Main

The C. elegans IIS pathway acts both cell autonomously and non-autonomously to control longevity, growth, dauer formation, metabolism, and reproduction5,6,7 through its regulation of the nuclear localization and transcriptional activation of DAF-16 (also known as FOXO). The canonical IIS/FOXO gene set, which identified primarily intestinal and hypodermal targets (Extended Data Fig. 1a, b)1,8,9, has been instructive in our understanding of how insulin signalling regulates a diverse range of activities, including metabolism, autophagy, stress resistance, and proteostasis. However, IIS mutants also exhibit daf-16-dependent neuronal phenotypes, including extended positive olfactory learning2, increased short- and long-term associative memory2, increased thermotaxis learning10, improved neuronal morphology maintenance11,12, and improved axon regeneration3. These phenotypes are unlikely to be regulated by the known intestinal and hypodermal IIS/FOXO targets1,8. Therefore, to understand how IIS daf-2 mutant animals extend behavioural functionality, we must identify the neuronal targets of FOXO/DAF-16.

We first profiled the expression of daf-16;daf-2 mutant worms with daf-16 rescued in specific tissues6 (Supplementary Table 1). Intestinal daf-16 rescue correlates best with whole-worm profiles (Extended Data Fig. 1a, c). By contrast, neuronal daf-16 rescue profiles are anti-correlated with the intestinal DAF-16 and whole-worm profiles (Extended Data Fig. 1a, c). Surprisingly, many genes induced by neuronal DAF-16 rescue are expressed (WormBase) or predicted to be expressed in non-neuronal tissues13 (Extended Data Fig. 1d), and have non-neuronal functions (for example, collagens14; Extended Data Fig. 1b, e, Supplementary Table 2). Thus, whole-worm transcriptional analyses of neuronally rescued DAF-16 failed to reveal targets that account for daf-16-dependent age-related behaviours of daf-2 mutants. Therefore, we needed to specifically examine transcription in IIS-mutant neurons.

The tough outer cuticle prevents dissociation of adult tissues15, thus the wild-type adult neuronal transcriptome has not been described. To solve this problem, we used rapid, chilled chemomechanical disruption followed immediately by fluorescence-activated cell sorting (FACS) to isolate neurons marked with green fluorescent protein (GFP) from wild-type worms, then RNA-sequenced these isolated cells (Fig. 1a–c, Extended Data Fig. 2a–c, f, g, Supplementary Table 3). This method is gentle enough to preserve the integrity of cells and some neurites (Extended Data Fig. 2a), does not involve cell culturing before FACS, in contrast to previous methods16, and does not affect transcription (as shown by actinomycin D treatment; Fig. 1b, Extended Data Fig. 2d, e, Supplementary Table 4). Downsampling analysis showed that sufficient sequencing depth was achieved (Extended Data Fig. 2h).

Figure 1: Identification of neuronal IIS/FOXO targets requires neuronal isolation.
figure 1

a, Volcano plot of neuron-expressed relative to whole-worm-expressed genes obtained by neuron-specific RNA sequencing of adult wild-type animals. N = 3 biological replicates (wild-type neurons) and 2 biological replicates (whole worm). b, Neuron-expressed and -enriched genes are not influenced by cell isolation: treatment with the transcription inhibitor actinomycin D affected only 0.22% of all neuronal genes (Supplementary Table 4). c, Tissue expression prediction of wild-type adult neuron-enriched genes. Mean ± s.e.m. d, GO terms highlight the neuronal characteristics of both all and previously uncharacterized neuron-enriched genes. e, Embryonic16, larval16 and adult neuron-enriched genes and significant GO terms transition from developmental to neuronal and behavioural functions (Supplementary Table 5); FDR <10% for all gene sets.

PowerPoint slide

We compared gene expression in isolated wild-type neurons with whole-worm expression to identify genes that are enriched in neurons (Fig. 1a–c). Of the 1,507 ‘neuron-enriched’ genes (false discovery rate (FDR) < 0.1; Supplementary Table 3; Fig. 1a, b), only 4% have previously described expression patterns exclusively in non-neuronal tissues, and ‘Neuron’ is the only significantly enriched tissue (Fig. 1c, Extended Data Fig. 2f), indicating that the method is highly selective for neuronal transcripts. Gene promoter–GFP tests of previously uncharacterized genes from our neuron-enriched list confirmed neuronal expression, with no bias for particular neuron types (Extended Data Fig. 3a). We also detected genes previously reported to be expressed only in single neurons or small subsets of neurons, including glr-3 (in the RIA neuron), ttx-3 (interneuron AIY/AIA) and npr-14 (neuron AIY) (WormBase).

The wild-type neuron-enriched set includes synaptic machinery, ion channels, neurotransmitters, and signalling components (Supplementary Table 3), as well as >700 previously uncharacterized genes; these genes are predicted to have ‘neuronal’-like character and function (Fig. 1d). Comparison of the wild-type embryonic and larval neuronal transcriptomes with the adult neuronal transcriptome at the same FDR revealed a shift in functional categories from developmental processes to neuronal function/behaviour in the adult neuronal transcriptome (Fig. 1e, Extended Data Fig. 3b, c, Supplementary Table 5), suggesting that previous isolation methods16, either due to early developmental stage isolation or to re-culturing, biased expression towards developmental genes rather than neuronal/behavioural genes.

To identify adult neuronal IIS/FOXO targets, we sequenced RNA from isolated daf-2 and daf-16;daf-2 mutant neurons on day 1 of adulthood (Fig. 2a, Extended Data Fig. 4, Supplementary Table 6, 8). The IIS/FOXO neuron-isolated gene set is enriched for neuronal expression: 86% and 92% of the up- and downregulated genes, respectively, are expressed in wild-type neurons. While several top Class I gene targets of DAF-16, including hil-1, sip-1, mtl-1, nnt-1, ins-6, and daf-16 itself, were upregulated in both daf-2 mutant neurons and daf-2 mutant whole worms (Group B; Fig. 2b), most of the IIS/FOXO neuronally regulated set differs from the canonical whole-worm IIS/FOXOs set1,8 (Fig. 2b). Specifically, in contrast to the metabolism-dominated functions of canonical whole-worm IIS/FOXO targets1,8, the neuronal IIS set gene ontology terms reflect neuron-like functions (Extended Data Fig. 5b): serpentine receptors, G protein-coupled receptors, syntaxin, globins, kinesins, insulins, ion channels, potassium channels, seven-transmembrane receptors, the NPR-1 neuropeptide receptor, and the SER-3 octopamine receptor are upregulated in daf-2 neurons (Supplementary Table 6). A few genes (fat-3 and crh-1, a CREB homologue) are upregulated in daf-2 neurons but downregulated in whole daf-2 animals.

Figure 2: RNA-seq transcriptional profile of isolated neurons reveals IIS/FOXO neuronal transcriptome.
figure 2

a, Volcano plot of daf-2-regulated, daf-16-dependent up- (red) and downregulated (green) neuronal genes (P < 0.05, N = 4 biological replicates per strain). b, Comparison of whole-worm (Class I)8 vs neuronal-IIS/FOXO targets. P values: hypergeometric distributions. ce, Short-term associative memory (STAM) assays. c, Schematic of STAM assay and chemotaxis profiles of daf-2 treated with sod-3 (c) or neuronal IIS/FOXO target gene RNAi (d, e). d, Learning indices relative to control RNAi at 3 h post-training of daf-2 treated with adult-only (green) or whole-life (blue) neuronal IIS/FOXO target gene RNAi. Mean ± s.e.m., *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, two-way repeated measures ANOVA, Bonferroni post hoc tests. At least 3 biological replicates were performed for all STAM assays.

PowerPoint slide

The IIS/FOXO downregulated set includes serpentine receptors, guanylate cyclases, signalling peptides and receptors (neuropeptide-like proteins, FMRF-like peptides and neuropeptides), and the vesicle trafficking G protein rab-28 (Supplementary Table 6). Expression of the sensory neuron cilia protein IFTA-2, which co-localizes with DAF-2 and whose loss increases lifespan17, is downregulated in daf-2 mutants, consistent with the longevity of daf-2 and ciliated sensory neuron mutants18. Similarly, sams-1 (S-adenosyl methionine synthetase), which is downregulated under long-lived dietary restriction conditions19, and sma-5 and dbl-1, components of TGF-beta pathways linked with IIS7,20, are downregulated, perhaps coordinating the longevity and reproductive output of these pathways.

Unlike canonical IIS/FOXO targets1, neuronal IIS/FOXO gene promoters are not enriched for the DBE (DAF-16 binding element, GTAAAt/cA), but the overlapping, upregulated (Group B) targets’ promoters contain twice as many DBEs (Extended Data Fig. 5a). The overlapping downregulated (Group F) targets are enriched for the PQM-1/DAE motif (CTTATCA, see refs 1, 8; Supplementary Table 7). DAF-16 may regulate neuronal activities indirectly through activation of ~60 IIS/FOXO-upregulated transcription factors (Supplementary Table 6).

We next tested the roles of top-scoring genes in daf-2-regulated neuronal phenotypes. Long-term and short-term associative memory are both extended in daf-2 mutants in a daf-16-dependent manner2 (Extended Data Fig. 6). The bZIP transcription factor CREB, which is required for long-term memory in many organisms, including C. elegans2, is upregulated by IIS/FOXO in neurons (Supplementary Table 6), correlating with the increased long-term memory of daf-2 mutants2,21. However, short-term associative memory (STAM; Fig. 2c) is CREB-independent2, and the genes that enable STAM extension in daf-2 mutants are unknown. While the DAF-16 non-neuronal target sod-3 had no effect on the extended STAM of daf-2 mutants (Fig. 2c, Extended Data Fig. 6b–d), knockdown of 8 of the 10 top-ranked, upregulated IIS/FOXO targets significantly decreased the STAM of daf-2(e1370) (Fig. 2d, e), both in whole-life and adult-only RNA interference (RNAi) tests. (Neuronal RNAi is effective in learning, STAM, and LTAM tests21.) The variety of genes (ion channels, transcription factors, G-proteins, vesicle fusion proteins) required for daf-2 mutants’ extended STAM suggests that decreased insulin signalling affects a broad network of memory extension genes. Several of these genes are also required for learning and memory in wild type (Extended Data Fig. 6g), suggesting that daf-2 mutants maintain neuronal function, rather than using an alternative short-term memory mechanism.

daf-2 mutants also maintain motor neuron axon regeneration ability with age in a daf-16-dependent manner3, and we found this is also true for mechanosensory neurons (Fig. 3a, b, Extended Data Fig. 7a–d). To identify factors that enable axon regeneration with age, we isolated and RNA-sequenced six adult mechanosensory neurons (Fig. 3c, Supplementary Table 9); this set includes 94 known larval regeneration genes from limited candidate screens22 (P ≤ 1.82 × 10−20). To find daf-2/daf-16-dependent axon regeneration candidates, we identified mechanosensory genes that are also regulated by neuronal IIS/FOXO (Fig. 3c, Supplementary Table 9; P < 0.002). The forkhead transcription factor FKH-9 is a neuronal IIS/FOXO target (Supplementary Table 6) and a canonical Class I target1, and is expressed in mechanosensory neurons (Supplementary Table 9). The fkh-9 promoter is occupied by DAF-16, which we confirmed by chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR; Fig. 3e, Extended Data Fig. 8a, b). FKH-9–GFP localized to nuclei, and neurons were the primary site of differential FKH-9–GFP levels in daf-2 mutants (Fig. 3f, Extended Data Fig. 8c), all suggesting a role for FKH-9 in daf-2/daf-16-mediated neuronal function.

Figure 3: FKH-9 is a direct target of DAF-16 and is expressed in mechanosensory neurons.
figure 3

a, b, daf-16 is required for enhanced day 5 axon regeneration in daf-2 mutants, mean ± s.e.m., *P < 0.05, Fisher’s exact test, N = 26 (wild-type), 36 (daf-2) and 16 (daf-16;daf-2), 2 biological replicates. c, Known larval regeneration genes are significantly enriched in the day 1 adult mechanosensory transcriptome. 63 genes are both DAF-16 targets and expressed in mechanosensory neurons (FDR < 5%; 3 biological replicates). d, fkh-9 messenger RNA levels are higher in aged daf-2 compared to wild type in a daf-16-dependent manner. N = 4 biological replicates, two-way ANOVA, Bonferroni post hoc tests. e, Chromatin immunoprecipitation of DAF-16–GFP worms with and without heat shock, which mobilizes DAF-16 into the nucleus. DAF-16 binds to the sod-3 promoter but not its 3′ UTR, and to the fkh-9 promoter at multiple locations (Extended Data Fig. 8). Fold enrichment relative to wild-type (not expressing DAF-16–GFP) is shown (mean ± s.e.m., two-tailed t-test, N = 3 biological replicates). f, Neuronal FKH-9–GFP (fkh-9p::fkh-9::gfp) expression in daf-2 compared to wild type. N = 25 animals. Mean ± s.e.m., two-tailed t-test. df, *P < 0.05, **P < 0.01, ***P < 0.001.

PowerPoint slide

While there is no effect on the first day of adulthood (Extended Data Fig. 7e, f), loss of fkh-9 severely impairs axon regeneration ability in aged (day 5) daf-2 mutants (Fig. 4a), correlating with an increased difference in fkh-9 expression levels between wild-type and daf-2 (Fig. 3d). Pan-neuronal fkh-9 expression rescues the ability of day 5 daf-2;fkh-9 worms to regenerate PLM axons (Fig. 4b, c). fkh-9 levels are critical for neuron morphology, as fkh-9 neuronal overexpression causes axonal defects (Extended Data Fig. 7g).

Figure 4: FKH-9 is required for improved axon regeneration, short-term associative memory and lifespan in daf-2 mutants.
figure 4

a, fkh-9 knockdown reduces axon regeneration of day 5 daf-2 mutants, as does daf-16 knockdown. Mean ± s.e.m., *P < 0.05, Fisher’s exact test, N = 34 (control), 33 (fkh-9) and 31 (daf-16), 4 biological replicates. b, c, Neuronally-expressed fkh-9 rescues day 5 axon regeneration in daf-2;fkh-9 mutants. Mean ± s.e.m., *P < 0.05, Fisher’s exact test, N = 20 (daf-2), 19 (daf-2; fkh-9) and 35 (daf-2; fkh-9; Punc-119::fkh-9B, 2 biological replicates. d, fkh-9 is required for enhanced memory in adult-only RNAi-treated daf-2 mutant worms. e, Neuronally-expressed fkh-9 rescues extended STAM in daf-2;fkh-9 mutants with defective learning and memory. Mean ± s.e.m., **P < 0.01, ***P < 0.001, ****P < 0.0001, two-way repeated measures ANOVA, Bonferroni post hoc tests. f, Adult-specific fkh-9 RNAi treatment reduces daf-2 mutant lifespan. Median lifespan: control RNAi 42 days, fkh-9 RNAi 21 days, daf-16 RNAi 21 days. P < 0.0001 for control RNAi vs daf-16 RNAi and control vs fkh-9 RNAi, log-rank test. N = 144 worms per strain. g, Integrative Multi-species Prediction (IMP; see ref. 30) network analysis of DAF-16 neuronal target genes with STAM phenotypes (red circles).

PowerPoint slide

Adult-specific and whole-life reduction of fkh-9 also severely impaired extended STAM of daf-2 mutants (Fig. 4d, Extended Data Fig. 9). daf-2;fkh-9 double mutants were defective in both STAM and learning, and neuronal fkh-9 expression rescued these defects (Fig. 4e, Extended Data Fig. 9d, e), suggesting that fkh-9 is required for extended memory and normal neuronal development in daf-2 mutants. Day 1 and 5 fkh-9 expression levels correlated with STAM and axon regeneration (Fig. 3d). fkh-9 reduction delayed development, and reduction during adulthood caused severe matricide (Extended Data Fig. 10a–c). fkh-9 knockdown in adult daf-2 worms treated with FUdR (5-fluoro-2′- deoxyuridine) to block matricide20 significantly shortened lifespan (40–50%; Fig. 4f). Pan-neuronal fkh-9 expression did not rescue lifespan (Extended Data Fig. 10d), suggesting that FKH-9 acts in non-neuronal tissues to regulate lifespan. Thus, IIS/FOXO-regulated FKH-9 function is important for both neuronal and non-neuronal growth and development, as well as adult memory and axon regeneration. Interestingly, the FKH-9 mammalian homologue FOXG1 is required for axon outgrowth23 and is the most highly-induced gene in spinal cords treated with radial glial cell transplant following spinal cord injury24.

Network analysis using fkh-9 and the other 8 neuronal DAF-16 STAM genes (Fig. 4g, Supplementary Table 10) identified casy-1, which is required for several forms of associative learning and memory2,25,26,27, apl-1, the C. elegans orthologue of amyloid precursor protein (APP) that can disrupt sensory plasticity28, and dlk-1, the only previously known regulator of age-dependent axon regeneration3,29. Additionally, genes involved in neuronal degeneration (mec-17), neuronal development (egl-44, sem-4), neuronal function (egl-21, rcn-1, vab-9, cysl-1), synaptic regulation and function (cab-1, hlb-1, magu-4, sph-1, unc-64), and axon outgrowth (unc-14) and regeneration (egl-8, fos-1, pmk-3), were connected to the STAM genes. PQM-1 (ref. 8), whose motif (DAE) is overrepresented in neuronal IIS target promoters, and other IIS (akt-2, dct-6, hlh-30), TGF-β (daf-14, sma-4, crm-1, sma-9, sma-1, sta-1), and MAPK pathway (vhp-1, pmk-3) components emerged in the network. Transcriptional regulation by IIS/FOXO and its targets may lead to broader, indirect transcriptional and non-transcriptional regulation of genes with important neuronal functions.

Plasticity in development, reproduction and longevity allows organisms to respond appropriately to nutrient availability and changes in their environment. The IIS pathway is a critical mediator of these decisions, with FOXO selecting transcriptional targets to execute specific biochemical functions in each tissue, including factors that maintain cognitive function with age. daf-2 mutant worms maintain neuronal behaviours with age by using a set of transcriptional targets that are distinct from previously identified metabolic and stress resistance targets expressed in other tissues. These genes may regulate additional neuronal targets through non-transcriptional mechanisms (Fig. 4g). The regulation of tissue-specific transcriptional programs is important to coordinate phenotypic responses, extending neuronal abilities in concert with the extended longevity and reproductive span of daf-2 mutants.

Methods

Adult cell isolation

Day 1 adult neuronally GFP-labelled worms (Punc119::GFP or Pmec-4::GFP) were prepared for cell isolation as previously described15 with modifications (Extended Data Fig. 2). Synchronized adult worms were washed with M9 buffer to remove excess bacteria. The pellet (~250 μl) was washed with 500 μl lysis buffer (200 mM DTT, 0.25% SDS, 20 mM HEPES pH 8.0, 3% sucrose) and resuspended in 1,000 μl lysis buffer. Worms were incubated in lysis buffer with gentle rocking for 6.5 min at room temperature. The pellet was washed 6× with M9 and resuspended in 20 mg ml−1 pronase from Streptomyces griseus (Sigma-Aldrich). Worms were incubated at room temperature (<20 min) with periodic mechanical disruption by pipetting every 2 min. When most worm bodies were dissociated, leaving only small debris and eggs, ice-cold PBS buffer containing 2% fetal bovine serum (Gibco) was added. RNA from FACS-sorted neurons was prepared for RNA-seq and subsequent analysis (see Extended Data for details).

No statistical methods were used to predetermine sample size. The experiments were not randomized, and the investigators were not blinded to allocation during experiments and outcome assessment.

Short-term associative memory assay

Memory assays were performed as described2.

Axon regeneration assays

In vivo laser axotomy of PLM neurons was performed as described22.