the 5-HT1AR antagonist NAD-299 did not influence social memory, but infusion of the specific 5-HT1BR antagonist NAS-181 impaired it (Fig. 4b). None of these manipulations had detectable effects on baseline sociability (Extended Data Fig. 8a). Because social memory was impaired by a 5-HT1BR antagonist, we next tested whether a 5-HT1B agonist (CP93129) might prolong social memory in a manner similar to increasing MS neuron activity (Fig. 1d). Indeed, although social memory was again absent at 2 h after the initial social interaction in control mice, MS infusion of CP93129 before the social interaction resulted in social memory at 2 h (Fig. 4c).

Fig. 4: Bidirectional modulation of social memory by MS 5-HT1B receptors.
figure 4

a, Schematic and infusion site labelled with ink. Scale bar, 500 μm. b, c, Left, duration that mice spent in the chamber with a familiar mouse or a novel mouse. Right, discrimination scores (b: F5,78 = 6.404, P < 0.0001; n = 14. c: t13 = 2.602, P = 0.0219; n = 14). d, Schematic of fibre photometry experiment. e, h, Schematic and GCaMP6f expressionΒ (CaMKIIG–CaMP6f (e); Ef1α–DIO–GCaMP6f (h). Scale bars, 500 μm. f, g, i, j, Left, time courses of average GCaMP6f transient z-scores event-locked to direct interactions (β€˜no’, novel object). Right, quantification of average during interaction (f: F3,16 =  = 6.392, P = 0.0047; n = 5; g: t4 = 5.103, P = 0.007; n = 5; i: F2,12 = 5.523, P = 0.0199; n = 5; j: t4 = 3.752, P = 0.0199; n = 5). k, Schematic of ex vivo electrophysiology set-up. l, Summary time course of IPSCs in response to CP93129 (5 μM) (n = 15/7 (cells/mice)). Top, representative tracesΒ recorded at indicated time points 1 and 2. Scale bars, 100 pA, 50 ms. m, Summary time course of EPSCs in response to CP93129 (5 μM) (n = 9/4). Top, representative traces. Scale bars, 100 pA, 10 ms. n, Average PSC (% of control) in the presence of CP93129 (t22 = 1.854, P = 0.0386; IPSC, n = 9; EPSC, n = 15). Statistical tests: b, duration; c, g, j: two-tailed paired Student’s t-test. b, Discrimination score; f, i: one-way ANOVA with Tukey’s post-hoc test. n, One-tailed unpaired Student’s t-test. NS, not significant; *P < 0.05, **P < 0.01, ***P < 0.001. Error bars and shading denote s.e.m.

Source data

Our findings predict that MS neuron activity increases during a novel social interaction in a 5-HT1BR-dependent manner. To test this hypothesis, we expressed GCaMP6f in MS glutamatergic neurons and recorded MS neuron activity while the subject mouse had sequential interactions with a novel mouse; the same, now familiar mouse; a novel inanimate object; and, finally, another novel mouse (Fig. 4d, e). Activity in MS neurons increased robustly during interactions with a novel mouse whereas interactions with a familiar mouse or a novel inanimate object generated a smaller increase in MS neuron activity (Fig. 4f). Administration of NAS-181 reduced the increase in MS neuron activity during the novel mouse interaction (Fig. 4g), demonstrating the necessity of 5-HT1BRs. Similarly, the activity of MS cells that project to dCA2, captured by expressing CAV-Cre in dCA2 and Cre-dependent GCaMP6f in MS, was significantly higher during interaction with a novel mouse versus a familiar mouse or novel object and was also diminished after administration of NAS-181 (Fig. 4h–j).

To address how 5-HT1BRs may contribute to increasing MS neuron activity during novel social interactions, we performed ex vivo whole-cell recordings from visually identified dCA2-projecting MS neurons that were labelled by expression of CAV-Cre in dCA2 and Cre-dependent EGFP in MS (Fig. 4k). Application of the 5-HT1BR agonist CP93129 (2.5 or 5 ¡M) generated an inward, depolarizing current in around 60–70% of dCA2-projecting MS neurons (Extended Data Fig. 8b–e). Because 5-HT1BRs are commonly presynaptic and inhibit neurotransmitter release25, we hypothesized that activation of 5-HT1BRs depresses inhibitory synaptic transmission and increases MS neuron activity through β€˜disinhibition’. Consistent with this prediction, CP93129 decreased evoked inhibitory postsynaptic currents (IPSCs) by around 50% (Fig. 4l) and also depressed EPSCs to a smaller degree than IPSCs (Fig. 4m, n). These effects of CP93129 were not due to β€˜rundown’ of synaptic currents, as EPSCs and IPSCs were stable for 45 min using identical recording conditions (Extended Data Fig. 8f–k). The depression of IPSCs is likely to be through a presynaptic action, as evidenced by a decrease in the frequency of spontaneous IPSCs and no effect on their amplitude (Extended Data Fig. 8l, m).

Modulation of social memory by MR 5-HT release

Our results suggest that social memory formation during a novel social interaction requires 5-HT release in the MS, which increases the activity of dCA2-projecting MS neurons through disinhibition. Consistent with the median raphe (MR) being a source of 5-HT in the MS26, expressing Cre-dependent EGFP in the MR of Sert-Cre mice revealed 5-HTergic axon innervation in the MS (Extended Data Fig. 9a). Using TRIO (tracing the relationship between input and output) monosynaptic rabies virus tracing27, we also observed labelling in the MR (Extended Data Fig. 9b), suggesting that there is direct synaptic input from the MR onto dCA2-projecting MS cells. To test the necessity of MR-mediated 5-HT release in the MS for social memory, we expressed Cre-dependent NpHR3.0–eYFP or eYFP alone in the MR of Sert-Cre mice and placed an optical fibre adjacent to the MS (Fig. 5a). Optogenetic inhibition of MR 5-HT inputs in the MS during the initial social interaction impaired social memory, which was normal in the control eYFP mice and when light was not applied to the NpHR mice (Fig. 5b, c). Baseline sociability and inanimate object memory were unaffected by these manipulations (Extended Data Fig. 9c–f).

Fig. 5: MR 5-HT release in the MS regulates social memory.
figure 5

a, d, Schematic and MR injection site (NpHR3.0–eYFP (a); ChR2–eYFP (d)). Scale bars, 500 μm. b, c, e, f, Left, duration that mice (NpHR mice (b) versus eYFP mice (c); ChR2 mice (e) versus eYFP mice (f)) spent in the chamber with a familiar mouse or a novel mouse. Right, discrimination scores (b: t9 = 2.27, P = 0.0493; n = 10; c: t9 = 0.4444, P = 0.6672; n = 10; e: t10 = 2.471, P = 0.033; n = 11; f: t9 = 0.2896, P = 0.7787; n = 10). g, Schematic and MS infusion site filled with ink. Scale bar, 500 μm. h, Left, duration in chamber with a familiar mouse or a novel mouse. Right, discrimination scores (t12 = 2.202, P = 0.048; saline, n = 14; CP, n = 13). i, Left, duration in chamber with a familiar object (fo) or a novel object (no). Right, discrimination scores (t13 = 0.3522, P = 0.7304; n = 14). Two-tailed paired Student’s t-test except for e, ChR2 off duration; i, saline duration: two-tailed Wilcoxon signed rank test. NS, not significant; *P < 0.05, **P < 0.01, ***P < 0.001. Error bars denote s.e.m.

Source data

To test whether enhancing 5-HT release in the MS influences the duration of social memory, we expressed ChR2–eYFP or eYFP in MR 5-HT neurons of Sert-Cre mice (Fig. 5d). Activation of 5-HT inputs in the MS during the initial novel social interaction generated social memory lasting 2 h, which was not present in the control conditions (Fig. 5e, f). Similar to previous results, these manipulations had no effects on baseline sociability (Extended Data Fig. 9g, h). To investigate whether our findings might have therapeutic implications, we generated mice that are haploinsufficient for neuroligins 1, 2 and 3 (Nlgn123hetΒ mice)28. Because Nlgn3 knockout or mutation impairs social memory29, but not sociability30, we predicted that Nlgn123het mice would exhibit the same behavioural phenotype. Indeed, these mice exhibited normal baseline sociability (Extended Data Fig. 9i) but lacked social memory (Fig. 5h). Infusion of CP93129 into the MS rescued this social memory deficit (Fig. 5g, h) but had no effect on inanimate object memory (Fig. 5i) or sociability (Extended Data Fig. 9i).

Discussion

Previous work has established that the hippocampal dCA2 subregion has a specific role in social memory through its projections to vCA13,5, which in turn sends projections to the nucleus accumbens that are also required for social memory6. Here we show that increased activity in dCA2-projecting glutamatergic MS neurons during the initial social interaction is critical for social memory formation, probably because the strength of MS to dCA2 pyramidal neuron synapses increases. Furthermore, we show that dCA2-projecting MS neuron activity during a novel social interaction requires the MR-mediated release of 5-HT, which influences the stability of social memory through actions on 5-HT1BRs in the MS (Extended Data Fig. 10). Particularly notable were the findings that infusion of a 5-HT1BR agonist into the MS prolongs the duration of social memory and rescues social memory deficits in an autism-associated mouse model. Given the role of neuroligins in synaptic function in CA1 pyramidal neurons31, we surmise that the impairment of social memory in Nlgn123het mice may be due to deficient strength and/or plasticity at MS to dCA2 pyramidal neuron synapses and that administration of CP93129 into MS enhances activity in dCA2-projecting MS neurons in a manner that compensates for their synaptic communication deficit.

Our current model (Extended Data Fig. 10) suggests that 5-HT in the MS primarily works presynaptically to preferentially reduce the inhibition of dCA2-projecting MS neurons, perhaps through volume transmission32,33 in a manner similar to the domain-overlap model proposed for dopamine34. However, we cannot rule out a role for 5-HT in acting directly on this population of neurons. Of note, none of our many manipulations of 5-HT action in the MS or MS neuron activity influenced basal sociability, suggesting that the appetitive value of a novel social interaction was not affected. Similar manipulations of 5-HT release in the nucleus accumbens have robust effects on sociability, also through 5-HT1BRs8,9. Furthermore, 5-HT action in other brain structures appears to be involved in aggression and perhaps impulsivity35,36. Thus, our results add to the growing evidence that global conclusions about the functions of neuromodulators such as 5-HT are not warranted. Instead, the function of 5-HT or other neuromodulators needs to be considered in the context of the brain region in which they are acting.

5-HT neurons in the MR are heterogeneous and consist of a variety of distinct molecular subtypes37,38, which may orchestrate different behaviours39. Therefore, it remains to be determined which specific subtypes of 5-HT MR neurons are critical for social memory and what role the release of other neurotransmitters from MR neurons, such as glutamate, might have40. It is also important to acknowledge that neuromodulators such as 5-HT do not act in isolation. The actions of both oxytocin41,42 and dopamine29 in structures other than the MS have been implicated in social memory, suggesting that a comprehensive understanding of how social memories form will require simultaneously examining the actions of multiple neuromodulators in a range of brain areas.

Methods

Mice

Female and male C57BL/6J (JAX, 664), TRAP2 (ref. 43) (a gift from L. Luo), TRAP2;Ai14 (a gift from L. Luo), B6.Cg-Tg(Amigo2-cre)1Sieg/J (Amigo2-Cre, JAX, 30215)3, Tg(Slc6a4-cre)ET33Gsat (Sert-Cre, JAX, 3836639) and Nlgn123het mice were used as experimental subjects. A breeding strategy for the transgenic lines was used, in which strictly transgene-carrying male mice were crossed with wild-type female C57BL/6J mice sourced commercially to produce heterozygous offspring. Nlgn123het mice were generated by breeding Nlgn123cKO28 (a gift from T. C. SΓΌdhof) male mice with B6.C-Tg(CMV-cre)1Cgn/J (JAX, 6054) females. Mice were weaned at 21 days old and housed with 2–5 mice per cage. Behavioural experiments were performed when mice were 7–14 weeks old. All mice were housed on a 12-h light–dark cycle with food and water ad libitum and behavioural experiments were conducted during the same circadian period (07:00–19:00). All procedures complied with the animal care standards set forth by the National Institutes of Health (NIH) and were approved by the Stanford University Administrative Panel on Laboratory Animal Care and Administrative Panel of Biosafety. No statistical methods were used to predetermine sample size, which was based on previous experience with the variance of the assays. All experiments were analysed without knowledge of the specific manipulation each mouse had undergone and performed without knowledge of the identity of the virus that had been injected or the transgene being expressed.

Stereotactic injections and cannula implantation

Mice, 4–7 weeks of age, were anaesthetized by intraperitoneal injection with a mixture of ketamine (75 mg per kg body weight) and dexmedetomidine (0.375 mg/kg body weight). Heads were fixed on a Kopf stereotaxic apparatus and holes were drilled bilaterally into the skull except for structures on the midline. For injections into specific brain regions the following bregma coordinates were used: dCA2, βˆ’1.6 mm anteroposterior (AP), Β±1.6 mm mediolateral (ML), 1.7 mm dorsoventral (DV) from dura; MS, 0.65 mm AP, 0 mm ML, 4 mm DV; NDB, 1.25 mm AP, 0 mm ML, 5 mm DV; MR, βˆ’4.4 mm AP, 0 mm ML, 4.6 mm DV. Glass cannulas filled with virus solution were lowered to the specified depth from the dura and 0.5–1.5 ¡l of virus solution was injected using a microinjection pump (Harvard Apparatus) at a flow rate of 0.1–0.25 ¡l minβˆ’1. After surgery, atipamezole (10 mg per kg body weight) was administered by intraperitoneal injection.

Adeno-associated viruses (AAVs) used for stereotactic injections were purchased from the Stanford Neuroscience Gene Vector and Virus Core and included: AAVDJ-hSyn-EGFP, AAVDJ-hSyn-hM4Di-mCherry, AAVDJ-hSyn-DIO-hM4D(Gi)-mCherry, AAVDJ-hSyn-fDIO-hM4D(Gi)-mCherry, AAVDJ-hSyn-hM3D(Gq)-mCherry, AAVDJ-hSyn-mCherry, AAV8-EF1Ξ±-fDIO-mCherry, AAV8-EF1Ξ±-NphR3.0-eYFP, AAVDJ-EF1Ξ±-DIO-eYFP, AAVDJ-EF1-ChETA-eYFP, AAVDJ-hSyn-ChR2-eYFP, AAVDJ-EF1Ξ±-DIO-tdTomato, AAVDJ-CAMKII-GCaMP6f, AAVDJ-EF1Ξ±-DIO-GCaMP6f, AAVDJ-EF1Ξ±-DIO-NpHR3.0-eYFP, AAVDJ-EF1Ξ±-DIO-ChR2-eYFP, AAVDJ-EF1Ξ±-DIO-eYFP, AAVDJ-CMV-DIO-EGFP and AAV-DJ-hSyn-FRT-mGFP-2A-Synaptophysin-mRuby44. AAV titres ranged from 1 Γ— 1012 to 2 Γ— 1013 gc mlβˆ’1. CAV2-Cre45 was purchased from E. Kremer (Institut de GΓ©nΓ©tique MolΓ©culaire de Montpellier). For monosynaptic tracing, synapsin-driven lentiviral NLS-GFP-Cre, AAV-CAG-FLEx-TC, AAV-CAG-FLEx-G and rabies virus were obtained from Janelia Research Campus. The rabies virus expressing Flp (SIR-Flp)15 was provided by K. Beier. Behavioural experiments involving manipulations of cell bodies were conducted 3–4 weeks after virus injections. The incubation time was 6–8 weeks for experiments involving manipulation of axon terminals.

For optogenetic behavioural experiments, optic fibres were implanted above the MS (coordinates: 0.65 AP, 0 ML, 3.5 DV) and dCA2 (βˆ’1.6 AP, Β±1.6 ML, 1.2 DV). Optical implants were self-manufactured using 1.25-mm-diameter multimode ceramic ferrules (ThorLab), 200-Β΅m fibre optic cable with numerical aperture (NA) 0.39 (ThorLabSA) and blue dye epoxy (Fibre Instrument Sales). For securing the optical implants to the skull, miniature screws (thread size 00–90 × 1/16, Antrin Miniature Specialties) and light-cured dental adhesive cement (Geristore A&B paste, DenMat) were applied. For drug infusions, a 26-gauge guide cannula (Plastics One) was first implanted bilaterally or unilaterally for midline structures into the brain region being studied. For dCA2 drug injection, the guide cannula was 0.9 mm in length from the cannula base and the infusers (essentially needles that fit into the guide cannula) for injections were 33-gauge and 2.4 mm in length. The cannulas for implantation above the vCA1 (coordinates: βˆ’3.16 AP, Β±3.1 ML, 4 DV) were 2.8 mm in length and the infusers were 4.3 mm. The cannula implants for MS were 3.2 mm in length and the infusers were 4.7 mm. On the basis of visual histological analysis and blinded to the experimental results of the subject mice, a small percentage of the mice (around 5% of more than 300 mice) were excluded from behavioural analysis according to the following criteria: (1) off-target transgene expression (substantial somatic expression of fluorophore outside the region of interest by visual inspection); (2) weak transgene expression; or (3) inaccurate implant placement.

Intraperitoneal injection and cannula infusion of drugs

4-hydroxytamoxifen (4-OHT, Sigma H6278-50MG) was delivered via intraperitoneal (IP) injection at 50 mg kgβˆ’1 45 min before behavioural experiments. Clozapine N-oxide (CNO, Tocris Biosciences 4936) was also given via IP injection at a concentration of 10 mg kgβˆ’1 45 min before behavioural assays. For cannula microinjection experiments, 500 pg of CNO was infused in a total volume of 200 nl per side at a speed of 200 nl minβˆ’1 through an injector cannula using a microinfusion pump (Harvard Apparatus). The following drugs were also administered via microinjections at the same speed in a total volume of 400 nl–2 μl: AP-5 (Tocris Biosciences 0106, 2.4 μg), a mixture of mecamylamine hydrochloride (Tocris Biosciences 2843, 2 μg) and scopolamine hydrobromide (Tocris Biosciences 1414, 3 μg), L-368,899 hydrochloride (OXTR-A, Tocris Biosciences 2641, 0.25 μg), methiothepin mesylate salt (Millipore Sigma M149, 0.2 μg), methysergide maleate (Tocris Biosciences 1064, 0.5 μg), NAD-229 (Tocris Biosciences 3282, 3.5 μg), NAS-181 (Tocris Biosciences 1413, 2 μg) and CP93129 dihydrochloride (Tocris Biosciences 1032, 0.5 μg). Injection infusers were removed 2 min after completion of the fusion and mice were returned to their home cages for 30 min before behavioural assays. NAS-181 (10 mg kgβˆ’1) was administered IP 30 min before fibre photometry experiments.

TRAP2 behavioural experiments

Mice were habituated for two days to IP injections (saline, 45 min prior) before placement in empty behavioural chambers (30 min, 23 cm length Γ— 23 cm height Γ— 25 cm width). On day three, female TRAP2;Ai14 mice (4–6 weeks old) were injected with 4-OHT (50 mg kgβˆ’1). Forty-five minutes later, mice were divided into three groups. Group 1 was placed in behavioural chambers with a novel female TRAP2;Ai14 mouse. Group 2 was placed in the chambers with an inanimate object (for example, a Lego toy as shown in Fig. 1a) and group 3 was placed into an empty chamber. After 4 h, mice were returned to their home cages and 10 days later perfused with 4% paraformaldehyde (PFA) in PBS. The brains were placed into 4% PFA overnight and then cut on a vibratome into 60-ΞΌm slices. Images were acquired with an Olympus automated VS120 slide scanner (10Γ— objective) and overlaid with images from Allen Brain Atlas for blinded manual counting of tdTomato-positive cells in specified brain regions.

Optogenetic stimulation

Mice were habituated for two days in the behavioural chambers with diodes (in off state) connected to their optic fibres. For photostimulation of NpHR3.0, optical implants were connected to a 532-nm laser diode (Shanghai Dream Lasers Technology) via a FC/PC adaptor and a fibre optic rotary joint (Doric Lenses). For photostimulation of ChR2, ferrules were connected to a 473-nm laser diode (OEM Laser Systems). A Master-8 pulse stimulator (A.M.P.I.) was used to control the laser, which was adjusted to around 8 mW for somatic stimulation and around 15 mW for axon terminal stimulation using a digital power meter console (ThorLabs). To avoid tissue overheating when stimulating NpHR3.0, a light on–off cycle of 8 s on and 2 s off was applied. For ChR2 stimulation, 5-ms light pulses at 1 Hz were applied.

Three-chamber behavioural tests

Test mice were habituated to IP injections or to the fibre optic patch cord, depending on the experiment, as described above. They were also habituated to the three-chamber apparatus, which contained two empty upside-down metal grid pencil cups (10 cm in diameter) in the outer two chambers, for 5 min on two consecutive days. The three-chamber apparatus (72 cm length Γ— 23 cm width Γ— 25 cm height) was constructed of 0.635-cm-thick sheets of clear extruded acrylic for walls, and Komatex for floors (white) and barrier walls (black; TAP Plastics), with two outer chambers (28 cm length Γ— 23 cm width) connected by a centre chamber (16 cm length Γ— 23 cm width). The chambers were divided by translucent acrylic walls with 6.5-cm-diameter holes, 1 cm from the chamber floor. These dividers were removed during optogenetic experiments to allow subject mice to move freely with their patch cords attached. Mice placed under the cup were also habituated to the cup by spending 5 min under the cup on two consecutive days. On the test day, subject mice were placed in the middle chamber for 1 min, at which time the dividers were lifted, and the mice were allowed to explore the other two chambers freely for 10 min. One chamber contained an inanimate object, and the other chamber contained a novel mouse of the same sex. In the case of male mice, the mouse under the cup was a juvenile (3–5 weeks old). This session is referred to as a sociability test and was performed similarly to previously described methods8. The location of novel mice and objects was counterbalanced between sessions. After an interval of 10 min, 2 h or 24 h, subject mice were placed back into the middle chamber for 1 min at which time the barriers were removed, and the mice were allowed to freely explore for 5 min. During this social memory assay6,7,46 one chamber contained a mouse under a cup from the previous session (familiar mouse) and the other chamber contained a novel mouse under a cup. Again, the location of novel mouse and familiar mouse was counterbalanced between sessions.

For analysis, a video tracking system (BIOBSERVE) was used to automatically track the location of the subject mouse during its free exploration. The sociability discrimination score was calculated as: [(time in novel mouse chamberβ€‰βˆ’β€‰time in object chamber)/(time in novel mouse chamber  +  time in object chamber)]. The social memory discrimination score was calculated as: [(time in novel mouse chamberβ€‰βˆ’β€‰time in familiar mouse chamber)/(time in novel mouse chamber  +  time in familiar mouse chamber)]. The object memory test was conducted in the same manner as the social memory test, with the difference that an object was placed into one chamber and the other chamber remained empty in the first session and the familiarized object was placed in one chamber and a novel object in another chamber in the second session. The same cohort of mice was used for social and object memory tests with the sequence of the two tests counterbalanced within the same cohort.

Female mice were used for the initial experiments illustrated in Figs. 1, 2a–g and Extended Data Figs. 1–3 to avoid potential confounds of aggressive behaviour between the subject mice and object mice under the cups. Beginning with the experiments illustrated in Fig. 2h and Extended Data Fig. 4, both male and female mice were used in all behavioural tasks and there was no sex difference between their discrimination scores in the sociability assays or social memory tests in any experimental condition. For all remaining experiments, both male and female subject mice were used in approximately equal numbers. Subject mice were excluded from the analysis (less than 2% of total), if they spent the entirety of the assay in only one chamber or if their velocity was 0 for more than 50% of the assay.

Five-trial social memory test

This test was performed as previously described3,47. Female C57BL/6J subject mice were individually housed for 7 days before the behavioural assay. On the day of testing, the mice were kept in their home cage and presented with a 10-week-old novel C57BL/6J female mouse for 4 successive 1-minute trials, interspersed by 10-min intervals. On the final trial, another novel mouse was presented. The trials were recorded, and the direct interaction time was scored manually and blinded to the experimental manipulation performed on the subject mouse.

Fear conditioning

Experiments were conducted in operant conditioning chambers (20 cm length Γ— 24 cm width Γ— 18 cm height, Med Associates) contained within sound-attenuating cubicles. The bottom of the chamber was composed of a metal grid, through which the foot shock was delivered. On day 1, mice received IP injections of CNO (10 mg kgβˆ’1) 45 min before the experiment. Mice were individually placed into the fear conditioning chamber, which was illuminated by white light. After 2 min of free exploration, three foot shocks of 0.35 mA (lasting 2 s) were administered with an interval of 1 min. The subject mouse remained in the chamber for 1 min before being placed in a separate cage. After all mice from one cage underwent fear conditioning, all subject mice were returned to their home cage. After 24 h, mice were placed individually into the same chamber and per cent time freezing was analysed using video tracking software (BIOBSERVE).

Conditioned place preference

The experimental protocol was similar to that previously described48. On day 1, mice were tested for baseline preference by placing them in an unbiased chamber containing two interchangeable halves of flooring made of two textures. The β€˜grid’ floor was composed of 13-cm acrylic rods (0.15-cm diameter) mounted 0.5 cm apart in acrylic rails (0.5 cm thick, surrounding all sides). The β€˜hole’ floor was made out of perforated acrylic sheet with 8-mm round holes, 1 cm apart mounted on acrylic rails. Both floors (23 cm length Γ— 14 cm width) were placed in the bottom of a black box (23 cm length Γ— 23 cm width Γ— 25 cm height). After 15 min of free exploration, mice were returned to their home cage. On day 2, mice were administered saline via IP injection 45 min before the conditioning. Each subject mouse explored the black box with either two hole or two grid flooring sections for 15 min. After 4 h, mice received an IP injection of CNO (15 mg kgβˆ’1) and were placed 45 min later into the chamber containing two sections of the alternate flooring for 15 min. On day 3, mice were placed into the chamber containing both flooring types and time spent on either type of flooring was scored using video tracking software (BIOBSERVE).

Immunohistochemistry

Mice were perfused with 4% PFA or 10% formalin and brains were removed and post-fixed overnight at 4 °C. For procedures not involving immunohistochemistry (IHC) and in situ hybridization, 60–75-ΞΌm coronal sections were prepared on a vibratome. For IHC, brains were transferred to 30% sucrose for 48 h to cryo-preserve, mounted on the cryostat mounting disk with Tissue-Tek O.C.T. Compound (Sakura) on the tissue rack at βˆ’20 °C and sectioned on a cryostat into 40-ΞΌm slices. Free-floating sections were processed for immunohistochemistry. After three 10-min washes in PBS on a shaker, the tissue was incubated with blocking solution (5% normal goat serum or donkey serum and 0.3% Triton X-100 in PBS) for 45 min and then incubated in primary antibodies overnight at 4 °C on a shaker. The primary antibodies used were: rabbit polyclonal anti-CaMKII 1:200 (Thermo Fisher Scientific, PA5-38239), rabbit polyclonal anti-choline acetyltransferase (CHAT) 1:200 (Millipore Sigma, AB143), chicken anti-GFP 1:1,000 (Aves labs, GFP-1020). After three washes of 10 min in PBS, secondary antibodies were added and incubated for 3 h at room temperature on a shaker. The secondary antibodies used were: goat anti-rabbit Alexa Fluor 546 1:500 (Thermo Fisher Scientific, A11035), donkey anti-chicken Alexa Fluor 488 (Jackson Immuno Research Labs, 703545155). After three more washes the slices were mounted with Fluoromount-G mounting medium (Southern Biotech) onto microscopy slides for visualization at 10Γ— using an Olympus automated VS120 slide scanner (UPLSAPO 10Γ— objective, NA 0.4, fluorescence light source: X-Cite 120LED light engine, filter cubes: DAPI, FITC, TRITC, Cy5, wavelength: 430–741 nm, XM10 camera, bit depth of images: 14 bit) or Nikon A1 confocal microscope (CFI Plan Apochromat Lambda 10Γ— objective, NA 0.45, lasers: LU-N4/N4S 4-laser unit, 405 nm, 488 nm, 561 nm, 640 nm, filter cubes: 450/50, 525/50, 600/50, 685/70, 700/75, wavelength: 425–738 nm, detector: A1-DU4-2 4 Detector Unit: 4 Multi-Alkali PMTs, bit depth of images: 12 bit).

In situ hybridization

The experiment was carried out using the RNAscope Multiplex Fluorescent v2 kit and was performed according to the ACD Bio manual49. Brains were collected, fresh-frozen on dry ice and stored at βˆ’80 °C until sectioning on the cryostat. Fifteen-micrometre slices were prepared and collected directly on Superfrost plus microscopy slides. Following a 15-min fixation in prechilled 4% PFA, slices were rinsed twice in PBS and dehydrated gradually in higher concentrated ethanol in the order: 50%, 70%, two times 100% ethanol (5 min each). Slides were air-dried for 5 min and a barrier was drawn with a hydrophobic pen around the slices. Slides were then placed into a humidity control tray and incubated with around 5 drops of RNAscope hydrogen peroxide each for 10 min. After one wash with distilled water, slices were incubated with Protease IV for 30 min. After two washes in PBS, slices underwent a hybridization step in a probe mix for 2 h at 40 °C in a HyEZ Oven, at which time they were washed twice at room temperature in wash buffer (50Γ— RNAscope Wash buffer diluted in distilled water). The following probes were used: RNAscope Probe-EGFP-C1 (400281), Mm-Gad2-C2 (415071-C2), Mm-Slc17a6-C3 (319171-C3), Mm-Chat-C4 (408731-C4). Probes were warmed up to 40 °C in a water bath for 10 min before use. 1 volume of C2, C3 and/or C4 were diluted in 50 volumes of C1. In the first amplification step, slides were incubated with around 6 drops of RNAscope Multiplex FL v2 Amp1 for 30 min at 40 °C in the HyEZ Oven and subsequently washed twice with wash buffer at room temperature This amplification step is repeated with RNAscope Multiplex FL v2 Amp2 and RNAscope Multiplex FL v2 Amp3, whereby the incubation time in Amp3 was only 15 min. For the development of the horse radish peroxidase (HRP) signal, slides were incubated with RNAscope Multiplex FL v2 HRP-C1 in the HyEZ Oven at 40 °C for 15 min and rinsed twice in wash buffer at room temperature. Afterwards, slides were incubated in 1:1,500 diluted Opal 520, 570, 620 or 690 (depending on the probe-dye combination) for 30 min at 40 °C and rinsed twice in wash buffer. This step was repeated with HRP-C2, HRP-C3 or HRP-C4 and different Opal dyes. In the final mounting step, Fluoromount-G mounting medium was dripped onto the slides and cover slips were placed over the slices. Slides were imaged on a Leica SP8 confocal (20Γ— HC PL APO IMM CORR CS2 objective, NA 0.75, Argon laser, 65 mW, filter cubes: 458, 476, 488, 496, 514 nm, wavelength: 470–670 nm, detector: 5-channel confocal detection: 3 Hybrid-GaAsP detectors for increased light sensitivity, 2 standard fluorescent photomultiplier tubes (PMT), bit depth of images: 8 bit).

IHC and in situ quantification

All samples were imaged with the same settings to allow comparison between samples and background subtraction and thresholds were set uniformly for all images. Based on a pixel-intensity threshold, images were binarized and merged with DAPI in ImageJ50. A signal in a given channel that colocalized with DAPI was manually identified as a positive cell. Counting was conducted by visual scanning and with a manual cell counter.

Ex vivo electrophysiology

For recording synaptic responses from dCA2 pyramidal neurons, 4–7 weeks after stereotactic injections of AAVDJ-EF1-ChETA-eYFP or AAVDJ-hSyn-Ch2-eYFP into the MSDJ AAVDJ-EF1Ξ±-DIO-tdTomato into dCA2, Amigo2-Cre mice were euthanized and brains were placed into sucrose cutting solution containing (in mM): 228 sucrose, 26 NaHCO3, 11 glucose, 2.5 KCl, 1.2 NaH2PO4, 7 MgCl2 and 0.5 CaCl2 and sliced on a Leica vibratome. Coronal MS and hippocampal slices (200–250 μm) were transferred into artificial cerebrospinal fluid (ACSF) containing (in mM): 119 NaCl, 26 NaHCO3, 11 glucose, 2.5 KCl, 1.2 NaH2PO4, 1.3 MgCl2 and 2.5 CaCl2 (osmolarity 289–295) for recovery at 32 °C for 30 min and then further 60 min of equilibration at room temperature. To evaluate the accuracy of injections, MS slices were placed in a recording chamber perfused with 28–30 °C ACSF equilibrated with 95% O2 and 5% CO2 and visualized with a 40Γ— water-immersion objective on an upright fluorescent microscope (BX51WI; Olympus) equipped with infrared-differential interference contrast video microscopy and epifluorescence (Olympus). If MS neurons expressed eYFP, the hippocampal slices from that mouse were then placed in the recording chamber for optical stimulation. Whole-cell voltage-clamp recordings were made with pipettes (3 –4 MΞ©) filled with (in mM): 140 CsMeSO4, 8 NaCl, 10 HEPES, 0.25 EGTA, 2 Mg2ATP, 0.3 Na3GTP, 0.1 spermine and 7 phosphocreatine (pH 7.29–7.36; osmolarity 290–295). Series and input resistance were continuously monitored with βˆ’4-mV, 70-ms pulse delivered through the recording pipette. Experiments were discarded if the series resistance varied by >15%. In some experiments CsMeSO4 was replaced with an equimolar concentration of KMeSO4 to perform current-clamp recordings or voltage-clamp recordings (for experiments in Extended Data Fig. 8b).

Whole-cell recordings were made from visually identified tdTomato-labelled pyramidal neurons in the CA2 while 473-nm 5-ms light pulses from the Opto Duet Laser system (IkeCool Corporation) were delivered to the whole slice via a 40Γ— water-immersion objective on an Olympus microscope (BX51WI) at 0.1 Hz, thereby stimulating ChETA or ChR2 in MS axons and/or terminals. For voltage-clamp recordings, baseline PSCs were recorded at βˆ’70 mV for 5 min, at which time NBQX (10 μM) was added to the ACSF and PSCs recorded until their amplitudes were stable for 5 min. Then mecamylamine hydrochloride (5 μM) and in some cases scopolamine hydrobromide (5 μM) were added and PSCs assayed until stable for 5 min. The holding membrane potential was then adjusted to -50 mV and picrotoxin (50 uM) was applied to assess the contribution of GABA-A receptors to the PSCs. In 40% of cells, the order in which drugs were applied was shuffled with no detectable differences in their effects. For calculation of AMPAR/NMDAR ratios, the ACSF contained picrotoxin, mecamylamine hydrocholoride and scopolamine hydrobromide. The peak amplitude of an averaged AMPAR EPSC (n = 27–29 consecutive EPSCS measured at βˆ’70 mV) was divided by the amplitude of an averaged NMDAR EPSC (n = 27–29 measured at 50 ms after the onset of the EPSC at +40 mV). Paired-pulse ratios of averaged AMPAR EPSCs (n = 17–24) were calculated using a 50-ms interstimulus interval. To assess the strength of MS inputs to CA2 neurons (Fig. 3d) the peak amplitude was measured from an average of 10 consecutive EPSPs evoked at each light power. To elicit LTD at MS to dCA2 pyramidal neuron synapses in slices (Extended Data Fig. 6c), we applied 300 5-ms light pulses at 1 Hz. Summary graphs were generated by averaging individual EPSC measurements in 1-min bins and expressing each point as a percentage of the average of a 5-min baseline EPSC.

MS neurons projecting to dCA2 were labelled with EGFP by injection of CAV-Cre into dCA2 and Cre-dependent EGFP in the MS. Evoked EPSCs and IPSCs were recorded from EGFP-positive MS cells with a bipolar stimulating electrode (fabricated from platinum/iridium wire) placed near the recording pipette at 0.1 and 0.05 Hz, respectively. IPSCs and spontaneous IPSCs (sIPSCs) were recorded at βˆ’70 mV in the presence of NBQX (10 μM) and D-AP5 (50 μM). EPSCs were recorded at βˆ’70 mV in the presence of picrotoxin (50 ¡M). Summary graphs of the effects of CP93129 (Fig. 4l, m) and experiments controlling for pipette solution on evoked PSCs stability over time (Extended Data Fig. 8f, g) were generated by averaging the raw data in 1-min bins and expressing each point as a percentage of the averaged 3-min control. To increase IPSC signal to noise ratio, MS cells were chloride loaded with a pipette solution containing (in mM): 80 CsCl, 65 CsMeSO4, 8 NaCl, 10 HEPES, 0.25 EGTA, 2 Mg2ATP, 0.3 Na3GTP, 0.1 spermine and 7 phosphocreatine (pH 7.34; osmolarity 308). Cumulative probability graphs of sIPSCs were generated from a random sampling of 200 events before and in CP93129. The events for each experimental condition were arranged in ascending order and averaged. The resulting means and s.e.m. were plotted against their cumulative probability in the dataset. Intrinsic cell excitability was assessed with a series of incremental rectangular depolarizing current pulses (20 pA, 500 ms) injected into MS cells while recording in current-clamp. Drug-induced changes in the number of action potentials were determined by the number of action potentials evoked in the presence of no applied holding current. In cells in which CP93129 increased excitability, a graph was constructed plotting action potential number versus injected current. Peak currents induced by CP93129 (2 or 5 μM) were defined as those that could be clearly distinguished (>10 pA) as an agonist-induced inward or outward event from baseline. Recordings were made using a MultiClamp 700B amplifier (Molecular Devices), digitized at 10 kHz with the Digidata 1320A or 1440A data acquisition system (Molecular Devices), and analysed using Axograph or custom software written for Igor Pro (Wavemetrics). sIPSCs were analysed with MiniAnalysis (Synaptosoft).

Fibre photometry

Fibre photometry was performed as previously described9. AAVDJ-CaMKII-GCaMP6f or AAVDJ-EF1Ξ±-DIO-GCaMP6f was injected into the MS and CAV-Cre was injected into the dCA2. Fibre optic implants (Doric) were inserted and secured above the MS (0.65 AP, 0 ML, 3.5 DV). After 2–3 weeks, mice were habituated to the behavioural set-up for 10 min and tested one day later with simultaneous video and fibre photometry acquisition. On the test day, mice were placed in one chamber of the three-chamber apparatus, which was barricaded to create a small single chamber. Direct interaction with a novel mouse, a familiar mouse and a novel object was monitored for 5 min with an interval of 5 min. The presentation sequence of mouse and object was counterbalanced between subject mice. Fibre photometry data were acquired using the Synapse software, which controls an RZ5P lock-in amplifier (Tucker-Davis Technologies). During acquisition GCaMP6f was excited by frequency-modulated 473- and 405-nm light-emitting diodes (Doric), to stimulate Ca2+-dependent and isosbestic emission, respectively. All optical signals were band-pass-filtered with a fluorescence mini cube (Doric), emission was measured with a femtowatt photoreceiver (2151; Newport), and signal was digitized at 6 kHz. MATLAB (MathWorks) was used for signal processing, including correction of motion artifact and fluorescent bleaching. Signals were debleached by fitting with a mono- or bi-exponential decay function, and the resulting fluorescence trace was z-scored. Videos were manually analysed and the time of every physical contact after approach from a distance of more than 2 cm between subject mouse and an object or mouse was determined. An average of 7-s non-overlapping epochs of fluorescence was used to construct peristimulus time histograms. The time of contact is defined as time = 0, positioned in between 3 s before and 4 s after contact time. For each subject mouse, 2–7 contacts occurred per session and these were averaged to obtain the z-score graph for that mouse. Peak z-scored fluorescence was the maximal z-score between 0 and 4 s.

Statistical methods and reproducibility

The experimenter was blinded to the virus injection the animals had received. All analyses were performed with the experimenter blinded to the manipulation that the subject mouse had received. All data were tested for normality of sample distributions and when violated, non-parametric statistical tests were used. One-way ANOVA with Tukey’s multiple comparison post-hoc test was performed to assess significance for multiple group comparisons and two-way ANOVA with Tukey’s or Sidak’s multiple comparison post-hoc test for multiple group comparisons across multiple time points. Whenever ANOVA is performed, P values in the figure legends indicate the ANOVA results of theΒ whole group, whereas theΒ asterisks between individual samples are the results of the multiple comparisonΒ post-hoc tests.Β Two-tailed paired Student’s t-tests were used for within-group comparison of two treatments and the unpaired test was used for comparison between two groups. When normal distributions were not assumed, the Wilcoxon signed rank tests was performed for within group comparisons of two treatments, Mann-Whitney for between group comparison, and Kruskal-Wallis with post-hoc Dunn’s test for multiple comparisons. NS, not significant. *P < 0.05, **P < 0.01, ***P < 0.001. In all figures, data are shown as mean + s.e.m.

Fig. 1b: 1 out of 12 mice; Fig. 1g: 1 out of 9 mice; Fig. 2h: 1 out of 15 mice; Fig. 4a: 1 out of 14 mice; Fig. 4e, h: 1 out of 5 mice; Fig. 5a: 1 out of 10 mice; Fig. 5d: 1 out of 11 mice; Fig. 5g: 1 out of 13 mice.

Reporting summary

Further information on research design is available in theΒ Nature Research Reporting Summary linked to this paper.